Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Virus Res ; 345: 199398, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38754786

RESUMEN

Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne nairovirus with a wide geographic spread that can cause severe and lethal disease. No specific medical countermeasures are approved to combat this illness. The CCHFV L protein contains an ovarian tumor (OTU) domain with a cysteine protease thought to modulate cellular immune responses by removing ubiquitin and ISG15 post-translational modifications from host and viral proteins. Viral deubiquitinases like CCHFV OTU are attractive drug targets, as blocking their activity may enhance cellular immune responses to infection, and potentially inhibit viral replication itself. We previously demonstrated that the engineered ubiquitin variant CC4 is a potent inhibitor of CCHFV replication in vitro. A major challenge of the therapeutic use of small protein inhibitors such as CC4 is their requirement for intracellular delivery, e.g., by viral vectors. In this study, we examined the feasibility of in vivo CC4 delivery by a replication-deficient recombinant adenovirus (Ad-CC4) in a lethal CCHFV mouse model. Since the liver is a primary target of CCHFV infection, we aimed to optimize delivery to this organ by comparing intravenous (tail vein) and intraperitoneal injection of Ad-CC4. While tail vein injection is a traditional route for adenovirus delivery, in our hands intraperitoneal injection resulted in higher and more widespread levels of adenovirus genome in tissues, including, as intended, the liver. However, despite promising in vitro results, neither route of in vivo CC4 treatment resulted in protection from a lethal CCHFV infection.


Asunto(s)
Adenoviridae , Modelos Animales de Enfermedad , Virus de la Fiebre Hemorrágica de Crimea-Congo , Fiebre Hemorrágica de Crimea , Replicación Viral , Animales , Virus de la Fiebre Hemorrágica de Crimea-Congo/genética , Fiebre Hemorrágica de Crimea/virología , Ratones , Adenoviridae/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Vectores Genéticos/genética , Antivirales/farmacología , Femenino , Hígado/virología , Humanos
2.
NPJ Vaccines ; 9(1): 88, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38782933

RESUMEN

Crimean-Congo hemorrhagic fever virus (CCHFV) can cause severe human disease and is considered a WHO priority pathogen due to the lack of efficacious vaccines and antivirals. A CCHF virus replicon particle (VRP) has previously shown protective efficacy in a lethal Ifnar-/- mouse model when administered as a single dose at least 3 days prior to challenge. Here, we determine that non-specific immune responses are not sufficient to confer short-term protection, since Lassa virus VRP vaccination 3 days prior to CCHFV challenge was not protective. We also investigate how CCHF VRP vaccination confers protective efficacy by examining viral kinetics, histopathology, clinical analytes and immunity early after challenge (3 and 6 days post infection) and compare to unvaccinated controls. We characterize how these effects differ based on vaccination period and correspond to previously reported CCHF VRP-mediated protection. Vaccinating Ifnar-/- mice with CCHF VRP 28, 14, 7, or 3 days prior to challenge, all known to confer complete protection, significantly reduced CCHFV viral load, mucosal shedding, and markers of clinical disease, with greater reductions associated with longer vaccination periods. Interestingly, there were no significant differences in innate immune responses, T cell activation, or antibody titers after challenge between groups of mice vaccinated a week or more before challenge, but higher anti-NP antibody avidity and effector function (ADCD) were positively associated with longer vaccination periods. These findings support the importance of antibody-mediated responses in VRP vaccine-mediated protection against CCHFV infection.

3.
Nat Commun ; 15(1): 1722, 2024 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-38409240

RESUMEN

Crimean-Congo hemorrhagic fever virus (CCHFV) is a WHO priority pathogen. Antibody-based medical countermeasures offer an important strategy to mitigate severe disease caused by CCHFV. Most efforts have focused on targeting the viral glycoproteins. However, glycoproteins are poorly conserved among viral strains. The CCHFV nucleocapsid protein (NP) is highly conserved between CCHFV strains. Here, we investigate the protective efficacy of a CCHFV monoclonal antibody targeting the NP. We find that an anti-NP monoclonal antibody (mAb-9D5) protected female mice against lethal CCHFV infection or resulted in a significant delay in mean time-to-death in mice that succumbed to disease compared to isotype control animals. Antibody protection is independent of Fc-receptor functionality and complement activity. The antibody bound NP from several CCHFV strains and exhibited robust cross-protection against the heterologous CCHFV strain Afg09-2990. Our work demonstrates that the NP is a viable target for antibody-based therapeutics, providing another direction for developing immunotherapeutics against CCHFV.


Asunto(s)
Virus de la Fiebre Hemorrágica de Crimea-Congo , Fiebre Hemorrágica de Crimea , Femenino , Animales , Ratones , Virus de la Fiebre Hemorrágica de Crimea-Congo/metabolismo , Proteínas de la Nucleocápside/metabolismo , Anticuerpos Monoclonales , Fiebre Hemorrágica de Crimea/prevención & control , Glicoproteínas/metabolismo , Anticuerpos Antivirales
4.
J Med Chem ; 66(12): 8159-8169, 2023 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-37262387

RESUMEN

We have recently reported on the use of aryl-fluorosulfates in designing water- and plasma-stable agents that covalently target Lys, Tyr, or His residues in the BIR3 domain of the inhibitor of the apoptosis protein (IAP) family. Here, we report further structural, cellular, and pharmacological characterizations of this agent, including the high-resolution structure of the complex between the Lys-covalent agent and its target, the BIR3 domain of X-linked IAP (XIAP). We also compared the cellular efficacy of the agent in two-dimensional (2D) and three-dimensional (3D) cell cultures, side by side with the clinical candidate reversible IAP inhibitor LCL161. Finally, in vivo pharmacokinetic studies indicated that the agent was long-lived and orally bioavailable. Collectively our data further corroborate that aryl-fluorosulfates, when incorporated correctly in a ligand, can result in Lys-covalent agents with pharmacodynamic and pharmacokinetic properties that warrant their use in the design of pharmacological probes or even therapeutics.


Asunto(s)
Proteínas Inhibidoras de la Apoptosis , Proteína Inhibidora de la Apoptosis Ligada a X , Unión Proteica , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Apoptosis
5.
Int J Mol Sci ; 24(5)2023 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-36901828

RESUMEN

Papain-like protease (PLpro) is critical to COVID-19 infection. Therefore, it is a significant target protein for drug development. We virtually screened a 26,193 compound library against the PLpro of SARS-CoV-2 and identified several drug candidates with convincing binding affinities. The three best compounds all had better estimated binding energy than those of the drug candidates proposed in previous studies. By analyzing the docking results for the drug candidates identified in this and previous studies, we demonstrate that the critical interactions between the compounds and PLpro proposed by the computational approaches are consistent with those proposed by the biological experiments. In addition, the predicted binding energies of the compounds in the dataset showed a similar trend as their IC50 values. The predicted ADME and drug-likeness properties also suggested that these identified compounds can be used for COVID-19 treatment.


Asunto(s)
COVID-19 , Humanos , Evaluación Preclínica de Medicamentos , SARS-CoV-2 , Tratamiento Farmacológico de COVID-19 , Papaína , Simulación del Acoplamiento Molecular , Inhibidores de Proteasas , Antivirales , Simulación de Dinámica Molecular
6.
Nat Commun ; 13(1): 7298, 2022 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-36435827

RESUMEN

Crimean-Congo Hemorrhagic Fever Virus (CCHFV) causes a life-threatening disease with up to a 40% mortality rate. With no approved medical countermeasures, CCHFV is considered a public health priority agent. The non-neutralizing mouse monoclonal antibody (mAb) 13G8 targets CCHFV glycoprotein GP38 and protects mice from lethal CCHFV challenge when administered prophylactically or therapeutically. Here, we reveal the structures of GP38 bound with a human chimeric 13G8 mAb and a newly isolated CC5-17 mAb from a human survivor. These mAbs bind overlapping epitopes with a shifted angle. The broad-spectrum potential of c13G8 and CC5-17 and the practicality of using them against Aigai virus, a closely related nairovirus were examined. Binding studies demonstrate that the presence of non-conserved amino acids in Aigai virus corresponding region prevent CCHFV mAbs from binding Aigai virus GP38. This information, coupled with in vivo efficacy, paves the way for future mAb therapeutics effective against a wide swath of CCHFV strains.


Asunto(s)
Virus de la Fiebre Hemorrágica de Crimea-Congo , Fiebre Hemorrágica de Crimea , Ratones , Humanos , Animales , Virus de la Fiebre Hemorrágica de Crimea-Congo/química , Fiebre Hemorrágica de Crimea/prevención & control , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Epítopos , Anticuerpos Monoclonales
7.
ACS Infect Dis ; 8(3): 596-611, 2022 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-35199517

RESUMEN

Over the last 20 years, both severe acute respiratory syndrome coronavirus-1 and severe acute respiratory syndrome coronavirus-2 have transmitted from animal hosts to humans causing zoonotic outbreaks of severe disease. Both viruses originate from a group of betacoronaviruses known as subgroup 2b. The emergence of two dangerous human pathogens from this group along with previous studies illustrating the potential of other subgroup 2b members to transmit to humans has underscored the need for antiviral development against them. Coronaviruses modify the host innate immune response in part through the reversal of ubiquitination and ISGylation with their papain-like protease (PLpro). To identify unique or overarching subgroup 2b structural features or enzymatic biases, the PLpro from a subgroup 2b bat coronavirus, BtSCoV-Rf1.2004, was biochemically and structurally evaluated. This evaluation revealed that PLpros from subgroup 2b coronaviruses have narrow substrate specificity for K48 polyubiquitin and ISG15 originating from certain species. The PLpro of BtSCoV-Rf1.2004 was used as a tool alongside PLpro of CoV-1 and CoV-2 to design 30 novel noncovalent drug-like pan subgroup 2b PLpro inhibitors that included determining the effects of using previously unexplored core linkers within these compounds. Two crystal structures of BtSCoV-Rf1.2004 PLpro bound to these inhibitors aided in compound design as well as shared structural features among subgroup 2b proteases. Screening of these three subgroup 2b PLpros against this novel set of inhibitors along with cytotoxicity studies provide new directions for pan-coronavirus subgroup 2b antiviral development of PLpro inhibitors.


Asunto(s)
COVID-19 , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo , Animales , Inhibidores de Proteasas , SARS-CoV-2 , Ubiquitina/metabolismo
8.
Molecules ; 26(24)2021 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-34946521

RESUMEN

An increasing number of studies have demonstrated the antiviral nature of polyphenols, and many polyphenols have been proposed to inhibit SARS-CoV or SARS-CoV-2. Our previous study revealed the inhibitory mechanisms of polyphenols against DNA polymerase α and HIV reverse transcriptase to show that polyphenols can block DNA elongation by competing with the incoming NTPs. Here we applied computational approaches to examine if some polyphenols can also inhibit RNA polymerase (RdRp) in SARS-CoV-2, and we identified some better candidates than remdesivir, the FDA-approved drug against RdRp, in terms of estimated binding affinities. The proposed compounds will be further examined to develop new treatments for COVID-19.


Asunto(s)
Antivirales/farmacología , Polifenoles/farmacología , SARS-CoV-2/efectos de los fármacos , Antocianinas/química , Antocianinas/farmacología , Antivirales/aislamiento & purificación , Simulación de Dinámica Molecular , Estructura Molecular , Polifenoles/química , ARN Polimerasa Dependiente del ARN , SARS-CoV-2/enzimología , Tratamiento Farmacológico de COVID-19
9.
Comput Struct Biotechnol J ; 19: 5371-5380, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34567475

RESUMEN

Although scientists around the world have put lots of effort into the development of new treatments for COVID-19 since the outbreak, no drugs except Veklury (remdesivir) have been approved by FDA. There is an urgent need to discover some alternative antiviral treatment for COVID-19. Because polyphenols have been shown to possess antiviral activities, here we conducted a large-scale virtual screening for more than 400 polyphenols. Several lead compounds such as Petunidin 3-O-(6″-p-coumaroyl-glucoside) were identified to have promising binding affinities and convincing binding mechanisms. Analyzing the docking results and ADME properties sheds light on the potential efficacy of the top-ranked drug candidates and pinpoints the key residues on the target proteins for the future of drug development.

10.
J Pharm Pharm Sci ; 24: 390-399, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34319871

RESUMEN

PURPOSE: SARS-CoV-2 infection is associated with substantial mortality and high morbidity. This study tested the effect of angiotensin II type I receptor blocker, losartan, on SARS-CoV-2 replication and inhibition of the papain-like protease of the virus. METHODS: The dose-dependent inhibitory effect of losartan, in concentrations from 1µM to 100µM as determined by quantitative cell analysis combining fluorescence microscopy, image processing, and cellular measurements (Cellomics analysis) on SARS-CoV-2 replication was investigated in Vero E6 cells. The impact of losartan on deubiquitination and deISGylation of SARS-CoV-2 papain-like protease (PLpro) were also evaluated.  Results: Losartan reduced PLpro cleavage of tetraUbiquitin to diUbiquitin.  It was less effective in inhibiting PLpro's cleavage of ISG15-AMC than Ubiquitin-AMC.  To determine if losartan inhibited SARS-CoV-2 replication, losartan treatment of SARS-CoV-2 infected Vero E6 was examined. Losartan treatment one hour prior to SARS-CoV-2 infection reduced levels of SARS-CoV-2 nuclear protein, an indicator of virus replication, by 80% and treatment one-hour post-infection decreased viral replication by 70%. CONCLUSION: Losartan was not an effective inhibitor of deubiquitinase or deISGylase activity of the PLpro but affected the SARS-CoV-2 replication of Vero E6 cells in vitro.  As losartan has a favorable safety profile and is currently available it has features necessary for efficacious drug repurposing and treatment of COVID-19.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Antivirales/farmacología , Losartán/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Chlorocebus aethiops , Biología Computacional , Proteasas Similares a la Papaína de Coronavirus/antagonistas & inhibidores , Proteasas Similares a la Papaína de Coronavirus/metabolismo , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Enzimas Desubicuitinizantes/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiología , Ubiquitina/metabolismo , Células Vero , Replicación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
11.
Acta Crystallogr D Struct Biol ; 77(Pt 7): 943-953, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-34196620

RESUMEN

Porcine epidemic diarrhea is a devastating porcine disease that is caused by the alphacoronavirus porcine epidemic diarrhea virus (PEDV). Like other members of the Coronaviridae family, PEDV encodes a multifunctional papain-like protease 2 (PLP2) that has the ability to process the coronavirus viral polyprotein to aid in RNA replication and antagonize the host innate immune response through cleavage of the regulatory proteins ubiquitin (Ub) and/or interferon-stimulated gene product 15 (ISG15) (deubiquitination and deISGylation, respectively). Because Betacoronavirus PLPs have been well characterized, it was sought to determine how PLP2 from the alphacoronavirus PEDV differentiates itself from its related counterparts. PEDV PLP2 was first biochemically characterized, and a 3.1 Šresolution crystal structure of PEDV PLP2 bound to Ub was then solved, providing insight into how Alphacoronavirus PLPs bind to their preferred substrate, Ub. It was found that PEDV PLP2 is a deubiquitinase and readily processes a variety of di-Ub linkages, in comparison with its Betacoronavirus counterparts, which have a narrower range of di-Ub activity but process both Ub and ISG15.


Asunto(s)
Infecciones por Coronavirus/virología , Proteasas Similares a la Papaína de Coronavirus/química , Proteasas Similares a la Papaína de Coronavirus/metabolismo , Virus de la Diarrea Epidémica Porcina/fisiología , Ubiquitina/metabolismo , Animales , Cristalografía por Rayos X , Unión Proteica , Conformación Proteica , Porcinos
12.
ACS Infect Dis ; 7(6): 1483-1502, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34019767

RESUMEN

Viral proteases are highly specific and recognize conserved cleavage site sequences of ∼6-8 amino acids. Short stretches of homologous host-pathogen sequences (SSHHPS) can be found spanning the viral protease cleavage sites. We hypothesized that these sequences corresponded to specific host protein targets since >40 host proteins have been shown to be cleaved by Group IV viral proteases and one Group VI viral protease. Using PHI-BLAST and the viral protease cleavage site sequences, we searched the human proteome for host targets and analyzed the hit results. Although the polyprotein and host proteins related to the suppression of the innate immune responses may be the primary targets of these viral proteases, we identified other cleavable host proteins. These proteins appear to be related to the virus-induced phenotype associated with Group IV viruses, suggesting that information about viral pathogenesis may be extractable directly from the viral genome sequence. Here we identify sequences cleaved by the SARS-CoV-2 papain-like protease (PLpro) in vitro within human MYH7 and MYH6 (two cardiac myosins linked to several cardiomyopathies), FOXP3 (an X-linked Treg cell transcription factor), ErbB4 (HER4), and vitamin-K-dependent plasma protein S (PROS1), an anticoagulation protein that prevents blood clots. Zinc inhibited the cleavage of these host sequences in vitro. Other patterns emerged from multispecies sequence alignments of the cleavage sites, which may have implications for the selection of animal models and zoonosis. SSHHPS/nsP is an example of a sequence-specific post-translational silencing mechanism.


Asunto(s)
Papaína , Péptido Hidrolasas , SARS-CoV-2/enzimología , Proteasas Virales/metabolismo , Secuencia de Aminoácidos , Miosinas Cardíacas/química , Factores de Transcripción Forkhead/química , Humanos , Cadenas Pesadas de Miosina/química , Papaína/metabolismo , Péptido Hidrolasas/metabolismo , Proteína S/química , Receptor ErbB-4/química
13.
Chem Res Toxicol ; 34(3): 804-816, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33538594

RESUMEN

The recent use of organophosphate nerve agents in Syria, Malaysia, Russia, and the United Kingdom has reinforced the potential threat of their intentional release. These agents act through their ability to inhibit human acetylcholinesterase (hAChE; E.C. 3.1.1.7), an enzyme vital for survival. The toxicity of hAChE inhibition via G-series nerve agents has been demonstrated to vary widely depending on the G-agent used. To gain insight into this issue, the structures of hAChE inhibited by tabun, sarin, cyclosarin, soman, and GP were obtained along with the inhibition kinetics for these agents. Through this information, the role of hAChE active site plasticity in agent selectivity is revealed. With reports indicating that the efficacy of reactivators can vary based on the nerve agent inhibiting hAChE, human recombinatorially expressed hAChE was utilized to define these variations for HI-6 among various G-agents. To identify the structural underpinnings of this phenomenon, the structures of tabun, sarin, and soman-inhibited hAChE in complex with HI-6 were determined. This revealed how the presence of G-agent adducts impacts reactivator access and placement within the active site. These insights will contribute toward a path of next-generation reactivators and an improved understanding of the innate issues with the current reactivators.


Asunto(s)
Acetilcolinesterasa/metabolismo , Inhibidores de la Colinesterasa/efectos adversos , Agentes Nerviosos/efectos adversos , Oximas/efectos adversos , Compuestos de Piridinio/efectos adversos , Acetilcolinesterasa/química , Acetilcolinesterasa/aislamiento & purificación , Inhibidores de la Colinesterasa/química , Humanos , Estructura Molecular , Agentes Nerviosos/química , Oximas/química , Compuestos de Piridinio/química
14.
Front Immunol ; 12: 794664, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35058932

RESUMEN

Post-translational modification of host and viral proteins by ubiquitin and ubiquitin-like proteins plays a key role in a host's ability to mount an effective immune response. Avian species lack a ubiquitin-like protein found in mammals and other non-avian reptiles; interferon stimulated gene product 15 (ISG15). ISG15 serves as a messenger molecule and can be conjugated to both host and viral proteins leading them to be stabilized, degraded, or sequestered. Structurally, ISG15 is comprised of a tandem ubiquitin-like domain (Ubl), which serves as the motif for post-translational modification. The 2'-5' oligoadenylate synthetase-like proteins (OASL) also encode two Ubl domains in series near its C-terminus which binds OASL to retinoic acid inducible gene-I (RIG-I). This protein-protein interaction increases the sensitivity of RIG-I and results in an enhanced production of type 1 interferons and a robust immune response. Unlike human and other mammalian OASL homologues, avian OASLs terminate their tandem Ubl domains with the same LRLRGG motif found in ubiquitin and ISG15, a motif required for their conjugation to proteins. Chickens, however, lack RIG-I, raising the question of structural and functional characteristics of chicken OASL (chOASL). By investigating chOASL, the evolutionary history of viruses with deubiquitinases can be explored and drivers of species specificity for these viruses may be uncovered. Here we show that the chOASL tandem Ubl domains shares structural characteristics with mammalian ISG15, and that chOASL can oligomerize and conjugate to itself. In addition, the ISG15-like features of avian OASLs and how they impact interactions with viral deubiquitinases and deISGylases are explored.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/química , 2',5'-Oligoadenilato Sintetasa/metabolismo , Inmunomodulación , Dominios y Motivos de Interacción de Proteínas , Ubiquitina/química , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Pollos , Humanos , Espectrometría de Masas , Modelos Biológicos , Unión Proteica , Conformación Proteica , Procesamiento Proteico-Postraduccional , Proteolisis , Relación Estructura-Actividad , Proteínas Virales/química , Proteínas Virales/metabolismo
15.
Acta Crystallogr D Struct Biol ; 76(Pt 11): 1114-1123, 2020 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-33135682

RESUMEN

Nairoviruses are arthropod-borne viruses with a nearly global geographical distribution. Several are known causative agents of human disease, including Crimean-Congo hemorrhagic fever virus (CCHFV), which has a case fatality rate that can exceed 30%. Nairoviruses encode an ovarian tumour domain protease (OTU) that can suppress the innate immune response by reversing post-translational modifications by ubiquitin (Ub) and/or interferon-stimulated gene product 15 (ISG15). As a result, the OTU has been identified as a potential target for the development of CCHFV therapeutics. Despite sharing the same general fold, nairoviral OTUs show structural and enzymatic diversity. The CCHFV OTU, for example, possesses activity towards both Ub and ISG15, while the Hazara virus (HAZV) OTU interacts exclusively with Ub. Virology studies focused on the OTU have mostly been restricted to CCHFV, which requires BSL-4 containment facilities. Although HAZV has been proposed as a BSL-2 alternative, differences in the engagement of substrates by CCHFV and HAZV OTUs may present complicating factors when trying to model one using the other. To understand the molecular underpinnings of the differences in activity, a 2.78 Šresolution crystal structure of HAZV OTU bound to Ub was solved. Using structure-guided site-directed mutagenesis, HAZV OTUs were engineered with altered or eliminated deubiquitinase activity, including one with an exclusive activity for ISG15. Additionally, analysis of the structure yielded insights into the difference in inhibition observed between CCHFV and HAZV OTUs with a Ub-based inhibitor. These new insights present opportunities to utilize HAZV as a model system to better understand the role of the OTU in the context of infection.


Asunto(s)
Nairovirus/enzimología , Péptido Hidrolasas , Ubiquitina , Proteínas Virales , Modelos Moleculares , Péptido Hidrolasas/química , Péptido Hidrolasas/metabolismo , Unión Proteica , Dominios Proteicos , Ubiquitina/química , Ubiquitina/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo
16.
Virus Res ; 286: 198036, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32492472

RESUMEN

Interferon (IFN)-stimulated gene product 15 (ISG15) is a ubiquitin-like protein critical for the control of microbial infections. ISG15 appears to serve a wide variety of functions, which regulate multiple cellular responses contributing to the development of an antiviral state. ISG15 is a versatile molecule directly modulating both host and virus protein function which regulate many signaling pathways, including its own synthesis. Here we review the various roles ISG15 plays in the antiviral immune response, and examine the mechanisms by which viruses attempt to mitigate or exploit ISG15 activity.


Asunto(s)
Citocinas/metabolismo , Inmunidad Innata/inmunología , Ubiquitinas/metabolismo , Virosis/inmunología , Replicación Viral/inmunología , Animales , Citocinas/genética , Humanos , Interferón Tipo I/inmunología , Macrófagos/inmunología , Ratones , Neutrófilos/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Ubiquitinas/genética , Proteínas Virales/metabolismo , Internalización del Virus
17.
J Am Chem Soc ; 142(20): 9147-9151, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-32364709

RESUMEN

We describe the synthesis of 10-aza-9-oxakalkitoxin, an N,N,O-trisubstituted hydroxylamine-based analog, or hydroxalog, of the cytotoxic marine natural product kalkitoxin in which the -NMe-O- moiety replaces a -CHMe-CH2- unit in the backbone of the natural product. 10-Aza-9-oxakalkitoxin displays potent and selective cytotoxicity (IC50 2.4 ng mL-1) comparable to that of kalkitoxin itself (IC50 3.2 ng mL-1) against the human hepato-carcinoma cell line HepG2 over both the human leukemia cell line CEM and the normal hematopoietic CFU-GM. Like kalkitoxin, and contrary to the common expectation for hydroxylamines, 10-aza-9-oxakalkitoxin is not mutagenic.


Asunto(s)
Antineoplásicos/farmacología , Productos Biológicos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Productos Biológicos/síntesis química , Productos Biológicos/química , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular
18.
ACS Infect Dis ; 6(8): 2099-2109, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32428392

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent for COVID-19, is a novel human betacoronavirus that is rapidly spreading worldwide. The outbreak currently includes over 3.7 million cases and 260,000 fatalities. As a betacoronavirus, SARS-CoV-2 encodes for a papain-like protease (PLpro) that is likely responsible for cleavage of the coronavirus (CoV) viral polypeptide. The PLpro is also responsible for suppression of host innate immune responses by virtue of its ability to reverse host ubiquitination and ISGylation events. Here, the biochemical activity of SARS-CoV-2 PLpro against ubiquitin (Ub) and interferon-stimulated gene product 15 (ISG15) substrates is evaluated, revealing that the protease has a marked reduction in its ability to process K48 linked Ub substrates compared to its counterpart in SARS-CoV. Additionally, its substrate activity more closely mirrors that of the PLpro from the Middle East respiratory syndrome coronavirus and prefers ISG15s from certain species including humans. Additionally, naphthalene based PLpro inhibitors are shown to be effective at halting SARS-CoV-2 PLpro activity as well as SARS-CoV-2 replication.


Asunto(s)
Betacoronavirus/enzimología , Infecciones por Coronavirus/virología , Cisteína Endopeptidasas/metabolismo , Citocinas/metabolismo , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Neumonía Viral/virología , Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , COVID-19 , Chlorocebus aethiops , Proteasas 3C de Coronavirus , Cisteína Endopeptidasas/química , Citocinas/antagonistas & inhibidores , Citocinas/química , Humanos , Naftalenos/farmacología , Pandemias , Unión Proteica , Conformación Proteica , SARS-CoV-2 , Especificidad por Sustrato , Ubiquitinas/antagonistas & inhibidores , Ubiquitinas/química , Células Vero , Proteínas no Estructurales Virales/química , Replicación Viral/efectos de los fármacos
19.
PLoS One ; 14(12): e0226415, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31869347

RESUMEN

Tick-borne nairoviruses (order Bunyavirales) encode an ovarian tumor domain protease (OTU) that suppresses the innate immune response by reversing the post-translational modification of proteins by ubiquitin (Ub) and interferon-stimulated gene product 15 (ISG15). Ub is highly conserved across eukaryotes, whereas ISG15 is only present in vertebrates and shows substantial sequence diversity. Prior attempts to address the effect of ISG15 diversity on viral protein-ISG15 interactions have focused on only a single species' ISG15 or a limited selection of nairovirus OTUs. To gain a more complete perspective of OTU-ISG15 interactions, we biochemically assessed the relative activities of 14 diverse nairovirus OTUs for 12 species' ISG15 and found that ISG15 activity is predominantly restricted to particular nairovirus lineages reflecting, in general, known virus-host associations. To uncover the underlying molecular factors driving OTUs affinity for ISG15, X-ray crystal structures of Kupe virus and Ganjam virus OTUs bound to sheep ISG15 were solved and compared to complexes of Crimean-Congo hemorrhagic fever virus and Erve virus OTUs bound to human and mouse ISG15, respectively. Through mutational and structural analysis seven residues in ISG15 were identified that predominantly influence ISG15 species specificity among nairovirus OTUs. Additionally, OTU residues were identified that influence ISG15 preference, suggesting the potential for viral OTUs to adapt to different host ISG15s. These findings provide a foundation to further develop research methods to trace nairovirus-host relationships and delineate the full impact of ISG15 diversity on nairovirus infection.


Asunto(s)
Citocinas/metabolismo , Especificidad del Huésped/genética , Nairovirus/enzimología , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , Dominios y Motivos de Interacción de Proteínas/genética , Ubiquitinas/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Cristalografía por Rayos X , Células HEK293 , Interacciones Huésped-Patógeno/genética , Humanos , Ratones , Modelos Moleculares , Nairovirus/clasificación , Nairovirus/genética , Péptido Hidrolasas/química , Filogenia , Unión Proteica/genética , Homología de Secuencia , Ovinos , Especificidad de la Especie , Ubiquitina/metabolismo , Proteínas Virales/química , Proteínas Virales/genética
20.
mBio ; 10(4)2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31337717

RESUMEN

Crimean-Congo hemorrhagic fever virus (CCHFV) infection can result in a severe hemorrhagic syndrome for which there are no antiviral interventions available to date. Certain RNA viruses, such as CCHFV, encode cysteine proteases of the ovarian tumor (OTU) family that antagonize interferon (IFN) production by deconjugating ubiquitin (Ub). The OTU of CCHFV, a negative-strand RNA virus, is dispensable for replication of the viral genome, despite being part of the large viral RNA polymerase. Here, we show that mutations that prevent binding of the OTU to cellular ubiquitin are required for the generation of recombinant CCHFV containing a mutated catalytic cysteine. Similarly, the high-affinity binding of a synthetic ubiquitin variant (UbV-CC4) to CCHFV OTU strongly inhibits viral growth. UbV-CC4 inhibits CCHFV infection even in the absence of intact IFN signaling, suggesting that its antiviral activity is not due to blocking the OTU's immunosuppressive function. Instead, the prolonged occupancy of the OTU with UbV-CC4 directly targets viral replication by interfering with CCHFV RNA synthesis. Together, our data provide mechanistic details supporting the development of antivirals targeting viral OTUs.IMPORTANCE Crimean-Congo hemorrhagic fever virus is an important human pathogen with a wide global distribution for which no therapeutic interventions are available. CCHFV encodes a cysteine protease belonging to the ovarian tumor (OTU) family which is involved in host immune suppression. Here we demonstrate that artificially prolonged binding of the OTU to a substrate inhibits virus infection. This provides novel insights into CCHFV OTU function during the viral replicative cycle and highlights the OTU as a potential antiviral target.


Asunto(s)
Proteasas de Cisteína/metabolismo , Enzimas Desubicuitinizantes/metabolismo , Virus de la Fiebre Hemorrágica de Crimea-Congo/enzimología , Ubiquitina/farmacología , Replicación Viral , Animales , Línea Celular Tumoral , Proteasas de Cisteína/genética , Citocinas/genética , Citocinas/metabolismo , Enzimas Desubicuitinizantes/genética , Femenino , Virus de la Fiebre Hemorrágica de Crimea-Congo/fisiología , Humanos , Ratones , Mutación , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Ubiquitinación , Ubiquitinas/genética , Ubiquitinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...