Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(2): 379-396.e38, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-35021063

RESUMEN

The liver is the largest solid organ in the body, yet it remains incompletely characterized. Here we present a spatial proteogenomic atlas of the healthy and obese human and murine liver combining single-cell CITE-seq, single-nuclei sequencing, spatial transcriptomics, and spatial proteomics. By integrating these multi-omic datasets, we provide validated strategies to reliably discriminate and localize all hepatic cells, including a population of lipid-associated macrophages (LAMs) at the bile ducts. We then align this atlas across seven species, revealing the conserved program of bona fide Kupffer cells and LAMs. We also uncover the respective spatially resolved cellular niches of these macrophages and the microenvironmental circuits driving their unique transcriptomic identities. We demonstrate that LAMs are induced by local lipid exposure, leading to their induction in steatotic regions of the murine and human liver, while Kupffer cell development crucially depends on their cross-talk with hepatic stellate cells via the evolutionarily conserved ALK1-BMP9/10 axis.


Asunto(s)
Evolución Biológica , Hepatocitos/metabolismo , Macrófagos/metabolismo , Proteogenómica , Animales , Núcleo Celular/metabolismo , Hígado Graso/genética , Hígado Graso/patología , Homeostasis , Humanos , Macrófagos del Hígado/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Lípidos/química , Hígado/metabolismo , Linfocitos/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Células Mieloides/metabolismo , Obesidad/patología , Proteoma/metabolismo , Transducción de Señal , Transcriptoma/genética
2.
Nature ; 615(7953): 678-686, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36922586

RESUMEN

Dengue is a major health threat and the number of symptomatic infections caused by the four dengue serotypes is estimated to be 96 million1 with annually around 10,000 deaths2. However, no antiviral drugs are available for the treatment or prophylaxis of dengue. We recently described the interaction between non-structural proteins NS3 and NS4B as a promising target for the development of pan-serotype dengue virus (DENV) inhibitors3. Here we present JNJ-1802-a highly potent DENV inhibitor that blocks the NS3-NS4B interaction within the viral replication complex. JNJ-1802 exerts picomolar to low nanomolar in vitro antiviral activity, a high barrier to resistance and potent in vivo efficacy in mice against infection with any of the four DENV serotypes. Finally, we demonstrate that the small-molecule inhibitor JNJ-1802 is highly effective against viral infection with DENV-1 or DENV-2 in non-human primates. JNJ-1802 has successfully completed a phase I first-in-human clinical study in healthy volunteers and was found to be safe and well tolerated4. These findings support the further clinical development of JNJ-1802, a first-in-class antiviral agent against dengue, which is now progressing in clinical studies for the prevention and treatment of dengue.


Asunto(s)
Antivirales , Virus del Dengue , Dengue , Primates , Proteínas no Estructurales Virales , Animales , Humanos , Ratones , Antivirales/efectos adversos , Antivirales/farmacología , Antivirales/uso terapéutico , Ensayos Clínicos Fase I como Asunto , Dengue/tratamiento farmacológico , Dengue/prevención & control , Dengue/virología , Virus del Dengue/clasificación , Virus del Dengue/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Farmacorresistencia Viral , Técnicas In Vitro , Terapia Molecular Dirigida , Primates/virología , Unión Proteica/efectos de los fármacos , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
3.
Nature ; 598(7881): 504-509, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34616043

RESUMEN

Dengue virus causes approximately 96 million symptomatic infections annually, manifesting as dengue fever or occasionally as severe dengue1,2. There are no antiviral agents available to prevent or treat dengue. Here, we describe a highly potent dengue virus inhibitor (JNJ-A07) that exerts nanomolar to picomolar activity against a panel of 21 clinical isolates that represent the natural genetic diversity of known genotypes and serotypes. The molecule has a high barrier to resistance and prevents the formation of the viral replication complex by blocking the interaction between two viral proteins (NS3 and NS4B), thus revealing a previously undescribed mechanism of antiviral action. JNJ-A07 has a favourable pharmacokinetic profile that results in outstanding efficacy against dengue virus infection in mouse infection models. Delaying start of treatment until peak viraemia results in a rapid and significant reduction in viral load. An analogue is currently in further development.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/clasificación , Virus del Dengue/efectos de los fármacos , Dengue/virología , Proteínas de la Membrana/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Antivirales/farmacocinética , Antivirales/uso terapéutico , Dengue/tratamiento farmacológico , Virus del Dengue/genética , Virus del Dengue/metabolismo , Modelos Animales de Enfermedad , Femenino , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , ARN Helicasas/antagonistas & inhibidores , ARN Helicasas/metabolismo , Serina Endopeptidasas/metabolismo , Carga Viral/efectos de los fármacos , Proteínas no Estructurales Virales/antagonistas & inhibidores , Viremia/tratamiento farmacológico , Viremia/virología , Replicación Viral/efectos de los fármacos
4.
Nature ; 590(7845): 320-325, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33260195

RESUMEN

The expanding pandemic of coronavirus disease 2019 (COVID-19) requires the development of safe, efficacious and fast-acting vaccines. Several vaccine platforms are being leveraged for a rapid emergency response1. Here we describe the development of a candidate vaccine (YF-S0) for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that uses live-attenuated yellow fever 17D (YF17D) vaccine as a vector to express a noncleavable prefusion form of the SARS-CoV-2 spike antigen. We assess vaccine safety, immunogenicity and efficacy in several animal models. YF-S0 has an excellent safety profile and induces high levels of SARS-CoV-2 neutralizing antibodies in hamsters (Mesocricetus auratus), mice (Mus musculus) and cynomolgus macaques (Macaca fascicularis), and-concomitantly-protective immunity against yellow fever virus. Humoral immunity is complemented by a cellular immune response with favourable T helper 1 polarization, as profiled in mice. In a hamster model2 and in macaques, YF-S0 prevents infection with SARS-CoV-2. Moreover, a single dose conferred protection from lung disease in most of the vaccinated hamsters within as little as 10 days. Taken together, the quality of the immune responses triggered and the rapid kinetics by which protective immunity can be attained after a single dose warrant further development of this potent SARS-CoV-2 vaccine candidate.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , COVID-19/prevención & control , Vectores Genéticos/genética , SARS-CoV-2/inmunología , Vacunas Atenuadas/inmunología , Vacuna contra la Fiebre Amarilla/genética , Animales , Vacunas contra la COVID-19/administración & dosificación , Vacunas contra la COVID-19/efectos adversos , Vacunas contra la COVID-19/genética , Cricetinae , Modelos Animales de Enfermedad , Femenino , Glicosilación , Macaca fascicularis/genética , Macaca fascicularis/inmunología , Macaca fascicularis/virología , Masculino , Mesocricetus/genética , Mesocricetus/inmunología , Mesocricetus/virología , Ratones , Seguridad , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/genética
5.
Nature ; 586(7830): 509-515, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32967005

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the aetiological agent of coronavirus disease 2019 (COVID-19), an emerging respiratory infection caused by the introduction of a novel coronavirus into humans late in 2019 (first detected in Hubei province, China). As of 18 September 2020, SARS-CoV-2 has spread to 215 countries, has infected more than 30 million people and has caused more than 950,000 deaths. As humans do not have pre-existing immunity to SARS-CoV-2, there is an urgent need to develop therapeutic agents and vaccines to mitigate the current pandemic and to prevent the re-emergence of COVID-19. In February 2020, the World Health Organization (WHO) assembled an international panel to develop animal models for COVID-19 to accelerate the testing of vaccines and therapeutic agents. Here we summarize the findings to date and provides relevant information for preclinical testing of vaccine candidates and therapeutic agents for COVID-19.


Asunto(s)
Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/prevención & control , Modelos Animales de Enfermedad , Pandemias/prevención & control , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/prevención & control , Animales , Betacoronavirus/efectos de los fármacos , Betacoronavirus/inmunología , COVID-19 , Vacunas contra la COVID-19 , Infecciones por Coronavirus/inmunología , Hurones/virología , Humanos , Mesocricetus/virología , Ratones , Neumonía Viral/inmunología , Primates/virología , SARS-CoV-2 , Vacunas Virales/inmunología
6.
Eur J Immunol ; 54(5): e2250133, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38571392

RESUMEN

Live-attenuated yellow fever vaccine (YF17D) was developed in the 1930s as the first ever empirically derived human vaccine. Ninety years later, it is still a benchmark for vaccines made today. YF17D triggers a particularly broad and polyfunctional response engaging multiple arms of innate, humoral and cellular immunity. This unique immunogenicity translates into an extraordinary vaccine efficacy and outstanding longevity of protection, possibly by single-dose immunization. More recently, progress in molecular virology and synthetic biology allowed engineering of YF17D as a powerful vector and promising platform for the development of novel recombinant live vaccines, including two licensed vaccines against Japanese encephalitis and dengue, even in paediatric use. Likewise, numerous chimeric and transgenic preclinical candidates have been described. These include prophylactic vaccines against emerging viral infections (e.g. Lassa, Zika and SARS-CoV-2) and parasitic diseases (e.g. malaria), as well as therapeutic applications targeting persistent infections (e.g. HIV and chronic hepatitis), and cancer. Efforts to overcome historical safety concerns and manufacturing challenges are ongoing and pave the way for wider use of YF17D-based vaccines. In this review, we summarize recent insights regarding YF17D as vaccine platform, and how YF17D-based vaccines may complement as well as differentiate from other emerging modalities in response to unmet medical needs and for pandemic preparedness.


Asunto(s)
Vacunas Atenuadas , Vacuna contra la Fiebre Amarilla , Virus de la Fiebre Amarilla , Humanos , Vacuna contra la Fiebre Amarilla/inmunología , Virus de la Fiebre Amarilla/inmunología , Vacunas Atenuadas/inmunología , Animales , Fiebre Amarilla/prevención & control , Fiebre Amarilla/inmunología , Vacunación/métodos
7.
Proc Natl Acad Sci U S A ; 117(43): 26955-26965, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33037151

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) rapidly spread around the globe after its emergence in Wuhan in December 2019. With no specific therapeutic and prophylactic options available, the virus has infected millions of people of which more than half a million succumbed to the viral disease, COVID-19. The urgent need for an effective treatment together with a lack of small animal infection models has led to clinical trials using repurposed drugs without preclinical evidence of their in vivo efficacy. We established an infection model in Syrian hamsters to evaluate the efficacy of small molecules on both infection and transmission. Treatment of SARS-CoV-2-infected hamsters with a low dose of favipiravir or hydroxychloroquine with(out) azithromycin resulted in, respectively, a mild or no reduction in virus levels. However, high doses of favipiravir significantly reduced infectious virus titers in the lungs and markedly improved lung histopathology. Moreover, a high dose of favipiravir decreased virus transmission by direct contact, whereas hydroxychloroquine failed as prophylaxis. Pharmacokinetic modeling of hydroxychloroquine suggested that the total lung exposure to the drug did not cause the failure. Our data on hydroxychloroquine (together with previous reports in macaques and ferrets) thus provide no scientific basis for the use of this drug in COVID-19 patients. In contrast, the results with favipiravir demonstrate that an antiviral drug at nontoxic doses exhibits a marked protective effect against SARS-CoV-2 in a small animal model. Clinical studies are required to assess whether a similar antiviral effect is achievable in humans without toxic effects.


Asunto(s)
Amidas/uso terapéutico , Antivirales/uso terapéutico , Betacoronavirus/efectos de los fármacos , Hidroxicloroquina/uso terapéutico , Pirazinas/uso terapéutico , Amidas/farmacocinética , Animales , Chlorocebus aethiops , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Cricetinae , Modelos Animales de Enfermedad , Transmisión de Enfermedad Infecciosa/prevención & control , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Hidroxicloroquina/farmacocinética , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/virología , Pirazinas/farmacocinética , SARS-CoV-2 , Resultado del Tratamiento , Células Vero , Carga Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
9.
PLoS Pathog ; 15(5): e1007760, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31071193

RESUMEN

Enterovirus A71 (EV-A71) is a non-polio neurotropic enterovirus with pandemic potential. There are no antiviral agents approved to prevent or treat EV-A71 infections. We here report on the molecular mechanism by which a novel class of tryptophan dendrimers inhibits (at low nanomolar to high picomolar concentration) EV-A71 replication in vitro. A lead compound in the series (MADAL385) prevents binding and internalization of the virus but does not, unlike classical capsid binders, stabilize the particle. By means of resistance selection, reverse genetics and cryo-EM, we map the binding region of MADAL385 to the 5-fold vertex of the viral capsid and demonstrate that a single molecule binds to each vertex. By interacting with this region, MADAL385 prevents the interaction of the virus with its cellular receptors PSGL1 and heparan sulfate, thereby blocking the attachment of EV-A71 to the host cells.


Asunto(s)
Antivirales/farmacología , Cápside/metabolismo , Infecciones por Enterovirus/metabolismo , Enterovirus/efectos de los fármacos , Heparitina Sulfato/metabolismo , Glicoproteínas de Membrana/metabolismo , Triptófano/farmacología , Antivirales/química , Cápside/efectos de los fármacos , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Dendrímeros/química , Dendrímeros/farmacología , Infecciones por Enterovirus/tratamiento farmacológico , Infecciones por Enterovirus/virología , Células HeLa , Heparitina Sulfato/antagonistas & inhibidores , Humanos , Glicoproteínas de Membrana/antagonistas & inhibidores , Conformación Proteica , Triptófano/química , Replicación Viral/efectos de los fármacos
10.
RNA Biol ; 18(5): 732-744, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33459164

RESUMEN

Flaviviruses such as the dengue (DENV) and the Zika virus (ZIKV) are important human pathogens causing around 100 million symptomatic infections each year. During infection, small subgenomic flavivirus RNAs (sfRNAs) are formed inside the infected host cell as a result of incomplete degradation of the viral RNA genome by cellular exoribonuclease XRN1. Although the full extent of sfRNA functions is to be revealed, these non-coding RNAs are key virulence factors and their detrimental effects on multiple cellular processes seem to consistently involve molecular interactions with RNA-binding proteins (RBPs). Discovery of such sfRNA-binding host-factors has followed established biochemical pull-down approaches skewed towards highly abundant proteins hampering proteome-wide coverage. Yeast three-hybrid (Y3H) systems represent an attractive alternative approach. To facilitate proteome-wide screens for RBP, we revisited and improved existing RNA-Y3H methodology by (1) implementing full-length ORF libraries in combination with (2) efficient yeast mating to increase screening depth and sensitivity, and (3) stringent negative controls to eliminate over-representation of non-specific RNA-binders. These improvements were validated employing the well-characterized interaction between DDX6 (DEAD-box helicase 6) and sfRNA of DENV as paradigm. Our advanced Y3H system was used to screen for human proteins binding to DENV and ZIKV sfRNA, resulting in a list of 69 putative sfRNA-binders, including several previously reported as well as numerous novel RBP host factors. Our methodology requiring no sophisticated infrastructure or analytic pipeline may be employed for the discovery of meaningful RNA-protein interactions at large scale in other fields.


Asunto(s)
Interacciones Huésped-Patógeno , Mapas de Interacción de Proteínas , ARN Viral/metabolismo , Proteínas de Unión al ARN/aislamiento & purificación , Células Cultivadas , Dengue/genética , Dengue/metabolismo , Virus del Dengue/genética , Genoma Humano , Humanos , Sistemas de Lectura Abierta/genética , Organismos Modificados Genéticamente , Unión Proteica , Estabilidad del ARN , ARN Viral/genética , Proteínas de Unión al ARN/análisis , Proteínas de Unión al ARN/metabolismo , Saccharomyces cerevisiae , Técnicas del Sistema de Dos Híbridos , Virus Zika/genética , Infección por el Virus Zika/genética , Infección por el Virus Zika/metabolismo
11.
J Proteome Res ; 19(7): 2529-2538, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32216351

RESUMEN

RNA-protein interactions are essential for the regulation of mRNA and noncoding RNA functions and are implicated in many diseases, such as cancer and neurodegenerative disorders. A method that can detect RNA-protein interactions in living mammalian cells on a proteome-wide scale will be an important asset to identify and study these interactions. Here we show that a combination of the mammalian two-hybrid protein-protein detection method KISS (kinase substrate sensor) and the yeast RNA three-hybrid method, utilizing the specific interaction between the MS2 RNA and MS2 coat protein, is capable of detecting RNA-protein interactions in living mammalian cells. For conceptional proof we used the subgenomic flavivirus RNA (sfRNA) of the dengue virus (DENV), a highly structured noncoding RNA derived from the DENV genome known to target host cell proteins involved in innate immunity and antiviral defense, as bait. Using RNA-KISS, we could confirm the previously established interaction between the RNA-binding domain of DDX6 and the DENV sfRNA. Finally, we performed a human proteome-wide screen for DENV sfRNA-binding host factors, identifying several known flavivirus host factors such as DDX6 and PACT, further validating the RNA-KISS method as a robust and high-throughput cell-based RNA-protein interaction screening tool.


Asunto(s)
Flavivirus , ARN Viral , Animales , ARN Helicasas DEAD-box , Flavivirus/genética , Humanos , Proteínas Proto-Oncogénicas , ARN no Traducido , ARN Viral/genética , Replicación Viral
12.
J Neuroinflammation ; 17(1): 315, 2020 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-33097065

RESUMEN

BACKGROUND: Japanese encephalitis virus (JEV) is the leading cause of viral encephalitis in Asia. JEV infection of mice and humans can lead to an uncontrolled inflammatory response in the central nervous system (CNS), resulting in a detrimental outcome. Pigs act as important amplification and reservoir hosts, and JEV infection of pigs is mostly subclinical. Information on virus spread in the CNS and immune responses controlling JEV infection in the CNS of pigs, however remains scarce. METHODS: Nine-week-old pigs were inoculated intranasal or intradermal with a relevant dose of 105 TCID50 of JEV genotype 3 Nakayama strain. Clinical signs were assessed daily, and viral spread was followed by RT-qPCR. mRNA expression profiles were determined to study immune responses in the CNS. RESULTS: Besides a delay of 2 days to reach the peak viremia upon intranasal compared to intradermal inoculation, the overall virus spread via both inoculation routes was highly similar. JEV appearance in lymphoid and visceral organs was in line with a blood-borne JEV dissemination. JEV showed a particular tropism to the CNS but without the induction of neurological signs. JEV entry in the CNS probably occurred via different hematogenous and neuronal pathways, but replication in the brain was mostly efficiently suppressed and associated with a type I IFN-independent activation of OAS1 expression. In the olfactory bulb and thalamus, where JEV replication was not completely controlled by this mechanism, a short but strong induction of chemokine gene expression was detected. An increased IFNy expression was simultaneously observed, probably originating from infiltrating T cells, correlating with a fast suppression of JEV replication. The chemokine response was however not associated with the induction of a strong inflammatory response, nor was an induction of the NLRP3 inflammasome observed. CONCLUSIONS: These findings indicate that an adequate antiviral response and an attenuated inflammatory response contribute to a favorable outcome of JEV infection in pigs and help to explain the limited neurological disease compared to other hosts. We show that the NLRP3 inflammasome, a key mediator of neurologic disease in mice, is not upregulated in pigs, further supporting its important role in JEV infections.


Asunto(s)
Sistema Nervioso Central/inmunología , Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis Japonesa/inmunología , Inmunidad/inmunología , Mediadores de Inflamación/inmunología , Animales , Sistema Nervioso Central/patología , Chlorocebus aethiops , Virus de la Encefalitis Japonesa (Especie)/aislamiento & purificación , Encefalitis Japonesa/patología , Inflamación/inmunología , Inflamación/patología , Mediadores de Inflamación/metabolismo , Porcinos , Células Vero
13.
Biologicals ; 64: 83-95, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32089431

RESUMEN

Rabies is a major neglected zoonotic disease and causes a substantial burden in the Asian region. Currently, Pacific Oceania is free of rabies but enzootic areas throughout southeast Asia represent a major risk of disease introduction to this region. On September 25-26, 2019, researchers, government officials and related stakeholders met at an IABS conference in Bangkok, Thailand to engage on the topic of human rabies mediated by dogs. The objective of the meeting was focused upon snowballing efforts towards achieving substantial progress in rabies prevention, control and elimination within Asia by 2030, and thereby to safeguard the Pacific region. Individual sessions focused upon domestic animal, wildlife and human vaccination; the production and evaluation of quality, safety and efficacy of existing rabies biologics; and the future development of new products. Participants reviewed the progress to date in eliminating canine rabies by mass vaccination, described supportive methods to parenteral administration by oral vaccine application, considered updated global and local approaches at human prophylaxis and discussed the considerable challenges ahead. Such opportunities provide continuous engagement on disease management among professionals at a trans-disciplinary level and promote new applied research collaborations in a modern One Health context.


Asunto(s)
Enfermedades de los Perros , Vacunas Antirrábicas/uso terapéutico , Rabia , Zoonosis , Animales , Congresos como Asunto , Enfermedades de los Perros/epidemiología , Enfermedades de los Perros/prevención & control , Perros , Humanos , Rabia/epidemiología , Rabia/prevención & control , Tailandia , Zoonosis/epidemiología , Zoonosis/prevención & control
14.
J Hepatol ; 64(3): 565-73, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26626494

RESUMEN

BACKGROUND & AIMS: Yearly, approximately 20million people become infected with the hepatitis E virus (HEV) resulting in over 3million cases of acute hepatitis. Although HEV-mediated hepatitis is usually self-limiting, severe cases of fulminant hepatitis as well as chronic infections have been reported, resulting annually in an estimated 60,000 deaths. We studied whether pluripotent stem cell (PSC)-derived hepatocytes, mesodermal and/or neuroprogenitor cells support HEV replication. METHODS: Human PSC were differentiated towards hepatocyte-like cells, mesodermal cells and neuroprogenitors and subsequently infected with HEV. Infection and replication of HEV was analyzed by qRT-PCR, RNA in situ hybridization, negative strand RT-PCR, production of infectious virions and transfection with a transient HEV reporter replicon. RESULTS: PSC-derived hepatocytes supported the complete replication cycle of HEV, as demonstrated by the intracellular presence of positive and negative strand HEV RNA and the production of infectious virions. The replication of the virus in these cells was inhibited by the antiviral drugs ribavirin and interferon-α2b. In contrast to PSC-derived hepatocytes, PSC-derived mesodermal cells and neuroprogenitors only supported HEV replication upon transfection with a HEV subgenomic replicon. CONCLUSION: We demonstrate that PSC can be used to study the hepatotropism of HEV infection. The complete replication cycle of HEV can be recapitulated in infected PSC-derived hepatocytes. By contrast other germ layer cells support intracellular replication but are not infectable with HEV. Thus the early steps in the viral cycle are the main determinant governing HEV tissue tropism. PSC-hepatocytes offer a physiological relevant tool to study the biology of HEV infection and replication and may aid in the design of therapeutic strategies.


Asunto(s)
Virus de la Hepatitis E/fisiología , Hepatocitos/virología , Células Madre Pluripotentes/citología , Replicación Viral , Células Hep G2 , Humanos , ARN Viral/análisis , Internalización del Virus
15.
J Hepatol ; 65(3): 499-508, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27174035

RESUMEN

BACKGROUND & AIMS: Ribavirin monotherapy is the preferred treatment for chronic hepatitis E, although occasional treatment failure occurs. We present a patient with chronic hepatitis E experiencing ribavirin treatment failure with a completely resistant phenotype. We aimed to identify viral mutations associated with treatment failure and explore the underlying mechanisms. METHODS: Viral genomes were deep-sequenced at different time points and the role of identified mutations was assessed in vitro using mutant replicons, antiviral assays, cell culture of patient-derived virus and deep-sequencing. RESULTS: Ribavirin resistance was associated with Y1320H, K1383N and G1634R mutations in the viral polymerase, but also an insertion in the hypervariable region comprising a duplication and a polymerase-derived fragment. Analysis of these genome alterations in vitro revealed replication-increasing roles for Y1320H and G1634R mutations and the hypervariable region insertion. In contrast, the K1383N mutation in the polymerase F1-motif suppressed viral replication and increased the in vitro sensitivity to ribavirin, contrary to the clinical phenotype. Analysis of the replication of mutant full-length virus and in vitro culturing of patient-derived virus confirmed that sensitivity to ribavirin was retained. Finally, deep-sequencing of hepatitis E virus genomes revealed that ribavirin is mutagenic to viral replication in vitro and in vivo. CONCLUSIONS: Mutations Y1320H, G1634R and the hypervariable region insertion compensated for K1383N-associated replication defects. The specific role of the K1383N mutation remains enigmatic, but it appears to be of importance for the ribavirin resistant phenotype in this patient. LAY SUMMARY: Ribavirin is the most common treatment for chronic hepatitis E and is mostly effective, although some cases of ribavirin treatment failure have been described. Here, we report on a particular case of ribavirin resistance and investigate the underlying causes of treatment failure. Mutations in the viral polymerase, an essential enzyme for viral replication, appear to be responsible.


Asunto(s)
Virus de la Hepatitis E , Antivirales , Farmacorresistencia Viral , Humanos , Mutación , Ribavirina , Insuficiencia del Tratamiento , Replicación Viral
16.
Rev Med Virol ; 25(4): 205-23, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25828437

RESUMEN

Dengue virus and other flaviviruses such as the yellow fever, West Nile, and Japanese encephalitis viruses are emerging vector-borne human pathogens that affect annually more than 100 million individuals and that may cause debilitating and potentially fatal hemorrhagic and encephalitic diseases. Currently, there are no specific antiviral drugs for the treatment of flavivirus-associated disease. A better understanding of the flavivirus-host interactions during the different events of the flaviviral life cycle may be essential when developing novel antiviral strategies. The flaviviral non-structural protein 4b (NS4b) appears to play an important role in flaviviral replication by facilitating the formation of the viral replication complexes and in counteracting innate immune responses such as the following: (i) type I IFN signaling; (ii) RNA interference; (iii) formation of stress granules; and (iv) the unfolded protein response. Intriguingly, NS4b has recently been shown to constitute an excellent target for the selective inhibition of flavivirus replication. We here review the current knowledge on NS4b.


Asunto(s)
Antivirales/aislamiento & purificación , Flavivirus/inmunología , Flavivirus/fisiología , Interacciones Huésped-Patógeno , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Humanos
17.
Gastroenterology ; 147(5): 1008-11.e7; quiz e15-6, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25181691

RESUMEN

We analyzed blood samples collected from 15 patients with chronic hepatitis E who were recipients of solid-organ transplants. All patients cleared the hepatitis E virus (HEV) except for 2 (nonresponders); 1 patient died. A G1634R mutation in viral polymerase was detected in the HEV RNA of the nonresponders; this mutation did not provide the virus with resistance to ribavirin in vitro. However, the mutant form of a subgenomic replicon of genotype 3 HEV replicated more efficiently in vitro than HEV without this mutation, and the same was true for infectious virus, including in competition assays. Similar results were obtained for genotype 1 HEV. The G1634R mutation therefore appears to increase the replicative capacity of HEV in the human liver and hence reduce the efficacy of ribavirin.


Asunto(s)
Antivirales/uso terapéutico , ARN Polimerasas Dirigidas por ADN/genética , Virus de la Hepatitis E/efectos de los fármacos , Hepatitis E/tratamiento farmacológico , Hepatitis Crónica/tratamiento farmacológico , Mutación , Trasplante de Órganos/efectos adversos , Ribavirina/uso terapéutico , Replicación Viral/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Farmacorresistencia Viral/genética , Femenino , Genotipo , Células Hep G2 , Hepatitis E/diagnóstico , Hepatitis E/mortalidad , Hepatitis E/virología , Virus de la Hepatitis E/enzimología , Virus de la Hepatitis E/genética , Virus de la Hepatitis E/crecimiento & desarrollo , Hepatitis Crónica/diagnóstico , Hepatitis Crónica/mortalidad , Hepatitis Crónica/virología , Humanos , Masculino , Mutagénesis Sitio-Dirigida , Fenotipo , Factores de Tiempo , Transfección , Insuficiencia del Tratamiento , Replicación Viral/genética
18.
Antimicrob Agents Chemother ; 58(1): 267-73, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24145541

RESUMEN

Hepatitis E virus (HEV) is a common cause of acute hepatitis that results in high mortality in pregnant women and may establish chronic infections in immunocompromised patients. We demonstrate for the first time that alpha interferon (IFN-α) and ribavirin inhibit in vitro HEV replication in both a subgenomic replicon and an infectious culture system based on a genotype 3 strain. IFN-α showed a moderate but significant synergism with ribavirin. These findings corroborate the reported clinical effectiveness of both drugs. In addition, the antiviral activity of ribavirin against wild-type genotype 1, 2, and 3 strains was confirmed by immunofluorescence staining. Furthermore, the in vitro activity of ribavirin depends on depletion of intracellular GTP pools, which is evident from the facts that (i) other GTP-depleting agents (5-ethynyl-1-ß-d-ribofuranosylimidazole-4-carboxamide [EICAR] and mycophenolic acid) inhibit viral replication, (ii) exogenously added guanosine reverses the antiviral effects, and (iii) a strong correlation (R(2) = 0.9998) exists between the antiviral activity and GTP depletion of ribavirin and other GTP-depleting agents.


Asunto(s)
Antivirales/farmacología , Guanosina Trifosfato/metabolismo , Virus de la Hepatitis E/efectos de los fármacos , Virus de la Hepatitis E/metabolismo , Interferón-alfa/farmacología , Ribavirina/farmacología , Replicación Viral/efectos de los fármacos , Línea Celular , Humanos
19.
J Antimicrob Chemother ; 69(10): 2770-84, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24951535

RESUMEN

OBJECTIVES: T-705, also known as favipiravir, is a small-molecule inhibitor that is currently in clinical development for the treatment of influenza virus infections. This molecule also inhibits the replication of a broad spectrum of other RNA viruses. The objective of this study was to investigate the antiviral effect of favipiravir on chikungunya virus (CHIKV) replication and to contribute to unravelling the molecular mechanism of action against this virus. METHODS: The anti-CHIKV effect of favipiravir was examined in cell culture and in a mouse model of lethal infection. A five-step protocol was used to select for CHIKV variants with reduced susceptibility to favipiravir. The resistant phenotype was confirmed in cell culture and the whole genome was sequenced. The identified mutations were reverse-engineered into an infectious clone to confirm their impact on the antiviral efficacy of favipiravir. RESULTS: Favipiravir inhibits the replication of laboratory strains and clinical isolates of CHIKV, as well as of a panel of other alphaviruses. Several favipiravir-resistant CHIKV variants were independently selected and all of them in particular acquired the unique K291R mutation in the RNA-dependent RNA polymerase (RdRp). Reverse-engineering of this K291R mutation into an infectious clone of CHIKV confirmed the link between the mutant genotype and the resistant phenotype. Interestingly, this particular lysine is also highly conserved in the RdRp of positive-stranded RNA viruses in general. CONCLUSIONS: This study provides an important insight into the precise molecular mechanism by which favipiravir exerts its antiviral activity against (alpha)viruses, which may be of help in designing other potent broad-spectrum antivirals.


Asunto(s)
Amidas/farmacología , Antivirales/farmacología , Virus Chikungunya/efectos de los fármacos , Virus Chikungunya/genética , Farmacorresistencia Viral/genética , Mutación , Pirazinas/farmacología , Proteínas no Estructurales Virales/genética , Amidas/química , Animales , Antivirales/química , Línea Celular , Fiebre Chikungunya/tratamiento farmacológico , Fiebre Chikungunya/virología , Efecto Citopatogénico Viral/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ratones , Pruebas de Sensibilidad Microbiana , Fenotipo , Pirazinas/química , Reproducibilidad de los Resultados , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA