Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 4855, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38844458

RESUMEN

Hepatitis E virus (HEV) is a long-neglected RNA virus and the major causative agent of acute viral hepatitis in humans. Recent data suggest that HEV has a very heterogeneous hypervariable region (HVR), which can tolerate major genomic rearrangements. In this study, we identify insertions of previously undescribed sequence snippets in serum samples of a ribavirin treatment failure patient. These insertions increase viral replication while not affecting sensitivity towards ribavirin in a subgenomic replicon assay. All insertions contain a predicted nuclear localization sequence and alanine scanning mutagenesis of lysine residues in the HVR influences viral replication. Sequential replacement of lysine residues additionally alters intracellular localization in a fluorescence dye-coupled construct. Furthermore, distinct sequence patterns outside the HVR are identified as viral determinants that recapitulate the enhancing effect. In conclusion, patient-derived insertions can increase HEV replication and synergistically acting viral determinants in and outside the HVR are described. These results will help to understand the underlying principles of viral adaptation by viral- and host-sequence snatching during the clinical course of infection.


Asunto(s)
Virus de la Hepatitis E , Hepatitis E , Ribavirina , Replicación Viral , Replicación Viral/genética , Virus de la Hepatitis E/genética , Virus de la Hepatitis E/fisiología , Virus de la Hepatitis E/efectos de los fármacos , Humanos , Hepatitis E/virología , Hepatitis E/tratamiento farmacológico , Ribavirina/farmacología , Mutagénesis Insercional , Antivirales/farmacología , ARN Viral/genética , Genoma Viral , Replicón/genética
2.
Antiviral Res ; 227: 105922, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38825018

RESUMEN

Hepatitis E is an underestimated disease, leading to estimated 20 million infections and up to 70,000 deaths annually. Infections are mostly asymptomatic, but can reach mortality rates up to 25% in pregnant women or become chronic in immunocompromised patients. Hepatitis E virus (HEV) infection have been associated with a range of extrahepatic manifestations, including a spectrum of neurological symptoms. Current therapy options are limited to non-specific antivirals like ribavirin, but recently, repurposed viral polymerase inhibitors like sofosbuvir and NITD008 were described to inhibit HEV replication. Here, we evaluated the efficacy of these drugs in various neuronal-derived cell lines to determine their potency outside the liver. Our findings indicate that both drugs, especially sofosbuvir, exhibited reduced efficacy in neuronal cells compared to hepatic cells. These results should be taken into account in the development of direct-acting antivirals for HEV and their potency at extrahepatic replication sites.


Asunto(s)
Antivirales , Virus de la Hepatitis E , Hepatitis E , Neuronas , Sofosbuvir , Replicación Viral , Sofosbuvir/farmacología , Antivirales/farmacología , Humanos , Virus de la Hepatitis E/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/virología , Línea Celular , Hepatitis E/tratamiento farmacológico , Hepatitis E/virología , Adenosina/análogos & derivados
3.
J Virol ; 98(6): e0029524, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38712945

RESUMEN

Hepatitis E virus is a single-strand, positive-sense RNA virus that can lead to chronic infection in immunocompromised patients. Virus-host recombinant variants (VHRVs) have been described in such patients. These variants integrate part of human genes into the polyproline-rich region that could introduce new post-translational modifications (PTMs), such as ubiquitination. The aim of this study was to characterize the replication capacity of different VHRVs, namely, RNF19A, ZNF787, KIF1B, EEF1A1, RNA18, RPS17, and RPL6. We used a plasmid encoding the Kernow strain, in which the fragment encoding the S17 insertion was deleted (Kernow p6 delS17) or replaced by fragments encoding the different insertions. The HEV RNA concentrations in the supernatants and the HepG2/C3A cell lysates were determined via RT-qPCR. The capsid protein ORF2 was immunostained. The effect of ribavirin was also assessed. The HEV RNA concentrations in the supernatants and the cell lysates were higher for the variants harboring the RNF19A, ZNF787, KIF1B, RPS17, and EEF1A1 insertions than for the Kernow p6 del S17, while it was not with RNA18 or RPL6 fragments. The number of ORF2 foci was higher for RNF19A, ZNF787, KIF1B, and RPS17 than for Kernow p6 del S17. VHRVs with replicative advantages were less sensitive to the antiviral effect of ribavirin. No difference in PTMs was found between VHRVs with a replicative advantage and those without. In conclusion, our study showed that insertions did not systematically confer a replicative advantage in vitro. Further studies are needed to determine the mechanisms underlying the differences in replicative capacity. IMPORTANCE: Hepatitis E virus (HEV) is a major cause of viral hepatitis. HEV can lead to chronic infection in immunocompromised patients. Ribavirin treatment is currently used to treat such chronic infections. Recently, seven virus-host recombinant viruses were characterized in immunocompromised patients. These viruses have incorporated a portion of a human gene fragment into their genome. We studied the consequences of these insertions on the replication capacity. We found that these inserted fragments could enhance virus replication for five of the seven recombinant variants. We also showed that the recombinant variants with replicative advantages were less sensitive to ribavirin in vitro. Finally, we found that the mechanisms leading to such a replicative advantage do not seem to rely on the post-translational modifications introduced by the human gene fragment that could have modified the function of the viral protein. The mechanisms involved in improving the replication of such recombinant viruses remain to be explored.


Asunto(s)
Virus de la Hepatitis E , Hepatitis E , ARN Viral , Ribavirina , Replicación Viral , Virus de la Hepatitis E/genética , Virus de la Hepatitis E/efectos de los fármacos , Humanos , Hepatitis E/virología , ARN Viral/genética , ARN Viral/metabolismo , Ribavirina/farmacología , Antivirales/farmacología , Células Hep G2 , Procesamiento Proteico-Postraduccional , Recombinación Genética
4.
Int J Mol Sci ; 23(23)2022 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-36498981

RESUMEN

The changes occurring in viral quasispecies populations during infection have been monitored using diversity indices, nucleotide diversity, and several other indices to summarize the quasispecies structure in a single value. In this study, we present a method to partition quasispecies haplotypes into four fractions according to their fitness: the master haplotype, rare haplotypes at two levels (those present at <0.1%, and those at 0.1−1%), and a fourth fraction that we term emerging haplotypes, present at frequencies >1%, but less than that of the master haplotype. We propose that by determining the changes occurring in the volume of the four quasispecies fitness fractions together with those of the Hill number profile we will be able to visualize and analyze the molecular changes in the composition of a quasispecies with time. To develop this concept, we used three data sets: a technical clone of the complete SARS-CoV-2 spike gene, a subset of data previously used in a study of rare haplotypes, and data from a clinical follow-up study of a patient chronically infected with HEV and treated with ribavirin. The viral response to ribavirin mutagenic treatment was selection of a rich set of synonymous haplotypes. The mutation spectrum was very complex at the nucleotide level, but at the protein (phenotypic/functional) level the pattern differed, showing a highly prevalent master phenotype. We discuss the putative implications of this observation in relation to mutagenic antiviral treatment.


Asunto(s)
Virus de la Hepatitis E , Hepatitis E , Ribavirina , Humanos , Estudios de Seguimiento , Mutágenos , Nucleótidos , Cuasiespecies/genética , Ribavirina/uso terapéutico , SARS-CoV-2/genética , Hepatitis E/tratamiento farmacológico , Virus de la Hepatitis E/efectos de los fármacos , Virus de la Hepatitis E/genética
5.
J Virol ; 96(6): e0190621, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35107380

RESUMEN

Hepatitis E virus (HEV) is a quasi-enveloped virus with a single-stranded positive-sense RNA genome belonging to the family Hepeviridae. Studies of the molecular aspects of HEV and drug screening have benefited from the discovery of bioluminescent reporter genes. However, the stability of large foreign genes is difficult to maintain after insertion into the viral genome. Currently, ribavirin is used to treat HEV-infected patients who require antiviral therapy. This has several major drawbacks. Thus, the development of novel anti-HEV drugs is of great importance. We developed a system consisting of recombinant infectious HEV harboring a small luciferase gene (nanoKAZ) in the hypervariable region (HVR) of the open reading frame 1 (ORF1) (HEV-nanoKAZ). It replicated efficiently in cultured cells, was genetically stable, and had morphological characteristics similar to those of the parental virus. Both membrane-associated (eHEV-nanoKAZ) and membrane-unassociated (neHEV-nanoKAZ) particles were infectious. HEV particles circulating in the bloodstream and attaching to hepatocytes in HEV-infected patients are membrane-associated; thus, eHEV-nanoKAZ was applied in drug screening. The eHEV-nanoKAZ system covers at least the inhibitor of HEV entry and inhibitor of HEV RNA replication. Four drugs with anti-HEV activity were identified. Their effectiveness in cultured cells was confirmed in naive and HEV-producing PLC/PRF/5 cells. Two hit drugs (azithromycin and ritonavir) strongly inhibited HEV production in culture supernatants, as well as intracellular expression of ORF2 protein, and may therefore be candidate novel anti-HEV drugs. The HEV-nanoKAZ system was developed and applied in drug screening and is expected to be useful for investigating the HEV life cycle. IMPORTANCE Bioluminescent reporter viruses are essential tools in molecular virological research. They have been widely used to investigate viral life cycles and in the development of antiviral drugs. For drug screening, the use of a bioluminescent reporter virus helps shorten the time required to perform the assay. A system, consisting of recombinant infectious HEV harboring the nanoKAZ gene in the HVR of ORF1 (HEV-nanoKAZ), was developed in this study and was successfully applied to drug screening in which four hit drugs with anti-HEV activity were identified. The results of this study provide evidence supporting the use of this system in more variable HEV studies. In addition, both forms of viral particles (eHEV-nanoKAZ and neHEV-nanoKAZ) are infectious, which will enable their application in HEV studies requiring both forms of viral particles, such as in the investigation of unknown HEV receptors and the elucidation of host factors important for HEV entry.


Asunto(s)
Antivirales , Evaluación Preclínica de Medicamentos , Virus de la Hepatitis E , Antivirales/farmacología , Virus de la Hepatitis E/efectos de los fármacos , Virus de la Hepatitis E/genética , Humanos , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
6.
Antiviral Res ; 197: 105228, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34929248

RESUMEN

Hepatitis E virus (HEV) infection can cause severe acute hepatitis in pregnant women and chronic infection in immunocompromised patients, promoting the development of effective antiviral therapies. In this study, we identified niclosamide, a widely used anthelmintic drug, as a potent inhibitor of HEV replication in a range of subgenomic and full-length HEV models, which are based on human cell lines and liver organoids harbouring genotype 1 and 3 HEV strains. Niclosamide is known to have multiple cellular targets including the inhibition of STAT3 and NFκB signaling pathways. Although HEV activates STAT3, we excluded its involvement in the anti-HEV activity of niclosamide. Interestingly, HEV infection activated NFκB and activation of NFκB promoted viral replication. Consistently, stable silencing of NFκB by lentiviral RNAi inhibited HEV replication. By targeting NFκB signaling, we further revealed its role in mediating the anti-HEV action of niclosamide. These results demonstrated that niclosamide potently inhibits HEV replication by inhibiting NFκB signaling but independent of STAT3. Our findings support the potential of repurposing niclosamide for treating HEV infection.


Asunto(s)
Antivirales/farmacología , Virus de la Hepatitis E/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Niclosamida/farmacología , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Células HEK293 , Hepatocitos/efectos de los fármacos , Hepatocitos/virología , Humanos , Organoides
8.
J Gen Virol ; 102(7)2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34242156

RESUMEN

Bactrian camel hepatitis E virus (HEV) is a novel HEV belonging to genotype 8 (HEV-8) in the Orthohepevirus A species of the genus Hepevirus in the family Hepeviridae. HEV-8 cross-transmits to cynomolgus monkeys and has a potential risk for zoonotic infection. Until now, neither a cell-culture system to grow the virus nor a reverse genetics system to generate the virus has been developed. To generate replication-competent HEV-8 and to establish a cell-culture system, we synthesized capped genomic HEV-8 RNAs by in vitro transcription and used them to transfect into PLC/PRF/5 cells. A HEV-8 strain, HEV-8M2, was recovered from the capped HEV-8 RNA-transfected cell-culture supernatants and subsequently passaged in the cells, demonstrating that PLC/PRF/5 cells were capable of supporting the replication of the HEV-8, and that a cell-culture system for HEV-8 was successfully established. In addition to PLC/PRF/5 cells, A549 and Caco-2 cells appeared to be competent for the replication, but HepG2 C3/A, Vero, Hela S3, HEp-2C, 293T and GL37 cells were incompetent. The HEV-8M2 strain was capable of infecting cynomolgus monkeys by an intravenous inoculation, indicating that HEV-8 was infectious and again carried a risk for zoonotic infection. In contrast, HEV-8 did not infect nude rats and BALB/c nude mice, suggesting that the reservoir of HEV-8 was limited. In addition, the replication of the HEV-8M2 strain was efficiently abrogated by ribavirin but not by favipiravir, suggesting that ribavirin is a drug candidate for therapeutic treatment of HEV-8-induced hepatitis. The infectious HEV-8 produced by a reverse genetics system would be useful to elucidate the mechanisms of HEV replication and the pathogenesis of type E hepatitis.


Asunto(s)
Virus de la Hepatitis E/genética , Virus de la Hepatitis E/fisiología , Hepatitis E/virología , Genética Inversa , Amidas/farmacología , Animales , Antivirales/farmacología , Proteínas de la Cápside/análisis , Línea Celular , Femenino , Genoma Viral , Virus de la Hepatitis E/efectos de los fármacos , Virus de la Hepatitis E/patogenicidad , Humanos , Macaca fascicularis , Masculino , Ratones , Ratones Desnudos , Pirazinas/farmacología , ARN Viral/genética , Ratas , Ribavirina/farmacología , Transfección , Replicación Viral/efectos de los fármacos
9.
Int J Antimicrob Agents ; 58(3): 106383, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34157403

RESUMEN

Hepatitis E virus (HEV) infection in immunocompromised patients, pregnant women and children requires treatment; however, no approved medication is currently available. The macrolide antibiotic azithromycin has been identified as a potent HEV inhibitor. Azithromycin inhibits HEV replication and viral protein expression in multiple cell culture models with genotype 1 and 3 strains. This is largely independent of its induction of an interferon-like response. Because it is safe and cheap, repurposing azithromycin for treating HEV infection is attractive, particularly in resource-limited settings.


Asunto(s)
Antibacterianos/farmacología , Azitromicina/farmacología , Células Cultivadas/efectos de los fármacos , Reposicionamiento de Medicamentos , Virus de la Hepatitis E/efectos de los fármacos , Hepatitis E/tratamiento farmacológico , Replicación Viral/efectos de los fármacos , Adolescente , Adulto , Antivirales/farmacología , Niño , Preescolar , Femenino , Humanos , Huésped Inmunocomprometido , Masculino , Embarazo
10.
mBio ; 12(3): e0143421, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34154410

RESUMEN

Progesterone is crucial for the maintenance of pregnancy. During pregnancy hepatitis E virus (HEV) infection is associated with increased fulminant hepatic failure and mortality rates. In this study, we determined whether progesterone modulates HEV replication and HEV-induced innate cytokine response in Huh7-S10-3 human liver cells. We first demonstrated that Huh7-S10-3 liver cells expressed SH3-domain-containing progesterone receptor membrane component (PGRMC)1/2 receptors involved in the progesterone nonclassical signaling pathway, while the classical progesterone receptor isoforms progesterone receptor-A and -B protein levels were undetectable. We showed that the genotype 3 HEV (strain P6) induced mRNA expression of type III interferon (IFN-λ1), but not other innate cytokines in Huh7-S10-3 cells. Pretreatment with progesterone at concentrations of 80 nM, 160 nM, or 480 nM, which are the physiological concentrations typically seen in the first- to third-trimester during pregnancy, significantly increased HEV replication in Huh7-S10-3 cells. However, pretreatment of cells with progesterone (80 nM) did not affect the level of HEV-induced IFN-λ1 mRNA expression. We further showed that loss of PGRMC1/2 receptors by small interfering RNA (siRNA) knockdown leads to an increase in HEV-induced IFN-λ1 expression levels at early time points via the extracellular signal-regulated kinase pathway and thus resulted in a reduced level of HEV replication. Collectively, the results indicated that progesterone-mediated modulation of HEV replication in human liver cells is plausibly through SH3-domain containing proteins such as PGRMC1/2, but not likely through immunomodulation of HEV-induced interferon response in liver cells. The results have important implications in understanding the underlying mechanisms of high mortality and fulminant hepatitis in HEV-infected pregnant women. IMPORTANCE Hepatitis E is usually a self-limiting acute disease; however, during pregnancy, a severe form of fulminant hepatic failure and high mortality rate are associated with hepatitis E virus (HEV) infection. Increased levels of progesterone and HEV RNA are observed in pregnant women with fulminant hepatic failures. Since progesterone is crucial for maintenance of pregnancy, we investigated the potential role of progesterone in HEV replication and disease pathogenesis. We demonstrated that progesterone at a concentration seen during pregnancy enhances HEV replication in human liver cells, but did not modulate HEV-induced interferon response in human liver cells. We also showed that loss of the progesterone nonclassical receptor, progesterone receptor membrane component (PGRMC)1/2, leads to a reduced level of HEV replication and an increased level of HEV-induced type III interferon (IFN-λ1) mRNA expression via the extracellular signal-regulated kinase pathway. The results from this study will aid our understanding of the underlying mechanism of pathogenesis and HEV-associated severe disease during pregnancy.


Asunto(s)
Virus de la Hepatitis E/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/virología , Progesterona/farmacología , Replicación Viral/efectos de los fármacos , Citocinas/inmunología , Femenino , Virus de la Hepatitis E/genética , Virus de la Hepatitis E/inmunología , Virus de la Hepatitis E/fisiología , Hepatocitos/inmunología , Humanos , Inmunidad Innata , Hígado/patología , Hígado/virología , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/virología , ARN Viral/genética , Replicación Viral/inmunología
11.
J Hepatol ; 75(3): 557-564, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33961939

RESUMEN

BACKGROUND & AIMS: HEV is a significant cause of acute hepatitis globally. Some genotypes establish persistent infection when immunity is impaired. Adaptive immune mechanisms that mediate resolution of infection have not been identified. Herein, the requirement for CD8+ T cells to control HEV infection was assessed in rhesus macaques, a model of acute and persistent HEV infection in humans. METHODS: Rhesus macaques were untreated or treated with depleting anti-CD8α monoclonal antibodies before challenge with an HEV genotype (gt)3 isolate derived from a chronically infected human patient. HEV replication, alanine aminotransferase, anti-capsid antibody and HEV-specific CD4+ and CD8+ T cell responses were assessed after infection. RESULTS: HEV control in untreated macaques coincided with the onset of a neutralizing IgG response against the ORF2 capsid and liver infiltration of functional HEV-specific CD4+ and CD8+ T cells. Virus control was delayed by 1 week in CD8+ T cell-depleted macaques. Infection resolved with onset of a neutralizing IgG antibody response and a much more robust expansion of CD4+ T cells with antiviral effector function. CONCLUSIONS: Liver infiltration of functional CD8+ T cells coincident with HEV clearance in untreated rhesus macaques, and a 1-week delay in HEV clearance in CD8+ T cell-depleted rhesus macaques, support a role for this subset in timely control of virus replication. Resolution of infection in the absence of CD8+ T cells nonetheless indicates that neutralizing antibodies and/or CD4+ T cells may act autonomously to inhibit HEV replication. HEV susceptibility to multiple adaptive effector mechanisms may explain why persistence occurs only with generalized immune suppression. The findings also suggest that neutralizing antibodies and/or CD4+ T cells should be considered as a component of immunotherapy for chronic infection. LAY SUMMARY: The hepatitis E virus (HEV) is a major cause of liver disease globally. Some genetic types (genotypes) of HEV persist in the body if immunity is impaired. Our objective was to identify immune responses that promote clearance of HEV. Findings indicate that HEV may be susceptible to multiple arms of the immune response that can act independently to terminate infection. They also provide a pathway to assess immune therapies for chronic HEV infection.


Asunto(s)
Hepatitis E/rehabilitación , Inmunoglobulina G/farmacología , Macaca mulatta/virología , Animales , Linfocitos T CD8-positivos/fisiología , Modelos Animales de Enfermedad , Haplorrinos , Virus de la Hepatitis E/efectos de los fármacos , Virus de la Hepatitis E/patogenicidad , Inmunoglobulina G/uso terapéutico , Hígado/virología
12.
Arch Virol ; 166(7): 2005-2010, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33987753

RESUMEN

We show that ivermectin, an FDA-approved anti-parasitic drug, effectively inhibits infection with hepatitis E virus (HEV) genotypes 1 and 3 in a range of cell culture models, including hepatic and extrahepatic cells. Long-term treatment showed no clear evidence of the development of drug resistance. Gene silencing of importin-α1, a cellular target of ivermectin and a key member of the host nuclear transport complex, inhibited viral replication and largely abolished the anti-HEV effect of ivermectin.


Asunto(s)
Virus de la Hepatitis E/efectos de los fármacos , Hepatitis E/tratamiento farmacológico , Ivermectina/farmacología , Replicación Viral/efectos de los fármacos , alfa Carioferinas/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Núcleo Celular/virología , Hepatitis E/metabolismo , Hepatitis E/virología , Humanos , Proteínas Nucleares/metabolismo
13.
Cell Mol Gastroenterol Hepatol ; 12(1): 159-180, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33601063

RESUMEN

BACKGROUND AND AIMS: The Hepatitis E virus hijacks the endosomal system for its release. These structures are highly dependent on cholesterol. Hence, this study investigates the impact of HEV on cholesterol-metabolism, the effect of intracellular cholesterol content on HEV-release and the potential of cholesterol-modulators to serve as antivirals. METHODS: Intracellular cholesterol-content of cells was modulated and impacts on HEV were monitored using qPCR, Western blot, microscopy, virus-titration and density-gradient centrifugation. Blood-lipids and HEV-RNA were routinely quantified in chronically infected patients during follow-up visits. RESULTS: In HEV-infected cells, decreased levels of cholesterol are found. In patients, HEV infection decreases serum-lipid concentrations. Importantly, statin treatment herein increases viral titers. Similarly, reduction of intracellular cholesterol via simvastatin treatment increases viral release in vitro. On the contrary, elevating intracellular cholesterol via LDL or 25-hydroxycholesterol strongly reduces viral release due to enhanced lysosomal degradation of HEV. Drug-induced elevation of intracellular cholesterol via fenofibrate or PSC833 impairs HEV release via the same mechanism. CONCLUSIONS: This study analyses the crosstalk between HEV and intracellular cholesterol. The results highlight the importance of an intact cholesterol homeostasis for HEV-release and thereby identify a potential target for antiviral strategies. Especially fenofibrate is considered a promising novel antiviral against HEV. Beyond this, the study may help clinicians evaluating co-treatments of HEV-infected patients with statins, as this may be counter indicated.


Asunto(s)
Antivirales/farmacología , Colesterol/metabolismo , Ciclosporinas/farmacología , Fenofibrato/farmacología , Virus de la Hepatitis E/efectos de los fármacos , Antivirales/química , Supervivencia Celular/efectos de los fármacos , Ciclosporinas/química , Fenofibrato/química , Humanos , Pruebas de Sensibilidad Microbiana , Células Tumorales Cultivadas , Replicación Viral/efectos de los fármacos
14.
Antiviral Res ; 185: 104997, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33326835

RESUMEN

Hepatitis E virus (HEV) causes 14 million infections and 60,000 deaths per year globally, with immunocompromised persons and pregnant women experiencing severe symptoms. Although ribavirin can be used to treat chronic hepatitis E, toxicity in pregnant patients and the emergence of resistant strains are major concerns. Therefore there is an imminent need for effective HEV antiviral agents. The aims of this study were to develop a drug screening platform and to discover novel approaches to targeting steps within the viral life cycle. We developed a screening platform for molecules inhibiting HEV replication and selected a candidate, isocotoin. Isocotoin inhibits HEV replication through interference with heat shock protein 90 (HSP90), a host factor not previously known to be involved in HEV replication. Additional work is required to understand the compound's translational potential, however this suggests that HSP90-modulating molecules, which are in clinical development as anti-cancer agents, may be promising therapies against HEV.


Asunto(s)
Antivirales/farmacología , Descubrimiento de Drogas , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Virus de la Hepatitis E/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Interacciones Microbiota-Huesped/efectos de los fármacos , Antivirales/aislamiento & purificación , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Proteínas HSP90 de Choque Térmico/metabolismo , Hepatitis E/tratamiento farmacológico , Virus de la Hepatitis E/química , Humanos , Unión Proteica , Replicación Viral/efectos de los fármacos
15.
Antiviral Res ; 184: 104967, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33137361

RESUMEN

Exposure to hepatitis E virus (HEV) bears a high risk of developing chronic infection in immunocompromised patients, including organ transplant recipients and cancer patients. We aim to identify effective anti-HEV therapies through screening and repurposing safe-in-human broad-spectrum antiviral agents. In this study, a safe-in-human broad-spectrum antiviral drug library comprising of 94 agents was used. Upon screening, we identified gemcitabine, a widely used anti-cancer drug, as a potent inhibitor of HEV replication. The antiviral effect was confirmed in a range of cell culture models with genotype 1 and 3 HEV strains. As a cytidine analog, exogenous supplementation of pyrimidine nucleosides effectively reversed the antiviral activity of gemcitabine, but the level of pyrimidine nucleosides per se does not affect HEV replication. Surprisingly, similar to interferon-alpha (IFNα) treatment, gemcitabine activates STAT1 phosphorylation. This subsequently triggers activation of interferon-sensitive response element (ISRE) and transcription of interferon-stimulated genes (ISGs). Cytidine or uridine effectively inhibits gemcitabine-induced activation of ISRE and ISGs. As expected, JAK inhibitor 1 blocked IFNα, but not gemcitabine-induced STAT1 phosphorylation, ISRE/ISG activation, and anti-HEV activity. These effects of gemcitabine were completely lost in STAT1 knockout cells. In summary, gemcitabine potently inhibits HEV replication by triggering interferon-like response through STAT1 phosphorylation but independent of Janus kinases. This represents a non-canonical antiviral mechanism, which utilizes the innate defense machinery that is distinct from the classical interferon response. These results support repurposing gemcitabine for treating hepatitis E, especially for HEV-infected cancer patients, leading to dual anti-cancer and antiviral effects.


Asunto(s)
Desoxicitidina/análogos & derivados , Virus de la Hepatitis E/efectos de los fármacos , Interferón-alfa , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT1/metabolismo , Antivirales/farmacología , Línea Celular , Desoxicitidina/farmacología , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Sinergismo Farmacológico , Regulación de la Expresión Génica , Hepatitis E/tratamiento farmacológico , Virus de la Hepatitis E/fisiología , Interacciones Microbiota-Huesped , Humanos , Interferón-alfa/farmacología , Quinasas Janus/metabolismo , Ácido Micofenólico/antagonistas & inhibidores , Nucleósidos de Pirimidina/farmacología , Elementos de Respuesta , Ribavirina/antagonistas & inhibidores , Transducción de Señal , Replicación Viral/efectos de los fármacos , Gemcitabina
16.
Food Environ Virol ; 12(4): 350-354, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32852672

RESUMEN

Hepatitis E virus (HEV) infection can cause acute and chronic hepatitis in humans. The zoonotic HEV genotype 3, which is highly prevalent in Europe, is mainly transmitted by consumption of raw meat and raw meat products produced from infected pigs or wild boars. High salt concentrations represent an important measure to preserve meat products and to inactivate foodborne pathogens. Here, an HEV preparation in phosphate-buffered saline (PBS) was subjected to different salt concentrations and the remaining infectivity was measured in a cell culture assay. Treatments with up to 20% sodium chloride for 24 h at 23 °C, with and without addition of 0.015% sodium nitrite or 0.03% sodium nitrate, did not lead to virus inactivation as compared to PBS only. Conditions usually applied for short-term and long-term fermented raw sausages were simulated by incubation at 22 °C for up to 6 days and at 16 °C for up to 8 weeks, respectively. Only 2% sodium chloride with 0.015% sodium nitrite showed a weak (< 1 log10), but significant, infectivity reduction after 2 and 4 days as compared to PBS only. Addition of 2% sodium chloride and 0.03% sodium nitrate showed a slight, but not significant, decrease in infectivity after 2 and 8 weeks as compared to PBS only. In conclusion, HEV is highly stable at high salt concentrations and at salt conditions usually applied to preserve raw meat products.


Asunto(s)
Conservación de Alimentos/métodos , Conservantes de Alimentos/farmacología , Virus de la Hepatitis E/efectos de los fármacos , Nitratos/farmacología , Cloruro de Sodio/farmacología , Nitrito de Sodio/farmacología , Animales , Conservantes de Alimentos/análisis , Hepatitis E/transmisión , Hepatitis E/virología , Virus de la Hepatitis E/fisiología , Humanos , Productos de la Carne/virología , Nitratos/análisis , Cloruro de Sodio/análisis , Nitrito de Sodio/análisis , Porcinos , Inactivación de Virus/efectos de los fármacos
17.
BMC Infect Dis ; 20(1): 522, 2020 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-32677900

RESUMEN

BACKGROUND: Hepatitis E virus (HEV) may be resistant to immunosuppression reduction and ribavirin treatment in kidney transplant recipients because of mutant strains and severe side effects of ribavirin which conduct to dose reduction. Sofosbuvir efficacy is controversial. Peg-interferon 2 alpha (PEG-IFN) is currently contraindicated due to a high risk of acute humoral and cellular rejection. The present study assessed, for the first time, the effect of PEG-IFN in a kidney transplant recipient infected with HEV. CASE PRESENTATION: The patient had chronic active HEV that was resistant to immunosuppression reduction and optimal ribavirin treatment. He developed significant liver fibrosis. PEG-IFN was administered for 10 months, and it was well tolerated and did not induce rejection. A sustained virological response was obtained. CONCLUSIONS: We conclude that prolonged treatment with PEG-IFN in kidney transplant recipients infected with HEV could be considered as a salvage option.


Asunto(s)
Farmacorresistencia Viral/efectos de los fármacos , Hepatitis E/tratamiento farmacológico , Hepatitis Crónica/tratamiento farmacológico , Interferón-alfa/uso terapéutico , Trasplante de Riñón , Polietilenglicoles/uso terapéutico , Ribavirina/uso terapéutico , Receptores de Trasplantes , Hepatitis E/virología , Virus de la Hepatitis E/efectos de los fármacos , Virus de la Hepatitis E/fisiología , Hepatitis Crónica/virología , Humanos , Masculino , Persona de Mediana Edad , Proteínas Recombinantes/uso terapéutico , Inducción de Remisión , Respuesta Virológica Sostenida , Resultado del Tratamiento
19.
J Cell Mol Med ; 24(12): 6988-6999, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32374474

RESUMEN

Outbreaks of infections with viruses like Sars-CoV-2, Ebola virus and Zika virus lead to major global health and economic problems because of limited treatment options. Therefore, new antiviral drug candidates are urgently needed. The promising new antiviral drug candidate silvestrol effectively inhibited replication of Corona-, Ebola-, Zika-, Picorna-, Hepatis E and Chikungunya viruses. Besides a direct impact on pathogens, modulation of the host immune system provides an additional facet to antiviral drug development because suitable immune modulation can boost innate defence mechanisms against the pathogens. In the present study, silvestrol down-regulated several pro- and anti-inflammatory cytokines (IL-6, IL-8, IL-10, CCL2, CCL18) and increased TNF-α during differentiation and activation of M1-macrophages, suggesting that the effects of silvestrol might cancel each other out. However, silvestrol amplified the anti-inflammatory potential of M2-macrophages by increasing expression of anti-inflammatory surface markers CD206, TREM2 and reducing release of pro-inflammatory IL-8 and CCL2. The differentiation of dendritic cells in the presence of silvestrol is characterized by down-regulation of several surface markers and cytokines indicating that differentiation is impaired by silvestrol. In conclusion, silvestrol influences the inflammatory status of immune cells depending on the cell type and activation status.


Asunto(s)
Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Citocinas/genética , Células Dendríticas/efectos de los fármacos , Factores Inmunológicos/farmacología , Macrófagos/efectos de los fármacos , Triterpenos/farmacología , Betacoronavirus/crecimiento & desarrollo , Betacoronavirus/inmunología , Diferenciación Celular/efectos de los fármacos , Virus Chikungunya/efectos de los fármacos , Virus Chikungunya/crecimiento & desarrollo , Virus Chikungunya/inmunología , Citocinas/clasificación , Citocinas/inmunología , Células Dendríticas/inmunología , Células Dendríticas/virología , Ebolavirus/efectos de los fármacos , Ebolavirus/crecimiento & desarrollo , Ebolavirus/inmunología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Virus de la Hepatitis E/efectos de los fármacos , Virus de la Hepatitis E/crecimiento & desarrollo , Virus de la Hepatitis E/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/virología , Especificidad de Órganos , Picornaviridae/efectos de los fármacos , Picornaviridae/crecimiento & desarrollo , Picornaviridae/inmunología , Cultivo Primario de Células , SARS-CoV-2 , Transducción de Señal , Virus Zika/efectos de los fármacos , Virus Zika/crecimiento & desarrollo , Virus Zika/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA