Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Stem Cell ; 29(2): 217-231.e8, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35032430

RESUMEN

Kidney failure is frequently observed during and after COVID-19, but it remains elusive whether this is a direct effect of the virus. Here, we report that SARS-CoV-2 directly infects kidney cells and is associated with increased tubule-interstitial kidney fibrosis in patient autopsy samples. To study direct effects of the virus on the kidney independent of systemic effects of COVID-19, we infected human-induced pluripotent stem-cell-derived kidney organoids with SARS-CoV-2. Single-cell RNA sequencing indicated injury and dedifferentiation of infected cells with activation of profibrotic signaling pathways. Importantly, SARS-CoV-2 infection also led to increased collagen 1 protein expression in organoids. A SARS-CoV-2 protease inhibitor was able to ameliorate the infection of kidney cells by SARS-CoV-2. Our results suggest that SARS-CoV-2 can directly infect kidney cells and induce cell injury with subsequent fibrosis. These data could explain both acute kidney injury in COVID-19 patients and the development of chronic kidney disease in long COVID.


Asunto(s)
COVID-19 , SARS-CoV-2 , COVID-19/complicaciones , Fibrosis , Humanos , Riñón , Organoides/patología , Síndrome Post Agudo de COVID-19
2.
Antiviral Res ; 197: 105223, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34856248

RESUMEN

Repurposing drugs is a promising strategy to identify therapeutic interventions against novel and re-emerging viruses. Posaconazole is an antifungal drug used to treat invasive aspergillosis and candidiasis. Recently, posaconazole and its structural analog, itraconazole were shown to inhibit replication of multiple viruses by modifying intracellular cholesterol homeostasis. Here, we show that posaconazole inhibits replication of the alphaviruses Semliki Forest virus (SFV), Sindbis virus and chikungunya virus with EC50 values ranging from 1.4 µM to 9.5 µM. Posaconazole treatment led to a significant reduction of virus entry in an assay using a temperature-sensitive SFV mutant, but time-of-addition and RNA transfection assays indicated that posaconazole also inhibits post-entry stages of the viral replication cycle. Virus replication in the presence of posaconazole was partially rescued by the addition of exogenous cholesterol. A transferrin uptake assay revealed that posaconazole considerably slowed down cellular endocytosis. A single point mutation in the SFV E2 glycoprotein, H255R, provided partial resistance to posaconazole as well as to methyl-ß-cyclodextrin, corroborating the effect of posaconazole on cholesterol and viral entry. Our results indicate that posaconazole inhibits multiple steps of the alphavirus replication cycle and broaden the spectrum of viruses that can be targeted in vitro by posaconazole, which could be further explored as a therapeutic agent against emerging viruses.


Asunto(s)
Alphavirus/efectos de los fármacos , Antivirales/farmacología , Reposicionamiento de Medicamentos/métodos , Triazoles/farmacología , Replicación Viral/efectos de los fármacos , Alphavirus/clasificación , Animales , Línea Celular , Virus Chikungunya/efectos de los fármacos , Chlorocebus aethiops , Cricetinae , Endocitosis/efectos de los fármacos , Humanos , Virus de los Bosques Semliki/efectos de los fármacos , Virus Sindbis/efectos de los fármacos , Células Vero , Internalización del Virus/efectos de los fármacos
3.
Viruses ; 13(2)2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-33670363

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged as a new human pathogen in late 2019 and it has infected over 100 million people in less than a year. There is a clear need for effective antiviral drugs to complement current preventive measures, including vaccines. In this study, we demonstrate that berberine and obatoclax, two broad-spectrum antiviral compounds, are effective against multiple isolates of SARS-CoV-2. Berberine, a plant-derived alkaloid, inhibited SARS-CoV-2 at low micromolar concentrations and obatoclax, which was originally developed as an anti-apoptotic protein antagonist, was effective at sub-micromolar concentrations. Time-of-addition studies indicated that berberine acts on the late stage of the viral life cycle. In agreement, berberine mildly affected viral RNA synthesis, but it strongly reduced infectious viral titers, leading to an increase in the particle-to-pfu ratio. In contrast, obatoclax acted at the early stage of the infection, which is in line with its activity to neutralize the acidic environment in endosomes. We assessed infection of primary human nasal epithelial cells that were cultured on an air-liquid interface and found that SARS-CoV-2 infection induced and repressed expression of specific sets of cytokines and chemokines. Moreover, both obatoclax and berberine inhibited SARS-CoV-2 replication in these primary target cells. We propose berberine and obatoclax as potential antiviral drugs against SARS-CoV-2 that could be considered for further efficacy testing.


Asunto(s)
Antivirales/farmacología , Berberina/farmacología , Indoles/farmacología , Pirroles/farmacología , SARS-CoV-2/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Adolescente , Animales , COVID-19/virología , Células Cultivadas , Chlorocebus aethiops , Células Epiteliales/virología , Humanos , Masculino , ARN Viral/genética , SARS-CoV-2/fisiología , Células Vero
4.
Mol Ecol ; 30(7): 1594-1611, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33432714

RESUMEN

Horizontal gene transfer from viruses to eukaryotic cells is a pervasive phenomenon. Somatic viral integrations are linked to persistent viral infection whereas integrations into germline cells are maintained in host genomes by vertical transmission and may be co-opted for host functions. In the arboviral vector Aedes aegypti, an endogenous viral element from a nonretroviral RNA virus (nrEVE) was shown to produce PIWI-interacting RNAs (piRNAs) to limit infection with a cognate virus. Thus, nrEVEs may constitute a heritable, sequence-specific mechanism for antiviral immunity, analogous to piRNA-mediated silencing of transposable elements. Here, we combine population genomics and evolutionary approaches to analyse the genomic architecture of nrEVEs in A. aegypti. We conducted a genome-wide screen for adaptive nrEVEs and searched for novel population-specific nrEVEs in the genomes of 80 individual wild-caught mosquitoes from five geographical populations. We show a dynamic landscape of nrEVEs in mosquito genomes and identified five novel nrEVEs derived from two currently circulating viruses, providing evidence of the environmental-dependent modification of a piRNA cluster. Overall, our results show that virus endogenization events are complex with only a few nrEVEs contributing to adaptive evolution in A. aegypti.


Asunto(s)
Aedes , Aedes/genética , Animales , Genómica , Metagenómica , Mosquitos Vectores/genética , ARN Interferente Pequeño/genética
5.
Viruses ; 10(3)2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29522475

RESUMEN

The power and ease of Drosophila genetics and the medical relevance of mosquito-transmitted viruses have made dipterans important model organisms in antiviral immunology. Studies of virus-host interactions at the molecular and population levels have illuminated determinants of resistance to virus infection. Here, we review the sources and nature of variation in antiviral immunity and virus susceptibility in model dipteran insects, specifically the fruit fly Drosophila melanogaster and vector mosquitoes of the genera Aedes and Culex. We first discuss antiviral immune mechanisms and describe the virus-specificity of these responses. In the following sections, we review genetic and microbiota-dependent variation in antiviral immunity. In the final sections, we explore less well-studied sources of variation, including abiotic factors, sexual dimorphism, infection history, and endogenous viral elements. We borrow from work on other pathogen types and non-dipteran species when it parallels or complements studies in dipterans. Understanding natural variation in virus-host interactions may lead to the identification of novel restriction factors and immune mechanisms and shed light on the molecular determinants of vector competence.


Asunto(s)
Drosophila melanogaster/virología , Variación Genética , Mosquitos Vectores/virología , Virosis/genética , Virosis/prevención & control , Animales , Drosophila melanogaster/inmunología , Drosophila melanogaster/microbiología , Ambiente , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Mosquitos Vectores/inmunología , Mosquitos Vectores/microbiología , Factores Sexuales
6.
Methods Mol Biol ; 1746: 197-213, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29492897

RESUMEN

Insects are the most abundant and diverse group of animals on earth, but our knowledge of their viruses is biased toward insect-borne viruses that cause disease in plants, animals, or humans. Recent metagenomic studies and systematic surveys of viruses in wild-caught insects have identified an unanticipated large repertoire of novel viruses and viral sequences. These include new members of existing clades, new clades, and even entirely new virus families. These studies greatly expand the known virosphere in insects, provide opportunities to study virus-host interactions, and generate new insights into virus evolution. In this chapter, we discuss the methods used to identify novel viruses in insects and highlight some notable surprises arising from these studies.


Asunto(s)
Genoma Viral , Virus de Insectos/clasificación , Virus de Insectos/genética , Insectos/virología , Metagenómica , Animales , Células Cultivadas , Interacciones Huésped-Patógeno , Filogenia
7.
Artículo en Inglés | MEDLINE | ID: mdl-27993855

RESUMEN

As new pathogenic viruses continue to emerge, it is paramount to have intervention strategies that target a common denominator in these pathogens. The fusion of viral and cellular membranes during viral entry is one such process that is used by many pathogenic viruses, including chikungunya virus, West Nile virus, and influenza virus. Obatoclax, a small-molecule antagonist of the Bcl-2 family of proteins, was previously determined to have activity against influenza A virus and also Sindbis virus. Here, we report it to be active against alphaviruses, like chikungunya virus (50% effective concentration [EC50] = 0.03 µM) and Semliki Forest virus (SFV; EC50 = 0.11 µM). Obatoclax inhibited viral entry processes in an SFV temperature-sensitive mutant entry assay. A neutral red retention assay revealed that obatoclax induces the rapid neutralization of the acidic environment of endolysosomal vesicles and thereby most likely inhibits viral fusion. Characterization of escape mutants revealed that the L369I mutation in the SFV E1 fusion protein was sufficient to confer partial resistance against obatoclax. Other inhibitors that target the Bcl-2 family of antiapoptotic proteins inhibited neither viral entry nor endolysosomal acidification, suggesting that the antiviral mechanism of obatoclax does not depend on its anticancer targets. Obatoclax inhibited the growth of flaviviruses, like Zika virus, West Nile virus, and yellow fever virus, which require low pH for fusion, but not that of pH-independent picornaviruses, like coxsackievirus A9, echovirus 6, and echovirus 7. In conclusion, obatoclax is a novel inhibitor of endosomal acidification that prevents viral fusion and that could be pursued as a potential broad-spectrum antiviral candidate.


Asunto(s)
Antivirales/farmacología , Virus Chikungunya/efectos de los fármacos , Endosomas/efectos de los fármacos , Lisosomas/efectos de los fármacos , Fusión de Membrana/efectos de los fármacos , Pirroles/farmacología , Virus de los Bosques Semliki/efectos de los fármacos , Animales , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/virología , Virus Chikungunya/genética , Virus Chikungunya/crecimiento & desarrollo , Cricetinae , Farmacorresistencia Viral/genética , Endosomas/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Expresión Génica , Hepatocitos/efectos de los fármacos , Hepatocitos/virología , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Indoles , Lisosomas/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Mutación , Rojo Neutro/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/crecimiento & desarrollo , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Virus del Nilo Occidental/efectos de los fármacos , Virus del Nilo Occidental/genética , Virus del Nilo Occidental/crecimiento & desarrollo , Virus de la Fiebre Amarilla/efectos de los fármacos , Virus de la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/crecimiento & desarrollo , Virus Zika/efectos de los fármacos , Virus Zika/genética , Virus Zika/crecimiento & desarrollo
8.
Sci Rep ; 6: 37124, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27845418

RESUMEN

Chikungunya virus (CHIKV), genus Alphavirus, family Togaviridae, has a positive-stand RNA genome approximately 12 kb in length. In infected cells, the genome is translated into non-structural polyprotein P1234, an inactive precursor of the viral replicase, which is activated by cleavages carried out by the non-structural protease, nsP2. We have characterized CHIKV nsP2 using both cell-free and cell-based assays. First, we show that Cys478 residue in the active site of CHIKV nsP2 is indispensable for P1234 processing. Second, the substrate requirements of CHIKV nsP2 are quite similar to those of nsP2 of related Semliki Forest virus (SFV). Third, substitution of Ser482 residue, recently reported to contribute to the protease activity of nsP2, with Ala has almost no negative effect on the protease activity of CHIKV nsP2. Fourth, Cys478 to Ala as well as Trp479 to Ala mutations in nsP2 completely abolished RNA replication in CHIKV and SFV trans-replication systems. In contrast, trans-replicases with Ser482 to Ala mutation were similar to wild type counterparts. Fifth, Cys478 to Ala as well as Trp479 to Ala mutations in nsP2 abolished the rescue of infectious virus from CHIKV RNA transcripts while Ser482 to Ala mutation had no effect. Thus, CHIKV nsP2 is a cysteine protease.


Asunto(s)
Fiebre Chikungunya/metabolismo , Virus Chikungunya/fisiología , Cisteína Endopeptidasas/metabolismo , Poliproteínas/metabolismo , Proteolisis , ARN Viral/metabolismo , Proteínas Virales/metabolismo , Replicación Viral/fisiología , Sustitución de Aminoácidos , Animales , Dominio Catalítico , Línea Celular , Fiebre Chikungunya/genética , Cricetinae , Cisteína Endopeptidasas/genética , Mutación Missense , Poliproteínas/genética , ARN Viral/genética , Proteínas Virales/genética
9.
Antimicrob Agents Chemother ; 60(12): 7382-7395, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27736770

RESUMEN

Chikungunya virus (CHIKV; genus Alphavirus) is the causative agent of chikungunya fever. CHIKV replication can be inhibited by some broad-spectrum antiviral compounds; in contrast, there is very little information about compounds specifically inhibiting the enzymatic activities of CHIKV replication proteins. These proteins are translated in the form of a nonstructural (ns) P1234 polyprotein precursor from the CHIKV positive-strand RNA genome. Active forms of replicase enzymes are generated using the autoproteolytic activity of nsP2. The available three-dimensional (3D) structure of nsP2 protease has made it a target for in silico drug design; however, there is thus far little evidence that the designed compounds indeed inhibit the protease activity of nsP2 and/or suppress CHIKV replication. In this study, a set of 12 compounds, predicted to interact with the active center of nsP2 protease, was designed using target-based modeling. The majority of these compounds were shown to inhibit the ability of nsP2 to process recombinant protein and synthetic peptide substrates. Furthermore, all compounds found to be active in these cell-free assays also suppressed CHIKV replication in cell culture, the 50% effective concentration (EC50) of the most potent inhibitor being ∼1.5 µM. Analysis of stereoisomers of one compound revealed that inhibition of both the nsP2 protease activity and CHIKV replication depended on the conformation of the inhibitor. Combining the data obtained from different assays also indicates that some of the analyzed compounds may suppress CHIKV replication using more than one mechanism.


Asunto(s)
Antivirales/síntesis química , Virus Chikungunya/efectos de los fármacos , Cisteína Endopeptidasas/metabolismo , Genoma Viral , Poliproteínas/antagonistas & inhibidores , Inhibidores de Proteasas/síntesis química , ARN Helicasas/antagonistas & inhibidores , Animales , Ácidos Carboxílicos/síntesis química , Ácidos Carboxílicos/farmacología , Dominio Catalítico , Línea Celular , Virus Chikungunya/enzimología , Virus Chikungunya/genética , Virus Chikungunya/crecimiento & desarrollo , Cricetinae , Cristalografía por Rayos X , Ciclopropanos/síntesis química , Ciclopropanos/farmacología , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/genética , Diseño de Fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Concentración 50 Inhibidora , Simulación del Acoplamiento Molecular , Poliproteínas/química , Poliproteínas/genética , Poliproteínas/metabolismo , Inhibidores de Proteasas/farmacología , ARN Helicasas/química , ARN Helicasas/genética , ARN Helicasas/metabolismo , Estereoisomerismo , Relación Estructura-Actividad , Replicación Viral/efectos de los fármacos
10.
J Virol ; 90(21): 9743-9757, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27535052

RESUMEN

Chikungunya virus (CHIKV) has infected millions of people in the tropical and subtropical regions since its reemergence in the last decade. We recently identified the nontoxic plant alkaloid berberine as an antiviral substance against CHIKV in a high-throughput screen. Here, we show that berberine is effective in multiple cell types against a variety of CHIKV strains, also at a high multiplicity of infection, consolidating the potential of berberine as an antiviral drug. We excluded any effect of this compound on virus entry or on the activity of the viral replicase. A human phosphokinase array revealed that CHIKV infection specifically activated the major mitogen-activated protein kinase (MAPK) signaling pathways extracellular signal-related kinase (ERK), p38 and c-Jun NH2-terminal kinase (JNK). Upon treatment with berberine, this virus-induced MAPK activation was markedly reduced. Subsequent analyses with specific inhibitors of these kinases indicated that the ERK and JNK signaling cascades are important for the generation of progeny virions. In contrast to specific MAPK inhibitors, berberine lowered virus-induced activation of all major MAPK pathways and resulted in a stronger reduction in viral titers. Further, we assessed the in vivo efficacy of berberine in a mouse model and measured a significant reduction of CHIKV-induced inflammatory disease. In summary, we demonstrate the efficacy of berberine as a drug against CHIKV and highlight the importance of the MAPK signaling pathways in the alphavirus infectious cycle. IMPORTANCE: Chikungunya virus (CHIKV) is a mosquito-borne virus that causes severe and persistent muscle and joint pain and has recently spread to the Americas. No licensed drug exists to counter this virus. In this study, we report that the alkaloid berberine is antiviral against different CHIKV strains and in multiple human cell lines. We demonstrate that berberine collectively reduced the virus-induced activation of cellular mitogen-activated protein kinase signaling. The relevance of these signaling cascades in the viral life cycle was emphasized by specific inhibitors of these kinase pathways, which decreased the production of progeny virions. Berberine significantly reduced CHIKV-induced inflammatory disease in a mouse model, demonstrating efficacy of the drug in vivo Overall, this work makes a strong case for pursuing berberine as a potential anti-CHIKV therapeutic compound and for exploring the MAPK signaling pathways as antiviral targets against alphavirus infections.


Asunto(s)
Alcaloides/farmacología , Antivirales/farmacología , Berberina/farmacología , Fiebre Chikungunya/tratamiento farmacológico , Virus Chikungunya/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular , Fiebre Chikungunya/metabolismo , Cricetinae , Células HEK293 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Células Vero , Activación Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
11.
Antiviral Res ; 126: 117-24, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26752081

RESUMEN

Chikungunya virus (CHIKV) is an arthritogenic arbovirus of the Alphavirus genus, which has infected millions of people after its re-emergence in the last decade. In this study, a BHK cell line containing a stable CHIKV replicon with a luciferase reporter was used in a high-throughput platform to screen approximately 3000 compounds. Following initial validation, 25 compounds were chosen as primary hits for secondary validation with wild type and reporter CHIKV infection, which identified three promising compounds. Abamectin (EC50 = 1.5 µM) and ivermectin (EC50 = 0.6 µM) are fermentation products generated by a soil dwelling actinomycete, Streptomyces avermitilis, whereas berberine (EC50 = 1.8 µM) is a plant-derived isoquinoline alkaloid. They inhibited CHIKV replication in a dose-dependent manner and had broad antiviral activity against other alphaviruses--Semliki Forest virus and Sindbis virus. Abamectin and ivermectin were also active against yellow fever virus, a flavivirus. These compounds caused reduced synthesis of CHIKV genomic and antigenomic viral RNA as well as downregulation of viral protein expression. Time of addition experiments also suggested that they act on the replication phase of the viral infectious cycle.


Asunto(s)
Alphavirus/efectos de los fármacos , Antivirales/farmacología , Fiebre Chikungunya/tratamiento farmacológico , Virus Chikungunya/efectos de los fármacos , Alphavirus/fisiología , Animales , Berberina/farmacología , Línea Celular , Línea Celular Tumoral , Fiebre Chikungunya/virología , Virus Chikungunya/fisiología , Cricetinae , ADN Viral/antagonistas & inhibidores , ADN Viral/biosíntesis , ADN Viral/efectos de los fármacos , Flavivirus/efectos de los fármacos , Humanos , Ivermectina/análogos & derivados , Ivermectina/farmacología , ARN Viral/antagonistas & inhibidores , ARN Viral/biosíntesis , ARN Viral/efectos de los fármacos , ARN Viral/genética , Replicón/efectos de los fármacos , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/biosíntesis , Proteínas Virales/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Virus de la Fiebre Amarilla/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA