Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.215
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(20): e2321260121, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38722807

RESUMEN

Protein capsids are a widespread form of compartmentalization in nature. Icosahedral symmetry is ubiquitous in capsids derived from spherical viruses, as this geometry maximizes the internal volume that can be enclosed within. Despite the strong preference for icosahedral symmetry, we show that simple point mutations in a virus-like capsid can drive the assembly of unique symmetry-reduced structures. Starting with the encapsulin from Myxococcus xanthus, a 180-mer bacterial capsid that adopts the well-studied viral HK97 fold, we use mass photometry and native charge detection mass spectrometry to identify a triple histidine point mutant that forms smaller dimorphic assemblies. Using cryoelectron microscopy, we determine the structures of a precedented 60-mer icosahedral assembly and an unexpected 36-mer tetrahedron that features significant geometric rearrangements around a new interaction surface between capsid protomers. We subsequently find that the tetrahedral assembly can be generated by triple-point mutation to various amino acids and that even a single histidine point mutation is sufficient to form tetrahedra. These findings represent a unique example of tetrahedral geometry when surveying all characterized encapsulins, HK97-like capsids, or indeed any virus-derived capsids reported in the Protein Data Bank, revealing the surprising plasticity of capsid self-assembly that can be accessed through minimal changes in the protein sequence.


Asunto(s)
Proteínas de la Cápside , Cápside , Microscopía por Crioelectrón , Mutación Puntual , Cápside/metabolismo , Cápside/química , Cápside/ultraestructura , Proteínas de la Cápside/genética , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Myxococcus xanthus/genética , Myxococcus xanthus/metabolismo , Modelos Moleculares
2.
Bioorg Chem ; 147: 107415, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38701597

RESUMEN

The tobacco mosaic virus coat protein (TMV-CP) is indispensable for the virus's replication, movement and transmission, as well as for the host plant's immune system to recognize it. It constitutes the outermost layer of the virus particle, and serves as an essential component of the virus structure. TMV-CP is essential for initiating and extending viral assembly, playing a crucial role in the self-assembly process of Tobacco Mosaic Virus (TMV). This research employed TMV-CP as a primary target for virtual screening, from which a library of 43,417 compounds was sourced and SH-05 was chosen as the lead compound. Consequently, a series of α-amide phosphate derivatives were designed and synthesized, exhibiting remarkable anti-TMV efficacy. The synthesized compounds were found to be beneficial in treating TMV, with compound 3g displaying a slightly better curative effect than Ningnanmycin (NNM) (EC50 = 304.54 µg/mL) at an EC50 of 291.9 µg/mL. Additionally, 3g exhibited comparable inactivation activity (EC50 = 63.2 µg/mL) to NNM (EC50 = 67.5 µg/mL) and similar protective activity (EC50 = 228.9 µg/mL) to NNM (EC50 = 219.7 µg/mL). Microscale thermal analysis revealed that the binding of 3g (Kd = 4.5 ± 1.9 µM) to TMV-CP showed the same level with NNM (Kd = 5.5 ± 2.6 µM). Results from transmission electron microscopy indicated that 3g could disrupt the structure of TMV virus particles. The toxicity prediction indicated that 3g was low toxicity. Molecular docking showed that 3g interacted with TMV-CP through hydrogen bond, attractive charge interaction and π-Cation interaction. This research provided a novel α-amide phosphate structure target TMV-CP, which may help the discovery of new anti-TMV agents in the future.


Asunto(s)
Antivirales , Proteínas de la Cápside , Fosfatos , Virus del Mosaico del Tabaco , Virus del Mosaico del Tabaco/efectos de los fármacos , Antivirales/farmacología , Antivirales/química , Antivirales/síntesis química , Fosfatos/química , Fosfatos/farmacología , Relación Estructura-Actividad , Estructura Molecular , Proteínas de la Cápside/antagonistas & inhibidores , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Diseño de Fármacos , Pruebas de Sensibilidad Microbiana , Amidas/química , Amidas/farmacología , Amidas/síntesis química , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Simulación del Acoplamiento Molecular
3.
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
4.
Viruses ; 16(5)2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38793552

RESUMEN

The HIV-1 capsid (CA) protein forms the outer shell of the viral core that is released into the cytoplasm upon infection. CA binds various cellular proteins, including CPSF6, that direct HIV-1 integration into speckle-associated domains in host chromatin. Upon HIV-1 infection, CPSF6 forms puncta in the nucleus. Here, we characterised these CPSF6 puncta further in HeLa cells, T-cells and macrophages and confirmed that integration and reverse transcription are not required for puncta formation. Indeed, we found that puncta formed very rapidly after infection, correlating with the time that CA entered the nucleus. In aphidicolin-treated HeLa cells and macrophages, puncta were detected for the length of the experiment, suggesting that puncta are only lost upon cell division. CA still co-localised with CPSF6 puncta at the latest time points, considerably after the peak of reverse transcription and integration. Intriguingly, the number of puncta induced in macrophages did not correlate with the MOI or the total number of nuclear speckles present in each cell, suggesting that CA/CPSF6 is only directed to a few nuclear speckles. Furthermore, we found that CPSF6 already co-localised with nuclear speckles in uninfected T-cells, suggesting that HIV-1 promotes a natural behaviour of CPSF6.


Asunto(s)
VIH-1 , Macrófagos , Linfocitos T , Factores de Escisión y Poliadenilación de ARNm , VIH-1/fisiología , Humanos , Factores de Escisión y Poliadenilación de ARNm/metabolismo , Factores de Escisión y Poliadenilación de ARNm/genética , Linfocitos T/virología , Linfocitos T/metabolismo , Células HeLa , Macrófagos/virología , Macrófagos/metabolismo , Integración Viral , Núcleo Celular/metabolismo , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/genética , Infecciones por VIH/virología , Infecciones por VIH/metabolismo , Cápside/metabolismo
5.
Plant Signal Behav ; 19(1): 2358270, 2024 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-38796845

RESUMEN

Trans-acting small interfering RNAs (tasiRNAs) are 21-nt phased (phased siRNAs) resulting from successive DCL-catalyzed processing from the end of a double-stranded RNA substrate originating from the RDR of an AGO-catalyzed cleaved RNA at a micro RNA target site. Plant tasiRNAs have been synthesized to produce synthetic tasiRNAs (syn-tasiRNAs) targeting viral RNAs that confer viral resistance. In this study, we engineered syn-tasiRNAs to target potato virus Y (PVY) infection by replacing five native siRNAs of TAS1c with 210-bp fragments from the coat protein (CP) region of the PVY genome. The results showed that the transient expression of syn-tasiR-CPpvy2 in Nicotiana benthamiana (N. benthamiana) plants conferred antiviral resistance, supported by the absence of PVY infection symptoms and viral accumulation. This indicated that syn-tasiR-CPpvy2 successfully targeted and silenced the PVY CP gene, effectively inhibiting viral infection. syn-tasiR-CPpvy1 displayed attenuated symptoms and decreased viral accumulation in these plants However, severe symptoms of PVY infection and a similar amount of viral accumulation as the control were observed in plants expressing syn-tasiR-CPpvy3. syn-tasiR-CPpvy/pvx, which targets both PVY and potato virus X (PVX), was engineered using a single precursor. After the transient expression of syn-tasiR-CPpvy/pvx3 and syn-tasiR-CPpvy/pvx5 in N. benthamiana, the plants were resistant to both PVY and PVX. These results suggested that engineered syn-tasiRNAs could not only specifically induce antiviral resistance against one target virus but could also be designed for multi-targeted silencing of different viruses, thereby preventing complex virus infection in plants.


Asunto(s)
Proteínas de la Cápside , Resistencia a la Enfermedad , Nicotiana , Enfermedades de las Plantas , Potyvirus , ARN Interferente Pequeño , Nicotiana/virología , Nicotiana/genética , Nicotiana/inmunología , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/genética , Potyvirus/fisiología , Enfermedades de las Plantas/virología , Enfermedades de las Plantas/inmunología , Enfermedades de las Plantas/genética , Resistencia a la Enfermedad/genética , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/genética , Plantas Modificadas Genéticamente/virología
6.
Virology ; 595: 110093, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38692134

RESUMEN

Oncolytic virotherapy stands out as a burgeoning and promising therapeutic paradigm, harnessing the intrinsic cytotoxicity of oncolytic viruses for selective replication and dissemination within tumors. The primary mode of action revolves around the direct eradication of tumor cells. In our previous investigations, we formulated an oncolytic herpes simplex virus type 2 (OH2) and substantiated its anti-tumor efficacy both in vivo and in vitro. Subsequently, we embarked on a phase I/II clinical trial in China (NMPA, 2018L02743) and the USA (FDA, IND 27137) to assess OH2's safety, biodistribution, and anti-tumor activity as a standalone agent in patients with advanced solid tumors. In this investigation, our primary focus was to comprehend the influence of the major capsid protein VP5 of OH2 on its efficacy as an antitumor agent. Our findings underscore that the VP5 protein significantly amplifies OH2's oncolytic impact on A549 cells. Additionally, we observed that VP5 actively promotes the induction of apoptosis in A549 cells, both in vivo and in vitro. Through comprehensive transcriptional sequencing, we further authenticated that the VP5 protein triggers apoptosis-related signaling pathways and Gene Ontology (GO) terms in A549 cells. Moreover, we scrutinized differentially expressed genes in the p53-dependent apoptosis pathway and conducted meticulous in vitro validation of these genes. Subsequently, we delved deeper into unraveling the functional significance of the TP53I3 gene and conclusively affirmed that the VP5 protein induces apoptosis in A549 cells through the TP53I3 gene. These revelations illuminate the underlying mechanisms of OH2's antitumor activity and underscore the pivotal role played by the VP5 protein. The outcomes of our study harbor promising implications for the formulation of effective oncolytic virotherapy strategies in cancer treatment.


Asunto(s)
Apoptosis , Herpesvirus Humano 2 , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Células A549 , Viroterapia Oncolítica/métodos , Animales , Herpesvirus Humano 2/fisiología , Herpesvirus Humano 2/genética , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Vet Res ; 55(1): 63, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760810

RESUMEN

The maintenance of viral protein homeostasis depends on the interaction between host cell proteins and viral proteins. As a molecular chaperone, heat shock protein 70 (HSP70) has been shown to play an important role in viral infection. Our results showed that HSP70 can affect translation, replication, assembly, and release during the life cycle of duck hepatitis A virus type 1 (DHAV-1). We demonstrated that HSP70 can regulate viral translation by interacting with the DHAV-1 internal ribosome entry site (IRES). In addition, HSP70 interacts with the viral capsid proteins VP1 and VP3 and promotes their stability by inhibiting proteasomal degradation, thereby facilitating the assembly of DHAV-1 virions. This study demonstrates the specific role of HSP70 in regulating DHAV-1 replication, which are helpful for understanding the pathogenesis of DHAV-1 infection and provide additional information about the role of HSP70 in infection by different kinds of picornaviruses, as well as the interaction between picornaviruses and host cells.


Asunto(s)
Proteínas HSP70 de Choque Térmico , Virus de la Hepatitis del Pato , Sitios Internos de Entrada al Ribosoma , Replicación Viral , Virus de la Hepatitis del Pato/fisiología , Virus de la Hepatitis del Pato/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Animales , Proteínas Estructurales Virales/metabolismo , Proteínas Estructurales Virales/genética , Patos , Enfermedades de las Aves de Corral/virología , Infecciones por Picornaviridae/veterinaria , Infecciones por Picornaviridae/virología , Infecciones por Picornaviridae/metabolismo , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/genética , Hepatitis Viral Animal/virología , Hepatitis Viral Animal/metabolismo , Biosíntesis de Proteínas
8.
J Biosci ; 492024.
Artículo en Inglés | MEDLINE | ID: mdl-38783793

RESUMEN

A high level of disorder in many viral proteins is a direct consequence of their small genomes, which makes interaction with multiple binding partners a necessity for infection and pathogenicity. A segment of the flaviviral capsid protein (C), also known as the molecular recognition feature (MoRF), undergoes a disorder-toorder transition upon binding to several protein partners. To understand their role in pathogenesis, MoRFs were identified and their occurrence across different flaviviral capsids were studied. Despite lack of sequence similarities, docking studies of Cs with the host proteins indicate conserved interactions involving MoRFs across members of phylogenetic subclades. Additionally, it was observed from the protein-protein networks that some MoRFs preferentially bind proteins that are involved in specialized functions such as ribosome biogenesis. The findings point to the importance of MoRFs in the flaviviral life cycle, with important consequences for disease progression and suppression of the host immune system. Potentially, they might have impacted the way flaviviruses evolved to infect varied hosts using multiple vectors.


Asunto(s)
Proteínas de la Cápside , Flavivirus , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/química , Flavivirus/patogenicidad , Flavivirus/genética , Flavivirus/fisiología , Flavivirus/metabolismo , Filogenia , Humanos , Unión Proteica , Cápside/metabolismo , Cápside/química , Infecciones por Flavivirus/virología , Infecciones por Flavivirus/metabolismo , Simulación del Acoplamiento Molecular , Secuencia de Aminoácidos
9.
Sci Rep ; 14(1): 10253, 2024 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704431

RESUMEN

The tegument protein pp150 of Human Cytomegalovirus (HCMV) is known to be essential for the final stages of virus maturation and mediates its functions by interacting with capsid proteins. Our laboratory has previously identified the critical regions in pp150 important for pp150-capsid interactions and designed peptides similar in sequence to these regions, with a goal to competitively inhibit capsid maturation. Treatment with a specific peptide (PepCR2 or P10) targeted to pp150 conserved region 2 led to a significant reduction in murine CMV (MCMV) growth in cell culture, paving the way for in vivo testing in a mouse model of CMV infection. However, the general pharmacokinetic parameters of peptides, including rapid degradation and limited tissue and cell membrane permeability, pose a challenge to their successful use in vivo. Therefore, we designed a biopolymer-stabilized elastin-like polypeptide (ELP) fusion construct (ELP-P10) to enhance the bioavailability of P10. Antiviral efficacy and cytotoxic effects of ELP-P10 were studied in cell culture, and pharmacokinetics, biodistribution, and antiviral efficacy were studied in a mouse model of CMV infection. ELP-P10 maintained significant antiviral activity in cell culture, and this conjugation significantly enhanced P10 bioavailability in mouse tissues. The fluorescently labeled ELP-P10 accumulated to higher levels in mouse liver and kidneys as compared to the unconjugated P10. Moreover, viral titers from vital organs of MCMV-infected mice indicated a significant reduction of virus load upon ELP-P10 treatment. Therefore, ELP-P10 has the potential to be developed into an effective antiviral against CMV infection.


Asunto(s)
Antivirales , Infecciones por Citomegalovirus , Elastina , Muromegalovirus , Péptidos , Fosfoproteínas , Proteínas de la Matriz Viral , Animales , Elastina/química , Elastina/metabolismo , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/virología , Ratones , Antivirales/farmacología , Antivirales/farmacocinética , Antivirales/química , Péptidos/farmacología , Péptidos/química , Muromegalovirus/efectos de los fármacos , Humanos , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/química , Citomegalovirus/efectos de los fármacos , Cápside/metabolismo , Cápside/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/farmacocinética , Modelos Animales de Enfermedad , Polipéptidos Similares a Elastina
10.
Methods Mol Biol ; 2807: 153-161, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743227

RESUMEN

Recent evidence has shown that uncoating and reverse transcription precede nuclear import. These recent breakthroughs have been made possible through the development of innovative biochemical and imaging techniques. This method outlines the biochemical assay used for detecting the presence of the HIV-1 core in the nuclear compartment. In this procedure, human cells are infected with HIV-1NL4-3, with or without the inclusion of PF74, a small molecule that inhibits core entry into the nuclear compartment. Subsequently, cells are separated into cytosolic and nuclear fractions. To assess whether the capsid protein has reached the nuclear compartment, cytosolic and nuclear fractions are subjected to Western blot analysis, utilizing antibodies specific to the HIV-1 capsid protein p24. To validate the true origin of these fractions, Western blot analysis employing antibodies against cytosolic and nuclear markers are also performed. In summary, this assay provides a reliable and efficient means to detect the presence of the HIV-1 capsid protein in the nucleus during infection under various conditions.


Asunto(s)
Cápside , Núcleo Celular , Infecciones por VIH , VIH-1 , Humanos , Núcleo Celular/metabolismo , Infecciones por VIH/virología , Infecciones por VIH/metabolismo , Cápside/metabolismo , Proteína p24 del Núcleo del VIH/metabolismo , Proteína p24 del Núcleo del VIH/análisis , Proteínas de la Cápside/metabolismo , Western Blotting/métodos , Fenilalanina/metabolismo , Fenilalanina/análogos & derivados , Línea Celular
11.
Nat Commun ; 15(1): 3780, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710714

RESUMEN

Recombinant adeno-associated viruses (rAAVs) have emerged as promising gene therapy vectors due to their proven efficacy and safety in clinical applications. In non-human primates (NHPs), rAAVs are administered via suprachoroidal injection at a higher dose. However, high doses of rAAVs tend to increase additional safety risks. Here, we present a novel AAV capsid (AAVv128), which exhibits significantly enhanced transduction efficiency for photoreceptors and retinal pigment epithelial (RPE) cells, along with a broader distribution across the layers of retinal tissues in different animal models (mice, rabbits, and NHPs) following intraocular injection. Notably, the suprachoroidal delivery of AAVv128-anti-VEGF vector completely suppresses the Grade IV lesions in a laser-induced choroidal neovascularization (CNV) NHP model for neovascular age-related macular degeneration (nAMD). Furthermore, cryo-EM analysis at 2.1 Å resolution reveals that the critical residues of AAVv128 exhibit a more robust advantage in AAV binding, the nuclear uptake and endosome escaping. Collectively, our findings highlight the potential of AAVv128 as a next generation ocular gene therapy vector, particularly using the suprachoroidal delivery route.


Asunto(s)
Neovascularización Coroidal , Dependovirus , Terapia Genética , Vectores Genéticos , Epitelio Pigmentado de la Retina , Animales , Dependovirus/genética , Vectores Genéticos/genética , Vectores Genéticos/administración & dosificación , Terapia Genética/métodos , Ratones , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/virología , Neovascularización Coroidal/terapia , Neovascularización Coroidal/genética , Conejos , Humanos , Técnicas de Transferencia de Gen , Degeneración Macular/terapia , Degeneración Macular/genética , Degeneración Macular/patología , Modelos Animales de Enfermedad , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Transducción Genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ratones Endogámicos C57BL , Retina/metabolismo , Retina/virología , Masculino , Células HEK293
12.
Genes (Basel) ; 15(4)2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38674401

RESUMEN

The variable domain of a heavy-chain antibody (VHH) has the potential to be used to redirect the cell tropism of adenoviral vectors. Here, we attempted to establish platforms to simplify the screening of VHHs for their specific targeting function when being incorporated into the fiber of adenovirus. Both fowl adenovirus 4 (FAdV-4) and simian adenovirus 1 (SAdV-1) have two types of fiber, one of which is dispensable for virus propagation and is a proper site for VHH display. An intermediate plasmid, pMD-FAV4Fs, was constructed as the start plasmid for FAdV-4 fiber2 modification. Foldon from phage T4 fibritin, a trigger for trimerization, was employed to bridge the tail/shaft domain of fiber2 and VHHs against human CD16A, a key membrane marker of natural killer (NK) cells. Through one step of restriction-assembly, the modified fiber2 was transferred to the adenoviral plasmid, which was linearized and transfected to packaging cells. Five FAdV-4 viruses carrying the GFP gene were finally rescued and amplified, with three VHHs being displayed. One recombinant virus, FAdV4FC21-EG, could hardly transduce human 293 or Jurkat cells. In contrast, when it was used at a multiplicity of infection of 1000 viral particles per cell, the transduction efficiency reached 51% or 34% for 293 or Jurkat cells expressing exogenous CD16A. Such a strategy of fiber modification was transplanted to the SAdV-1 vector to construct SAdV1FC28H-EG, which moderately transduced primary human NK cells while the parental virus transduced none. Collectively, we reformed the strategy of integrating VHH to fiber and established novel platforms for screening VHHs to construct adenoviral vectors with a specific tropism.


Asunto(s)
Vectores Genéticos , Tropismo Viral , Humanos , Vectores Genéticos/genética , Células HEK293 , Cadenas Pesadas de Inmunoglobulina/genética , Aviadenovirus/genética , Aviadenovirus/inmunología , Animales , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/metabolismo
13.
J Virol ; 98(5): e0006824, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38661364

RESUMEN

The portal protein of tailed bacteriophage plays essential roles in various aspects of capsid assembly, motor assembly, genome packaging, connector formation, and infection processes. After DNA packaging is complete, additional proteins are assembled onto the portal to form the connector complex, which is crucial as it bridges the mature head and tail. In this study, we report high-resolution cryo-electron microscopy (cryo-EM) structures of the portal vertex from bacteriophage lambda in both its prohead and mature virion states. Comparison of these structures shows that during head maturation, in addition to capsid expansion, the portal protein undergoes conformational changes to establish interactions with the connector proteins. Additionally, the independently assembled tail undergoes morphological alterations at its proximal end, facilitating its connection to the head-tail joining protein and resulting in the formation of a stable portal-connector-tail complex. The B-DNA molecule spirally glides through the tube, interacting with the nozzle blade region of the middle-ring connector protein. These insights elucidate a mechanism for portal maturation and DNA translocation within the phage lambda system. IMPORTANCE: The tailed bacteriophages possess a distinct portal vertex that consists of a ring of 12 portal proteins associated with a 5-fold capsid shell. This portal protein is crucial in multiple stages of virus assembly and infection. Our research focused on examining the structures of the portal vertex in both its preliminary prohead state and the fully mature virion state of bacteriophage lambda. By analyzing these structures, we were able to understand how the portal protein undergoes conformational changes during maturation, the mechanism by which it prevents DNA from escaping, and the process of DNA spirally gliding.


Asunto(s)
Bacteriófago lambda , Proteínas de la Cápside , Cápside , Microscopía por Crioelectrón , Virión , Ensamble de Virus , Bacteriófago lambda/fisiología , Bacteriófago lambda/genética , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/química , Virión/metabolismo , Virión/ultraestructura , Cápside/metabolismo , Cápside/ultraestructura , ADN Viral/genética , ADN Viral/metabolismo , Empaquetamiento del ADN , Modelos Moleculares , Conformación Proteica
14.
Sci Total Environ ; 930: 172307, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38599392

RESUMEN

For canine parvovirus -2 (CPV-2), a zoonotic virus capable of cross-species transmission in animals, the amino acid changes of capsid protein VP2 are key factors when binding to other species' transferrin receptors (TfR). CPV-2 variants can spread from felines and canines, for example, to Carnivora, Artiodactyla, and Pholidota species, and CPV-2c variants are essential to spread from Carnivora to Artiodactyla and Pholidota species in particular. In our study, a CPV-2a variant maintained a relatively stable trend, and the proportion of CPV-2c gradually rose from 1980 to 2021. The VP2 amino acid sequence analysis showed that five amino acid mutations at 426E/D, 305H/D, and 297S may be necessary for the virus to bind to different host receptors. Meanwhile, receptor-binding loop regions and amino acid sites 87 L, 93 N, 232I, and 305Y were associated with CPV-2 cross-species transmission. The homology of TfRs in different hosts infected with CPV-2 ranged from 77.2 % to 99.0 %, and from pig to feline, canine, and humans was 80.7 %, 80.4 %, and 77.2 %, respectively. The amino acid residues of TfRs involved in the viral binding in those hosts are highly conserved, which suggests that CPV-2 may be capable of pig-to-human transmission. Our analysis of the origin, evolutionary trend, cross-species transmission dynamics, and genetic characteristics of CPV-2 when binding to host receptors provides a theoretical basis for further research on CPV-2's mechanism of cross-species transmission and for establishing an early warning and monitoring mechanism for the possible threat of CPV-2 to animal-human public security.


Asunto(s)
Parvovirus Canino , Parvovirus Canino/genética , Animales , Perros , Humanos , Infecciones por Parvoviridae/veterinaria , Infecciones por Parvoviridae/transmisión , Gatos , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/genética , Zoonosis/virología , Zoonosis/transmisión , Receptores de Transferrina/metabolismo , Receptores de Transferrina/genética
15.
J Virol ; 98(5): e0035024, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38591900

RESUMEN

Feline calicivirus (FCV) is one of the few members of the Caliciviridae family that grows well in cell lines and, therefore, serves as a surrogate to study the biology of other viruses in the family. Conley et al. (14) demonstrated that upon the receptor engagement to the capsid, FCV VP2 forms a portal-like assembly, which might provide a channel for RNA release. However, the process of calicivirus RNA release is not yet fully understood. Our findings suggest that the separation of the FCV capsid from its genome RNA (gRNA) occurs rapidly in the early endosomes of infected cells. Using a liposome model decorated with the FCV cell receptor fJAM-A, we demonstrate that FCV releases its gRNA into the liposomes by penetrating membranes under low pH conditions. Furthermore, we found that VP2, which is rich in hydrophobic residues at its N-terminus, functions as the pore-forming protein. When we substituted the VP2 N-terminal hydrophobic residues, the gRNA release efficacy of the FCV mutants decreased. In conclusion, our results suggest that in the acidic environment of early endosomes, FCV VP2 functions as the pore-forming protein to mediate gRNA release into the cytoplasm of infected cells. This provides insight into the mechanism of calicivirus genome release.IMPORTANCEResearch on the biology and pathogenicity of certain caliciviruses, such as Norovirus and Sapovirus, is hindered by the lack of easy-to-use cell culture system. Feline calicivirus (FCV), which grows effectively in cell lines, is used as a substitute. At present, there is limited understanding of the genome release mechanism in caliciviruses. Our findings suggest that FCV uses VP2 to pierce the endosome membrane for genome release and provide new insights into the calicivirus gRNA release mechanism.


Asunto(s)
Calicivirus Felino , Proteínas de la Cápside , Endosomas , Genoma Viral , ARN Viral , Calicivirus Felino/genética , Calicivirus Felino/metabolismo , Calicivirus Felino/fisiología , Gatos , Endosomas/virología , Endosomas/metabolismo , Animales , ARN Viral/metabolismo , ARN Viral/genética , Línea Celular , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/genética , Infecciones por Caliciviridae/virología , Infecciones por Caliciviridae/metabolismo , Liberación del Virus , Cápside/metabolismo , Liposomas/metabolismo
16.
J Virol ; 98(5): e0019724, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38593321

RESUMEN

Noroviruses are major causative agents of acute nonbacterial gastroenteritis in humans. There are neither antiviral therapeutic agents nor vaccines for noroviruses at this time. To evaluate the potential usefulness of two previously isolated human monoclonal antibody fragments, CV-1A1 and CV-2F5, we first conducted a single-particle analysis to determine the cryo-electron microscopy structure of virus-like particles (VLPs) from the genogroup I genotype 4 (GI.4) Chiba strain uniformly coated with CV-1A1 fragments. The results revealed that the GI.4-specific CV-1A1 antibody bound to the P2 subdomain, in which amino acids are less conserved and variable. Interestingly, a part of the CV-1A1 intrudes into the histo-blood group antigen-binding site, suggesting that this antibody might exert neutralizing activity. Next, we determined the crystal structure of the protruding (P) domain of the capsid protein in the complex form with the CV-2F5 antibody fragment. Consistent with the cross-reactivity, the CV-2F5 bound to the P1 subdomain, which is rich in amino acids conserved among the GI strains, and moreover induced a disruption of Chiba VLPs. These results suggest that the broadly reactive CV-2F5 antibody can be used as both a universal detection reagent and an antiviral drug for GI noroviruses. IMPORTANCE: We conducted the structural analyses of the VP1 protein from the GI.4 Chiba norovirus to identify the binding sites of the previously isolated human monoclonal antibodies CV-1A1 and CV-2F5. The cryo-electron microscopy of the Chiba virus-like particles (VLPs) complexed with the Fv-clasp forms of GI.4-specific CV-1A1 revealed that this antibody binds to the highly variable P2 subdomain, suggesting that this antibody may have neutralizing ability against the GI.4 strains. X-ray crystallography revealed that the CV-2F5 antibody bound to the P1 subdomain, which is rich in conserved amino acids. This result is consistent with the ability of the CV-2F5 antibody to react with a wide variety of GI norovirus strains. It is also found that the CV-2F5 antibody caused a disruption of VLPs. Our findings, together with previous reports on the structures of VP1 proteins and VLPs, are expected to open a path for the structure-based development of antivirals and vaccines against norovirus disease.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Antivirales , Proteínas de la Cápside , Microscopía por Crioelectrón , Norovirus , Norovirus/inmunología , Microscopía por Crioelectrón/métodos , Humanos , Anticuerpos Monoclonales/inmunología , Cristalografía por Rayos X , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Sitios de Unión , Anticuerpos Antivirales/inmunología , Anticuerpos Neutralizantes/inmunología , Modelos Moleculares
17.
Viruses ; 16(4)2024 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-38675835

RESUMEN

Many protein expression systems are primarily utilised to produce a single, specific recombinant protein. In contrast, most biological processes such as virus assembly rely upon a complex of several interacting proteins rather than the activity of a sole protein. The high complexity of the baculovirus genome, coupled with a multiphase replication cycle incorporating distinct transcriptional steps, made it the ideal system to manipulate for high-level expression of a single, or co-expression of multiple, foreign proteins within a single cell. We have developed and utilised a series of recombinant baculovirus systems to unravel the sequential assembly process of a complex non-enveloped model virus, bluetongue virus (BTV). The high protein yields expressed by the baculovirus system not only facilitated structure-function analysis of each viral protein but were also advantageous to crystallography studies and supported the first atomic-level resolution of a recombinant viral protein, the major BTV capsid protein. Further, the formation of recombinant double-shelled virus-like particles (VLPs) provided insights into the structure-function relationships among the four major structural proteins of the BTV whilst also representing a potential candidate for a viral vaccine. The baculovirus multi-gene expression system facilitated the study of structurally complex viruses (both non-enveloped and enveloped viruses) and heralded a new generation of viral vaccines.


Asunto(s)
Baculoviridae , Proteínas Recombinantes , Baculoviridae/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Animales , Expresión Génica , Virus de la Lengua Azul/genética , Vectores Genéticos/genética , Ensamble de Virus , Proteínas Virales/genética , Proteínas Virales/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/química
18.
Viruses ; 16(4)2024 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-38675855

RESUMEN

The foot-and-mouth disease virus is a highly contagious and economically devastating virus of cloven-hooved animals, including cattle, buffalo, sheep, and goats, causing reduced animal productivity and posing international trade restrictions. For decades, chemically inactivated vaccines have been serving as the most effective strategy for the management of foot-and-mouth disease. Inactivated vaccines are commercially produced in cell culture systems, which require successful propagation and adaptation of field isolates, demanding a high cost and laborious time. Cell culture adaptation is chiefly indebted to amino acid substitutions in surface-exposed capsid proteins, altering the necessity of RGD-dependent receptors to heparan sulfate macromolecules for virus binding. Several amino acid substations in VP1, VP2, and VP3 capsid proteins of FMDV, both at structural and functional levels, have been characterized previously. This literature review combines frequently reported amino acid substitutions in virus capsid proteins, their critical roles in virus adaptation, and functional characterization of the substitutions. Furthermore, this data can facilitate molecular virologists to develop new vaccine strains against the foot-and-mouth disease virus, revolutionizing vaccinology via reverse genetic engineering and synthetic biology.


Asunto(s)
Sustitución de Aminoácidos , Proteínas de la Cápside , Virus de la Fiebre Aftosa , Fiebre Aftosa , Tropismo Viral , Virus de la Fiebre Aftosa/genética , Virus de la Fiebre Aftosa/metabolismo , Animales , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/química , Fiebre Aftosa/virología , Receptores Virales/metabolismo , Receptores Virales/genética , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/metabolismo , Técnicas de Cultivo de Célula
19.
Viruses ; 16(4)2024 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-38675928

RESUMEN

The higher-order structure (HOS) is a critical quality attribute of recombinant adeno-associated viruses (rAAVs). Evaluating the HOS of the entire rAAV capsid is challenging because of the flexibility and/or less folded nature of the VP1 unique (VP1u) and VP1/VP2 common regions, which are structural features essential for these regions to exert their functions following viral infection. In this study, hydrogen/deuterium exchange mass spectrometry (HDX-MS) was used for the structural analysis of full and empty rAAV8 capsids. We obtained 486 peptides representing 85% sequence coverage. Surprisingly, the VP1u region showed rapid deuterium uptake even though this region contains the phospholipase A2 domain composed primarily of α-helices. The comparison of deuterium uptake between full and empty capsids showed significant protection from hydrogen/deuterium exchange in the full capsid at the channel structure of the 5-fold symmetry axis. This corresponds to cryo-electron microscopy studies in which the extended densities were observed only in the full capsid. In addition, deuterium uptake was reduced in the VP1u region of the full capsid, suggesting the folding and/or interaction of this region with the encapsidated genome. This study demonstrated HDX-MS as a powerful method for probing the structure of the entire rAAV capsid.


Asunto(s)
Proteínas de la Cápside , Cápside , Dependovirus , Dependovirus/química , Dependovirus/genética , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/genética , Cápside/química , Cápside/metabolismo , Serogrupo , Medición de Intercambio de Deuterio , Espectrometría de Masas de Intercambio de Hidrógeno-Deuterio/métodos , Humanos , Deuterio/química , Espectrometría de Masas , Microscopía por Crioelectrón , Modelos Moleculares
20.
Cell Host Microbe ; 32(4): 441-442, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38604120

RESUMEN

The size of the nuclear pore should, in principle, prevent HIV-1 entry. However, HIV-1 capsid is able to gain nuclear pore entry. In a recent issue of Nature, Fu et al. and Dickson et al. demonstrate that the HIV-1 capsid mimics the nuclear transport protein karyopherins to access host nuclei.


Asunto(s)
Infecciones por VIH , Poro Nuclear , Humanos , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Infecciones por VIH/metabolismo , Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA