Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Microbiol ; 13: 943877, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36532428

RESUMEN

COVID-19 has a wide range of clinical presentations, and the susceptibility to SARS-CoV-2 infection and the mortality rate also vary by region and ethnicity. Here, we found that rs12329760 in the TMPRSS2 gene, a missense variant common in East Asian populations, contributes to protection against SARS-CoV-2 infection. TMPRSS2 is a protease responsible for SARS-CoV-2 entry and syncytium formation. rs12329760 (c.478G>A, p. V160M) was associated with a reduced risk of moderate symptoms. The enzymatic activity of Met160-TMPRSS2 was lower than that of Val160-TMPRSS2, and thus the viral entry and the syncytium formation of SARS-CoV-2 were impaired. Collectively, these results indicate that the genetic variation in TMPRSS2, which is common in East Asians, is one of the molecular determinants of COVID-19 susceptibility.

2.
J Med Chem ; 65(4): 2926-2939, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-34313428

RESUMEN

The novel coronavirus, SARS-CoV-2, has been identified as the causative agent for the current coronavirus disease (COVID-19) pandemic. 3CL protease (3CLpro) plays a pivotal role in the processing of viral polyproteins. We report peptidomimetic compounds with a unique benzothiazolyl ketone as a warhead group, which display potent activity against SARS-CoV-2 3CLpro. The most potent inhibitor YH-53 can strongly block the SARS-CoV-2 replication. X-ray structural analysis revealed that YH-53 establishes multiple hydrogen bond interactions with backbone amino acids and a covalent bond with the active site of 3CLpro. Further results from computational and experimental studies, including an in vitro absorption, distribution, metabolism, and excretion profile, in vivo pharmacokinetics, and metabolic analysis of YH-53 suggest that it has a high potential as a lead candidate to compete with COVID-19.


Asunto(s)
Antivirales/farmacología , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/farmacología , Cetonas/farmacología , Peptidomiméticos/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Antivirales/síntesis química , Antivirales/química , COVID-19/metabolismo , Chlorocebus aethiops , Proteasas 3C de Coronavirus/aislamiento & purificación , Proteasas 3C de Coronavirus/metabolismo , Inhibidores de Cisteína Proteinasa/síntesis química , Inhibidores de Cisteína Proteinasa/química , Humanos , Cetonas/química , Masculino , Pruebas de Sensibilidad Microbiana , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Modelos Moleculares , Conformación Molecular , Peptidomiméticos/síntesis química , Peptidomiméticos/química , Ratas , Ratas Wistar , SARS-CoV-2/enzimología , Células Vero , Tratamiento Farmacológico de COVID-19
3.
J Biol Chem ; 296: 100346, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33524396

RESUMEN

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has developed into a global pandemic since its first outbreak in the winter of 2019. An extensive investigation of SARS-CoV-2 is critical for disease control. Various recombinant monoclonal antibodies of human origin that neutralize SARS-CoV-2 infection have been isolated from convalescent patients and will be applied as therapies and prophylaxis. However, the need for dedicated monoclonal antibodies suitable for molecular pathology research is not fully addressed. Here, we produced six mouse anti-SARS-CoV-2 spike monoclonal antibodies that not only exhibit robust performance in immunoassays including western blotting, ELISA, immunofluorescence, and immunoprecipitation, but also demonstrate neutralizing activity against SARS-CoV-2 infection to VeroE6/TMPRSS2 cells. Due to their mouse origin, our monoclonal antibodies are compatible with the experimental immunoassay setups commonly used in basic molecular biology research laboratories, providing a useful tool for future research. Furthermore, in the hope of applying the antibodies of clinical setting, we determined the variable regions of the antibodies and used them to produce recombinant human/mouse chimeric antibodies.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , COVID-19/prevención & control , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/aislamiento & purificación , Anticuerpos Antivirales/química , Anticuerpos Antivirales/aislamiento & purificación , Sitios de Unión , COVID-19/inmunología , COVID-19/virología , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Ratones , Pruebas de Neutralización , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Subunidades de Proteína/administración & dosificación , Subunidades de Proteína/genética , Subunidades de Proteína/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/administración & dosificación , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunación
4.
Sci Immunol ; 4(40)2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31653718

RESUMEN

The respiratory epithelium is exposed to the environment and initiates inflammatory responses to exclude pathogens. Influenza A virus (IAV) infection triggers inflammatory responses in the respiratory mucosa, but the mechanisms of inflammasome activation are poorly understood. We identified MxA as a functional inflammasome sensor in respiratory epithelial cells that recognizes IAV nucleoprotein and triggers the formation of ASC (apoptosis-associated speck-like protein containing a CARD) specks via interaction of its GTPase domain with the PYD domain of ASC. ASC specks were present in bronchiolar epithelial cells of IAV-infected MxA-transgenic mice, which correlated with early IL-1ß production and early recruitment of granulocytes in the lungs of infected mice. Collectively, these results demonstrate that MxA contributes to IAV resistance by triggering a rapid inflammatory response in infected respiratory epithelial cells.


Asunto(s)
Inflamasomas/inmunología , Gripe Humana/inmunología , Proteínas de Resistencia a Mixovirus/inmunología , Mucosa Respiratoria/inmunología , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mucosa Respiratoria/virología
5.
Front Microbiol ; 9: 1164, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29910790

RESUMEN

Lentinula edodes mycelia (LEM) solid culture extracts contain many bioactive compounds with diverse pharmacological activities such as antitumor, antiviral, and immunopotentiating effects. In this study, we examined the anti-influenza virus activity of LEM in vitro and in vivo. LEM directly inhibited influenza virus growth in vitro at early phases of infection, possibly at the entry process of viral particles to host cells. We also found that the nasal administration of LEM increased the survival rate of infected mice, and this was likely due to the direct action of LEM on the viral growth. The oral administration of LEM showed prolonged median survival time of infected mice. Histological analysis revealed that the moderate bronchiolitis was observed in infected mice by the oral administration with LEM, and the extent of alveolitis was dramatically reduced. The orally LEM-administered mice showed a rapid activation of IFN-ß gene expression upon influenza virus infection. These results suggest that the immunopotentiation activity of LEM on type I IFN pathway represses the virus spread to distal alveolar regions from peribronchiolar regions which are primary infection sites in the mouse model. We propose that LEM has anti-influenza virus activities through the direct action on viral growth and stimulatory activity of innate immunity.

6.
J Virol ; 92(14)2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29743359

RESUMEN

Respiratory epithelial cell death by influenza virus infection is responsible for the induction of inflammatory responses, but the exact cell death mechanism is not understood. Here we showed that influenza virus infection induces apoptosis and pyroptosis in normal or precancerous human bronchial epithelial cells. Apoptosis was induced only in malignant tumor cells infected with influenza virus. In human precancerous respiratory epithelial cells (PL16T), the number of apoptotic cells increased at early phases of infection, but pyroptotic cells were observed at late phases of infection. These findings suggest that apoptosis is induced at early phases of infection but the cell death pathway is shifted to pyroptosis at late phases of infection. We also found that the type I interferon (IFN)-mediated JAK-STAT signaling pathway promotes the switch from apoptosis to pyroptosis by inhibiting apoptosis possibly through the induced expression of the Bcl-xL anti-apoptotic gene. Further, the inhibition of JAK-STAT signaling repressed pyroptosis but enhanced apoptosis in infected PL16T cells. Collectively, we propose that type I IFN signaling pathway triggers pyroptosis but not apoptosis in the respiratory epithelial cells in a mutually exclusive manner to initiate proinflammatory responses against influenza virus infection.IMPORTANCE Respiratory epithelium functions as a sensor of infectious agents to initiate inflammatory responses along with cell death. However, the exact cell death mechanism responsible for inflammatory responses by influenza virus infection is still unclear. We showed that influenza virus infection induced apoptosis and pyroptosis in normal or precancerous human bronchial epithelial cells. Apoptosis was induced at early phases of infection, but the cell death pathway was shifted to pyroptosis at late phases of infection under the regulation of type I IFN signaling to promote proinflammatory cytokine production. Taken together, our results indicate that the type I IFN signaling pathway plays an important role to induce pyroptosis but represses apoptosis in the respiratory epithelial cells to initiate proinflammatory responses against influenza virus infection.


Asunto(s)
Apoptosis , Gripe Humana/patología , Interferón Tipo I/metabolismo , Páncreas/patología , Lesiones Precancerosas/patología , Piroptosis , Mucosa Respiratoria/patología , Proteínas Reguladoras de la Apoptosis/metabolismo , Células Cultivadas , Humanos , Virus de la Influenza A/patogenicidad , Gripe Humana/metabolismo , Gripe Humana/virología , Páncreas/metabolismo , Páncreas/virología , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/virología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/virología , Transducción de Señal
8.
Bioorg Med Chem Lett ; 26(4): 1218-23, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26810265

RESUMEN

Sumoylation involves the enzymatic conjugation of small ubiquitin-like modifier (SUMO) protein to their substrate proteins. Sumoylation is not only crucial for maintaining normal cellular physiology but also implicated in the development of several diseases including cancer. SUMO E1, the first protein in sumoylation pathway is of particular significance due to its confirmed role in tumorogenesis. However, notwithstanding its role as potential drug target, only a few small molecule inhibitors of SUMO E1 have been identified. Here, we report the identification of pyrazole and thiazole urea containing compounds as inhibitors of SUMO E1. We have utilized 3D-shape matching, electrostatic potential similarity evaluations and molecular docking to scaffold hop from previously known aryl urea scaffold with SUMO E1 activity to thiazole and pyrazole urea based scaffolds. These two classes of compounds were found to have moderate SUMO E1 inhibitory activity and can be used as starting points for the development of highly potent lead compounds against cancer.


Asunto(s)
Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/antagonistas & inhibidores , Urea/análogos & derivados , Sitios de Unión , Humanos , Concentración 50 Inhibidora , Conformación Molecular , Simulación del Acoplamiento Molecular , Estructura Terciaria de Proteína , Pirazoles/química , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Relación Estructura-Actividad , Tiazoles/química , Urea/metabolismo
9.
PLoS Pathog ; 11(11): e1005284, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26575487

RESUMEN

Influenza virus particles are assembled at the plasma membrane in concert with incorporation of the virus genome, but the details of its spatio-temporal regulation are not understood. Here we showed that influenza virus infection induces the assembly of pericentrosomal endocytic recycling compartment (ERC) through the activation of Rab11a GTPase and cell cycle-independent maturation of centrosome by YB-1, a multifunctional protein that is involved in mitotic division, RNA metabolism and tumorigenesis. YB-1 is recruited to the centrosome in infected cells and is required for anchoring microtubules to the centrosome. We also found that viral infection accumulates cholesterol in ERC and is dependent on YB-1. Depletion of YB-1 shows reduced cholesterol-enriched ERC and prevented budozone formation at the plasma membrane. These results suggest that cholesterol in recycling endosomes, which are emanated from ERC, may trigger the virus assembly concomitantly with the packaging of the virus genome. We propose that the virus genome is transported to the plasma membrane by cholesterol-enriched recycling endosomes through cell cycle-independent activation of the centrosome by YB-1.


Asunto(s)
Centrosoma/metabolismo , Colesterol/metabolismo , Endosomas/metabolismo , Orthomyxoviridae , Transporte Biológico , Ciclo Celular , Membrana Celular/metabolismo , Endocitosis/fisiología , Humanos , Microtúbulos/metabolismo
10.
J Chem Inf Model ; 54(10): 2784-93, 2014 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-25191977

RESUMEN

Sumoylation is a post-translational modification that plays an important role in a wide range of cellular processes. Among the proteins involved in the sumoylation pathway, Ubc9 is the sole E2-conjugating enzyme required for sumoylation and plays a central role by interacting with almost all of the partners required for sumoylation. Ubc9 has been implicated in a variety of human malignancies. In order to exploit the therapeutic potential of Ubc9, we have identified the potential site to target for rational drug design using molecular modeling approaches. The structural information derived was then used to prioritize hits from a small-molecule library for biological assay using a virtual screening protocol that involves shape matching with a known inhibitor inhibitors and docking of a small-molecule library utilizing computational approaches that incorporate both ligand and protein flexibility. Nineteen compounds were acquired from different chemical vendors and were tested for Ubc9 inhibitory activity. Five compounds showed inhibitory activity against Ubc9, out of which one compound was selected for further optimization. A similarity search was then carried out to retrieve commercially available derivatives, which were further acquired and assayed, resulting in two compounds with acceptable potency. These two compounds can be used as starting points for the development of more potent inhibitors of Ubc9 targeting the predicted site.


Asunto(s)
Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Proteínas Activadoras de GTPasa/antagonistas & inhibidores , Procesamiento Proteico-Postraduccional , Bibliotecas de Moléculas Pequeñas/química , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Secuencia de Aminoácidos , Bioensayo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Proteínas Activadoras de GTPasa/química , Proteínas Activadoras de GTPasa/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad , Sumoilación/efectos de los fármacos , Enzimas Ubiquitina-Conjugadoras/química , Enzimas Ubiquitina-Conjugadoras/metabolismo , Interfaz Usuario-Computador
12.
Anal Biochem ; 448: 92-4, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24333278

RESUMEN

SUMOylation is a posttranslational process that attaches a small ubiquitin-like modifier (SUMO) to its target proteins covalently. SUMOylation controls multiple cellular processes through the recognition of SUMO by a SUMO-interacting motif (SIM). In this study, we developed assay systems for detecting noncovalent interactions between SUMO and SIM in cells using split-luciferase complementation. We applied a version of this assay to the detection of in vitro SUMO-SIM interactions using a bacterial expression system. These novel assays enable screening of inhibitors of SUMO-dependent protein-protein interactions, either in vivo or in vitro, in a high-throughput manner.


Asunto(s)
Proteínas Portadoras/metabolismo , Luciferasas/metabolismo , Proteína SUMO-1/metabolismo , Espectrometría de Fluorescencia , Secuencia de Aminoácidos , Proteínas Portadoras/genética , Escherichia coli/metabolismo , Colorantes Fluorescentes/química , Genes Reporteros , Humanos , Luz , Luciferasas/química , Luciferasas/genética , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína SUMO-1/genética
13.
ACS Chem Biol ; 8(12): 2635-42, 2013 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-24143955

RESUMEN

Conjugation of small ubiquitin-like modifier (SUMO) to protein (SUMOylation) regulates multiple biological systems by changing the functions and fates of a large number of proteins. Consequently, abnormalities in SUMOylation have been linked to multiple diseases, including breast cancer. Using an in situ cell-based screening system, we have identified spectomycin B1 and related natural products as novel SUMOylation inhibitors. Unlike known SUMOylation inhibitors such as ginkgolic acid, spectomycin B1 directly binds to E2 (Ubc9) and selectively blocks the formation of the E2-SUMO intermediate; that is, Ubc9 is the direct target of spectomycin B1. Importantly, either spectomycin B1 treatment or Ubc9 knockdown inhibited estrogen-dependent proliferation of MCF7 human breast-cancer cells. Our findings suggest that Ubc9 inhibitors such as spectomycin B1 have potential as therapeutic agents against hormone-dependent breast cancers.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Procesamiento Proteico-Postraduccional , Espectinomicina/farmacología , Enzimas Ubiquitina-Conjugadoras/metabolismo , Línea Celular Tumoral , Femenino , Ensayos Analíticos de Alto Rendimiento , Humanos , Cinética , Unión Proteica , Salicilatos/química , Salicilatos/aislamiento & purificación , Salicilatos/farmacología , Transducción de Señal , Espectinomicina/química , Sumoilación , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Enzimas Ubiquitina-Conjugadoras/genética
14.
Bioorg Med Chem Lett ; 23(18): 5145-9, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23920437

RESUMEN

SUMO activating enzyme 1 (SUMO E1) is the first enzyme in sumoylation pathway and an important cancer drug target. However, only a few inhibitors were reported up to now that includes three natural products, semi-synthetic protein inhibitors and one AMP mimic. Here, we report the identification of quinazolinyloxy biaryl urea as a new class of SUMO E1 inhibitors. The most active compound of this class inhibited the in vitro sumoylation with an IC50 of 13.4 µM. This compound inhibits sumoylation by blocking the formation of SUMOE1-SUMO thioester intermediate. The biological activity of the most active compound is comparable to previously reported inhibitors with properties suitable for medicinal chemistry optimization for potency and druggability.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Quinazolinas/farmacología , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Urea/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estructura Molecular , Quinazolinas/síntesis química , Quinazolinas/química , Relación Estructura-Actividad , Enzimas Activadoras de Ubiquitina/metabolismo , Urea/análogos & derivados , Urea/química
15.
J Chem Inf Model ; 53(4): 809-20, 2013 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-23544417

RESUMEN

SUMO activating enzyme 1 (SUMO E1) is responsible for the activation of SUMO in the first step of the sumoylation cascade. SUMO E1 is linked to many human diseases including cancer, thus making it a potential therapeutic target. There are few reported SUMO E1 inhibitors including several natural products. To identify small molecule inhibitors of SUMO E1 with better drug-like properties for potential therapeutic studies, we have used structure-based virtual screening to identify hits from the Maybridge small molecule library for biological assay. Our virtual screening protocol involves fast docking of the entire small molecule library with rigid protein and ligands followed by redocking of top hits using a method that incorporates both ligand and protein flexibility. Subsequently, the top-ranking compounds were prioritized using the molecular dynamics simulation-based binding free energy calculation. Out of 24 compounds that were acquired and tested using in vitro sumoylation assay, four of them showed more than 85% inhibition of sumoylation with the most active compound showing an IC50 of 14.4 µM. A similarity search with the most active compound in the ZINC database has identified three more compounds with improved potency. These compounds share a common phenyl urea scaffold and have been confirmed to inhibit SUMO E1 by in vitro SUMO-1 thioester bond formation assay. Our study suggests that these phenyl urea compounds could be used as a starting point for the development of novel therapeutic agents.


Asunto(s)
Inhibidores Enzimáticos/química , Compuestos de Fenilurea/química , Bibliotecas de Moléculas Pequeñas/química , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Interfaz Usuario-Computador , Algoritmos , Pruebas de Enzimas , Ensayos Analíticos de Alto Rendimiento , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Relación Estructura-Actividad Cuantitativa , Sumoilación , Enzimas Activadoras de Ubiquitina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA