Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
1.
Viruses ; 13(12)2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34960671

RESUMEN

Rhinoviruses (RV), like many other viruses, modulate programmed cell death to their own advantage. The viral protease, 3C has an integral role in the modulation, and we have shown that RVA-16 3C protease cleaves Receptor-interacting protein kinase-1 (RIPK1), a key host factor that modulates various cell death and cell survival pathways. In the current study, we have investigated whether this cleavage is conserved across selected RV strains. RIPK1 was cleaved in cells infected with strains representing diversity across phylogenetic groups (A and B) and receptor usage (major and minor groups). The cleavage was abrogated in the presence of the specific 3C protease inhibitor, Rupintrivir. Interestingly, there appears to be involvement of another protease (maybe 2A protease) in RIPK1 cleavage in strains belonging to genotype B. Our data show that 3C protease from diverse RV strains cleaves RIPK1, highlighting the importance of the cleavage to the RV lifecycle.


Asunto(s)
Proteasas Virales 3C/metabolismo , Infecciones por Picornaviridae/enzimología , Rhinovirus/enzimología , Proteasas Virales 3C/genética , Antivirales/química , Antivirales/farmacología , Apoptosis/efectos de los fármacos , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Isoxazoles/química , Isoxazoles/farmacología , Fenilalanina/análogos & derivados , Fenilalanina/química , Fenilalanina/farmacología , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/virología , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Pirrolidinonas/química , Pirrolidinonas/farmacología , Rhinovirus/química , Rhinovirus/efectos de los fármacos , Rhinovirus/genética , Valina/análogos & derivados , Valina/química , Valina/farmacología
2.
Virology ; 562: 128-141, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34315103

RESUMEN

Picornavirus family members cause disease in humans. Human rhinoviruses (RV), the main causative agents of the common cold, increase the severity of asthma and COPD; hence, effective agents against RVs are required. The 2A proteinase (2Apro), found in all enteroviruses, represents an attractive target; inactivating mutations in poliovirus 2Apro result in an extension of the VP1 protein preventing infectious virion assembly. Variations in sequence and substrate specificity on eIF4G isoforms between RV 2Apro of genetic groups A and B hinder 2Apro as drug targets. Here, we demonstrate that although RV-A2 and RV-B4 2Apro cleave the substrate GAB1 at different sites, the 2Apro from both groups cleave equally efficiently an artificial site containing P1 methionine. We determined the RV-A2 2Apro structure complexed with zVAM.fmk, containing P1 methionine. Analysis of this first 2Apro-inhibitor complex reveals a conserved hydrophobic P4 pocket among enteroviral 2Apro as a potential target for broad-spectrum anti-enteroviral inhibitors.


Asunto(s)
Antivirales/química , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Rhinovirus/enzimología , Proteínas Virales/química , Proteínas Virales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Cisteína Endopeptidasas/genética , Enterovirus/química , Enterovirus/enzimología , Factor 4G Eucariótico de Iniciación/metabolismo , Variación Genética , Células HeLa , Humanos , Conformación Proteica , Rhinovirus/química , Rhinovirus/genética , Especificidad por Sustrato , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética
3.
Bioorg Med Chem ; 42: 116219, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34077853

RESUMEN

Covalent target modulation with small molecules has been emerging as a promising strategy for drug discovery. However, covalent inhibitory antibody remains unexplored due to the lack of efficient strategies to engineer antibody with desired bioactivity. Herein, we developed an intracellular selection method to generate covalent inhibitory antibody against human rhinovirus 14 (HRV14) 3C protease through unnatural amino acid mutagenesis along the heavy chain complementarity-determining region 3 (CDR-H3). A library of antibody mutants was thus constructed and screened in vivo through co-expression with the target protease. Using this screening strategy, six covalent antibodies with proximity-enabled bioactivity were identified, which were shown to covalently target HRV14-3C protease with high inhibitory potency and exquisite selectivity. Compared to structure-based rational design, this library-based screening method provides a simple and efficient way for the discovery and engineering of covalent antibody for enzyme inhibition.


Asunto(s)
Proteasas Virales 3C/antagonistas & inhibidores , Anticuerpos/farmacología , Regiones Determinantes de Complementariedad/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/farmacología , Rhinovirus/enzimología , Proteasas Virales 3C/metabolismo , Anticuerpos/química , Inhibidores de Cisteína Proteinasa/química , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad
4.
Science ; 370(6521)2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33093214

RESUMEN

Immune sensor proteins are critical to the function of the human innate immune system. The full repertoire of cognate triggers for human immune sensors is not fully understood. Here, we report that human NACHT, LRR, and PYD domains-containing protein 1 (NLRP1) is activated by 3C proteases (3Cpros) of enteroviruses, such as human rhinovirus (HRV). 3Cpros directly cleave human NLRP1 at a single site between Glu130 and Gly131 This cleavage triggers N-glycine-mediated degradation of the autoinhibitory NLRP1 N-terminal fragment via the cullinZER1/ZYG11B complex, which liberates the activating C-terminal fragment. Infection of primary human airway epithelial cells by live human HRV triggers NLRP1-dependent inflammasome activation and interleukin-18 secretion. Our findings establish 3Cpros as a pathogen-derived trigger for the human NLRP1 inflammasome and suggest that NLRP1 may contribute to inflammatory diseases of the airway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Cisteína Endopeptidasas/metabolismo , Inmunidad Innata , Inflamasomas/metabolismo , Mucosa Respiratoria/virología , Rhinovirus/enzimología , Proteínas Virales/metabolismo , Proteasas Virales 3C , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Reguladoras de la Apoptosis/química , Proteínas de Ciclo Celular/metabolismo , Proteínas Cullin/metabolismo , Glutamina/química , Glutamina/metabolismo , Glicina/química , Glicina/metabolismo , Células HEK293 , Células HeLa , Humanos , Interleucina-18/metabolismo , Proteínas NLR , Proteolisis
5.
ACS Chem Biol ; 15(1): 63-73, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31613083

RESUMEN

Human rhinovirus 3C protease (HRV 3C-P) is a high-value commercial cysteine protease that could specifically recognize the short peptide sequence of LEVLFQ↓GP. In here, a strategy based on our previous Yeast Endoplasmic Reticulum Sequestration Screening (YESS) approach was developed in Saccharomyces cerevisiae, a model microorganism, to fully characterize the substrate specificity of a typical human virus protease, HRV 3C-P, in a quantitative and fast manner. Our results demonstrated that HRV 3C-P had very high specificity at P1 and P1' positions, only recognizing Gln/Glu at the P1 position and Gly/Ala/Cys/Ser at the P1' position, respectively. Comparably, it exhibited efficient recognition of most residues at the P2' position, except Trp. Further biochemical characterization through site mutagenesis, enzyme structural modeling, and comparison with other 3C proteases indicated that the S1 pocket of HRV 3C-P was constituted by neutral and basic amino acids, in which His160 and Thr141 specifically interacted with Gln or Glu residues at the substrate P1 position. Additionally, the stringent S1' pocket determined its unique property of only accommodating residues without or with short side chains. Based on our characterization, LEVLFQ↓GM was identified as a more favorable substrate than the original LEVLFQ↓GP at high temperature, which might be caused by the conversion of random coils to ß-turns in HRV 3C-P along with the temperature increase. Our studies prompted a further understanding of the substrate specificity and recognition mechanism of HRV 3C-P. Besides, the YESS-PSSC combined with the enzyme modeling strategy in this study provides a general strategy for deciphering the substrate specificities of proteases.


Asunto(s)
Cisteína Endopeptidasas/química , Péptidos/química , Rhinovirus/enzimología , Proteínas Virales/química , Proteasas Virales 3C , Secuencia de Aminoácidos , Sitios de Unión , Cisteína Endopeptidasas/genética , Regulación de la Expresión Génica , Humanos , Modelos Moleculares , Mutagénesis , Unión Proteica , Conformación Proteica , Saccharomyces cerevisiae/genética , Relación Estructura-Actividad , Especificidad por Sustrato , Temperatura , Termodinámica , Proteínas Virales/genética
6.
Curr Microbiol ; 76(12): 1477-1486, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31612259

RESUMEN

Expression and secretion of recombinant proteins in the endotoxin-free bacterium, Bacillus subtilis, has been thoroughly studied, but overexpression in the cytoplasm has been limited to only a few proteins. Here, we used the robust IPTG-inducible promoter, Pgrac212, to overexpress human rhinovirus 3C protease (HRV3C) in the cytoplasm of B. subtilis cells. A novel solubility tag, the N-terminal domain of the lysS gene of B. subtilis coding for a lysyl-tRNA synthetase was placed at the N terminus with a cleavage site for the endoprotease HRV3C, followed by His-HRV3C or His-GST-HRV3C. The recombinant protease was purified by using a Ni-NTA column. In this study, the His-HRV3C and His-GST-HRV3C proteases were overexpressed in the cytoplasm of B. subtilis at 11% and 16% of the total cellular proteins, respectively. The specific protease activities were 8065 U/mg for His-HRV3C and 3623 U/mg for His-GST-HRV3C. The purified enzymes were used to cleave two different substrates followed by purification of the two different protein targets, the green fluorescent protein and the beta-galactosidase. In conclusion, the combination of an inducible promoter Pgrac212 and a solubility tag allowed the overexpression of the HRV3C protease in the cytoplasm of B. subtilis. The resulting fusion protein was purified using a nickel column and was active in cleaving target proteins to remove the fusion tags. This study offers an effective method for producing recombinant proteins in the cytoplasm of endotoxin-free bacteria.


Asunto(s)
Bacillus subtilis/genética , Cisteína Endopeptidasas/genética , Citoplasma/metabolismo , Microbiología Industrial/métodos , Regiones Promotoras Genéticas/genética , Proteínas Recombinantes de Fusión/genética , Rhinovirus/enzimología , Proteínas Virales/genética , Proteasas Virales 3C , Bacillus subtilis/metabolismo , Clonación Molecular , Cisteína Endopeptidasas/aislamiento & purificación , Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Isopropil Tiogalactósido/farmacología , Lisina-ARNt Ligasa/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Rhinovirus/genética , Solubilidad , Proteínas Virales/aislamiento & purificación , beta-Galactosidasa/genética
7.
Molecules ; 24(10)2019 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-31117169

RESUMEN

The engineering of enzymes for the purpose of controlling their activity represents a valuable approach to address challenges in both fundamental and applied research. Here, we describe and compare different design strategies for the generation of a human rhinovirus-14 (HRV14) 3C protease-inducible caspase-3 (CASP3). We exemplify the application potential of the resulting protease by controlling the activity of a synthetic enzyme cascade, which represents an important motif for the design of artificial signal transduction networks. In addition, we use our engineered CASP3 to characterize the effect of aspartate mutations on enzymatic activity. Besides the identification of mutations that render the enzyme inactive, we find the CASP3-D192E mutant (aspartate-to-glutamate exchange at position 192) to be inaccessible for 3C protease-mediated cleavage. This indicates a structural change of CASP3 that goes beyond a slight misalignment of the catalytic triad. This study could inspire the design of additional engineered proteases that could be used to unravel fundamental research questions or to expand the collection of biological parts for the design of synthetic signaling pathways.


Asunto(s)
Caspasa 3/genética , Cisteína Endopeptidasas/genética , Ingeniería de Proteínas , Rhinovirus/enzimología , Proteínas Virales/genética , Proteasas Virales 3C , Ácido Aspártico/metabolismo , Caspasa 3/química , Caspasa 3/metabolismo , Dominio Catalítico/genética , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Ácido Glutámico/metabolismo , Humanos , Mutación , Proteínas Virales/química , Proteínas Virales/metabolismo
8.
Enzyme Microb Technol ; 123: 15-20, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30686346

RESUMEN

Human rhinovirus 3C protease (HRV 3C protease) is commonly used as a tool to remove fusion tags from recombinant proteins in gene engineering due to its distinguished specificity and high activity at low temperature. This paper is aimed to simplify the strategy of removing epitope tags from target proteins with HRV 3C protease. Fusion proteins composed of a xylanase from Bacillus sp. HBP8 (xylHB) and double tags (MBP/Nus and 6×His, with an HRV 3C protease recognition site between them) were applied as substrates. To perform the cleavage and purification, strains expressing HRV 3C protease and the substrates were mixed before (co-fermentation method) or after (post-fermentation method) inducing with IPTG, followed by cell disruption and incubation at 4℃, overnight for cleavage. The soluble cytoplasmic fraction was added to Ni-NTA resin to recover the cleaved target protein. Because the process was carried out in the cell lysate, it was named as cell lysate purification system based on HRV 3C protease (CLP3C). Our data indicated small number of cells expressing HRV 3C protease was enough to remove the fusion tags efficiently with both co-fermentation and post-fermentation methods. More importantly, the tags were cleaved precisely with no obvious non-specific degradation to the target protein. Hence, active xylanase was recovered easily with this strategy.


Asunto(s)
Bacillus/enzimología , Cisteína Endopeptidasas/metabolismo , Endo-1,4-beta Xilanasas/aislamiento & purificación , Proteínas Recombinantes de Fusión/aislamiento & purificación , Rhinovirus/enzimología , Proteínas Virales/metabolismo , Proteasas Virales 3C , Bacillus/genética , Endo-1,4-beta Xilanasas/genética , Endo-1,4-beta Xilanasas/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Humanos , Proteínas de Unión a Maltosa/genética , Proteínas de Unión a Maltosa/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo
9.
Bioorg Med Chem Lett ; 28(14): 2533-2538, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29866517

RESUMEN

Picornaviruses are non-enveloped viruses that represent a large family of positive-sense single-stranded RNA viruses including a number of causative agents of many human and animal diseases such as coxsackievirus B3 (CVB3) and rhinoviruses (HRV). In this study, we performed a high-throughput screening of a compound library composed of ∼6000 small molecules in search of potential picornavirus 3C protease (3Cpro) inhibitors. As results, we identified quinone analogues that effectively inhibited both CVB3 3Cpro and HRV 3Cpro with IC50 values in low micromolar range. Together with predicted binding modes of these compounds to the active site of the viral protease, it is implied that structural features of these non-peptidic inhibitors may act as useful scaffold for further anti-picornavirus drug design and development.


Asunto(s)
Antivirales/farmacología , Benzoquinonas/farmacología , Inhibidores de Proteasas/farmacología , Rhinovirus/efectos de los fármacos , Proteínas Virales/antagonistas & inhibidores , Proteasas Virales 3C , Antivirales/síntesis química , Antivirales/química , Benzoquinonas/síntesis química , Benzoquinonas/química , Cisteína Endopeptidasas/metabolismo , Relación Dosis-Respuesta a Droga , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química , Rhinovirus/enzimología , Relación Estructura-Actividad , Proteínas Virales/metabolismo
10.
Nat Chem ; 10(6): 599-606, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29760414

RESUMEN

Rhinoviruses (RVs) are the pathogens most often responsible for the common cold, and are a frequent cause of exacerbations in asthma, chronic obstructive pulmonary disease and cystic fibrosis. Here we report the discovery of IMP-1088, a picomolar dual inhibitor of the human N-myristoyltransferases NMT1 and NMT2, and use it to demonstrate that pharmacological inhibition of host-cell N-myristoylation rapidly and completely prevents rhinoviral replication without inducing cytotoxicity. The identification of cooperative binding between weak-binding fragments led to rapid inhibitor optimization through fragment reconstruction, structure-guided fragment linking and conformational control over linker geometry. We show that inhibition of the co-translational myristoylation of a specific virus-encoded protein (VP0) by IMP-1088 potently blocks a key step in viral capsid assembly, to deliver a low nanomolar antiviral activity against multiple RV strains, poliovirus and foot and-mouth disease virus, and protection of cells against virus-induced killing, highlighting the potential of host myristoylation as a drug target in picornaviral infections.


Asunto(s)
Aciltransferasas/antagonistas & inhibidores , Antivirales/farmacología , Cápside/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Rhinovirus/efectos de los fármacos , Ensamble de Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Antivirales/química , Inhibidores Enzimáticos/química , Células HeLa , Humanos , Concentración 50 Inhibidora , Estructura Molecular , Rhinovirus/enzimología , Rhinovirus/fisiología
11.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 4): 255-261, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29633974

RESUMEN

The majority of outbreaks of the common cold are caused by rhinoviruses. The 2A protease (2Apro) of human rhinoviruses (HRVs) is known to play important roles in the propagation of the virus and the modulation of host signal pathways to facilitate viral replication. The 2Apro from human rhinovirus C15 (HRV-C15) has been expressed in Escherichia coli and purified by affinity chromatography, ion-exchange chromatography and gel-filtration chromatography. The crystals diffracted to 2.6 Šresolution. The structure was solved by molecular replacement using the structure of 2Apro from coxsackievirus A16 (CVA16) as the search model. The structure contains a conserved His-Asp-Cys catalytic triad and a Zn2+-binding site. Comparison with other 2Apro structures from enteroviruses reveals that the substrate-binding cleft of 2Apro from HRV-C15 exhibits a more open conformation, which presumably favours substrate binding.


Asunto(s)
Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Rhinovirus/enzimología , Proteínas Virales/química , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , Cristalización , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Conformación Proteica , Rhinovirus/aislamiento & purificación , Homología de Secuencia , Especificidad por Sustrato
12.
Bioorg Med Chem Lett ; 28(5): 906-909, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29433930
13.
Cell Death Dis ; 9(3): 272, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29449668

RESUMEN

Apoptosis and programmed necrosis (necroptosis) determine cell fate, and antagonize infection. Execution of these complementary death pathways involves the formation of receptor-interacting protein kinase 1 (RIPK1) containing complexes. RIPK1 binds to adaptor proteins, such as TRIF (Toll-IL-1 receptor-domain-containing-adaptor-inducing interferon-beta factor), FADD (Fas-associated-protein with death domain), NEMO (NF-κB regulatory subunit IKKγ), SQSTM1 (sequestosome 1/p62), or RIPK3 (receptor-interacting protein kinase 3), which are involved in RNA sensing, NF-κB signaling, autophagosome formation, apoptosis, and necroptosis. We report that a range of rhinoviruses impair apoptosis and necroptosis in epithelial cells late in infection. Unlike the double-strand (ds) RNA mimetic poly I:C (polyinosinic:polycytidylic acid), the exposure of dsRNA to toll-like receptor 3 (TLR3) in rhinovirus-infected cells did not lead to apoptosis execution. Accordingly, necroptosis and the production of ROS (reactive oxygen species) were not observed late in infection, when RIPK3 was absent. Instead, a virus-induced alternative necrotic cell death pathway proceeded, which led to membrane rupture, indicated by propidium iodide staining. The impairment of dsRNA-induced apoptosis late in infection was controlled by the viral 3C-protease (3Cpro), which disrupted RIPK1-TRIF/FADD /SQSTM1 immune-complexes. 3Cpro and 3C precursors were found to coimmuno-precipitate with RIPK1, cleaving the RIPK1 death-domain, and generating N-terminal RIPK1 fragments. The depletion of RIPK1 or chemical inhibition of its kinase at the N-terminus did not interfere with virus progeny formation or cell fate. The data show that rhinoviruses suppress apoptosis and necroptosis, and release progeny by an alternative cell death pathway, which is controlled by viral proteases modifying innate immune complexes.


Asunto(s)
Apoptosis , Cisteína Endopeptidasas/metabolismo , Células Epiteliales/virología , Mucosa Nasal/virología , Necroptosis , Rhinovirus/enzimología , Neoplasias del Cuello Uterino/virología , Proteínas Virales/metabolismo , Proteasas Virales 3C , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Complejo Antígeno-Anticuerpo/metabolismo , Células Epiteliales/enzimología , Células Epiteliales/inmunología , Células Epiteliales/ultraestructura , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Femenino , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Mucosa Nasal/enzimología , Mucosa Nasal/inmunología , Mucosa Nasal/ultraestructura , Especies Reactivas de Oxígeno/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Rhinovirus/inmunología , Rhinovirus/patogenicidad , Proteína Sequestosoma-1/metabolismo , Transducción de Señal , Neoplasias del Cuello Uterino/enzimología , Neoplasias del Cuello Uterino/ultraestructura
14.
Sci Rep ; 8(1): 1569, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29371673

RESUMEN

Human Rhinovirus (HRV) is a pathogen of significant medical importance, being a major cause of upper respiratory tract infections (common colds) as well as causing the majority of virus-induced asthma exacerbations. We investigated whether HRV could modulate apoptosis, an innate antiviral response. Apoptotic signals are generated either extrinsically or intrinsically and are propagated via caspase cascades that lead to cell death, reducing viral replication, which relies on cellular machinery. Using HRV16 infected cells, in combination with chemical inducers and inhibitors of extrinsic apoptosis we show that HRV16 3C protease cleaves a key intermediate in extrinsic apoptosis. Receptor-interacting protein kinase-1 (RIPK1), an extrinsic apoptosis adaptor protein, was cleaved by caspase 8, as expected, during chemical induction of apoptosis. RIPK1 was cleaved in HRV infection albeit at a different site. Caspase 8 activation, which is associated with extrinsic apoptosis, was concurrent with HRV 3C protease mediated cleavage of RIPK1, and potentially increased the accessibility of the HRV 3C cleavage site within RIPK1 in-vitro. The caspase 8 mediated RIPK1 cleavage product has a pro-apoptotic function, and further cleavage of this pro-apoptotic cleavage product by HRV 3C may provide a mechanism by which HRV limits apoptosis.


Asunto(s)
Apoptosis , Caspasa 8/metabolismo , Cisteína Endopeptidasas/metabolismo , Interacciones Huésped-Patógeno , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Rhinovirus/enzimología , Proteínas Virales/metabolismo , Proteasas Virales 3C , Células A549 , Humanos , Hidrólisis , Rhinovirus/crecimiento & desarrollo
15.
Mol Biotechnol ; 59(9-10): 407-424, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28801725

RESUMEN

Human rhinovirus (HRV) 3C protease is widely used in recombinant protein production for various applications such as biochemical characterization and structural biology projects to separate recombinant fusion proteins from their affinity tags in order to prevent interference between these tags and the target proteins. Herein, we report the optimization of expression and purification conditions of glutathione S-transferase (GST)-tagged HRV 3C protease by statistically designed experiments. Soluble expression of GST-HRV 3C protease was initially optimized by response surface methodology (RSM), and a 5.5-fold increase in enzyme yield was achieved. Subsequently, we developed a new incomplete factorial (IF) design that examines four variables (bacterial strain, expression temperature, induction time, and inducer concentration) in a single experiment. The new design called Incomplete Factorial-Strain/Temperature/Time/Inducer (IF-STTI) was validated using three GST-tagged proteins. In all cases, IF-STTI resulted in only 10% lower expression yields than those obtained by RSM. Purification of GST-HRV 3C was optimized by an IF design that examines simultaneously the effect of the amount of resin, incubation time of cell lysate with resin, and glycerol and DTT concentration in buffers, and a further 15% increase in protease recovery was achieved. Purified GST-HRV 3C protease was active at both 4 and 25 °C in a variety of buffers.


Asunto(s)
Cisteína Endopeptidasas/genética , Proteínas Recombinantes de Fusión/genética , Rhinovirus/genética , Proteínas Virales/genética , Proteasas Virales 3C , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/aislamiento & purificación , Regulación Viral de la Expresión Génica , Glutatión Transferasa/química , Glutatión Transferasa/genética , Humanos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación , Rhinovirus/enzimología , Proteínas Virales/química , Proteínas Virales/aislamiento & purificación
16.
Bioorg Med Chem Lett ; 27(17): 4061-4065, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28778471

RESUMEN

The human rhinovirus (HRV) is the most significant cause of the common cold all over the world. The maturation and replication of this virus entirely depend on the activity of a virus-encoded 3C protease. Due to the high conservation among different serotypes and the minimal homology existing between 3C protease and known mammalian enzymes, 3C protease has been regarded as an attractive target for the treatment of HRV infections. In this study, we identified a novel (4R,5R)-N4-(2-((3-methoxyphenyl)amino)ethyl)-2,2-dimethyl-N5-(naphthalen-2-yl)-1,3-dioxolane-4,5-dicarboxamide (7a) to be a HRV 3C protease inhibitor via virtual screening. Further research has been focused on the design, synthesis and in vitro biological evaluation of 7a derivatives. The studies revealed that compound 7d has an IC50 value of 2.50±0.7µM against HRV 3C protease, and it thus could serve as a promising compound for the development of novel anti-rhinoviral medicines.


Asunto(s)
Dioxolanos/farmacología , Diseño de Fármacos , Inhibidores de Proteasas/farmacología , Rhinovirus/enzimología , Proteínas Virales/antagonistas & inhibidores , Proteasas Virales 3C , Cisteína Endopeptidasas/metabolismo , Dioxolanos/síntesis química , Dioxolanos/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química , Relación Estructura-Actividad , Proteínas Virales/metabolismo
17.
Virology ; 511: 123-134, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28843814

RESUMEN

In enteroviruses, the inhibition of protein synthesis from capped host cell mRNA is catalyzed by the virally encoded 2A proteinase (2Apro), which cleaves eukaryotic initiation factors (eIF) 4GI and 4GII. Despite much investigation, the exact mechanism of 2Apro cleavage remains however unclear. Here, we identify the domains responsible for the eIF4E/HRV2 2Apro interaction using molecular modelling and describe mutations that impair this interaction and delay in vitro cleavage of eIF4G isoforms. Furthermore, we produced HRV1A viruses bearing the mutation L17R, Y32A or Y86A in the 2Apro sequence. All three viruses showed reduced yield and were appreciably delayed during infection in eIF4GI cleavage. Thus, we propose for genetic group A HRVs that the eIF4E/2Apro interaction is essential for successful viral replication. In contrast, HRV4 2Apro and coxsackievirus B4 2Apro failed to form complexes with eIF4E, suggesting that the mechanism of eIF4G isoform cleavage in these and related viruses is different.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Factor 4G Eucariótico de Iniciación/metabolismo , Interacciones Huésped-Patógeno , Mapeo de Interacción de Proteínas , Rhinovirus/enzimología , Proteínas Virales/metabolismo , Análisis Mutacional de ADN , Genotipo , Humanos , Hidrólisis , Modelos Moleculares , Unión Proteica , Rhinovirus/genética , Rhinovirus/patogenicidad
18.
Electrophoresis ; 38(19): 2530-2535, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28683171

RESUMEN

Herein, a novel assay has been developed for monitoring PreScission protease (His-PSP) mediated enzyme cleavage of ATTO 590 labeled peptide substrate (ATTO-LEV). This novel method is based on combining the use of capillary electrophoresis and fluorescence detection (CE-FL) to dynamically monitor the enzyme cleavage activity. A multivalent peptide substrate was first constructed by immobilizing His-tagged ATTO 590 labeled peptide substrate (ATTO-LEVH6) onto the surface of CdSe/ZnS quantum dots (QDs). Once successfully immobilized, the novel multivalent peptide substrate resulted in the Förster resonance energy transfer (FRET) from QDs to ATTO 590. The ATTO-LEVH6-QD assembly was then incubated with His-PSP to study the proteolytic cleavage of surface bound ATTO-LEVH6 by CE-FL. Our data suggests that PreScission-mediated proteolytic cleavage is enzyme concentration- and incubation time-dependent. By combining capillary electrophoresis, QDs and FRET, our study herein not only provides a new method for the detection and dynamically monitoring of PSP enzyme cleavage activity, but also can be extended to the detection of many other enzymes and proteases.


Asunto(s)
Electroforesis Capilar/métodos , Péptido Hidrolasas/química , Péptidos/química , Puntos Cuánticos/química , Proteínas Virales/química , Transferencia Resonante de Energía de Fluorescencia/métodos , Colorantes Fluorescentes/química , Proteolisis , Rhinovirus/enzimología
19.
J Virol ; 91(8)2017 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-28179529

RESUMEN

The RNA rhinoviruses (RV) encode 2A proteases (2Apro) that contribute essential polyprotein processing and host cell shutoff functions during infection, including the cleavage of Phe/Gly-containing nucleoporin proteins (Nups) within nuclear pore complexes (NPC). Within the 3 RV species, multiple divergent genotypes encode diverse 2Apro sequences that act differentially on specific Nups. Since only subsets of Phe/Gly motifs, particularly those within Nup62, Nup98, and Nup153, are recognized by transport receptors (karyopherins) when trafficking large molecular cargos through the NPC, the processing preferences of individual 2Apro predict RV genotype-specific targeting of NPC pathways and cargos. To test this idea, transformed HeLa cell lines were created with fluorescent cargos (mCherry) for the importin α/ß, transportin 1, and transportin 3 import pathways and the Crm1-mediated export pathway. Live-cell imaging of single cells expressing recombinant RV 2Apro (A16, A45, B04, B14, B52, C02, and C15) showed disruption of each pathway with measurably different efficiencies and reaction rates. The B04 and B52 proteases preferentially targeted Nups in the import pathways, while B04 and C15 proteases were more effective against the export pathway. Virus-type-specific trends were also observed during infection of cells with A16, B04, B14, and B52 viruses or their chimeras, as measured by NF-κB (p65/Rel) translocation into the nucleus and the rates of virus-associated cytopathic effects. This study provides new tools for evaluating the host cell response to RV infections in real time and suggests that differential 2Apro activities explain, in part, strain-dependent host responses and diverse RV disease phenotypes.IMPORTANCE Genetic variation among human rhinovirus types includes unexpected diversity in the genes encoding viral proteases (2Apro) that help these viruses achieve antihost responses. When the enzyme activities of 7 different 2Apro were measured comparatively in transformed cells programed with fluorescent reporter systems and by quantitative cell imaging, the cellular substrates, particularly in the nuclear pore complex, used by these proteases were indeed attacked at different rates and with different affinities. The importance of this finding is that it provides a mechanistic explanation for how different types (strains) of rhinoviruses may elicit different cell responses that directly or indirectly lead to distinct disease phenotypes.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Interacciones Huésped-Patógeno , Proteínas de Complejo Poro Nuclear/metabolismo , Rhinovirus/enzimología , Rhinovirus/patogenicidad , Proteínas Virales/metabolismo , Células HeLa , Humanos , Microscopía Fluorescente , Transporte de Proteínas
20.
Bioorg Med Chem Lett ; 26(14): 3248-3252, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27265257

RESUMEN

Human rhinovirus (HRV) is a primary cause of common cold and is linked to exacerbation of underlying respiratory diseases such as asthma and COPD. HRV 3C protease, which is responsible for cleavage of viral polyprotein in to proteins essential for viral life-cycle, represents an important target. We have designed proline- and azetidine-based analogues of Rupintrivir that target the P2 pocket of the binding site. Potency optimization, aided with X-ray crystallography and quantum mechanical calculations, led to compounds with activity against a broad spectrum of HRV serotypes. Altogether, these compounds represent alternative starting points to identify promising leads in our continual efforts to treat HRV infections.


Asunto(s)
Antivirales/farmacología , Azetidinas/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Diseño de Fármacos , Prolina/farmacología , Rhinovirus/efectos de los fármacos , Proteínas Virales/antagonistas & inhibidores , Proteasas Virales 3C , Antivirales/síntesis química , Antivirales/química , Azetidinas/síntesis química , Azetidinas/química , Cristalografía por Rayos X , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/síntesis química , Inhibidores de Cisteína Proteinasa/química , Relación Dosis-Respuesta a Droga , Humanos , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Molecular , Prolina/síntesis química , Prolina/química , Teoría Cuántica , Rhinovirus/enzimología , Relación Estructura-Actividad , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA