Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 347
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 98(3): e0151523, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38323812

RESUMEN

Adeno-associated virus (AAV) requires co-infection with helper virus for efficient replication. We previously reported that Human Bocavirus 1 (HBoV1) genes, including NP1, NS2, and BocaSR, were critical for AAV2 replication. Here, we first demonstrate the essential roles of the NP1 protein in AAV2 DNA replication and protein expression. We show that NP1 binds to single-strand DNA (ssDNA) at least 30 nucleotides (nt) in length in a sequence-independent manner. Furthermore, NP1 colocalized with the BrdU-labeled AAV2 DNA replication center, and the loss of the ssDNA-binding ability of NP1 by site-directed mutation completely abolished AAV2 DNA replication. We used affinity-tagged NP1 protein to identify host cellular proteins associated with NP1 in cells cotransfected with the HBoV1 helper genes and AAV2 duplex genome. Of the identified proteins, we demonstrate that NP1 directly binds to the DBD-F domain of the RPA70 subunit with a high affinity through the residues 101-121. By reconstituting the heterotrimer protein RPA in vitro using gel filtration, we demonstrate that NP1 physically associates with RPA to form a heterologous complex characterized by typical fast-on/fast-off kinetics. Following a dominant-negative strategy, we found that NP1-RPA complex mainly plays a role in expressing AAV2 capsid protein by enhancing the transcriptional activity of the p40 promoter. Our study revealed a novel mechanism by which HBoV1 NP1 protein supports AAV2 DNA replication and capsid protein expression through its ssDNA-binding ability and direct interaction with RPA, respectively.IMPORTANCERecombinant adeno-associated virus (rAAV) vectors have been extensively used in clinical gene therapy strategies. However, a limitation of these gene therapy strategies is the efficient production of the required vectors, as AAV alone is replication-deficient in the host cells. HBoV1 provides the simplest AAV2 helper genes consisting of NP1, NS2, and BocaSR. An important question regarding the helper function of HBoV1 is whether it provides any direct function that supports AAV2 DNA replication and protein expression. Also of interest is how HBoV1 interplays with potential host factors to constitute a permissive environment for AAV2 replication. Our studies revealed that the multifunctional protein NP1 plays important roles in AAV2 DNA replication via its sequence-independent ssDNA-binding ability and in regulating AAV2 capsid protein expression by physically interacting with host protein RPA. Our findings present theoretical guidance for the future application of the HBoV1 helper genes in the rAAV vector production.


Asunto(s)
Proteínas de la Cápside , Cápside , ADN de Cadena Simple , ADN Viral , Proteínas de Unión al ADN , Dependovirus , Bocavirus Humano , Proteínas Virales , Humanos , Cápside/metabolismo , Proteínas de la Cápside/biosíntesis , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Dependovirus/genética , Dependovirus/crecimiento & desarrollo , Dependovirus/metabolismo , ADN de Cadena Simple/biosíntesis , ADN de Cadena Simple/metabolismo , ADN Viral/biosíntesis , ADN Viral/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Viral de la Expresión Génica , Bocavirus Humano/genética , Bocavirus Humano/metabolismo , Cinética , Mutagénesis Sitio-Dirigida , Mutación , Regiones Promotoras Genéticas , Unión Proteica , Dominios Proteicos , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
2.
Protein Expr Purif ; 220: 106502, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38754753

RESUMEN

Adeno-associated Virus (AAV) is a promising vector for gene therapy. However, few studies have focused on producing virus-like particles (VLPs) of AAV in cells, especially in E. coli. In this study, we describe a method to produce empty VP3-only VLPs of AAV2 in E. coli by co-expressing VP3 and assembly-activating protein (AAP) of AAV2. Although the yields of VLPs produced with our method were low, the VLPs were able to self-assemble in E. coli without the need of in vitro capsid assembly. The produced VLPs were characterized by immunological detection and transmission electron microscopy (TEM). In conclusion, this study demonstrated that capsid assembly of AAV2 is possible in E. coli, and E. coli may be a candidate system for production of VLPs of AAV.


Asunto(s)
Proteínas de la Cápside , Dependovirus , Escherichia coli , Escherichia coli/genética , Escherichia coli/metabolismo , Dependovirus/genética , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/biosíntesis , Virión/genética , Virión/metabolismo , Ensamble de Virus , Vectores Genéticos/metabolismo , Vectores Genéticos/genética , Vectores Genéticos/química , Parvovirinae/genética , Humanos
3.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33384338

RESUMEN

Human adenovirus species D (HAdV-D) types are currently being explored as vaccine vectors for coronavirus disease 2019 (COVID-19) and other severe infectious diseases. The efficacy of such vector-based vaccines depends on functional interactions with receptors on host cells. Adenoviruses of different species are assumed to enter host cells mainly by interactions between the knob domain of the protruding fiber capsid protein and cellular receptors. Using a cell-based receptor-screening assay, we identified CD46 as a receptor for HAdV-D56. The function of CD46 was validated in infection experiments using cells lacking and overexpressing CD46, and by competition infection experiments using soluble CD46. Remarkably, unlike HAdV-B types that engage CD46 through interactions with the knob domain of the fiber protein, HAdV-D types infect host cells through a direct interaction between CD46 and the hexon protein. Soluble hexon proteins (but not fiber knob) inhibited HAdV-D56 infection, and surface plasmon analyses demonstrated that CD46 binds to HAdV-D hexon (but not fiber knob) proteins. Cryoelectron microscopy analysis of the HAdV-D56 virion-CD46 complex confirmed the interaction and showed that CD46 binds to the central cavity of hexon trimers. Finally, soluble CD46 inhibited infection by 16 out of 17 investigated HAdV-D types, suggesting that CD46 is an important receptor for a large group of adenoviruses. In conclusion, this study identifies a noncanonical entry mechanism used by human adenoviruses, which adds to the knowledge of adenovirus biology and can also be useful for development of adenovirus-based vaccine vectors.


Asunto(s)
Adenovirus Humanos , Vacunas contra la COVID-19 , Proteínas de la Cápside , Regulación Viral de la Expresión Génica , SARS-CoV-2/genética , Internalización del Virus , Adenovirus Humanos/genética , Adenovirus Humanos/metabolismo , Vacunas contra la COVID-19/genética , Vacunas contra la COVID-19/metabolismo , Proteínas de la Cápside/biosíntesis , Proteínas de la Cápside/genética , Línea Celular , Humanos
4.
RNA ; 25(5): 645-655, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30803999

RESUMEN

External guide sequences (EGSs) signify the short RNAs that induce ribonuclease P (RNase P), an enzyme responsible for processing the 5' termini of tRNA, to specifically cleave a target mRNA by forming a precursor tRNA-like complex. Hence, the EGS technology may serve as a potential strategy for gene-targeting therapy. Our previous studies have revealed that engineered EGS variants induced RNase P to efficiently hydrolyze target mRNAs. In the present research, an EGS variant was designed to be complementary to the mRNA coding for human cytomegalovirus (HCMV) major capsid protein (MCP), which is vital to form the viral capsid. In vitro, the EGS variant was about 80-fold more efficient in inducing human RNase P-mediated cleavage of the target mRNA than a natural tRNA-derived EGS. Moreover, the expressed variant and natural tRNA-originated EGSs led to a decrease of MCP expression by 98% and 73%-74% and a decrease of viral growth by about 10,000- and 200-fold in cells infected with HCMV, respectively. These results reveal direct evidence that the engineered EGS variant has higher efficiency in blocking the expression of HCMV genes and viral growth than the natural tRNA-originated EGS. Therefore, our findings imply that the EGS variant can be a potent candidate agent for the treatment of infections caused by HCMV.


Asunto(s)
Proteínas de la Cápside/genética , Citomegalovirus/genética , ARN Guía de Kinetoplastida/genética , ARN Mensajero/genética , ARN de Transferencia de Serina/genética , ARN Viral/genética , Ribonucleasa P/metabolismo , Emparejamiento Base , Proteínas de la Cápside/biosíntesis , Línea Celular Transformada , Línea Celular Tumoral , Citomegalovirus/metabolismo , Fibroblastos/metabolismo , Fibroblastos/virología , Regulación Viral de la Expresión Génica , Marcación de Gen/métodos , Ingeniería Genética/métodos , Interacciones Huésped-Patógeno/genética , Humanos , Terapia Molecular Dirigida , Neuroglía/metabolismo , Neuroglía/virología , Conformación de Ácido Nucleico , Cultivo Primario de Células , División del ARN , ARN Guía de Kinetoplastida/química , ARN Guía de Kinetoplastida/metabolismo , ARN Mensajero/química , ARN Mensajero/metabolismo , ARN de Transferencia de Serina/química , ARN de Transferencia de Serina/metabolismo , ARN Viral/química , ARN Viral/metabolismo , Ribonucleasa P/química , Ribonucleasa P/genética , Replicación Viral/fisiología
5.
RNA ; 25(4): 431-452, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30659060

RESUMEN

Noncanonical translation, and particularly initiation on non-AUG codons, are frequently used by viral and cellular mRNAs during virus infection and disease. The Sindbis virus (SINV) subgenomic mRNA (sgRNA) constitutes a unique model system to analyze the translation of a capped viral mRNA without the participation of several initiation factors. Moreover, sgRNA can initiate translation even when the AUG initiation codon is replaced by other codons. Using SINV replicons, we examined the efficacy of different codons in place of AUG to direct the synthesis of the SINV capsid protein. The substitution of AUG by CUG was particularly efficient in promoting the incorporation of leucine or methionine in similar percentages at the amino terminus of the capsid protein. Additionally, valine could initiate translation when the AUG is replaced by GUG. The ability of sgRNA to initiate translation on non-AUG codons was dependent on the integrity of a downstream stable hairpin (DSH) structure located in the coding region. The structural requirements of this hairpin to signal the initiation site on the sgRNA were examined in detail. Of interest, a virus bearing CUG in place of AUG in the sgRNA was able to infect cells and synthesize significant amounts of capsid protein. This virus infects the human haploid cell line HAP1 and the double knockout variant that lacks eIF2A and eIF2D. Collectively, these findings indicate that leucine-tRNA or valine-tRNA can participate in the initiation of translation of sgRNA by a mechanism dependent on the DSH. This mechanism does not involve the action of eIF2, eIF2A, or eIF2D.


Asunto(s)
Codón Iniciador/genética , Biosíntesis de Proteínas , ARN Mensajero/genética , ARN Viral/genética , Transducción de Señal/genética , Virus Sindbis/genética , Proteínas de la Cápside/biosíntesis , Proteínas de la Cápside/genética , Línea Celular Tumoral , Codón Iniciador/metabolismo , Factor 2 Eucariótico de Iniciación/deficiencia , Factor 2 Eucariótico de Iniciación/genética , Fibroblastos/metabolismo , Fibroblastos/virología , Regulación de la Expresión Génica , Haploidia , Interacciones Huésped-Patógeno/genética , Humanos , Secuencias Invertidas Repetidas , Leucina/genética , Leucina/metabolismo , Metionina/genética , Metionina/metabolismo , Conformación de Ácido Nucleico , ARN Mensajero/metabolismo , ARN de Transferencia de Leucina/genética , ARN de Transferencia de Leucina/metabolismo , ARN de Transferencia de Valina/genética , ARN de Transferencia de Valina/metabolismo , ARN Viral/metabolismo , Replicón , Virus Sindbis/metabolismo , Valina/genética , Valina/metabolismo
6.
J Virol ; 94(15)2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32404528

RESUMEN

Caliciviruses have a positive-strand RNA genome with a length of about 7.5 kb that contains 2, 3, or 4 functional open reading frames (ORFs). A subgenomic mRNA (sg-RNA) is transcribed in the infected cell, and both major capsid protein viral protein 1 (VP1) and minor capsid protein VP2 are translated from the sg-RNA. Translation of proteins from the genomic RNA (g-RNA) and from the sg-RNA is mediated by the RNA-linked viral protein VPg (virus protein, genome linked). Most of the calicivirus genera have translation mechanisms leading to VP1 expression from the g-RNA. VP1 is part of the polyprotein for sapoviruses, lagoviruses, and neboviruses, and a termination/reinitiation mechanism was described for noroviruses. Vesiviruses have no known mechanism for the expression of VP1 from the g-RNA, and the Vesivirus genus is the only genus of the Caliciviridae that generates VP1 via a precursor capsid leader protein (LC-VP1). Analyses of feline calicivirus (FCV) g-RNA translation showed a low level of VP1 expression with an initiation downstream of the original start codon of LC-VP1, leading to a smaller, truncated LC-VP1 (tLC-VP1) protein. Deletion and substitution analyses of the region surrounding the LC-VP1 start codon allowed the identification of sequences within the leader protein coding region of FCV that have an impact on VP1 translation frequency from the g-RNA. Introduction of such mutations into the virus showed an impact of strongly reduced tLC-VP1 levels translated from the g-RNA on viral replication.IMPORTANCE Caliciviruses are a cause of important diseases in humans and animals. It is crucial to understand the prerequisites of efficient replication of these viruses in order to develop strategies for prevention and treatment of these diseases. It was shown before that all caliciviruses except vesiviruses have established mechanisms to achieve major capsid protein (VP1) translation from the genomic RNA. Here, we show for the first time that a member of the genus Vesivirus also has a translation initiation mechanism by which a precursor protein of the VP1 protein is expressed from the genomic RNA. This finding clearly points at a functional role of the calicivirus VP1 capsid protein in early replication, and we provide experimental data supporting this hypothesis.


Asunto(s)
Calicivirus Felino/metabolismo , Proteínas de la Cápside/biosíntesis , Regulación Viral de la Expresión Génica , Genoma Viral , Biosíntesis de Proteínas , ARN Viral/metabolismo , Animales , Calicivirus Felino/genética , Proteínas de la Cápside/genética , Gatos , Línea Celular , Cricetinae , ARN Viral/genética
7.
J Virol ; 94(2)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31666379

RESUMEN

Human bocavirus 1 (HBoV1), which belongs to the genus Bocaparvovirus of the Parvoviridae family, causes acute respiratory tract infections in young children. In vitro, HBoV1 infects polarized primary human airway epithelium (HAE) cultured at an air-liquid interface (HAE-ALI). HBoV1 encodes a small nonstructural protein, nuclear protein 1 (NP1), that plays an essential role in the maturation of capsid protein (VP)-encoding mRNAs and viral DNA replication. In this study, we determined the broad interactome of NP1 using the proximity-dependent biotin identification (BioID) assay combined with mass spectrometry (MS). We confirmed that two host mRNA processing factors, DEAH-box helicase 15 (DHX15) and cleavage and polyadenylation specificity factor 6 (CPSF6; also known as CFIm68), a subunit of the cleavage factor Im complex (CFIm), interact with HBoV1 NP1 independently of any DNA or mRNAs. Knockdown of CPSF6 significantly decreased the expression of capsid protein but not that of DHX15. We further demonstrated that NP1 directly interacts with CPSF6 in vitro and colocalizes within the virus replication centers. Importantly, we revealed a novel role of CPSF6 in the nuclear import of NP1, in addition to the critical role of CPSF6 in NP1-facilitated maturation of VP-encoding mRNAs. Thus, our study suggests that CPSF6 interacts with NP1 to escort NP1 imported into the nucleus for its function in the modulation of viral mRNA processing and viral DNA replication.IMPORTANCE Human bocavirus 1 (HBoV1) is one of the significant pathogens causing acute respiratory tract infections in young children worldwide. HBoV1 encodes a small nonstructural protein (NP1) that plays an important role in the maturation of viral mRNAs encoding capsid proteins as well as in viral DNA replication. Here, we identified a critical host factor, CPSF6, that directly interacts with NP1, mediates the nuclear import of NP1, and plays a role in the maturation of capsid protein-encoding mRNAs in the nucleus. The identification of the direct interaction between viral NP1 and host CPSF6 provides new insights into the mechanism by which a viral small nonstructural protein facilitates the multiple regulation of viral gene expression and replication and reveals a novel target for potent antiviral drug development.


Asunto(s)
Proteínas de la Cápside/biosíntesis , Núcleo Celular , Regulación Viral de la Expresión Génica , Bocavirus Humano/metabolismo , Proteínas Nucleares/metabolismo , ARN Mensajero , ARN Viral , Factores de Escisión y Poliadenilación de ARNm/metabolismo , Transporte Activo de Núcleo Celular , Proteínas de la Cápside/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/virología , Células HEK293 , Bocavirus Humano/genética , Humanos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/genética , ARN Helicasas/genética , ARN Helicasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Factores de Escisión y Poliadenilación de ARNm/genética
8.
Biotechnol Bioeng ; 118(4): 1707-1720, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33484156

RESUMEN

Expression of viral capsomeres in bacterial systems and subsequent in vitro assembly into virus-like particles is a possible pathway for affordable future vaccines. However, purification is challenging as viral capsomeres show poor binding to chromatography media. In this study, the behavior of capsomeres in unfractionated bacterial lysate was compared with that for purified capsomeres, with or without added microbial DNA, to better understand reasons for poor bioprocess behavior. We show that aggregates or complexes form through the interaction between viral capsomeres and DNA, especially in bacterial lysates rich in contaminating DNA. The formation of these complexes prevents the target protein capsomeres from accessing the pores of chromatography media. We find that protein-DNA interactions can be modulated by controlling the ionic strength of the buffer and that at elevated ionic strengths the protein-DNA complexes dissociate. Capsomeres thus released show enhanced bind-elute behavior on salt-tolerant chromatography media. DNA could therefore be efficiently removed. We believe this is the first report of the use of an optimized salt concentration that dissociates capsomere-DNA complexes yet enables binding to salt-tolerant media. Post purification, assembly experiments indicate that DNA-protein interactions can play a negative role during in vitro assembly, as DNA-protein complexes could not be assembled into virus-like particles, but formed worm-like structures. This study reveals that the control over DNA-protein interaction is a critical consideration during downstream process development for viral vaccines.


Asunto(s)
Proteínas de la Cápside , ADN Bacteriano/química , Escherichia coli , Vacunas de Partículas Similares a Virus , Proteínas de la Cápside/biosíntesis , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/aislamiento & purificación , Cromatografía Liquida , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/metabolismo , Vacunas de Partículas Similares a Virus/biosíntesis , Vacunas de Partículas Similares a Virus/química , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/aislamiento & purificación
9.
Proc Natl Acad Sci U S A ; 115(28): 7344-7349, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29941585

RESUMEN

Most double-stranded RNA (dsRNA) viruses transcribe RNA plus strands within a common innermost capsid shell. This process requires coordinated efforts by RNA-dependent RNA polymerase (RdRp) together with other capsid proteins and genomic RNA. Here we report the near-atomic resolution structure of the RdRp protein VP2 in complex with its cofactor protein VP4 and genomic RNA within an aquareovirus capsid using 200-kV cryoelectron microscopy and symmetry-mismatch reconstruction. The structure of these capsid proteins enabled us to observe the elaborate nonicosahedral structure within the double-layered icosahedral capsid. Our structure shows that the RdRp complex is anchored at the inner surface of the capsid shell and interacts with genomic dsRNA and four of the five asymmetrically arranged N termini of the capsid shell proteins under the fivefold axis, implying roles for these N termini in virus assembly. The binding site of the RNA end at VP2 is different from the RNA cap binding site identified in the crystal structure of orthoreovirus RdRp λ3, although the structures of VP2 and λ3 are almost identical. A loop, which was thought to separate the RNA template and transcript, interacts with an apical domain of the capsid shell protein, suggesting a mechanism for regulating RdRp replication and transcription. A conserved nucleoside triphosphate binding site was localized in our RdRp cofactor protein VP4 structure, and interactions between the VP4 and the genomic RNA were identified.


Asunto(s)
Proteínas de la Cápside/biosíntesis , ARN Polimerasas Dirigidas por ADN/metabolismo , Genoma Viral , ARN Viral/biosíntesis , Reoviridae/fisiología , Transcripción Genética/fisiología , Ensamble de Virus/fisiología , Animales , Proteínas de la Cápside/genética , Carpas , Línea Celular , ARN Viral/genética
10.
Biochem Biophys Res Commun ; 533(4): 983-987, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33008586

RESUMEN

JC polyomavirus (JCPyV) causes progressive multifocal leukoencephalopathy (PML), a demyelinating disease of the central nervous system, in immunocompromised patients. Although PML used to be rare, recently the incidence of PML has risen due to an increase in immunosuppressive therapy. An in vitro JCPyV infection system could be used for anti-drug screening and investigation of tropism changes, but study of JCPyV in vitro has been limited due to the difficulty of efficiently propagating the virus in cultured cells. PML-type JCPyV efficiently propagates in primary human fetal and progenitor cell-derived astrocytes, but the preparation of cells from human fetuses is associated with severe ethical problems. In this study, human iPS cell-derived astrocytes were exposed to PML-type JCPyV. Infection, replication, and VP1 and T antigens of JCPyV were detected and confirmed in this culture. The non-coding control region (NCCR) of M1-IMRb was conserved in infected cells without point mutations. In addition, PML-type JCPyV genomic DNA in infected cells was detected as a single band of approximately 5.1 kbp, with no deletions. This is the first demonstration that human iPS cell-derived astrocytes efficiently support replication of PML-type JCPyV without production of defective interfering particles. These findings indicated that a culture system using human iPS cell-derived astrocyte would be useful for studies of PML, especially for screening anti-JCPyV drugs.


Asunto(s)
Astrocitos/virología , Células Madre Pluripotentes Inducidas/virología , Virus JC/fisiología , Leucoencefalopatía Multifocal Progresiva/virología , Animales , Antígenos Virales/biosíntesis , Antígenos Virales de Tumores/biosíntesis , Astrocitos/patología , Células COS , Proteínas de la Cápside/biosíntesis , Proteínas de la Cápside/inmunología , Diferenciación Celular , Línea Celular , Chlorocebus aethiops , ADN Viral/genética , Genoma Viral , Humanos , Células Madre Pluripotentes Inducidas/patología , Virus JC/genética , Virus JC/patogenicidad , Leucoencefalopatía Multifocal Progresiva/etiología , Leucoencefalopatía Multifocal Progresiva/patología , Células-Madre Neurales/patología , Cultivo de Virus/métodos , Replicación Viral
11.
Protein Expr Purif ; 174: 105679, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32534017

RESUMEN

The applications of viral protein cages have expanded rapidly into the fields of bionanotechnology and materials science. However, the low-cost production of viral capsid proteins (CPs) on a large scale is always a challenge. Herein, we develop a highly efficient expression system by constructing recombinant Pichia pastoris cells as a "factory" for the secretion of soluble cowpea chlorotic mottle virus (CCMV) CPs. Under optimal induction conditions (0.9 mg/mL of methanol concentration at 30 °C for 96 h), a high yield of approximately 95 mg/L of CCMV CPs was harvested from the fermentation supernatant with CPs purity >90%, which has significantly simplified the rest of the purification process. The resultant CPs are employed to encapsulate Ruthenium (Ru) nanoparticles (NPs) via in-vitro self-assembly to prepare hybrid nanocatalyst, i.e. Ru@virus-like particles (VLPs). The catalytic activity over Ru@VLPs was evaluated by reducing 4-nitrophenol (4-NP) to 4-aminophenol (4-AP). The results indicate that, with the protection of protein cages, Ru NPs were highly stabilized during the catalytic reaction. This results in enhanced catalytic activity (reaction rate constant k = 0.14 min-1) in comparison with unsupported citrate-stabilized Ru NPs (Ru-CA) (k = 0.08 min-1). Additionally, comparatively lower activation energy over Ru@VLPs (approximately 32 kJ/mol) than that over Ru-CA (approximately 39 kJ/mol) could be attributed to the synergistic effect between Ru NPs and some functional groups such as amino groups (-NH2) on CPs that weakened the activation barrier of 4-NP reduction. Therefore, enhanced activity and decreased activation energy over Ru@VLPs demonstrated the superiority of Ru@VLPs to unsupported Ru-CA.


Asunto(s)
Bromovirus/genética , Proteínas de la Cápside , Nanopartículas del Metal/química , Rutenio/química , Saccharomycetales , Proteínas de la Cápside/biosíntesis , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/aislamiento & purificación , Cápsulas , Catálisis , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Saccharomycetales/genética , Saccharomycetales/crecimiento & desarrollo
12.
Protein Expr Purif ; 175: 105690, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32681956

RESUMEN

The capsid of human papillomavirus (HPV) consists of two capsid proteins - the major capsid protein L1 and the minor capsid protein L2. Assembled virus-like particles, which only consist of L1 proteins, are successfully applied as prophylactic vaccines against HPV infections. The capsid subunits are L1-pentamers, which are also reported to protect efficiently against HPV infections in animals. The recombinant production of L1 has been previously shown in E. coli, yeast, insect cells, plants and mammalian cell culture. Principally, in E. coli-based expression system L1 shows high expression yields but the protein is largely insoluble. In order to overcome this problem reported strategies address fusion proteins and overexpression of bacterial chaperones. However, an insufficient cleavage of the fusion proteins and removal of co-purified chaperones can hamper subsequent down streaming. We report a significant improvement in the production of soluble L1-pentamers by combining (I) a fusion of a N-terminal SUMO-tag to L1, (II) the heterologous co-expression of the chaperon system GroEL/ES and (III) low expression temperature. The fusion construct was purified in a 2-step protein purification including efficient removal of GroEL/ES and complete removal of the N-terminal SUMO-tag. The expression strategy was transferred to process-controlled high-cell-density fermentation with defined media according to the guidelines of good manufacturing practice. The produced L1 protein is highly pure (>95%), free of DNA (260:280 = 0.5) and pentameric. The production strategy yielded 5.73 mg of purified L1-pentamers per gram dry biomass. The optimized strategy is a suitable alternative for high yield L1-pentamer production and purification as a cheaper process for vaccine production.


Asunto(s)
Proteínas de la Cápside , Papillomavirus Humano 16/genética , Proteínas Oncogénicas Virales , Multimerización de Proteína , Proteínas Recombinantes de Fusión , Proteínas de la Cápside/biosíntesis , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/aislamiento & purificación , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas Oncogénicas Virales/biosíntesis , Proteínas Oncogénicas Virales/química , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/aislamiento & purificación , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación
13.
Protein Expr Purif ; 175: 105687, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32681952

RESUMEN

A promising candidate for tumor targeted toxins is the chicken anemia-derived protein apoptin that induces tumor-specific apoptosis. It was aimed to design a novel apoptin-based targeted toxin by genetic fusion of apoptin with the tumor-directed ligand epidermal growth factor (EGF) using Escherichia coli as expression host. However, apoptin is highly hydrophobic and tends to form insoluble aggregates. Therefore, three different apoptin-EGF variants were generated. The fusion protein hexa-histidine (His)-apoptin-EGF (HAE) was expressed in E. coli and purified under denaturing conditions due to inclusion bodies. The protein solubility was improved by maltose-binding protein (MBP) or glutathione S-transferase. The protein MBP-apoptin-EGFHis (MAEH) was found favorable as a targeted toxin regarding final yield (4-6 mg/L) and stability. MBP was enzymatically removed using clotting factor Xa, which resulted in low yield and poor separation. MAEH was tested on target and non-target cell lines. The targeted tumor cell line A431 showed significant toxicity with an IC50 of 69.55 nM upon incubation with MAEH while fibroblasts and target receptor-free cells remained unaffected. Here we designed a novel EGF receptor targeting drug with high yield, purity and stability.


Asunto(s)
Antineoplásicos , Proteínas de la Cápside , Factor de Crecimiento Epidérmico , Neoplasias/tratamiento farmacológico , Proteínas Recombinantes de Fusión , Animales , Antineoplásicos/aislamiento & purificación , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Proteínas de la Cápside/biosíntesis , Proteínas de la Cápside/genética , Proteínas de la Cápside/aislamiento & purificación , Proteínas de la Cápside/farmacología , Línea Celular Tumoral , Factor de Crecimiento Epidérmico/biosíntesis , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/aislamiento & purificación , Factor de Crecimiento Epidérmico/farmacología , Humanos , Ratones , Células 3T3 NIH , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/farmacología
14.
Mol Cell Probes ; 52: 101556, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32126262

RESUMEN

The present study was aimed at comparing different E. coli strains in expressing the capsid protein of Porcine Circovirus 2 (PCV2). Full length capsid protein could be expressed only in Rosetta-gami 2 (DE3) pLysS strain using pET32b (+) vector. This confirmed that only those strains which possess tRNAs for rare codons can express the full length capsid protein. Purification of full length capsid protein could not be achieved even after several attempts using native and denaturing conditions. Subsequently, an attempt was made for expression of N-terminal truncated capsid protein using the same expression system. Truncated capsid protein was successfully expressed, purified and characterized by western blotting. The truncated capsid protein was also shown to be efficacious in testing serum samples using an optimized indirect ELISA, wherein a diagnostic sensitivity of 88.89% and specificity of 90.82% was obtained as compared to commercially available GreenSpring® porcine circovirus (PCV2) ELISA test kit. Thus, the expressed truncated capsid protein appears to be a promising diagnostic agent for PCV2. The comparative analysis suggests that cluster of arginine residues at N-terminal of capsid protein not only affects its expression in some E. coli strains but also its purification by Ni-NTA chromatography, when expressed as a histidine tagged fusion protein.


Asunto(s)
Proteínas de la Cápside/biosíntesis , Circovirus/metabolismo , Escherichia coli/metabolismo , Proteínas Recombinantes/biosíntesis , Animales , Antígenos Virales/metabolismo , Proteínas de la Cápside/aislamiento & purificación , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Sistemas de Lectura Abierta/genética , Curva ROC , Proteínas Recombinantes/aislamiento & purificación , Porcinos
15.
Arch Virol ; 165(12): 2829-2835, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33000310

RESUMEN

The swine pathogen porcine circovirus type 2 (PCV2) causes significant economic damage worldwide. The PCV2 capsid (CP) residues 169-STIDYFQPNNKR-180 have been identified as a decoy epitope that diverts the host immune response away from protective epitopes. However, the decoy epitope may include important linear or conformational protective epitopes against PCV2. In this study, we used the baculovirus system to express recombinant complete CP (1-233) and mutant CP (Δ169-180), in which the decoy epitope was deleted, and evaluated the immune response to these in mice. Immunization with mutant CP (Δ169-180) protein, which formed very low level of virus-like particles (VLPs), elicited significantly lower levels of PCV2 CP-specific IgG antibodies and a slightly lower neutralizing activity than immunization with the complete CP (1-233) protein. This finding suggests that the complete CP is important for efficient VLP assembly and induction of PCV2-specific IgG antibodies and neutralizing antibodies in mice. This study may provide useful information for next-generation vaccine design for PCV2 control.


Asunto(s)
Proteínas de la Cápside/inmunología , Circovirus/inmunología , Epítopos/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Animales , Anticuerpos Neutralizantes/análisis , Anticuerpos Antivirales/análisis , Proteínas de la Cápside/biosíntesis , Proteínas de la Cápside/genética , Circovirus/genética , Epítopos/biosíntesis , Epítopos/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Síndrome Multisistémico de Emaciación Posdestete Porcino/inmunología , Síndrome Multisistémico de Emaciación Posdestete Porcino/prevención & control , Porcinos , Vacunación , Vacunas de Partículas Similares a Virus/genética
16.
Arch Virol ; 165(6): 1441-1444, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32239294

RESUMEN

Bovine papillomavirus type 9 (BPV9) is a causative agent of severe teat papillomatosis. Considering the lack of efficient BPV culture methods, recombinant proteins such as virus-like particles developed through genetic engineering may serve as a useful tool for developing effective vaccines against BPV infection. In this study, we successfully produced immunogenic particles composed of recombinant L1 protein of BPV9 (rBPV9-L1), using a baculovirus expression system. rBPV9-L1-immunized mice produced BPV9-specific IgG, which did not cross-react with BPV type 6, which is another causative agent of teat papillomatosis. Hence, immunogenic rBPV9-L1 is potentially applicable as a vaccine candidate for teat papillomatosis.


Asunto(s)
Proteínas de la Cápside/inmunología , Enfermedades de los Bovinos/prevención & control , Papillomaviridae/inmunología , Infecciones por Papillomavirus/veterinaria , Vacunas de Partículas Similares a Virus/inmunología , Animales , Proteínas de la Cápside/biosíntesis , Bovinos , Enfermedades de los Bovinos/virología , Femenino , Genotipo , Ratones , Papillomaviridae/genética , Infecciones por Papillomavirus/prevención & control , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología , Vacunación
17.
Arch Virol ; 165(10): 2301-2309, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32757056

RESUMEN

Porcine circovirus type 2 (PCV2) is a major pathogen associated with swine diseases. It is the smallest single-stranded DNA virus, and its genome contains four major open reading frames (ORFs). ORF2 encodes the major structural protein Cap, which can self-assemble into virus-like particles (VLPs) in vitro and contains the primary antigenic determinants. In this study, we developed a high-efficiency method for obtaining VLPs and optimized the purification conditions. In this method, we expressed the protein Cap with a 6× His tag using baculovirus-infected silkworm larvae as well as the E. coli BL21(DE3) prokaryotic expression system. The PCV2 Cap proteins produced by the silkworm larvae and E. coli BL21(DE3) were purified. Cap proteins purified from silkworm larvae self-assembled into VLPs in vitro, while the Cap proteins purified from bacteria were unable to self-assemble. Transmission electron microscopy confirmed the self-assembly of VLPs. The immunogenicity of the VLPs produced using the baculovirus system was demonstrated using an enzyme-linked immunosorbent assay (ELISA). Furthermore, the purification process was optimized. The results demonstrated that the expression system using baculovirus-infected silkworm larvae is a good choice for obtaining VLPs of PCV2 and has potential for the development of a low-cost and efficient vaccine.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Baculoviridae/genética , Bombyx/virología , Proteínas de la Cápside/inmunología , Circovirus/inmunología , Vacunas de Partículas Similares a Virus/biosíntesis , Vacunas Virales/biosíntesis , Animales , Antígenos Virales/química , Antígenos Virales/inmunología , Baculoviridae/inmunología , Proteínas de la Cápside/biosíntesis , Proteínas de la Cápside/genética , Infecciones por Circoviridae/inmunología , Infecciones por Circoviridae/prevención & control , Infecciones por Circoviridae/virología , Circovirus/genética , Epítopos/química , Epítopos/inmunología , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Histidina/genética , Histidina/inmunología , Sueros Inmunes/química , Inmunogenicidad Vacunal , Larva/virología , Ratones , Oligopéptidos/genética , Oligopéptidos/inmunología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/prevención & control , Enfermedades de los Porcinos/virología , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/aislamiento & purificación , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Vacunas Virales/aislamiento & purificación
18.
J Am Chem Soc ; 141(9): 3875-3884, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30730722

RESUMEN

Site-specific protein modification is a widely used strategy to attach drugs, imaging agents, or other useful small molecules to protein carriers. N-terminal modification is particularly useful as a high-yielding, site-selective modification strategy that can be compatible with a wide array of proteins. However, this modification strategy is incompatible with proteins with buried or sterically hindered N termini, such as virus-like particles (VLPs) composed of the well-studied MS2 bacteriophage coat protein. To assess VLPs with improved compatibility with these techniques, we generated a targeted library based on the MS2-derived protein cage with N-terminal proline residues followed by three variable positions. We subjected the library to assembly, heat, and chemical selections, and we identified variants that were modified in high yield with no reduction in thermostability. Positive charge adjacent to the native N terminus is surprisingly beneficial for successful extension, and over 50% of the highest performing variants contained positive charge at this position. Taken together, these studies described nonintuitive design rules governing N-terminal extensions and identified successful extensions with high modification potential.


Asunto(s)
Bacteriófagos/metabolismo , Proteínas de la Cápside/biosíntesis , Nanopartículas/metabolismo , Ingeniería de Proteínas , Bacteriófagos/química , Proteínas de la Cápside/química , Estructura Molecular , Nanopartículas/química
19.
Plant Biotechnol J ; 17(9): 1751-1759, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30791210

RESUMEN

Porcine circovirus type 2 (PCV-2) is the main causative agent associated with a group of diseases collectively known as porcine circovirus-associated disease (PCAD). There is a significant economic strain on the global swine industry due to PCAD and the production of commercial PCV-2 vaccines is expensive. Plant expression systems are increasingly regarded as a viable technology to produce recombinant proteins for use as pharmaceutical agents and vaccines. However, successful production and purification of PCV-2 capsid protein (CP) from plants is an essential first step towards the goal of a plant-produced PCV-2 vaccine candidate. In this study, the PCV-2 CP was transiently expressed in Nicotiana benthamiana plants via agroinfiltration and PCV-2 CP was successfully purified using sucrose gradient ultracentrifugation. The CP self-assembled into virus-like particles (VLPs) resembling native virions and up to 6.5 mg of VLPs could be purified from 1 kg of leaf wet weight. Mice immunized with the plant-produced PCV-2 VLPs elicited specific antibody responses to PCV-2 CP. This is the first report describing the expression of PCV-2 CP in plants, the confirmation of its assembly into VLPs and the demonstration of their use to elicit a strong immune response in a mammalian model.


Asunto(s)
Proteínas de la Cápside/inmunología , Circovirus , Inmunogenicidad Vacunal , Vacunas de Partículas Similares a Virus/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Proteínas de la Cápside/biosíntesis , Ratones , Plantas Modificadas Genéticamente , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología , Porcinos , Nicotiana/genética , Nicotiana/metabolismo
20.
J Virol ; 92(22)2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30135129

RESUMEN

Human bocavirus 1 (HBoV1) encodes a genus-specific protein, NP1, which regulates viral alternative pre-mRNA processing. Similar to NP1 of the related bocavirus minute virus of canine (MVC), HBoV1 NP1 suppressed cleavage and polyadenylation of RNAs at the viral internal polyadenylation site (pA)p. HBoV1 (pA)p is a complex region. It contains 5 significant cleavage and polyadenylation sites, and NP1 was found to regulate only the three of these sites that are governed by canonical AAUAAA hexamer signals. HBoV1 NP1 also facilitated splicing of the upstream intron adjacent to (pA)p. Alternative polyadenylation and splicing of the upstream intron were independent of each other, functioned efficiently within an isolated transcription unit, and were responsive independent of NP1. Characterization of HBoV1 NP1 generalizes its function within the genus Bocaparvovirus, uncovers important differences, and provides important comparisons with MVC NP1 for mechanistic and evolutionary considerations.IMPORTANCE The Parvovirinae are small nonenveloped icosahedral viruses that are important pathogens in many animal species, including humans. The NP1 protein of human bocavirus 1 (HBoV1), similar to NP1 of the bocavirus minute virus of canine (MVC), regulates viral alternative RNA processing by both suppressing polyadenylation at an internal site, (pA)p, and facilitating splicing of an upstream adjacent intron. These effects allow both extension into the capsid gene and splicing of the viral pre-mRNA that correctly registers the capsid gene open reading frame. Characterization of HBoV1 NP1 generalizes this central mode of parvovirus gene regulation to another member of the bocavirus genus and uncovers both important similarities and differences in function compared to MVC NP1 that will be important for future comparative studies.


Asunto(s)
Empalme Alternativo/genética , Proteínas de la Cápside/genética , Regulación Viral de la Expresión Génica/genética , Bocavirus Humano/genética , ARN Viral/genética , Proteínas no Estructurales Virales/metabolismo , Cápside/metabolismo , Proteínas de la Cápside/biosíntesis , Línea Celular , Células HEK293 , Bocavirus Humano/metabolismo , Humanos , Poliadenilación , Replicación Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA