Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Biochem Biophys Res Commun ; 534: 980-987, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33131770

RESUMEN

Virosomes as membranous vesicles with viral fusion protein in their membrane are versatile vehicles for cargo delivery. The vesicular stomatitis virus glycoprotein (VSV-G) is a common fusogenic protein used in virosome preparation. This glycoprotein has been used in liposomal systems so far, but in this study, we have tried to use the niosomal form instead of liposome for. Niosomes are vesicular systems composed of non-ionic surfactants. Niosomes were constructed by the thin-film hydration method. VSV-G gene in pMD2.G plasmid was expressed in the HEK293T cell line and then was reconstituted in the niosome bilayer. The formation of niosomal virosomes was confirmed with different methods such as SDS-PAGE gel, western blotting, and transmission electron microscopy (TEM). The efficiency of niosomal virosome was investigated with the pmCherry reporter gene. SDS-PAGE and western blotting proved the expression and successful insertion of protein into the bilayer. The TEM images showed the spike projection of VSV-G on the surface of niosomes. The transfection results showed high efficiency of niosomal virosomes as a novel carrier. This report has verified that niosome could be used as an efficient bilayer instead of liposome to construct virosomes.


Asunto(s)
Técnicas de Transferencia de Gen , Genes Reporteros , Glicoproteínas/genética , Vesiculovirus/genética , Proteínas Virales/genética , Virosomas/genética , Expresión Génica , Glicoproteínas/química , Células HEK293 , Humanos , Liposomas/química , Plásmidos/administración & dosificación , Plásmidos/genética , Transfección , Estomatitis Vesicular/virología , Vesiculovirus/química , Proteínas Virales/química , Virosomas/química
2.
J Microencapsul ; 38(5): 263-275, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33719838

RESUMEN

AIM: The present work involves encapsulation of herbal drug nanocurcumin into the virosomes and compared with a liposome in terms of their in vitro anti-proliferative, anti-inflammatory, and anti-migratory efficacy. METHODS: The anti-proliferative, anti-inflammatory, and anti-migratory efficacy of virosome and liposome were compared in HepG2 and CaCo2 cells by using MTT, Nitric oxide scavenging, and Wound healing assay, respectively. RESULTS: Size of the optimised NC-Virosome and NC-Liposome was 70.06 ± 1.63 and 265.80 ± 1.64 nm, respectively. The prepared NC-Virosome can be stored at -4 °C up to six months. The drug encapsulation efficiency of NC-Virosome and NC-Liposome was found to be 84.66 ± 1.67 and 62.15 ± 1.75% (w/w). The evaluated minimum inhibitory concentration (IC50 value) for NC-Virosome was 102.7 µg/ml and 108.1 µg/ml, while NC-Liposome showed 129.2 µg/ml and 160.1 µg/ml for HepG2 and CaCo2 cells, respectively. Morphological examination depicts detachment of the cells from substratum after exposure to NC-Virosome for 48 h. CONCLUSION: The prepared NC-Virosome provides remarkable in vitro efficacy in both the cell lines with site-specific drug-targeting potential as compared to the liposome, results proved its potential as a drug delivery vehicle for future therapy with reduced toxicity.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Liposomas/química , Virosomas/química , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/uso terapéutico , Antineoplásicos Fitogénicos/administración & dosificación , Células CACO-2 , Movimiento Celular/efectos de los fármacos , Curcumina/administración & dosificación , Curcumina/uso terapéutico , Sistemas de Liberación de Medicamentos , Excipientes , Células Hep G2 , Humanos , Pruebas de Sensibilidad Microbiana , Neoplasias/tratamiento farmacológico , Óxido Nítrico/química , Sales de Tetrazolio , Tiazoles , Cicatrización de Heridas/efectos de los fármacos
3.
J Virol ; 91(15)2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28515293

RESUMEN

Virus-like vesicles (VLVs) are membrane-enclosed vesicles that resemble native enveloped viruses in organization but lack the viral capsid and genome. During the productive infection of tumor-associated gammaherpesviruses, both virions and VLVs are produced and are released into the extracellular space. However, studies of gammaherpesvirus-associated VLVs have been largely restricted by the technical difficulty of separating VLVs from mature virions. Here we report a strategy of selectively isolating VLVs by using a Kaposi's sarcoma-associated herpesvirus (KSHV) mutant that is defective in small capsid protein and is unable to produce mature virions. Using mass spectrometry analysis, we found that VLVs contained viral glycoproteins required for cellular entry, as well as tegument proteins involved in regulating lytic replication, but lacked capsid proteins. Functional analysis showed that VLVs induced the expression of the viral lytic activator RTA, initiating KSHV lytic gene expression. Furthermore, employing RNA sequencing, we performed a genomewide analysis of cellular responses triggered by VLVs and found that PRDM1, a master regulator in cell differentiation, was significantly upregulated. In the context of KSHV replication, we demonstrated that VLV-induced upregulation of PRDM1 was necessary and sufficient to reactivate KSHV by activating its RTA promoter. In sum, our study systematically examined the composition of VLVs and demonstrated their biological roles in manipulating host cell responses and facilitating KSHV lytic replication.IMPORTANCE Cells lytically infected with tumor-associated herpesviruses produce a high proportion of virus-like vesicles (VLVs). The composition and function of VLVs have not been well defined, largely due to the inability to efficiently isolate VLVs that are free of virions. Using a cell system capable of establishing latent KSHV infection and robust reactivation, we successfully isolated VLVs from a KSHV mutant defective in the small capsid protein. We quantitatively analyzed proteins and microRNAs in VLVs and characterized the roles of VLVs in manipulating host cells and facilitating viral infection. More importantly, we demonstrated that by upregulating PRDM1 expression, VLVs triggered differentiation signaling in targeted cells and facilitated viral lytic infection via activation of the RTA promoter. Our study not only demonstrates a new strategy for isolating VLVs but also shows the important roles of KSHV-associated VLVs in intercellular communication and the viral life cycle.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/fisiología , Interacciones Huésped-Patógeno , Proteínas Represoras/biosíntesis , Transducción de Señal , Virosomas/química , Replicación Viral , Diferenciación Celular , Línea Celular , Herpesvirus Humano 8/química , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Transactivadores/metabolismo , Regulación hacia Arriba
4.
Biomacromolecules ; 19(9): 3738-3746, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30092631

RESUMEN

Viral protein cages, with their regular and programmable architectures, are excellent platforms for the development of functional nanomaterials. The ability to transform a virus into a material with intended structure and function relies on the existence of a well-understood model system, a noninfectious virus-like particle (VLP) counterpart. Here, we study the factors important to the ability of P22 VLP to retain or release various protein cargo molecules depending on the nature of the cargo, the capsid morphology, and the environmental conditions. Because the interaction between the internalized scaffold protein (SP) and the capsid coat protein (CP) is noncovalent, we have studied the efficiency with which a range of SP variants can dissociate from the interior of different P22 VLP morphologies and exit by traversing the porous capsid. Understanding the types of cargos that are either retained or released from the P22 VLP will aid in the rational design of functional nanomaterials.


Asunto(s)
Cápside/química , Virosomas/química , Proteínas de la Cápside/química , Liberación de Fármacos , Proteínas del Núcleo Viral/química
5.
J Virol ; 90(3): 1169-77, 2016 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-26537684

RESUMEN

UNLABELLED: Chikungunya virus is a positive-stranded RNA alphavirus. Structures of chikungunya virus-like particles in complex with strongly neutralizing antibody Fab fragments (8B10 and 5F10) were determined using cryo-electron microscopy and X-ray crystallography. By fitting the crystallographically determined structures of these Fab fragments into the cryo-electron density maps, we show that Fab fragments of antibody 8B10 extend radially from the viral surface and block receptor binding on the E2 glycoprotein. In contrast, Fab fragments of antibody 5F10 bind the tip of the E2 B domain and lie tangentially on the viral surface. Fab 5F10 fixes the B domain rigidly to the surface of the virus, blocking exposure of the fusion loop on glycoprotein E1 and therefore preventing the virus from becoming fusogenic. Although Fab 5F10 can neutralize the wild-type virus, it can also bind to a mutant virus without inhibiting fusion or attachment. Although the mutant virus is no longer able to propagate by extracellular budding, it can, however, enter the next cell by traveling through junctional complexes without being intercepted by a neutralizing antibody to the wild-type virus, thus clarifying how cell-to-cell transmission can occur. IMPORTANCE: Alphaviral infections are transmitted mainly by mosquitoes. Chikungunya virus (CHIKV), which belongs to the Alphavirus genus, has a wide distribution in the Old World that has expanded in recent years into the Americas. There are currently no vaccines or drugs against alphaviral infections. Therefore, a better understanding of CHIKV and its associated neutralizing antibodies will aid in the development of effective treatments.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Virus Chikungunya/inmunología , Virus Chikungunya/ultraestructura , Virosomas/inmunología , Virosomas/ultraestructura , Virus Chikungunya/química , Virus Chikungunya/fisiología , Microscopía por Crioelectrón , Cristalografía por Rayos X , Humanos , Fragmentos Fab de Inmunoglobulinas/metabolismo , Modelos Moleculares , Unión Proteica , Virosomas/química , Acoplamiento Viral
6.
J Liposome Res ; 27(2): 83-89, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26981843

RESUMEN

Insect-derived cell lines are used extensively to produce recombinant proteins because they are capable of performing a range of post-translational modifications. Due to their significance in biotechnological applications, various methods have been developed to transfect them. In this study, we introduce a virosome constructed from vesicular stomatitis virus (VSV) as a new delivery system for sf9 cells. We labeled these VSV virosomes by fluorescent probe Rhodamine B chloride (R18). By fluorescence microscope observation and conducting a fusion assay, we confirmed the uptake of VSV virosomes via endocytosis by sf9 cells and their fusion with the endosomal membrane. Moreover, we incubated cationic VSV virosomes with a GFP-expressing bacmid and transfected sf9 cells, after 24 h some cells expressed GFP indicating the ability of VSV virosomes to deliver heterologous DNA to these cells. This is the first report of a virosome-based delivery system introduced for an insect cell line.


Asunto(s)
Técnicas de Transferencia de Gen , Virus de la Estomatitis Vesicular Indiana/química , Animales , Cationes/química , Células Cultivadas , Células Sf9 , Spodoptera , Virosomas/química
7.
J Virol ; 90(5): 2306-15, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26656716

RESUMEN

UNLABELLED: Human parainfluenza virus type 3 (HPIV3) belongs to the Paramyxoviridae family. Its three internal viral proteins, the nucleoprotein (N), the phosphoprotein (P), and the polymerase (L), form the ribonucleoprotein (RNP) complex, which encapsidates the viral genome and associates with the matrix protein (M) for virion assembly. We previously showed that the M protein expressed alone is sufficient to assemble and release virus-like particles (VLPs) and a mutant with the L305A point mutation in the M protein (ML305A) has a VLP formation ability similar to that of wild-type M protein. In addition, recombinant HPIV3 (rHPIV3) containing the ML305A mutation (rHPIV3-ML305A) could be successfully recovered. In the present study, we found that the titer of rHPIV3-ML305A was at least 10-fold lower than the titer of rHPIV3. Using VLP incorporation and coimmunoprecipitation assays, we found that VLPs expressing the M protein (M-VLPs) can efficiently incorporate N and P via an N-M or P-M interaction and ML305A-VLPs had an ability to incorporate P via a P-M interaction similar to that of M-VLPs but were unable to incorporate N and no longer interacted with N. Furthermore, we found that the incorporation of P into ML305A-VLPs but not M-VLPs was inhibited in the presence of N. In addition, we provide evidence that the C-terminal region of P is involved in its interaction with both N and M and N binding to the C-terminal region of P inhibits the incorporation of P into ML305A-VLPs. Our findings provide new molecular details to support the idea that the N-M interaction and not the P-M interaction is critical for packaging N and P into infectious viral particles. IMPORTANCE: Human parainfluenza virus type 3 (HPIV3) is a nonsegmented, negative-sense, single-stranded RNA virus that belongs to the Paramyxoviridae family and can cause lower respiratory tract infections in infants and young children as well as elderly or immunocompromised individuals. However, no effective vaccine has been developed or licensed. We used virus-like particle (VLP) incorporation and coimmunoprecipitation assays to determine how the M protein assembles internal viral proteins. We demonstrate that both nucleoprotein (N) and phosphoprotein (P) can incorporate into M-VLPs and N inhibits the M-P interaction via the binding of N to the C terminus of P. We also provide additional evidence that the N-M interaction but not the P-M interaction is critical for the regulation of HPIV3 assembly. Our studies provide a more complete characterization of HPIV3 virion assembly and substantiation that N interaction with M regulates internal viral organization.


Asunto(s)
Nucleoproteínas/metabolismo , Virus de la Parainfluenza 3 Humana/fisiología , Proteínas de la Matriz Viral/metabolismo , Ensamble de Virus , Western Blotting , Línea Celular , Humanos , Inmunoprecipitación , Unión Proteica , Multimerización de Proteína , Virosomas/química , Virosomas/metabolismo
8.
J Virol ; 90(5): 2664-75, 2015 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-26699644

RESUMEN

UNLABELLED: The capsid protein (VP1) of all caliciviruses forms an icosahedral particle with two principal domains, shell (S) and protruding (P) domains, which are connected via a flexible hinge region. The S domain forms a scaffold surrounding the nucleic acid, while the P domains form a homodimer that interacts with receptors. The P domain is further subdivided into two subdomains, termed P1 and P2. The P2 subdomain is likely an insertion in the P1 subdomain; consequently, the P domain is divided into the P1-1, P2, and P1-2 subdomains. In order to investigate capsid antigenicity, N-terminal (N-term)/S/P1-1 and P2/P1-2 were switched between two sapovirus genotypes GI.1 and GI.5. The chimeric VP1 constructs were expressed in insect cells and were shown to self-assemble into virus-like particles (VLPs) morphologically similar to the parental VLPs. Interestingly, the chimeric VLPs had higher levels of cross-reactivities to heterogeneous antisera than the parental VLPs. In order to better understand the antigenicity from a structural perspective, we determined an intermediate-resolution (8.5-Å) cryo-electron microscopy (cryo-EM) structure of a chimeric VLP and developed a VP1 homology model. The cryo-EM structure revealed that the P domain dimers were raised slightly (∼5 Å) above the S domain. The VP1 homology model allowed us predict the S domain (67-229) and P1-1 (229-280), P2 (281-447), and P1-2 (448-567) subdomains. Our results suggested that the raised P dimers might expose immunoreactive S/P1-1 subdomain epitopes. Consequently, the higher levels of cross-reactivities with the chimeric VLPs resulted from a combination of GI.1 and GI.5 epitopes. IMPORTANCE: We developed sapovirus chimeric VP1 constructs and produced the chimeric VLPs in insect cells. We found that both chimeric VLPs had a higher level of cross-reactivity against heterogeneous VLP antisera than the parental VLPs. The cryo-EM structure of one chimeric VLP (Yokote/Mc114) was solved to 8.5-Å resolution. A homology model of the VP1 indicated for the first time the putative S and P (P1-1, P2, and P1-2) domains. The overall structure of Yokote/Mc114 contained features common among other caliciviruses. We showed that the P2 subdomain was mainly involved in the homodimeric interface, whereas a large gap between the P1 subdomains had fewer interactions.


Asunto(s)
Microscopía por Crioelectrón , Sapovirus/química , Sapovirus/ultraestructura , Virosomas/química , Virosomas/ultraestructura , Secuencia de Aminoácidos , Anticuerpos Antivirales/inmunología , Antígenos Virales/genética , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Antígenos Virales/ultraestructura , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/ultraestructura , Reacciones Cruzadas , Datos de Secuencia Molecular , Multimerización de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Recombinación Genética , Sapovirus/genética , Sapovirus/inmunología , Virosomas/genética , Virosomas/inmunología
9.
J Virol ; 88(15): 8386-96, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24829339

RESUMEN

UNLABELLED: Interaction between E and prM proteins in flavivirus-infected cells is a major factor for virus-like particle (VLP) production. The prM helical (prM-H) domain is topologically close to and may interact with domain II of the E protein (EDII). In this study, we investigated prM-H domain amino acid residues facing Japanese encephalitis virus EDII using site-directed mutagenesis to determine their roles in prM-E interaction and VLP production. Our results indicate that negatively charged prM-E125 residue at the prM-H domain affected VLP production via one or more interactions with positively charged E-K93 and E-H246 residues at EDII. Exchanges of oppositely charged residue side chains at prM-E125/E-K93 and prM-E125/E-H246 are recoverable for VLP production. The prM-E125 and E-H246 residues are conserved and that the positive charge of the E-K93 residue is preserved in different flavivirus groups. These findings suggest that the electrostatic attractions of prM-E125, E-K93, and E-H246 residues are important to flavivirus VLP production and that inhibiting these interactions is a potential strategy for blocking flavivirus infections. IMPORTANCE: Molecular interaction between E and prM proteins of Japanese encephalitis virus is a major driving force for virus-like particle (VLP) production. The current high-resolution structures available for prM-E complexes do not include the membrane proximal stem region of prM. The prM stem region contains an N-terminal loop and a helix domain (prM-H). Since the prM-H domain is topologically close to domain II of the E protein (EDII), this study was to determine molecular interactions between the prM-H domain and EDII. We found that the molecular interactions between prM-E125 residue and positively charged E-K93 and E-H246 residues at EDII are critical for VLP production. More importantly, the prM-E125 and E-H246 residues are conserved and the positive charge of the E-K93 residue is preserved in different flavivirus groups. Our findings help refine the structure and molecular interactions on the flavivirus surface and reveal a potential strategy for blocking flavivirus infections by inhibiting these electrostatic interactions.


Asunto(s)
Aminoácidos/metabolismo , Virus de la Encefalitis Japonesa (Especie)/fisiología , Glicoproteínas de Membrana/metabolismo , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Proteínas del Envoltorio Viral/metabolismo , Ensamble de Virus , Animales , Línea Celular , Análisis Mutacional de ADN , Virus de la Encefalitis Japonesa (Especie)/química , Humanos , Glicoproteínas de Membrana/genética , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas del Envoltorio Viral/genética , Virosomas/química
10.
J Virol ; 85(10): 4691-7, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21367906

RESUMEN

Bombyx mori densovirus 1 (BmDNV-1), a major pathogen of silkworms, causes significant losses to the silk industry. The structure of the recombinant BmDNV-1 virus-like particle has been determined at 3.1-Å resolution using X-ray crystallography. It is the first near-atomic-resolution structure of a virus-like particle within the genus Iteravirus. The particles consist of 60 copies of the 55-kDa VP3 coat protein. The capsid protein has a ß-barrel "jelly roll" fold similar to that found in many diverse icosahedral viruses, including archaeal, bacterial, plant, and animal viruses, as well as other parvoviruses. Most of the surface loops have little structural resemblance to other known parvovirus capsid proteins. In contrast to vertebrate parvoviruses, the N-terminal ß-strand of BmDNV-1 VP3 is positioned relative to the neighboring 2-fold related subunit in a "domain-swapped" conformation, similar to findings for other invertebrate parvoviruses, suggesting domain swapping is an evolutionarily conserved structural feature of the Densovirinae.


Asunto(s)
Bombyx/virología , Densovirinae/química , Animales , Cristalografía por Rayos X , Sustancias Macromoleculares/química , Modelos Moleculares , Estructura Cuaternaria de Proteína , Virosomas/química
11.
J Virol ; 84(18): 9070-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20610725

RESUMEN

HIV-1 is known to package several small cellular RNAs in addition to its genome. Previous work consistently demonstrated that the host structural RNA 7SL is abundant in HIV-1 virions but has yielded conflicting results regarding whether 7SL is present in minimal, assembly-competent virus-like particles (VLPs). Here, we demonstrate that minimal HIV-1 VLPs retain 7SL RNA primarily as an endoribonucleolytic fragment, referred to as 7SL remnant (7SLrem). Nuclease mapping showed that 7SLrem is a 111-nucleotide internal portion of 7SL, with 5' and 3' ends corresponding to unpaired loops in the 7SL two-dimensional structure. Analysis of VLPs comprised of different subsets of Gag domains revealed that all NC-positive VLPs contained intact 7SL while the presence of 7SLrem correlated with the absence of the NC domain. Because 7SLrem, which maps to the 7SL S domain, was not detectable in infected cells, we propose a model whereby the species recruited to assembling VLPs is intact 7SL RNA, with 7SLrem produced by an endoribonuclease in the absence of NC. Since recruitment of 7SL RNA was a conserved feature of all tested minimal VLPs, our model further suggests that 7SL's recruitment is mediated, either directly or indirectly, through interactions with conserved features of all tested VLPs, such as the C-terminal domain of CA.


Asunto(s)
VIH-1/química , ARN Citoplasmático Pequeño/análisis , Partícula de Reconocimiento de Señal/análisis , Virión/química , Virosomas/química , Secuencia de Bases , Línea Celular , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Ribonucleasas/metabolismo
12.
Sci Rep ; 11(1): 368, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33432002

RESUMEN

The present study represents a formulation of nanocurcumin based hybrid virosomes (NC-virosome) to deliver drugs at targeted sites. Curcumin is a bioactive component derived from Curcuma longa and well-known for its medicinal property, but it exhibits poor solubility and rapid metabolism, which led to low bioavailability and hence limits its applications. Nanocurcumin was prepared to increase the aqueous solubility and to overcome all the limitations associated with curcumin. Influenza virosomes were prepared by solubilization of the viral membrane with 1,2-distearoyl-sn-glycerol-3-phosphocholine (DSPC). During membrane reconstitution, the hydrophilic nanocurcumin was added to the solvent system, followed by overnight dialysis to obtain NC-virosomes. The same was characterized using a transmission electron microscope (TEM) and scanning electron microscope (SEM), MTT assay was used to evaluate it's in vitro-cytotoxicity using MDA-MB231 and Mesenchyme stem cells (MSCs). The results showed NC-virosomes has spherical morphology with size ranging between 60 and 90 nm. It showed 82.6% drug encapsulation efficiency. The viability of MDA-MB231 cells was significantly inhibited by NC-virosome in a concentration-dependent manner at a specific time. The IC50 for nanocurcumin and NC-virosome was 79.49 and 54.23 µg/ml, respectively. The site-specific drug-targeting, high efficacy and non- toxicity of NC-virosomes proves its future potential as drug delivery vehicles.


Asunto(s)
Curcumina/administración & dosificación , Portadores de Fármacos/síntesis química , Virosomas/síntesis química , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Células Cultivadas , Curcuma/química , Curcumina/efectos adversos , Curcumina/química , Curcumina/farmacocinética , Portadores de Fármacos/efectos adversos , Portadores de Fármacos/química , Composición de Medicamentos/métodos , Sistemas de Liberación de Medicamentos/efectos adversos , Liberación de Fármacos , Sinergismo Farmacológico , Humanos , Subtipo H1N1 del Virus de la Influenza A/química , Ensayo de Materiales , Nanopartículas/administración & dosificación , Nanopartículas/efectos adversos , Nanopartículas/química , Nanopartículas/metabolismo , Virosomas/efectos adversos , Virosomas/química , Inactivación de Virus
13.
Pharm Res ; 27(3): 400-20, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19998056

RESUMEN

Over the last two decades, small interfering RNA (siRNA)-mediated gene silencing has quickly become one of the most powerful techniques used to study gene function in vitro and a promising area for new therapeutics. Delivery remains a significant impediment to realizing the therapeutic potential of siRNA, a problem that is also tied to immunogenicity and toxicity. Numerous delivery vehicles have been developed, including some that can be categorized as pseudovirions: these are vectors that are directly derived from viruses but whose viral coding sequences have been eliminated, preventing their classification as viral vectors. Characteristics of the pseudovirions discussed in this review, namely phagemids, HSV amplicons, SV40 in vitro-packaged vectors, influenza virosomes, and HVJ-Envelope vectors, make them attractive for the delivery of siRNA-based therapeutics. Pseudovirions were shown to deliver siRNA effector molecules and bring about RNA interference (RNAi) in various cell types in vitro, and in vivo using immune-deficient and immune-competent mouse models. Levels of silencing were not always determined directly, but the duration of siRNA-induced knockdown lasted at least 3 days. We present examples of the use of pseudovirions for the delivery of synthetic siRNA as well as the delivery and expression of DNA-directed siRNA.


Asunto(s)
ARN Interferente Pequeño/administración & dosificación , Virosomas/química , Virus/química , Animales , Humanos , Virosomas/genética , Virosomas/metabolismo , Virus/genética , Virus/metabolismo
14.
Hum Vaccin ; 6(5): 407-19, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20953154

RESUMEN

Cervarix™ is a prophylactic human papillomavirus (HPV)-16/18 vaccine developed for the prevention of cervical cancer. The vaccine antigens are HPV-16 and HPV-18 L1 virus-like particles (VLPs) made from baculovirus expression vector system (BEVS)-produced HPV-16 and HPV-18 L1 proteins, respectively. In this study, we demonstrate that truncation of the nuclear targeting and DNA binding signals at the C-terminus of the HPV-16 and HPV-18 L1 proteins prevented intranuclear formation of the VLPs in the host cells and led to cytoplasmic localization of the L1 proteins as shown by in situ immunogold detection and electron microscopy. Following purification, these L1 proteins were able to form VLPs. The characteristics of these HPV-16 and HPV-18 L1 VLPs were studied using various physicochemical and immunological techniques. Amino acid analysis, SDS-PAGE and western blotting demonstrated the high purity of the L1 proteins and batch-to-batch consistency. The structure of the VLPs was shown to be similar to that reported for the native virions, as evaluated by microscopic observations, protein tomography and disc centrifugation experiments. The presence of important conformation-dependent neutralizing epitopes, such as U4, V5 and J4, was confirmed by ELISA and surface plasmon resonance. Structural robustness and consistency among batches was also observed by differential scanning calorimetry and electron microscopy. Moreover, adsorption to aluminum was shown not to impair VLP structure. In conclusion, the BEVS-produced HPV-16 and HPV-18 L1 VLPs display key structural and immunological features, which contribute to the efficacy of Cervarix™ vaccination.


Asunto(s)
Vacunas contra Papillomavirus/química , Virosomas/química , Virosomas/ultraestructura , Aminoácidos/análisis , Western Blotting , Proteínas de la Cápside/química , Proteínas de la Cápside/ultraestructura , Dicroismo Circular , Citoplasma/química , Citoplasma/ultraestructura , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Humanos , Sustancias Macromoleculares/química , Sustancias Macromoleculares/ultraestructura , Microscopía Inmunoelectrónica , Proteínas Oncogénicas Virales/química , Proteínas Oncogénicas Virales/ultraestructura , Conformación Proteica , Vacunas de Virosoma/química
15.
J Clin Microbiol ; 46(5): 1734-40, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18305135

RESUMEN

RNase-resistant, noninfectious virus-like particles containing exogenous RNA sequences (armored RNA) are good candidates as RNA controls and standards in RNA virus detection. However, the length of RNA packaged in the virus-like particles with high efficiency is usually less than 500 bases. In this study, we describe a method for producing armored L-RNA. Armored L-RNA is a complex of MS2 bacteriophage coat protein and RNA produced in Escherichia coli by the induction of a two-plasmid coexpression system in which the coat protein and maturase are expressed from one plasmid and the target RNA sequence with modified MS2 stem-loop (pac site) is transcribed from another plasmid. A 3V armored L-RNA of 2,248 bases containing six gene fragments-hepatitis C virus, severe acute respiratory syndrome coronavirus (SARS-CoV1, SARS-CoV2, and SARS-CoV3), avian influenza virus matrix gene (M300), and H5N1 avian influenza virus (HA300)-was successfully expressed by the two-plasmid coexpression system and was demonstrated to have all of the characteristics of armored RNA. We evaluated the 3V armored L-RNA as a calibrator for multiple virus assays. We used the WHO International Standard for HCV RNA (NIBSC 96/790) to calibrate the chimeric armored L-RNA, which was diluted by 10-fold serial dilutions to obtain samples containing 10(6) to 10(2) copies. In conclusion, the approach we used for armored L-RNA preparation is practical and could reduce the labor and cost of quality control in multiplex RNA virus assays. Furthermore, we can assign the chimeric armored RNA with an international unit for quantitative detection.


Asunto(s)
Sustancias Macromoleculares/metabolismo , Reacción en Cadena de la Polimerasa/normas , ARN Viral/metabolismo , Ribonucleasas/metabolismo , Virosomas/biosíntesis , Virosomas/química , Escherichia coli/genética , Hepacivirus/genética , Subtipo H5N1 del Virus de la Influenza A/genética , Levivirus/genética , ARN Viral/genética , Estándares de Referencia , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Proteínas del Núcleo Viral/biosíntesis , Proteínas del Núcleo Viral/genética , Virosomas/efectos de los fármacos , Virosomas/genética
16.
Chem Biol ; 14(5): 577-87, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17524988

RESUMEN

The circumsporozoite protein (CSP) of Plasmodium falciparum is a leading candidate antigen for inclusion in a malaria subunit vaccine. We describe here the design of a conformationally constrained synthetic peptide, designated UK-39, which has structural and antigenic similarity to the NPNA-repeat region of native CSP. NMR studies on the antigen support the presence of helical turn-like structures within consecutive NPNA motifs in aqueous solution. Intramuscular delivery of UK-39 to mice and rabbits on the surface of reconstituted influenza virosomes elicited high titers of sporozoite crossreactive antibodies. Influenza virus proteins were crucially important for the immunostimulatory activity of the virosome-based antigen delivery system, as a liposomal formulation of UK-39 was not immunogenic. IgG antibodies elicited by UK-39 inhibited invasion of hepatocytes by P. falciparum sporozoites, but not by antigenically distinct P. yoelii sporozoites. Our approach to optimized virosome-formulated synthetic peptide vaccines should be generally applicable for other infectious and noninfectious diseases.


Asunto(s)
Anticuerpos Antiprotozoarios/biosíntesis , Anticuerpos Antiprotozoarios/inmunología , Vacunas contra la Malaria/química , Vacunas contra la Malaria/inmunología , Proteínas Protozoarias/química , Proteínas Protozoarias/inmunología , Virosomas/química , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Química Farmacéutica , Reacciones Cruzadas , Diseño de Fármacos , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Hibridomas/inmunología , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Datos de Secuencia Molecular , Fosfatidiletanolaminas/química , Plasmodium falciparum/inmunología , Plasmodium falciparum/patogenicidad , Plasmodium yoelii/inmunología , Plasmodium yoelii/patogenicidad , Conejos , Relación Estructura-Actividad
17.
J Virol Methods ; 148(1-2): 237-43, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18242720

RESUMEN

Ebola virus is a filovirus that causes hemorrhagic fever in humans and is associated with case fatality rates of up to 90%. The lack of therapeutic interventions in combination with the threat of weaponizing this organism has enhanced research investigations. The expression of key viral proteins and the production of virus-like particles in mammalian systems are often pursued for characterization and functional studies. Common practice is to express these proteins through transient transfection of mammalian cells. Unfortunately the transfection reagents are expensive and the process is time consuming and labour intensive. This work describes utilizing an ecdysone inducible mammalian expression system to create stable cell lines that express the Ebola virus transmembrane glycoprotein (GP), the soluble glycoprotein (sGP) and the matrix protein (VP40) individually as well as GP and VP40 simultaneously (for the production of virus like particles). These products were the same as those expressed by the transient system, by Western blot analysis and electron microscopy. The inducible system proved to be an improvement of the current technology by enhancing the cost effectiveness and simplifying the process.


Asunto(s)
Ecdisona/farmacología , Proteínas Estructurales Virales/biosíntesis , Proteínas Estructurales Virales/genética , Virosomas/biosíntesis , Acholeplasmataceae , Western Blotting , Línea Celular , Humanos , Microscopía Electrónica de Transmisión , Virosomas/química , Virosomas/ultraestructura
18.
Anal Bioanal Chem ; 391(8): 2717-27, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18351325

RESUMEN

We previously reported that transferrin (Tf)-modified liposomes (Tf-L) additionally modified with a cholesterylated pH-sensitive fusogenic peptide (Chol-GALA) can release an encapsulated aqueous phase marker to cytosol via endosomal membrane fusion. However, further obstacles need to be overcome to bring the Tf-L to the level of a viral-like gene delivery system. In this study, we developed a novel packaging method to encapsulate condensed plasmid DNA into PEgylated Tf-L (Tf-PEG-L) to form a core-shell-type nanoparticle. The most difficult challenge was to provide a mechanism of escape for the condensed core from endosome to cytosol in the presence of polyethylene glycol (PEG). We hypothesized that a membrane-introduced Chol-GALA and a PEgylated GALA would interact synergistically to induce membrane fusion between liposome and endosome. By simultaneously incorporating Chol-GALA into the membrane of Tf-PEG-L and GALA at tips of PEG chains, a condensed core was released into cytosol, and transfection activity increased 100-fold. We concluded that topological control was responsible for the synergistic effect of GALA derivatives introduced on Tf-PEG-L.


Asunto(s)
Endosomas , Técnicas de Transferencia de Gen , Fusión de Membrana , Nanopartículas/química , Péptidos/química , Virosomas/química , Colesterol/química , Endosomas/química , Endosomas/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Células K562 , Péptidos/metabolismo , Polietilenglicoles/química , Transferrina/química , Transferrina/metabolismo , Virosomas/metabolismo
19.
Biotechnol J ; 13(4): e1700645, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29278302

RESUMEN

Induction of CD8+ cytotoxic T cells (CTLs) to conserved internal influenza antigens, such as nucleoprotein (NP), is a promising strategy for the development of cross-protective influenza vaccines. However, influenza NP protein alone cannot induce CTL immunity due to its low capacity to activate antigen-presenting cells (APCs) and get access to the MHC class I antigen processing pathway. To facilitate the generation of NP-specific CTL immunity the authors develop a novel influenza vaccine consisting of virosomes with the Toll-like receptor 4 (TLR4) ligand monophosphoryl lipid A (MPLA) and the metal-ion-chelating lipid DOGS-NTA-Ni incorporated in the membrane. In vitro, virosomes with incorporated MPLA induce stronger activation of APCs than unadjuvanted virosomes. Virosomes modified with DOGS-NTA-Ni show high conjugation efficacy for his-tagged proteins and facilitate efficient uptake of conjugated proteins by APCs. Immunization of mice with MPLA-adjuvanted virosomes with attached NP results in priming of NP-specific CTLs while MPLA-adjuvanted virosomes with admixed NP are inefficient in priming CTLs. Both vaccines induce equally high titers of NP-specific antibodies. When challenged with heterosubtypic influenza virus, mice immunized with virosomes with attached or admixed NP are protected from severe weight loss. Yet, unexpectedly, they show more weight loss and more severe disease symptoms than mice immunized with MPLA-virosomes without NP. Taken together, these results indicate that virosomes with conjugated antigen and adjuvant incorporated in the membrane are effective in priming of CTLs and eliciting antigen-specific antibody responses in vivo. However, for protection from influenza infection NP-specific immunity appears not to be advantageous.


Asunto(s)
Adyuvantes Inmunológicos/química , Lípido A/análogos & derivados , Proteínas de Unión al ARN/inmunología , Proteínas del Núcleo Viral/inmunología , Virosomas/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Vacunas contra la Influenza/inmunología , Lípido A/química , Ratones , Níquel/química , Proteínas de la Nucleocápside , Células RAW 264.7 , Linfocitos T Citotóxicos/metabolismo , Virosomas/química
20.
Curr Med Chem ; 14(29): 3152-6, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18220748

RESUMEN

Influenza virosomes have proven to be effective vehicles for the delivery of antigens in the vaccination of humans against a number of pathogens. However, their potential as a means for gene delivery has yet to be realized. Chemical modification of viruses is emerging as a new strategy for production of safe and efficient gene delivery systems. Influenza virosomes exhibit many of the features of the virus, such as for cell binding, uptake and endosomal escape, which can be easily engineered into designer delivery vehicles capable of safe, efficient and cell-specific cargo delivery. This review focuses on the next generation of influenza virosomes and highlights aspects of their modification that may lead to simple but effective gene delivery vehicles.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética , Orthomyxoviridae , Virosomas , Animales , Humanos , Concentración de Iones de Hidrógeno , Polietilenglicoles , Virosomas/química , Virosomas/inmunología , Virosomas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA