Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Intervalo de año de publicación
1.
mBio ; 15(3): e0228723, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38349185

RESUMEN

Since the beginning of the coronavirus disease 2019 (COVID-19) pandemic, much effort has been dedicated to identifying effective antivirals against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A number of calpain inhibitors show excellent antiviral activities against SARS-CoV-2 by targeting the viral main protease (Mpro), which plays an essential role in processing viral polyproteins. In this study, we found that calpain inhibitors potently inhibited the infection of a chimeric vesicular stomatitis virus (VSV) encoding the SARS-CoV-2 spike protein but not Mpro. In contrast, calpain inhibitors did not exhibit antiviral activities toward the wild-type VSV with its native glycoprotein. Genetic knockout of calpain-2 by CRISPR/Cas9 conferred resistance of the host cells to the chimeric VSV-SARS-CoV-2 virus and a clinical isolate of wild-type SARS-CoV-2. Mechanistically, calpain-2 facilitates SARS-CoV-2 spike protein-mediated cell attachment by positively regulating the cell surface levels of ACE2. These results highlight an Mpro-independent pathway targeted by calpain inhibitors for efficient viral inhibition. We also identify calpain-2 as a novel host factor and a potential therapeutic target responsible for SARS-CoV-2 infection at the entry step. IMPORTANCE: Many efforts in small-molecule screens have been made to counter SARS-CoV-2 infection by targeting the viral main protease, the major element that processes viral proteins after translation. Here, we discovered that calpain inhibitors further block SARS-CoV-2 infection in a main protease-independent manner. We identified the host cysteine protease calpain-2 as an important positive regulator of the cell surface levels of SARS-CoV-2 cellular receptor ACE2 and, thus, a facilitator of viral infection. By either pharmacological inhibition or genetic knockout of calpain-2, the SARS-CoV-2 binding to host cells is blocked and viral infection is decreased. Our findings highlight a novel mechanism of ACE2 regulation, which presents a potential new therapeutic target. Since calpain inhibitors also potently interfere with the viral main protease, our data also provide a mechanistic understanding of the potential use of calpain inhibitors as dual inhibitors (entry and replication) in the clinical setting of COVID-19 diseases. Our findings bring mechanistic insights into the cellular process of SARS-CoV-2 entry and offer a novel explanation to the mechanism of activities of calpain inhibitors.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Calpaína/metabolismo , Calpaína/farmacología , Enzima Convertidora de Angiotensina 2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Antivirales/farmacología , Internalización del Virus
2.
bioRxiv ; 2022 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-36482976

RESUMEN

Since the beginning of the coronavirus disease 2019 (COVID-19) pandemic, much effort has been dedicated to identifying effective antivirals against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A number of calpain inhibitors show excellent antiviral activities against SARS-CoV-2 by targeting the viral main protease (M pro ), which plays an essential role in processing viral polyproteins. In this study, we found that calpain inhibitors potently inhibited the infection of a chimeric vesicular stomatitis virus (VSV) encoding the SARS-CoV-2 spike protein, but not M pro . In contrast, calpain inhibitors did not exhibit antiviral activities towards the wild-type VSV with its native glycoprotein. Genetic knockout of calpain-2 by CRISPR/Cas9 conferred resistance of the host cells to the chimeric VSV-SARS-CoV-2 virus and a clinical isolate of wild-type SARS-CoV-2. Mechanistically, calpain-2 facilitates SARS-CoV-2 spike protein-mediated cell attachment by positively regulating the cell surface levels of ACE2. These results highlight an M pro -independent pathway targeted by calpain inhibitors for efficient viral inhibition. We also identify calpain-2 as a novel host factor and a potential therapeutic target responsible for SARS-CoV-2 infection at the entry step.

3.
Cell Rep ; 36(2): 109364, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34214467

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) variants govern transmissibility, responsiveness to vaccination, and disease severity. In a screen for new models of SARS-CoV-2 infection, we identify human H522 lung adenocarcinoma cells as naturally permissive to SARS-CoV-2 infection despite complete absence of angiotensin-converting enzyme 2 (ACE2) expression. Remarkably, H522 infection requires the E484D S variant; viruses expressing wild-type S are not infectious. Anti-S monoclonal antibodies differentially neutralize SARS-CoV-2 E484D S in H522 cells as compared to ACE2-expressing cells. Sera from vaccinated individuals block this alternative entry mechanism, whereas convalescent sera are less effective. Although the H522 receptor remains unknown, depletion of surface heparan sulfates block H522 infection. Temporally resolved transcriptomic and proteomic profiling reveal alterations in cell cycle and the antiviral host cell response, including MDA5-dependent activation of type I interferon signaling. These findings establish an alternative SARS-CoV-2 host cell receptor for the E484D SARS-CoV-2 variant, which may impact tropism of SARS-CoV-2 and consequently human disease pathogenesis.


Asunto(s)
COVID-19/inmunología , COVID-19/metabolismo , Receptores Virales , Glicoproteína de la Espiga del Coronavirus/metabolismo , Sustitución de Aminoácidos , Enzima Convertidora de Angiotensina 2 , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Ciclo Celular , Línea Celular Tumoral , Chlorocebus aethiops , Perfilación de la Expresión Génica , Heparitina Sulfato/metabolismo , Humanos , Interferón Tipo I/metabolismo , Helicasa Inducida por Interferón IFIH1/metabolismo , Modelos Biológicos , Unión Proteica , Dominios Proteicos , Proteómica , Receptores Virales/metabolismo , SARS-CoV-2 , Serina Endopeptidasas/metabolismo , Transducción de Señal , Glicoproteína de la Espiga del Coronavirus/genética , Células Vero , Internalización del Virus , Replicación Viral
4.
bioRxiv ; 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33688646

RESUMEN

Established in vitro models for SARS-CoV-2 infection are limited and include cell lines of non-human origin and those engineered to overexpress ACE2, the cognate host cell receptor. We identified human H522 lung adenocarcinoma cells as naturally permissive to SARS-CoV-2 infection despite complete absence of ACE2. Infection of H522 cells required the SARS-CoV-2 spike protein, though in contrast to ACE2-dependent models, spike alone was not sufficient for H522 infection. Temporally resolved transcriptomic and proteomic profiling revealed alterations in cell cycle and the antiviral host cell response, including MDA5-dependent activation of type-I interferon signaling. Focused chemical screens point to important roles for clathrin-mediated endocytosis and endosomal cathepsins in SARS-CoV-2 infection of H522 cells. These findings imply the utilization of an alternative SARS-CoV-2 host cell receptor which may impact tropism of SARS-CoV-2 and consequently human disease pathogenesis.

5.
Viruses ; 11(7)2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31319455

RESUMEN

Moloney leukemia virus 10 (MOV10) is an RNA helicase that has been shown to affect the replication of several viruses. The effect of MOV10 on Hepatitis B virus (HBV) infection is not known and its role on the replication of this virus is poorly understood. We investigated the effect of MOV10 down-regulation and MOV10 over-expression on HBV in a variety of cell lines, as well as in an infection system using a replication competent virus. We report that MOV10 down-regulation, using siRNA, shRNA, and CRISPR/Cas9 gene editing technology, resulted in increased levels of HBV DNA, HBV pre-genomic RNA, and HBV core protein. In contrast, MOV10 over-expression reduced HBV DNA, HBV pre-genomic RNA, and HBV core protein. These effects were consistent in all tested cell lines, providing strong evidence for the involvement of MOV10 in the HBV life cycle. We demonstrated that MOV10 does not interact with HBV-core. However, MOV10 binds HBV pgRNA and this interaction does not affect HBV pgRNA decay rate. We conclude that the restriction of HBV by MOV10 is mediated through effects at the level of viral RNA.


Asunto(s)
Virus de la Hepatitis B/fisiología , Hepatitis B/virología , Interacciones Huésped-Patógeno , Interacciones Microbianas , Virus de la Leucemia Murina de Moloney/fisiología , Replicación Viral , Animales , Línea Celular , Células Cultivadas , Regulación Viral de la Expresión Génica , Humanos , Ratones , Unión Proteica , ARN , ARN Helicasas/metabolismo , ARN Viral , Proteínas Virales/metabolismo
6.
ACS Infect Dis ; 5(5): 750-758, 2019 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-30582687

RESUMEN

An estimated 240 million are chronically infected with hepatitis B virus (HBV), which can lead to liver disease, cirrhosis, and hepatocellular carcinoma. Currently, HBV treatment options include only nucleoside reverse transcriptase inhibitors and the immunomodulatory agent interferon alpha, and these treatments are generally not curative. New treatments with novel mechanisms of action, therefore, are highly desired for HBV therapy. The viral core protein (Cp) has gained attention as a possible therapeutic target because of its vital roles in the HBV life cycle. Several classes of capsid assembly effectors (CAEs) have been described in detail, and these compounds all increase capsid assembly rate but inhibit HBV replication by different mechanisms. In this study, we have developed a thermal shift-based screening method for CAE discovery and characterization, filling a much-needed gap in high-throughput screening methods for capsid-targeting molecules. Using this approach followed by cell-based screening, we identified the compound HF9C6 as a CAE with low micromolar potency against HBV replication. HF9C6 caused large multicapsid aggregates when capsids were assembled in vitro and analyzed by transmission electron microscopy. Interestingly, when HBV-expressing cells were treated with HF9C6, Cp was excluded from cell nuclei, suggesting that this compound may inhibit nuclear entry of Cp and capsids. Furthermore, mutational scanning of Cp suggested that HF9C6 binds the known CAE binding pocket, indicating that key Cp-compound interactions within this pocket have a role in determining the CAE mechanism of action.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Virus de la Hepatitis B/efectos de los fármacos , Proteínas del Núcleo Viral/antagonistas & inhibidores , Internalización del Virus/efectos de los fármacos , Células Hep G2 , Virus de la Hepatitis B/fisiología , Hepatocitos/efectos de los fármacos , Hepatocitos/virología , Humanos , Ensamble de Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
7.
Viruses ; 10(10)2018 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-30274333

RESUMEN

We have recently developed the first microscopy-based strategy that enables simultaneous multiplex detection of viral RNA (vRNA), viral DNA (vDNA), and viral protein. Here, we used this approach to study the kinetics of latency reactivation in cells infected with the human immunodeficiency virus (HIV). We showed the transcription of nascent vRNA from individual latently integrated and reactivated vDNA sites appearing earlier than viral protein. We further demonstrated that this method can be used to quantitatively assess the efficacy of a variety of latency reactivating agents. Finally, this microscopy-based strategy was augmented with a flow-cytometry-based approach, enabling the detection of transcriptional reactivation of large numbers of latently infected cells. Hence, these approaches are shown to be suitable for qualitative and quantitative studies of HIV-1 latency and reactivation.


Asunto(s)
Linfocitos T CD4-Positivos/virología , ADN Viral/análisis , Infecciones por VIH/virología , VIH-1/fisiología , ARN Viral/análisis , Latencia del Virus/inmunología , Anticuerpos Monoclonales/inmunología , Linfocitos T CD4-Positivos/inmunología , Línea Celular Tumoral , ADN Viral/inmunología , Productos del Gen gag/inmunología , VIH-1/inmunología , Humanos , Cinética , ARN Viral/inmunología , Activación Viral/inmunología
8.
mSphere ; 3(2)2018 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-29669885

RESUMEN

Heteroaryldihydropyrimidines (HAPs) are compounds that inhibit hepatitis B virus (HBV) replication by modulating viral capsid assembly. While their biophysical effects on capsid assembly in vitro have been previously studied, the effect of HAP treatment on capsid protein (Cp) in individual HBV-infected cells remains unknown. We report here that the HAP Bay 38-7690 promotes aggregation of recombinant Cp in vitro and causes a time- and dose-dependent decrease of Cp in infected cells, consistent with previously studied HAPs. Interestingly, immunofluorescence analysis showed Cp aggregating in nuclear foci of Bay 38-7690-treated infected cells in a time- and dose-dependent manner. We found these foci to be associated with promyelocytic leukemia (PML) nuclear bodies (NBs), which are structures that affect many cellular functions, including DNA damage response, transcription, apoptosis, and antiviral responses. Cp aggregation is not an artifact of the cell system used, as it is observed in HBV-expressing HepAD38 cells, in HepG2 cells transfected with an HBV-expressing plasmid, and in HepG2-NTCP cells infected with HBV. Use of a Cp overexpression vector without HBV sequences shows that aggregation is independent of viral replication, and use of an HBV-expressing plasmid harboring a HAP resistance mutation in Cp abrogated the aggregation, demonstrating that the effect is due to direct compound-Cp interactions. These studies provide novel insight into the effects of HAP-based treatment at a single-cell level.IMPORTANCE Despite the availability of effective vaccines and treatments, HBV remains a significant global health concern, with more than 240 million individuals chronically infected. Current treatments are highly effective at controlling viral replication and disease progression but rarely cure infections. Therefore, much emphasis is being placed on finding therapeutics with new drug targets, such as viral gene expression, covalently closed circular DNA formation and stability, capsid formation, and host immune modulators, with the ultimate goal of an HBV cure. Understanding the mechanisms by which novel antiviral agents act will be imperative for the development of curative HBV therapies.


Asunto(s)
Antivirales/farmacología , Proteínas de la Cápside/química , Virus de la Hepatitis B/efectos de los fármacos , Cuerpos de Inclusión Viral/química , Agregado de Proteínas/efectos de los fármacos , Piridinas/farmacología , Pirimidinas/farmacología , Cápside/química , Cápside/efectos de los fármacos , Proteínas de la Cápside/genética , Técnica del Anticuerpo Fluorescente , Células Hep G2 , Hepatitis B/tratamiento farmacológico , Virus de la Hepatitis B/fisiología , Humanos , Proteínas Recombinantes/química , Ensamble de Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
9.
Rev. peru. med. exp. salud publica ; 30(2): 241-245, abr.-jun. 2013. ilus, graf, tab
Artículo en Español | LILACS, LIPECS | ID: lil-680989

RESUMEN

Con el objetivo de detectar y caracterizar molecularmente las metalo-ß-lactamasas (MßL) en aislamientos clínicos de Pseudomonas aeruginosa, se realizó un estudio trasversal en seis hospitales de referencia de Lima (Perú) en agosto de 2011. Se evaluó 51 aislamientos de P. aeruginosa, resistentes a ceftazidima y con sensibilidad reducida a carbapenémicos. El ensayo fenotípico se realizó con el método de aproximación de discos con sustratos (ceftazidima, imipenem y meropenem) y con ácido etilendiaminotetraacético (EDTA). La detección de genes MßL se realizó mediante la técnica de reacción en cadena de polimerasa multiplex. A través del método fenotípico se detectaron MßL en el 15,7% de los aislamientos, en todos ellos la detección de genes mostró la presencia del gen blaIMP. La descripción del primer reporte de MßL en aislamientos de P. aeruginosa en el Perú debería alertar a los equipos de vigilancia epidemiológica intrahospitalaria para promover su control y prevenir su diseminación.


The aim of this study was to detect and characterize molecularly metallo-ß-lactamase (MßL) in clinical isolates of Pseudomonas aeruginosa. We carry out a cross sectional study in six publics hospital in Lima on August 2011. 51 isolates of P. aeruginosa resistant to ceftazidime and reduced susceptibility to carbapenemes were evaluated.The phenotypic assay was performed using the approximation method with substrate disks (ceftazidime, imipenem and meropenem) and ethylenediaminetetraacetic acid (EDTA). MßL gene detection was performed using the technique of polymerase chain reaction (PCR) multiplex. Through MßL detected phenotypic method in 15.7% of isolates. Detection of genes revealed the presence of the gene in the 8 isolates blaIMP. The first report of MßL in P. aeruginosa in Peru was described, this should alert the monitoring equipment in the institutions to promote control their spread.


Asunto(s)
Humanos , Pseudomonas aeruginosa/enzimología , beta-Lactamasas/aislamiento & purificación , Antibacterianos/farmacología , Estudios Transversales , Pruebas de Sensibilidad Microbiana , Perú , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA