Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 609
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 20(4): e1012174, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38630801

RESUMEN

As a type of parasitic agent, satellite RNAs (satRNAs) rely on cognate helper viruses to achieve their replication and transmission. During the infection of satRNAs, helper virus RNAs serve as templates for synthesizing viral proteins, including the replication proteins essential for satRNA replication. However, the role of non-template functions of helper virus RNAs in satRNA replication remains unexploited. Here we employed the well-studied model that is composed of cucumber mosaic virus (CMV) and its associated satRNA. In the experiments employing the CMV trans-replication system, we observed an unexpected phenomenon the replication proteins of the mild strain LS-CMV exhibited defective in supporting satRNA replication, unlike those of the severe strain Fny-CMV. Independent of translation products, all CMV genomic RNAs could enhance satRNA replication, when combined with the replication proteins of CMV. This enhancement is contingent upon the recruitment and complete replication of helper virus RNAs. Using the method developed for analyzing the satRNA recruitment, we observed a markedly distinct ability of the replication proteins from both CMV strains to recruit the positive-sense satRNA-harboring RNA3 mutant for replication. This is in agreement with the differential ability of both 1a proteins in binding satRNAs in plants. The discrepancies provide a convincing explanation for the variation of the replication proteins of both CMV strains in replicating satRNAs. Taken together, our work provides compelling evidence that the non-template functions of helper virus RNAs create an optimal replication environment to enhance satRNA proliferation.


Asunto(s)
Cucumovirus , Virus Helper , Satélite de ARN , ARN Viral , Replicación Viral , Virus Helper/genética , Virus Helper/fisiología , Cucumovirus/genética , Cucumovirus/metabolismo , Cucumovirus/fisiología , Satélite de ARN/metabolismo , Satélite de ARN/genética , ARN Viral/genética , ARN Viral/metabolismo , Enfermedades de las Plantas/virología , Nicotiana/virología , Nicotiana/metabolismo , Nicotiana/genética , Proteínas Virales/metabolismo , Proteínas Virales/genética
2.
Phytopathology ; 114(5): 1126-1136, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38451582

RESUMEN

Sugar beet (Beta vulgaris) is grown in temperate regions around the world as a source of sucrose used for natural sweetening. Sugar beet is susceptible to a number of viral diseases, but identification of the causal agent(s) under field conditions is often difficult due to mixtures of viruses that may be responsible for disease symptoms. In this study, the application of RNAseq to RNA extracted from diseased sugar beet roots obtained from the field and from greenhouse-reared plants grown in soil infested with the virus disease rhizomania (causal agent beet necrotic yellow vein virus; BNYVV) yielded genome-length sequences from BNYVV, as well as beet soil-borne virus (BSBV). The nucleotide identities of the derived consensus sequence of BSBV RNAs ranged from 99.4 to 96.7% (RNA1), 99.3 to 95.3% (RNA2), and 98.3 to 95.9% (RNA3) compared with published BSBV sequences. Based on the BSBV genome consensus sequence, clones of the genomic RNAs 1, 2, and 3 were obtained to produce RNA copies of the genome through in vitro transcription. Capped RNA produced from the clones was infectious when inoculated into leaves of Chenopodium quinoa and B. vulgaris, and extracts from transcript-infected C. quinoa leaves could infect sugar beet seedling roots through a vortex inoculation method. Subsequent exposure of these infected sugar beet seedling roots to aviruliferous Polymyxa betae, the protist vector of both BNYVV and BSBV, confirmed that BSBV derived from the infectious clones could be transmitted by the vector. Co-inoculation of BSBV synthetic transcripts with transcripts of a cloned putative satellite virus designated Beta vulgaris satellite virus 1A (BvSat1A) resulted in the production of lesions on leaves of C. quinoa similar to those produced by inoculation with BSBV alone. Nevertheless, accumulation of genomic RNA and the encoded protein of the satellite virus in co-inoculated leaves was readily detected on Northern and Western blots, respectively, whereas no accumulation of satellite virus products occurred when satellite virus RNA was inoculated alone. The predicted sequence of the detected protein encoded by BvSat1A bears hallmarks of coat proteins of other satellite viruses, and virions of a size consistent with a satellite virus were observed in samples testing positive for the virus. The results demonstrate that BSBV is a helper virus for the novel satellite virus BvSat1A.


Asunto(s)
Beta vulgaris , Enfermedades de las Plantas , Virus de Plantas , Virus Satélites , Beta vulgaris/virología , Enfermedades de las Plantas/virología , Virus Satélites/genética , Virus Satélites/fisiología , Virus de Plantas/genética , Virus de Plantas/fisiología , Virus Helper/genética , Virus Helper/fisiología , ARN Viral/genética , Raíces de Plantas/virología , Genoma Viral/genética , Microbiología del Suelo
3.
ISME J ; 17(12): 2381-2388, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37907733

RESUMEN

Satellites are mobile genetic elements that are dependent upon the replication machinery of their helper viruses. Bacteriophages have provided many examples of satellite nucleic acids that utilize their helper morphogenic genes for propagation. Here we describe two novel satellite-helper phage systems, Mulch and Flayer, that infect Streptomyces species. The satellites in these systems encode for encapsidation machinery but have an absence of key replication genes, thus providing the first example of bacteriophage satellite viruses. We also show that codon usage of the satellites matches the tRNA gene content of the helpers. The satellite in one of these systems, Flayer, does not appear to integrate into the host genome, which represents the first example of a virulent satellite phage. The Flayer satellite has a unique tail adaptation that allows it to attach to its helper for simultaneous co-infection. These findings demonstrate an ever-increasing array of satellite strategies for genetic dependence on their helpers in the evolutionary arms race between satellite and helper phages.


Asunto(s)
Bacteriófagos , Streptomyces , Virus Satélites/genética , Streptomyces/genética , Virulencia , Virus Helper/genética , Bacteriófagos/genética
4.
Virol J ; 20(1): 228, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37817259

RESUMEN

Adeno-associated virus (AAV) differs from most other viruses, as it requires the simultaneous presence of a helper virus for an active infection. Up to 80% of the human population is seropositive for AAV antibodies. AAV has been known to be a non-pathogenic virus and an inhibitor of carcinogenesis caused by coinfecting viruses. However, the recent reports associating AAV infection with hepatocellular carcinoma development and the mysterious cases of acute severe hepatitis in children have challenged the idea that AAV is a harmless virus. Herein, we explore the usefulness of AAV in gene therapy and the importance of AAV as a protector or perpetrator in human carcinogenesis, ultimately reflecting on the dual role of AAV in human health.


Asunto(s)
Dependovirus , Neoplasias Hepáticas , Niño , Humanos , Dependovirus/genética , Replicación Viral , Virus Helper/genética , Carcinogénesis
5.
ACS Synth Biol ; 11(10): 3285-3295, 2022 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-36219557

RESUMEN

Recombinant adeno-associated viruses (rAAV) are important gene delivery vehicles for gene therapy applications. Their production relies on plasmid transfection or virus infection of producer cells, which pose a challenge in process scale-up. Here, we describe a template for a transfection-free, helper virus-free rAAV producer cell line using a synthetic biology approach. Three modules were integrated into HEK293 cells including an rAAV genome and multiple inducible promoters controlling the expression of AAV Rep, Cap, and helper coding sequences. The synthetic cell line generated infectious rAAV vectors upon induction. Independent control over replication and packaging activities allowed for manipulation of the fraction of capsid particles containing viral genomes, affirming the feasibility of tuning gene expression profiles in a synthetic cell line for enhancing the quality of the viral vector produced. The synthetic biology approach for rAAV production presented in this study can be exploited for scalable biomanufacturing.


Asunto(s)
Dependovirus , Biología Sintética , Humanos , Dependovirus/genética , Células HEK293 , Vectores Genéticos/genética , Virus Helper/genética , Virus Helper/metabolismo
6.
Int J Mol Sci ; 23(16)2022 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-36012656

RESUMEN

Viral satellite RNAs (satRNAs) are small subviral particles that are associated with the genomic RNA of a helper virus (HV). Their replication, encapsidation, and movement depend on the HV. In this paper, we performed a global analysis of the satRNAs associated with different isolates of tomato black ring virus (TBRV). We checked the presence of satRNAs in 42 samples infected with TBRV, performed recombination and genetic diversity analyses, and examined the selective pressure affecting the satRNAs population. We identified 18 satRNAs in total that differed in length and the presence of point mutations. Moreover, we observed a strong effect of selection operating upon the satRNA population. We also constructed infectious cDNA clones of satRNA and examined the viral load of different TBRV isolates in the presence and absence of satRNAs, as well as the accumulation of satRNA molecules on infected plants. Our data provide evidence that the presence of satRNAs significantly affects viral load; however, the magnitude of this effect differs among viral isolates and plant hosts. We also showed a positive correlation between the number of viral genomic RNAs (gRNAs) and satRNAs for two analysed TBRV isolates.


Asunto(s)
Satélite de ARN , ARN Viral , Variación Genética , Virus Helper/genética , Nepovirus , Enfermedades de las Plantas/genética , Plantas/genética , Satélite de ARN/genética , ARN Viral/genética , Replicación Viral/genética
7.
Viruses ; 14(1)2022 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-35062311

RESUMEN

Human hepatitis D virus (HDV) depends on hepatitis B virus co-infection and its glycoproteins for infectious particle formation. HDV was the sole known deltavirus for decades and believed to be a human-only pathogen. However, since 2018, several groups reported finding HDV-like agents from various hosts but without co-infecting hepadnaviruses. In vitro systems enabling helper virus-independent replication are key for studying the newly discovered deltaviruses. Others and we have successfully used constructs containing multimers of the deltavirus genome for the replication of various deltaviruses via transfection in cell culture. Here, we report the establishment of deltavirus infectious clones with 1.2× genome inserts bearing two copies of the genomic and antigenomic ribozymes. We used Swiss snake colony virus 1 as the model to compare the ability of the previously reported "2× genome" and the "1.2× genome" infectious clones to initiate replication in cell culture. Using immunofluorescence, qRT-PCR, immuno- and northern blotting, we found the 2× and 1.2× genome clones to similarly initiate deltavirus replication in vitro and both induced a persistent infection of snake cells. The 1.2× genome constructs enable easier introduction of modifications required for studying deltavirus replication and cellular interactions.


Asunto(s)
Boidae/virología , Células Clonales , Coinfección/genética , Virus de la Hepatitis Delta/genética , Replicación Viral , Animales , Boidae/genética , Genoma Viral , Virus Helper/genética , Hepadnaviridae/genética , Hepatitis B/genética , Virus de la Hepatitis B/genética , Hepatitis D/virología , ARN Catalítico , ARN Viral/genética , Transfección
8.
Int J Mol Sci ; 22(24)2021 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-34948288

RESUMEN

The killer phenotype of Torulaspora delbrueckii (Td) and Saccharomyces cerevisiae (Sc) is encoded in the genome of medium-size dsRNA viruses (V-M). Killer strains also contain a helper large size (4.6 kb) dsRNA virus (V-LA) which is required for maintenance and replication of V-M. Another large-size (4.6 kb) dsRNA virus (V-LBC), without known helper activity to date, may join V-LA and V-M in the same yeast. T. delbrueckii Kbarr1 killer strain contains the killer virus Mbarr1 in addition to two L viruses, TdV-LAbarr1 and TdV-LBCbarr1. In contrast, the T. delbrueckii Kbarr2 killer strain contains two M killer viruses (Mbarr1 and M1) and a LBC virus (TdV-LBCbarr2), which has helper capability to maintain both M viruses. The genomes of TdV-LBCbarr1 and TdV-LBCbarr2 were characterized by high-throughput sequencing (HTS). Both RNA genomes share sequence identity and similar organization with their ScV-LBC counterparts. They contain all conserved motifs required for translation, packaging, and replication of viral RNA. Their Gag-Pol amino-acid sequences also contain the features required for cap-snatching and RNA polymerase activity. However, some of these motifs and features are similar to those of LA viruses, which may explain that at least TdV-LBCbarr2 has a helper ability to maintain M killer viruses. Newly sequenced ScV-LBC genomes contained the same motifs and features previously found in LBC viruses, with the same genome location and secondary structure. Sequence comparison showed that LBC viruses belong to two clusters related to each species of yeast. No evidence for associated co-evolution of specific LBC with specific M virus was found. The presence of the same M1 virus in S. cerevisiae and T. delbrueckii raises the possibility of cross-species transmission of M viruses.


Asunto(s)
Virus ARN Bicatenario/genética , Genoma Viral/genética , Virus Helper/genética , ARN Bicatenario/genética , Torulaspora/genética , Vino/microbiología , Vino/virología , Secuencia de Aminoácidos , Secuencia de Bases , Cápside , ARN Viral/genética , Saccharomyces cerevisiae/genética
9.
Viruses ; 13(9)2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34578277

RESUMEN

The genetic diversity of baculoviruses provides a sustainable agronomic solution when resistance to biopesticides seems to be on the rise. This genetic diversity promotes insect infection by several genotypes (i.e., multiple infections) that are more likely to kill the host. However, the mechanism and regulation of these virus interactions are still poorly understood. In this article, we focused on baculoviruses infecting the codling moth, Cydia pomonella: two Cydia pomonella granulovirus genotypes, CpGV-M and CpGV-R5, and Cryptophlebia peltastica nucleopolyhedrovirus (CrpeNPV). The influence of the order of ingestion of the virus genotypes, the existence of an ingestion delay between the genotypes and the specificity of each genotype involved in the success of multiple infection were studied in the case of Cydia pomonella resistance. To obtain a multiple infection in resistant insects, the order of ingestion is a key factor, but the delay for ingestion of the second virus is not. CrpeNPV cannot substitute CpGV-R5 to allow replication of CpGV-M.


Asunto(s)
Conducta Alimentaria , Granulovirus/genética , Granulovirus/fisiología , Virus Helper/fisiología , Mariposas Nocturnas/virología , Replicación Viral , Animales , Variación Genética , Virus Helper/genética
10.
Viruses ; 13(9)2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34578372

RESUMEN

Human rotaviruses (HuRVAs) are highly important causes of acute gastroenteritis in infants and young children worldwide. A lack of reliable and reproducible reverse genetics systems for HuRVAs has limited a proper understanding of HuRVA biology and also the rational design of live-attenuated vaccines. Since the development of the first reverse genetics system for RVAs (partially plasmid-based reverse genetics system) in 2006, there have been many efforts with the goal of generating infectious recombinant HuRVAs entirely from cloned cDNAs. However, the establishment of a HuRVA reverse genetics system was very challenging until 2019. This review article provides an overview of the historical background of the recent development of long-awaited HuRVA reverse genetics systems, beginning with the generation of recombinant human-simian reassortant RVAs with the aid of a helper virus in 2006 and the generation of recombinant animal (simian) RVAs in a helper virus-free manner in 2017, and culminating in the generation of recombinant HuRVAs entirely from plasmid cDNAs in 2019. Notably, the original HuRVA reverse genetics system has already been optimized to increase the efficiency of virus generation. Although the application of HuRVA reverse genetics systems has only just been initiated, these technologies will help to answer HuRVA research questions regarding viral replication and pathogenicity that could not be addressed before, and to develop next-generation vaccines and intestine-specific rotaviral vectors.


Asunto(s)
Genoma Viral , Plásmidos/genética , Genética Inversa/métodos , Rotavirus/genética , Replicación Viral/genética , Virus Helper/genética , Humanos , ARN Viral/genética , Infecciones por Rotavirus/virología , Proteínas no Estructurales Virales/genética
11.
Sci Rep ; 11(1): 10400, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-34002008

RESUMEN

The lateral hypothalamus (LH) is critically involved in the regulation of homeostatic energy balance. Some neurons in the LH express receptors for leptin (LepRb), a hormone known to increase energy expenditure and decrease energy intake. However, the neuroanatomical inputs to LepRb-expressing LH neurons remain unknown. We used rabies virus tracing technology to map these inputs, but encountered non-specific tracing. To optimize this technology for a minor cell population (LepRb is not ubiquitously expressed in LH), we used LepRb-Cre mice and assessed how different titers of the avian tumor virus receptor A (TVA) helper virus affected rabies tracing efficiency and specificity. We found that rabies expression is dependent on TVA receptor expression, and that leakiness of TVA receptors is dependent on the titer of TVA virus used. We concluded that a titer of 1.0-3.0 × 107 genomic copies per µl of the TVA virus is optimal for rabies tracing. Next, we successfully applied modified rabies virus tracing technology to map inputs to LepRb-expressing LH neurons. We discovered that other neurons in the LH itself, the periventricular hypothalamic nucleus (Pe), the posterior hypothalamic nucleus (PH), the bed nucleus of the stria terminalis (BNST), and the paraventricular hypothalamic nucleus (PVN) are the most prominent input areas to LepRb-expressing LH neurons.


Asunto(s)
Conectoma/métodos , Hipotálamo/diagnóstico por imagen , Imagen Molecular/métodos , Neuronas/metabolismo , Receptores de Leptina/análisis , Animales , Proteínas Aviares/genética , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Virus Helper/genética , Hipotálamo/citología , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Virus de la Rabia/genética , Receptores de Leptina/metabolismo , Receptores Virales/genética , Núcleos Septales/citología , Núcleos Septales/diagnóstico por imagen , Núcleos Septales/metabolismo , Técnicas Estereotáxicas
12.
J Virol ; 95(13): e0048621, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-33853961

RESUMEN

Wild-type adeno-associated virus (AAV) can only replicate in the presence of helper factors, which can be provided by coinfecting helper viruses such as adenoviruses and herpesviruses. The AAV genome consists of a linear, single-stranded DNA (ssDNA), which is converted into different molecular structures within the host cell. Using high-throughput sequencing, we found that herpes simplex virus 1 (HSV-1) coinfection leads to a shift in the type of AAV genome end recombination. In particular, open-end inverted terminal repeat (ITR) recombination was enhanced, whereas open-closed ITR recombination was reduced in the presence of HSV-1. We demonstrate that the HSV-1 protein ICP8 plays an essential role in HSV-1-mediated interference with AAV genome end recombination, indicating that the previously described ICP8-driven mechanism of HSV-1 genome recombination may be underlying the observed changes. We also provide evidence that additional factors, such as products of true late genes, are involved. Although HSV-1 coinfection significantly changed the type of AAV genome end recombination, no significant change in the amount of circular AAV genomes was identified. IMPORTANCE Adeno-associated virus (AAV)-mediated gene therapy represents one of the most promising approaches for the treatment of genetic diseases. Currently, various GMP-compatible production methods can be applied to manufacture clinical-grade vector, including methods that employ helper factors derived from herpes simplex virus 1 (HSV-1). Yet, to date, we do not fully understand how HSV-1 interacts with AAV. We observed that HSV-1 modulates AAV genome ends similarly to the genome recombination events observed during HSV-1 replication and postulate that further improvements of the HSV-1 production platform may enhance packaging of the recombinant AAV particles.


Asunto(s)
Dependovirus/crecimiento & desarrollo , Dependovirus/genética , Genoma Viral/genética , Virus Helper/genética , Herpesvirus Humano 1/genética , Recombinación Genética/genética , Animales , Línea Celular , Chlorocebus aethiops , Coinfección/patología , Células HEK293 , Células HeLa , Herpes Simple/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Infecciones por Parvoviridae/patología , Secuencias Repetidas Terminales/genética , Células Vero , Interferencia Viral/genética , Replicación Viral/genética
13.
J Mol Biol ; 433(9): 166896, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33639215

RESUMEN

Vaccinia virus (VACV)-based vectors are in extensive use as vaccines and cancer immunotherapies. VACV engineering has traditionally relied on homologous recombination between a parental viral genome and a transgene-bearing transfer plasmid, an inefficient process that necessitates the use of a selection or screening marker to isolate recombinants. Recent extensions of this approach have sought to enhance the recovery of transgene-bearing viruses through the use of CRISPR-Cas9 engineering to cleave the viral genome in infected cells. However, these methods do not completely eliminate the generation of WT viral progeny and thus continue to require multiple rounds of viral propagation and plaque purification. Here, we describe MAVERICC (marker-free vaccinia virus engineering of recombinants through in vitroCRISPR/Cas9 cleavage), a new strategy to engineer recombinant VACVs in a manner that overcomes current limitations. MAVERICC also leverages the CRISPR/Cas9 system but requires no markers and yields essentially pure preparations of the desired recombinants in a single step. We used this approach to introduce point mutations, insertions, and deletions at multiple locations in the VACV genome, both singly and in combination. The efficiency and versatility of MAVERICC make it an ideal choice for generating mutants and mutant libraries at arbitrarily selected locations in the viral genome to build complex VACV vectors, effect vector improvements, and facilitate the study of poxvirus biology.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , ADN Recombinante/genética , Edición Génica/métodos , Virus Vaccinia/genética , Virus Vaccinia/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Epítopos/genética , Epítopos/inmunología , Genes Virales/genética , Marcadores Genéticos/genética , Vectores Genéticos/genética , Genoma Viral/genética , Virus Helper/genética , Fusión de Membrana , Virión/genética , Internalización del Virus
14.
Mol Ther ; 29(5): 1808-1820, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33571680

RESUMEN

The immunosuppressive tumor microenvironment (TME) is a formidable barrier to the success of adoptive cell therapies for solid tumors. Oncolytic immunotherapy with engineered adenoviruses (OAd) may disrupt the TME by infecting tumor cells, as well as surrounding stroma, to improve the functionality of tumor-directed chimeric antigen receptor (CAR)-T cells, yet efficient delivery of OAds to solid tumors has been challenging. Here we describe how mesenchymal stromal cells (MSCs) can be used to systemically deliver a binary vector containing an OAd together with a helper-dependent Ad (HDAd; combinatorial Ad vector [CAd]) that expresses interleukin-12 (IL-12) and checkpoint PD-L1 (programmed death-ligand 1) blocker. CAd-infected MSCs deliver and produce functional virus to infect and lyse lung tumor cells while stimulating CAR-T cell anti-tumor activity by release of IL-12 and PD-L1 blocker. The combination of this approach with administration of HER.2-specific CAR-T cells eliminates 3D tumor spheroids in vitro and suppresses tumor growth in two orthotopic lung cancer models in vivo. Treatment with CAd MSCs increases the overall numbers of human T cells in vivo compared to CAR-T cell only treatment and enhances their polyfunctional cytokine secretion. These studies combine the predictable targeting of CAR-T cells with the advantages of cancer cell lysis and TME disruption by systemic MSC delivery of oncolytic virotherapy: incorporation of immunostimulation by cytokine and checkpoint inhibitor production through the HDAd further enhances anti-tumor activity.


Asunto(s)
Anticuerpos Monoclonales/genética , Dependovirus/fisiología , Virus Helper/fisiología , Interleucina-12/metabolismo , Neoplasias Pulmonares/terapia , Células Madre Mesenquimatosas/virología , Receptores de Antígenos de Linfocitos T/metabolismo , Células A549 , Animales , Anticuerpos Monoclonales/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Línea Celular Tumoral , Terapia Combinada , Dependovirus/genética , Virus Helper/genética , Humanos , Inmunoterapia Adoptiva , Interleucina-12/antagonistas & inhibidores , Interleucina-12/genética , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Viroterapia Oncolítica , Receptor ErbB-2/inmunología , Microambiente Tumoral , Tropismo Viral , Ensayos Antitumor por Modelo de Xenoinjerto
15.
BMB Rep ; 53(11): 565-575, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32958121

RESUMEN

Gene therapy is emerging as a treatment option for inherited genetic diseases. The success of this treatment approach greatly depends upon gene delivery vectors. Researchers have attempted to harness the potential of viral vectors for gene therapy applications over many decades. Among the viral vectors available, gutless adenovirus (GLAd) has been recognized as one of the most promising vectors for in vivo gene delivery. GLAd is constructed by deleting all the viral genes from an adenovirus. Owing to this structural feature, the production of GLAd requires a helper that supplies viral proteins in trans. Conventionally, the helper is an adenovirus. Although the helper adenovirus efficiently provides helper functions, it remains as an unavoidable contaminant and also generates replicationcompetent adenovirus (RCA) during the production of GLAd. These two undesirable contaminants have raised safety concerns and hindered the clinical applications of GLAd. Recently, we developed helper virus-free gutless adenovirus (HF-GLAd), a new version of GLAd, which is produced by a helper plasmid instead of a helper adenovirus. Utilization of this helper plasmid eliminated the helper adenovirus and RCA contamination in the production of GLAd. HF-GLAd, devoid of helper adenovirus and RCA contaminants, will facilitate its clinical applications. In this review, we discuss the characteristics of adenoviruses, the evolution and production of adenoviral vectors, and the unique features of HF-GLAd as a new platform for gene therapy. Furthermore, we highlight the potential applications of HF-GLAd as a gene delivery vector for the treatment of various inherited genetic diseases. [BMB Reports 2020; 53(11): 565-575].


Asunto(s)
Adenoviridae/genética , Adenoviridae/metabolismo , Terapia Genética/métodos , Línea Celular , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Virus Helper/genética , Virus Helper/metabolismo , Humanos , Integrasas/genética , Plásmidos/genética , Proteínas Virales/genética
16.
Viruses ; 12(9)2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32867300

RESUMEN

P4 is a mobile genetic element (MGE) that can exist as a plasmid or integrated into its Escherichia coli host genome, but becomes packaged into phage particles by a helper bacteriophage, such as P2. P4 is the original example of what we have termed "molecular piracy", the process by which one MGE usurps the life cycle of another for its own propagation. The P2 helper provides most of the structural gene products for assembly of the P4 virion. However, when P4 is mobilized by P2, the resulting capsids are smaller than those normally formed by P2 alone. The P4-encoded protein responsible for this size change is called Sid, which forms an external scaffolding cage around the P4 procapsids. We have determined the high-resolution structure of P4 procapsids, allowing us to build an atomic model for Sid as well as the gpN capsid protein. Sixty copies of Sid form an intertwined dodecahedral cage around the T = 4 procapsid, making contact with only one out of the four symmetrically non-equivalent copies of gpN. Our structure provides a basis for understanding the sir mutants in gpN that prevent small capsid formation, as well as the nms "super-sid" mutations that counteract the effect of the sir mutations, and suggests a model for capsid size redirection by Sid.


Asunto(s)
Bacteriófagos/química , Proteínas de la Cápside/química , Cápside/química , Bacteriófagos/genética , Bacteriófagos/metabolismo , Cápside/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Virus Helper/química , Virus Helper/genética , Virus Helper/metabolismo , Mutación , Conformación Proteica , Virus Satélites/química , Virus Satélites/genética , Virus Satélites/metabolismo
17.
Virus Res ; 286: 198075, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32592818

RESUMEN

Reverse genetics technology allows one to engineer replication-competent viruses from cloned cDNAs at will. Since the establishment of the initial reverse genetics system for species A rotaviruses (RVAs) requiring a helper virus in 2006, attempts have been successfully made to improve this technology. Efficient generation of replication-competent RVAs is now possible from just 11 T7-driven plasmids encoding an RVA genome when the quantity ratio of the two rescue T7-driven plasmids for the NSP2 and NSP5 segments is increased by 3-fold in relation to that of the other nine plasmids (11 plasmid-only system). Further, it is now possible to generate recombinant RVAs even with severely less efficient infectivity by using the 11 plasmid-only system, which has not been possible with the existing approaches. More importantly, the 11 plasmid-only system does not need any helper expression plasmid, and thus this simplest and robust system has a clear advantage over the existing systems in terms of safety. This 11 plasmid-only system should contribute to the development of safe next-generation vaccines and vaccine vectors.


Asunto(s)
Genoma Viral , Genética Inversa , Rotavirus/genética , Animales , Línea Celular , ADN Complementario/genética , Virus Helper/genética , Humanos , Ratones , ARN Viral/genética , Proteínas no Estructurales Virales/genética , Replicación Viral
18.
mBio ; 11(2)2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32184255

RESUMEN

Satellite viruses, most commonly found in plants, rely on helper viruses to complete their replication cycle. The only known example of a human satellite virus is the hepatitis D virus (HDV), and it is generally thought to require hepatitis B virus (HBV) to form infectious particles. Until 2018, HDV was the sole representative of the genus Deltavirus and was thought to have evolved in humans, the only known HDV host. The subsequent identification of HDV-like agents in birds, snakes, fish, amphibians, and invertebrates indicated that the evolutionary history of deltaviruses is likely much longer than previously hypothesized. Interestingly, none of the HDV-like agents were found in coinfection with an HBV-like agent, suggesting that these viruses use different helper virus(es). Here we show, using snake deltavirus (SDeV), that HBV and hepadnaviruses represent only one example of helper viruses for deltaviruses. We cloned the SDeV genome into a mammalian expression plasmid, and by transfection could initiate SDeV replication in cultured snake and mammalian cell lines. By superinfecting persistently SDeV-infected cells with reptarenaviruses and hartmaniviruses, or by transfecting their surface proteins, we could induce production of infectious SDeV particles. Our findings indicate that deltaviruses can likely use a multitude of helper viruses or even viral glycoproteins to form infectious particles. This suggests that persistent infections, such as those caused by arenaviruses and orthohantaviruses used in this study, and recurrent infections would be beneficial for the spread of deltaviruses. It seems plausible that further human or animal disease associations with deltavirus infections will be identified in the future.IMPORTANCE Deltaviruses need a coinfecting enveloped virus to produce infectious particles necessary for transmission to a new host. Hepatitis D virus (HDV), the only known deltavirus until 2018, has been found only in humans, and its coinfection with hepatitis B virus (HBV) is linked with fulminant hepatitis. The recent discovery of deltaviruses without a coinfecting HBV-like agent in several different taxa suggested that deltaviruses could employ coinfection by other enveloped viruses to complete their life cycle. In this report, we show that snake deltavirus (SDeV) efficiently utilizes coinfecting reptarena- and hartmaniviruses to form infectious particles. Furthermore, we demonstrate that cells expressing the envelope proteins of arenaviruses and orthohantaviruses produce infectious SDeV particles. As the envelope proteins are responsible for binding and infecting new host cells, our findings indicate that deltaviruses are likely not restricted in their tissue tropism, implying that they could be linked to animal or human diseases other than hepatitis.


Asunto(s)
Virus de la Hepatitis Delta/genética , Virus de la Hepatitis Delta/patogenicidad , Serpientes/virología , Proteínas del Envoltorio Viral/genética , Animales , Línea Celular Tumoral , Coinfección/virología , Genoma Viral , Virus Helper/genética , Virus de la Hepatitis B/genética , Virus de la Hepatitis Delta/clasificación , Humanos , ARN Viral/genética , Tropismo Viral , Replicación Viral
19.
Mol Brain ; 13(1): 5, 2020 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-31931837

RESUMEN

Neurotropic viral transsynaptic tracing is an increasingly powerful technique for dissecting the structure and function of neural circuits. Herpes simplex virus type 1 strain H129 has been widely used as an anterograde tracer. However, HSV tracers still have several shortcomings, including high toxicity, low sensitivity and non-specific retrograde labeling. Here, we aimed to construct high-brightness HSV anterograde tracers by increasing the expression of exogenous genes carried by H129 viruses. Using a Trojan horse-like strategy, a HSV/AAV (adeno-associated virus) chimaera termed H8 was generated to enhance the expression of a fluorescent marker. In vitro and in vivo assays showed that the exogenous gene was efficiently replicated and amplified by the synergism of the HSV vector and introduced AAV replication system. H8 reporting fluorescence was brighter than that of currently available H129 tracers, and H8 could be used for fast and effective anterograde tracing without additional immunostaining. These results indicated that foreign gene expression in HSV tracers could be enhanced by integrating HSV with AAV replication system. This approach may be useful as a general enhanced expression strategy for HSV-based tracing tools or gene delivery vectors.


Asunto(s)
Transporte Axonal/fisiología , Encéfalo/citología , Virus Defectuosos/fisiología , Dependovirus/fisiología , Proteínas Fluorescentes Verdes/análisis , Virus Helper/fisiología , Herpesvirus Humano 1/fisiología , Vías Nerviosas/ultraestructura , Técnicas de Trazados de Vías Neuroanatómicas/métodos , Trazadores del Tracto Neuronal/análisis , Neuronas/ultraestructura , Virus Reordenados/fisiología , Animales , Línea Celular , Núcleo Celular/virología , Virus Defectuosos/genética , Dependovirus/genética , Genes Reporteros , Genes Sintéticos , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Virus Helper/genética , Herpesvirus Humano 1/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/virología , Virus Reordenados/genética , Proteinas del Complejo de Replicasa Viral/genética , Replicación Viral
20.
RNA ; 25(11): 1432-1438, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31434792

RESUMEN

R2 is a long interspersed element (LINE) found in a specific sequence of the 28S rDNA among a wide variety of animals. Recently, we observed that R2Ol isolated from medaka fish, Oryzias latipes, retrotransposes sequence specifically into the target sequence of zebrafish. Because the 28S target and flanking regions are widely conserved among vertebrates, we examined whether R2Ol can also integrate in a sequence-specific manner in human cells. Using adenovirus-mediated expression of R2Ol constructs, we confirmed an accurate insertion of R2Ol into the 28S target of human 293T cells. However, the R2Ol mutant devoid of endonuclease (EN) activity showed no retrotransposition ability, suggesting that the sequence-specific integration of R2Ol into 28S rDNA occurs via the cleavage activity of EN. By introducing both R2Ol helper virus and donor plasmid in human cells, we succeeded in retrotransposing an exogenous EGFP gene into the 28S target site by the trans-complementation system, which enabled simplification of specific gene knock-in in a time-efficient manner. We believe that R2Ol may provide an alternative targeted gene knock-in method for practical applications such as gene therapy in future.


Asunto(s)
ADN Ribosómico/genética , Elementos de Nucleótido Esparcido Largo , ARN Ribosómico 28S/genética , Retroelementos , Adenoviridae/genética , Técnicas de Sustitución del Gen , Prueba de Complementación Genética , Células HEK293 , Virus Helper/genética , Humanos , Plásmidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA