Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 280
Filtrar
1.
Cell Rep ; 43(4): 114050, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38564336

RESUMO

Seo et al.1 shed light on virus-host interactions as they reveal how poxvirus A51R stabilizes microtubules in infected cells, which impacts vaccinia virus virulence in mice by potentially inhibiting reactive-oxygen-species-dependent antiviral responses in macrophages.


Assuntos
Microtúbulos , Vaccinia virus , Microtúbulos/metabolismo , Animais , Virulência , Vaccinia virus/patogenicidade , Vaccinia virus/fisiologia , Humanos , Camundongos , Macrófagos/virologia , Macrófagos/metabolismo , Poxviridae/patogenicidade , Poxviridae/genética , Poxviridae/fisiologia
2.
J Fish Dis ; 47(6): e13934, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38421376

RESUMO

Carp oedema virus (CEV) has distinct molecularly identified genogroups of viral mutations, denoted as I, IIa, and IIb. Failure to propagate CEV in vitro limits studies towards understanding its interactions with host cells. Here, virus isolates belonging to genogroup I collected during natural outbreaks in the Czech Republic were employed for routine CEV cultivation in monolayers of carp-derived primary cells, common carp brain (CCB) cells, and epithelioma papulosum cyprinid (EPC) cells. Induction of cytopathic effects (CPEs) was observed and recorded in affected cells. Cell survival rate was evaluated under serial dilutions of the CEV inoculum. Virus cell entry was quantified and visualized by qPCR and transmission electron microscopy, respectively. Study findings indicate primary gills epithelia likely present the most suitable matrix for CEV growth in vitro. Cells of the head kidney and spleen facilitate virus entry with microscopically confirmed CPEs and the presence of cytoplasmic pleomorphic virus particles. Cells of the trunk kidney and gonads are unlikely to permit virus cell entry and CPEs development. Although CEV cultivation in cell lines was inconclusive, EPC cells were CEV permissible. Monolayers of carp-derived primary cells show promise for CEV cultivation that could enable elaborate study of mechanisms underlying cellular binding and responses.


Assuntos
Carpas , Doenças dos Peixes , Poxviridae , Animais , Carpas/virologia , Poxviridae/fisiologia , Poxviridae/genética , Doenças dos Peixes/virologia , Infecções por Poxviridae/veterinária , Infecções por Poxviridae/virologia , Cultura de Vírus/métodos , Linhagem Celular , República Tcheca , Células Cultivadas , Genótipo
3.
J Gen Virol ; 104(10)2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37792576

RESUMO

Poxviruses (family Poxviridae) have long dsDNA genomes and infect a wide range of hosts, including insects, birds, reptiles and mammals. These viruses have substantial incidence, prevalence and disease burden in humans and in other animals. Nucleotide and dinucleotide composition, mostly CpG and TpA, have been largely studied in viral genomes because of their evolutionary and functional implications. We analysed here the nucleotide and dinucleotide composition, as well as codon usage bias, of a set of representative poxvirus genomes, with a very diverse host spectrum. After correcting for overall nucleotide composition, entomopoxviruses displayed low overall GC content, no enrichment in TpA and large variation in CpG enrichment, while chordopoxviruses showed large variation in nucleotide composition, no obvious depletion in CpG and a weak trend for TpA depletion in GC-rich genomes. Overall, intergenome variation in dinucleotide composition in poxviruses is largely accounted for by variation in overall genomic GC levels. Nonetheless, using vaccinia virus as a model, we found that genes expressed at the earliest times in infection are more CpG-depleted than genes expressed at later stages. This observation has parallels in betahepesviruses (also large dsDNA viruses) and suggests an antiviral role for the innate immune system (e.g. via the zinc-finger antiviral protein ZAP) in the early phases of poxvirus infection. We also analysed codon usage bias in poxviruses and we observed that it is mostly determined by genomic GC content, and that stratification after host taxonomy does not contribute to explaining codon usage bias diversity. By analysis of within-species diversity, we show that genomic GC content is the result of mutational biases. Poxvirus genomes that encode a DNA ligase are significantly AT-richer than those that do not, suggesting that DNA repair systems shape mutation biases. Our data shed light on the evolution of poxviruses and inform strategies for their genetic manipulation for therapeutic purposes.


Assuntos
Poxviridae , Animais , Humanos , Poxviridae/genética , Nucleotídeos , Códon/genética , Evolução Molecular , Mamíferos/genética , Fosfatos de Dinucleosídeos , Antivirais
4.
Cancer Gene Ther ; 30(7): 1029-1039, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37016144

RESUMO

cGAMP-specific nucleases (poxins) are a recently described family of proteins dedicated to obstructing cyclic GMP-AMP synthase signaling (cGAS), an important sensor triggered by cytoplasmic viral replication that activates type I interferon (IFN) production. The B2R gene of vaccinia viruses (VACV) codes for one of these nucleases. Here, we evaluated the effects of inactivating the VACV B2 nuclease in the context of an oncolytic VACV. VACV are widely used as anti-cancer vectors due to their capacity to activate immune responses directed against tumor antigens. We aimed to elicit robust antitumor immunity by preventing viral inactivation of the cGAS/STING/IRF3 pathway after infection of cancer cells. Activation of such a pathway is associated with a dominant T helper 1 (Th1) cell differentiation of the response, which benefits antitumor outcomes. Deletion of the B2R gene resulted in enhanced IRF3 phosphorylation and type I IFN expression after infection of tumor cells, while effective VACV replication remained unimpaired, both in vitro and in vivo. In syngeneic mouse tumor models, the absence of the VACV cGAMP-specific nuclease translated into improved antitumor activity, which was associated with antitumor immunity directed against tumor epitopes.


Assuntos
Interferon Tipo I , Poxviridae , Camundongos , Animais , Poxviridae/genética , Nucleotídeos Cíclicos , Vaccinia virus/genética , Vaccinia virus/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Interferon Tipo I/genética , Imunidade , Imunidade Inata/genética
5.
PLoS Pathog ; 18(9): e1010316, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36103568

RESUMO

The evolutionarily successful poxviruses possess effective and diverse strategies to circumvent or overcome host defense mechanisms. Poxviruses encode many immunoregulatory proteins to evade host immunity to establish a productive infection and have unique means of inhibiting DNA sensing-dependent type 1 interferon (IFN-I) responses, a necessity given their dsDNA genome and exclusively cytoplasmic life cycle. We found that the key DNA sensing inhibition by poxvirus infection was dominant during the early stage of poxvirus infection before DNA replication. In an effort to identify the poxvirus gene products which subdue the antiviral proinflammatory responses (e.g., IFN-I response), we investigated the function of one early gene that is the known host range determinant from the highly conserved poxvirus host range C7L superfamily, myxoma virus (MYXV) M062. Host range factors are unique features of poxviruses that determine the species and cell type tropism. Almost all sequenced mammalian poxviruses retain at least one homologue of the poxvirus host range C7L superfamily. In MYXV, a rabbit-specific poxvirus, the dominant and broad-spectrum host range determinant of the C7L superfamily is the M062R gene. The M062R gene product is essential for MYXV infection in almost all cells tested from different mammalian species and specifically inhibits the function of host Sterile α Motif Domain-containing 9 (SAMD9), as M062R-null (ΔM062R) MYXV causes abortive infection in a SAMD9-dependent manner. In this study we investigated the immunostimulatory property of the ΔM062R. We found that the replication-defective ΔM062R activated host DNA sensing pathway during infection in a cGAS-dependent fashion and that knocking down SAMD9 expression attenuated proinflammatory responses. Moreover, transcriptomic analyses showed a unique feature of the host gene expression landscape that is different from the dsDNA alone-stimulated inflammatory state. This study establishes a link between the anti-neoplastic function of SAMD9 and the regulation of innate immune responses.


Assuntos
Interferon Tipo I , Myxoma virus , Infecções por Poxviridae , Poxviridae , Animais , Especificidade de Hospedeiro/genética , Humanos , Interferon Tipo I/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macrófagos/metabolismo , Mamíferos , Monócitos/metabolismo , Myxoma virus/genética , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Poxviridae/genética , Poxviridae/metabolismo , Infecções por Poxviridae/genética , Coelhos , Transcriptoma , Vaccinia virus/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo
6.
Microbiol Spectr ; 10(3): e0027222, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35583360

RESUMO

Poxviruses have large DNA genomes, and they are able to infect multiple vertebrate and invertebrate animals, including humans. Despite the eradication of smallpox, poxvirus infections still remain a significant public health concern. Vaccinia virus (VV) is the prototypic member in the poxviridae family and it has been used extensively for different prophylactic applications, including the generation of vaccines against multiple infectious diseases and/or for oncolytic treatment. Many attempts have been pursued to develop novel attenuated forms of VV with improved safety profiles for their implementation as vaccines and/or vaccines vectors. We and others have previously demonstrated how RNA viruses encoding codon-deoptimized viral genes are attenuated, immunogenic and able to protect, upon a single administration, against challenge with parental viruses. In this study, we employed the same experimental approach based on the use of misrepresented codons for the generation of a recombinant (r)VV encoding a codon-deoptimized A24R gene, which is a key component of the viral RNA polymerase. Similar to our previous studies with RNA viruses, the A24R codon-deoptimized rVV (v-A24cd) was highly attenuated in vivo but able to protect, after a single intranasal dose administration, against an otherwise lethal challenge with parental VV. These results indicate that poxviruses can be effectively attenuated by synonymous codon deoptimization and open the possibility of using this methodology alone or in combination with other experimental approaches for the development of attenuated vaccines for the treatment of poxvirus infection, or to generate improved VV-based vectors. Moreover, this approach could be applied to other DNA viruses. IMPORTANCE The family poxviridae includes multiple viruses of medical and veterinary relevance, being vaccinia virus (VV) the prototypic member in the family. VV was used during the smallpox vaccination campaign to eradicate variola virus (VARV), which is considered a credible bioterrorism threat. Because of novel innovations in genetic engineering and vaccine technology, VV has gained popularity as a viral vector for the development of vaccines against several infectious diseases. Several approaches have been used to generate attenuated VV for its implementation as vaccine and/or vaccine vector. Here, we generated a rVV containing a codon-deoptimized A24R gene (v-A24cd), which encodes a key component of the viral RNA polymerase. v-A24cd was stable in culture cells and highly attenuated in vivo but able to protect against a subsequent lethal challenge with parental VV. Our findings support the use of this approach for the development of safe, stable, and protective live-attenuated VV and/or vaccine vectors.


Assuntos
Poxviridae , Varíola , Vacinas Virais , Vírus , Animais , Códon , Poxviridae/genética , Desenvolvimento de Vacinas , Vacinas Atenuadas/genética , Vaccinia virus/genética , Proteínas do Complexo da Replicase Viral , Vacinas Virais/genética , Vírus/genética
7.
Front Immunol ; 13: 1050250, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36713447

RESUMO

Poxvirus vectors represent versatile modalities for engineering novel vaccines and cancer immunotherapies. In addition to their oncolytic capacity and immunogenic influence, they can be readily engineered to express multiple large transgenes. However, the integration of multiple payloads into poxvirus genomes by traditional recombination-based approaches can be highly inefficient, time-consuming and cumbersome. Herein, we describe a simple, cost-effective approach to rapidly generate and purify a poxvirus vector with multiple transgenes. By utilizing a simple, modular CRISPR/Cas9 assisted-recombinant vaccinia virus engineering (CARVE) system, we demonstrate generation of a recombinant vaccinia virus expressing three distinct transgenes at three different loci in less than 1 week. We apply CARVE to rapidly generate a novel immunogenic vaccinia virus vector, which expresses a bacterial diadenylate cyclase. This novel vector, STINGPOX, produces cyclic di-AMP, a STING agonist, which drives IFN signaling critical to the anti-tumor immune response. We demonstrate that STINGPOX can drive IFN signaling in primary human cancer tissue explants. Using an immunocompetent murine colon cancer model, we demonstrate that intratumoral administration of STINGPOX in combination with checkpoint inhibitor, anti-PD1, promotes survival post-tumour challenge. These data demonstrate the utility of CRISPR/Cas9 in the rapid arming of poxvirus vectors with therapeutic payloads to create novel immunotherapies.


Assuntos
Neoplasias , Poxviridae , Humanos , Animais , Camundongos , Vetores Genéticos/genética , Vaccinia virus , Poxviridae/genética , Imunoterapia
8.
Sci Rep ; 11(1): 23476, 2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34873256

RESUMO

African swine fever virus (ASFV) causes a highly contagious hemorrhagic disease that affects domestic pig and Eurasian wild boar populations. To date, no safe and efficacious treatment or vaccine against ASF is available. Nevertheless, there are several reports of protection elicited by experimental vaccines based on live attenuated ASFV and some levels of protection and reduced viremia in other approaches such as DNA, adenovirus, baculovirus, and vaccinia-based vaccines. Current ASF subunit vaccine research focuses mainly on delivering protective antigens and antigen discovery within the ASFV genome. However, due to the complex nature of ASFV, expression vectors need to be optimized to improve their immunogenicity. Therefore, in the present study, we constructed several recombinant MVA vectors to evaluate the efficiency of different promoters and secretory signal sequences in the expression and immunogenicity of the p30 protein from ASFV. Overall, the natural poxvirus PrMVA13.5L promoter induced high levels of both p30 mRNA and specific anti-p30 antibodies in mice. In contrast, the synthetic PrS5E promoter and the S E/L promoter linked to a secretory signal showed lower mRNA levels and antibodies. These findings indicate that promoter selection may be as crucial as the antigen used to develop ASFV subunit vaccines using MVA as the delivery vector.


Assuntos
Vírus da Febre Suína Africana/genética , Poxviridae/genética , Vacinas de Subunidades Antigênicas/genética , Proteínas Virais/genética , Vacinas Virais/genética , Febre Suína Africana/virologia , Animais , Anticorpos Antivirais/genética , Baculoviridae/genética , Linhagem Celular , Chlorocebus aethiops , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Regiões Promotoras Genéticas/genética , Suínos , Vacinas Atenuadas/genética , Células Vero , Replicação Viral/genética
9.
Virol J ; 18(1): 167, 2021 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-34391449

RESUMO

BACKGROUND: Poxviruses within the Capripoxvirus, Orthopoxvirus, and Parapoxvirus genera can infect livestock, with the two former having zoonotic importance. In addition, they induce similar clinical symptoms in common host species, creating a challenge for diagnosis. Although endemic in the country, poxvirus infections of small ruminants and cattle have received little attention in Botswana, with no prior use of molecular tools to diagnose and characterize the pathogens. METHODS: A high-resolution melting (HRM) assay was used to detect and differentiate poxviruses in skin biopsy and skin scab samples from four cattle, one sheep, and one goat. Molecular characterization of capripoxviruses and parapoxviruses was undertaken by sequence analysis of RPO30 and GPCR genes. RESULTS: The HRM assay revealed lumpy skin disease virus (LSDV) in three cattle samples, pseudocowpox virus (PCPV) in one cattle sample, and orf virus (ORFV) in one goat and one sheep sample. The phylogenetic analyses, based on the RPO30 and GPCR multiple sequence alignments showed that the LSDV sequences of Botswana were similar to common LSDV field isolates encountered in Africa, Asia, and Europe. The Botswana PCPV presented unique features and clustered between camel and cattle PCPV isolates. The Botswana ORFV sequence isolated from goat differed from the ORFV sequence isolated from sheep. CONCLUSIONS: This study is the first report on the genetic characterization of poxvirus diseases circulating in cattle, goats, and sheep in Botswana. It shows the importance of molecular methods to differentially diagnose poxvirus diseases of ruminants.


Assuntos
Vírus da Doença Nodular Cutânea/genética , Vírus do Orf , Infecções por Poxviridae , Poxviridae , Vírus da Pseudovaríola das Vacas , Animais , Botsuana/epidemiologia , Bovinos , Cabras , Vírus do Orf/genética , Filogenia , Poxviridae/genética , Infecções por Poxviridae/epidemiologia , Infecções por Poxviridae/veterinária , Vírus da Pseudovaríola das Vacas/genética , Ruminantes , Ovinos , Doenças dos Ovinos/epidemiologia
11.
Viruses ; 12(12)2020 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-33291286

RESUMO

The presence of carp edema virus (CEV) was confirmed in imported ornamental koi in Chiang Mai province, Thailand. The koi showed lethargy, loss of swimming activity, were lying at the bottom of the pond, and gasping at the water's surface. Some clinical signs such as skin hemorrhages and ulcers, swelling of the primary gill lamella, and necrosis of gill tissue, presented. Clinical examination showed co-infection by opportunistic pathogens including Dactylogyrus sp., Gyrodactylus sp. and Saprolegnia sp. on the skin and gills. Histopathologically, the gill of infected fish showed severe necrosis of epithelial cells and infiltrating of eosinophilic granular cells. Electron microscope examination detected few numbers of virions were present in the cytoplasm of gill tissue which showed an electron dense core with surface membranes worn by surface globular units. Molecular detection of CEV DNA from gill samples of fish was performed by polymerase chain reaction (PCR) and confirmed by nested-PCR. Phylogenetic analyses revealed that CEV isolate had 99.8% homology with the CEV isolated from South Korea (KY946715) and Germany (KY550420), and was assigned to genogroup IIa. In conclusion, this report confirmed the presence of CEV infection of koi Cyprinus carpio in Chiang Mai province, Thailand using pathological and molecular approaches.


Assuntos
Carpas/virologia , Doenças dos Peixes/virologia , Poxviridae , Animais , Biópsia , Genes Virais , Brânquias/patologia , Brânquias/virologia , Filogenia , Reação em Cadeia da Polimerase , Poxviridae/classificação , Poxviridae/genética , Poxviridae/ultraestrutura , Tailândia
12.
Curr Opin Virol ; 44: 183-190, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33242829

RESUMO

The potential of viruses as biotechnology platforms is becoming more appealing due to technological advances in synthetic biology techniques and to the increasing accessibility of means to manipulate virus genomes. Among viral systems, poxviruses, and their prototype member Vaccinia Virus, are one of the outstanding choices for different biotechnological and medical applications based on heterologous gene expression, recombinant vaccines or oncolytic viruses. The refinement of genetic engineering methods on Vaccinia Virus over the last decades have contributed to facilitate the manipulation of the genomes of poxviruses, and may aid in the improvement of virus variants designed for different goals through reverse genetic approaches. Targeted genetic changes are usually performed by homologous recombination with the viral genome. In addition to the classic approach, recent methodological advances that may assist new strategies for the mutation or edition of poxvirus genomes are reviewed.


Assuntos
Expressão Gênica , Engenharia Genética/métodos , Vetores Genéticos , Genoma Viral , Poxviridae/genética , Animais , Edição de Genes/métodos , Camundongos , Mutação , Poxviridae/classificação , Recombinação Genética , Vacinas Sintéticas/genética
13.
Mol Cell Probes ; 54: 101668, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33059011

RESUMO

Cyprinid herpesvirus 1 (CyHV-1) is the causative agent of carp pox characterized by epidermal papillomas in common carp and other cyprinids. In this study, we identified CyHV-1 in koi (Cyprinus carpio) from Iran in 2017 and 2019, showing clinical signs of the carp pox disease. Histopathology showed severe epidermal hyperplasia and the absence of club and goblet cells. Degenerative changes, including spongiosis and single-cell necrosis, were also observed. Keratinocyte dysplasia and a moderate lymphocytic infiltration were observed within the epidermis. PCR of the extracted DNA from skin lesions of affected koi from both outbreaks showed CyHV-1 specific TK amplicons, with high sequence identity (above 99%) among themselves and with other CyHV-1 isolates belong to Cluster I, as well as show 97% similarity to Cluster II isolates. To the best of our knowledge, this is the first report of Carp pox disease (CyHV-1) of koi in Iran and the Middle East.


Assuntos
Carpas/virologia , Infecções por Poxviridae/patologia , Infecções por Poxviridae/virologia , Poxviridae/genética , Poxviridae/isolamento & purificação , Sequência de Aminoácidos , Animais , Sequência de Bases , Irã (Geográfico) , Filogenia , Reação em Cadeia da Polimerase , Infecções por Poxviridae/veterinária , Pele/patologia , Pele/virologia
14.
J Vis Exp ; (159)2020 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-32510495

RESUMO

Vaccinia virus (VACV) was instrumental in eradicating variola virus (VARV), the causative agent of smallpox, from nature. Since its first use as a vaccine, VACV has been developed as a vector for therapeutic vaccines and as an oncolytic virus. These applications take advantage of VACV's easily manipulated genome and broad host range as an outstanding platform to generate recombinant viruses with a variety of therapeutic applications. Several methods have been developed to generate recombinant VACV, including marker selection methods and transient dominant selection. Here, we present a refinement of a host range selection method coupled with visual identification of recombinant viruses. Our method takes advantage of selective pressure generated by the host antiviral protein kinase R (PKR) coupled with a fluorescent fusion gene expressing mCherry-tagged E3L, one of two VACV PKR antagonists. The cassette, including the gene of interest and the mCherry-E3L fusion is flanked by sequences derived from the VACV genome. Between the gene of interest and mCherry-E3L is a smaller region that is identical to the first ~150 nucleotides of the 3' arm, to promote homologous recombination and loss of the mCherry-E3L gene after selection. We demonstrate that this method permits efficient, seamless generation of rVACV in a variety of cell types without requiring drug selection or extensive screening for mutant viruses.


Assuntos
Vetores Genéticos/administração & dosagem , Proteínas de Fluorescência Verde/metabolismo , Rim/metabolismo , Infecções por Poxviridae/metabolismo , Poxviridae/genética , eIF-2 Quinase/metabolismo , Animais , Células Cultivadas , Especificidade de Hospedeiro , Humanos , Rim/citologia , Rim/virologia , Infecções por Poxviridae/virologia , Coelhos , Vaccinia virus/genética
15.
Vopr Virusol ; 65(1): 49-56, 2020.
Artigo em Russo | MEDLINE | ID: mdl-32496721

RESUMO

INTRODUCTION: Currently, new directions in cancer therapy are actively developing, one of which is oncolytic immunotherapy. This approach would be to use of viruses as cancer specific cytolytic agents capable of stimulating both the tumor-specific and non-specific immune response. The objective paper was obtain a recombinant vaccinia virus containing genes encoding immunostimulating molecules and study oncolytic and immunostimulating properties of recombinant virus. MATERIAL AND METHODS: MTT test, ELISA, methods of transient dominant selection. RESULTS: The recombinant vaccinia virus (L-IVP_oncoB) were obtained with deletion of the gene encoding thymidine kinase and had an integrated gene encoding GM-CSF. Also the virus have deletion of the gene encoding viral growth factor and integrated genes encoding synthetic tumor-specific polyepitopic immunogens. It was shown that the modifications made to the viral genome did not affect the growth characteristics of the virus when cultured on CV-1 and 4647 cell cultures, and the cytopathogenic efficacy of the virus was determined in relation to cancer cultures of cells of various genesis. In in vivo experiment, it was revealed that the polyepitopic construct in the genome L-IVP_oncoB is able to initiate a change in the profile of cytokines. DISCUSSION: The obtained data characterized L-IVP_oncoB as a promising cytopathogenic and immunostimulating agent and showed the need for further study of its properties as means of oncolytic immunotherapy. CONCLUSION: The basic experiments on the evaluation of the biological properties of the obtained L-IVP_oncoB, which are necessary for the characterization of the oncolytic virus, have been carried out.


Assuntos
Neoplasias da Mama/terapia , Vírus Oncolíticos/genética , Vaccinia virus/genética , Replicação Viral/genética , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/virologia , Linhagem Celular Tumoral , Feminino , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Humanos , Imunoterapia , Orthopoxvirus/genética , Poxviridae/genética , Replicação Viral/imunologia
16.
Cancer Gene Ther ; 27(3-4): 125-135, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31209267

RESUMO

Oncolytic viruses have shown excellent safety profiles in preclinical and clinical studies; however, in most cases therapeutic benefits have been modest. We have previously reported the generation of a chimeric poxvirus (CF33), with significantly improved oncolytic characteristics, through chimerization among different poxviruses. Here we report the sequence analysis of CF33 and oncolytic potential of a GFP-encoding CF33 virus (CF33-GFP) with a J2R deletion in lung cancer models. Replication of CF33-GFP and the resulting cytotoxicity were higher in cancer cell lines compared to a normal cell line, in vitro. After infection with virus, cancer cells expressed markers for immunogenic cell death in vitro. Furthermore, CF33-GFP was safe and exerted potent anti-tumor effects at a dose as low as 1000 plaque forming units in both virus-injected and un-injected distant tumors in A549 tumor xenograft model in mice. Likewise, in a syngeneic model of lung cancer in mice, the virus showed significant anti-tumor effect and was found to increase tumor infiltration by CD8+ T cells. Collectively, these data warrant further investigation of this novel chimeric poxvirus for its potential use as a cancer bio-therapeutic.


Assuntos
Neoplasias Pulmonares/terapia , Terapia Viral Oncolítica/métodos , Poxviridae/genética , Células A549 , Animais , Linfócitos T CD8-Positivos/imunologia , Chlorocebus aethiops , Feminino , Deleção de Genes , Genes Reporter/genética , Proteínas de Fluorescência Verde/genética , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Poxviridae/imunologia , Timidina Quinase/genética , Proteínas Virais/genética , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Virol ; 94(4)2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31776279

RESUMO

On-site translation of mRNAs provides an efficient means of subcellular protein localization. In eukaryotic cells, the transport of cellular mRNAs to membraneless sites usually occurs prior to translation and involves specific sequences known as zipcodes that interact with RNA binding and motor proteins. Poxviruses replicate in specialized cytoplasmic factory regions where DNA synthesis, transcription, translation, and virion assembly occur. Some poxviruses embed infectious virus particles outside of factories in membraneless protein bodies with liquid gel-like properties known as A-type inclusions (ATIs) that are comprised of numerous copies of the viral 150-kDa ATI protein. Here, we demonstrate by fluorescent in situ hybridization that these inclusions are decorated with ATI mRNA. On-site translation is supported by the localization of a translation initiation factor eIF4E and by ribosome-bound nascent chain ribopuromycylation. Nascent peptide-mediated anchoring of ribosome-mRNA translation complexes to the inclusions is suggested by release of the mRNA by puromycin, a peptide chain terminator. Following puromycin washout, relocalization of ATI mRNA at inclusions depends on RNA and protein synthesis but requires neither microtubules nor actin polymerization. Further studies show that the ATI mRNAs remain near the sites of transcription in the factory regions when stop codons are introduced near the N terminus of the ATI or large truncations are made at the N or C termini. Instead of using a zipcode, we propose that ATI mRNA localization is mediated by ribosome-bound nascent ATI polypeptides that interact with ATI protein in inclusions and thereby anchor the complex for multiple rounds of mRNA translation.IMPORTANCE Poxvirus genome replication, transcription, translation, and virion assembly occur at sites within the cytoplasm known as factories. Some poxviruses sequester infectious virions outside of the factories in inclusion bodies comprised of numerous copies of the 150-kDa ATI protein, which can provide stability and protection in the environment. We provide evidence that ATI mRNA is anchored by nascent peptides and translated at the inclusion sites rather than in virus factories. Association of ATI mRNA with inclusion bodies allows multiple rounds of local translation and prevents premature ATI protein aggregation and trapping of virions within the factory.


Assuntos
Vaccinia virus/metabolismo , Proteínas Virais/metabolismo , Replicação Viral/genética , Citoplasma/metabolismo , Replicação do DNA , Fator de Iniciação 4E em Eucariotos/metabolismo , Células HeLa , Humanos , Corpos de Inclusão/metabolismo , Corpos de Inclusão Viral/virologia , Poxviridae/genética , Poxviridae/metabolismo , Proteínas com Motivo de Reconhecimento de RNA/genética , RNA Mensageiro/genética , Motivos de Ligação ao RNA/genética , Ribossomos/metabolismo , Vaccinia virus/genética , Proteínas Virais/genética , Vírion/metabolismo , Montagem de Vírus/genética
18.
Front Immunol ; 10: 2005, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31497029

RESUMO

Seasonal influenza viruses cause significant morbidity and mortality in the global population every year. Although seasonal vaccination limits disease, mismatches between the circulating strain and the vaccine strain can severely impair vaccine effectiveness. Because of this, there is an urgent need for a universal vaccine that induces broad protection against drifted seasonal and emerging pandemic influenza viruses. Targeting the conserved stalk region of the influenza virus hemagglutinin (HA), the major glycoprotein on the surface of the virus, results in the production of broadly protective antibody responses. Furthermore, replication deficient viral vectors based on Chimpanzee Adenovirus Oxford 1 (ChAdOx1) and modified vaccinia Ankara (MVA) virus expressing the influenza virus internal antigens, the nucleoprotein (NP) and matrix 1 (M1) protein, can induce strong heterosubtypic influenza virus-specific T cell responses in vaccinated individuals. Here, we combine these two platforms to evaluate the efficacy of a viral vectored vaccination regimen in protecting ferrets from H3N2 influenza virus infection. We observed that viral vectored vaccines expressing both stalk-targeting, chimeric HA constructs, and the NP+M1 fusion protein, in a prime-boost regimen resulted in the production of antibodies toward group 2 HAs, the HA stalk, NP and M1, as well as in induction of influenza virus-specific-IFNγ responses. The immune response induced by this vaccination regime ultimately reduced viral titers in the respiratory tract of influenza virus infected ferrets. Overall, these results improve our understanding of vaccination platforms capable of harnessing both cellular and humoral immunity with the goal of developing a universal influenza virus vaccine.


Assuntos
Antígenos Virais/genética , Hemaglutininas/genética , Vírus da Influenza A Subtipo H3N2 , Vacinas contra Influenza , Infecções por Orthomyxoviridae/prevenção & controle , Proteínas de Ligação a RNA/genética , Proteínas do Core Viral/genética , Proteínas da Matriz Viral/genética , Adenoviridae/genética , Animais , Antígenos Virais/imunologia , Linhagem Celular , Embrião de Galinha , Cães , Furões , Vetores Genéticos , Hemaglutininas/imunologia , Humanos , Vírus da Influenza A Subtipo H3N2/imunologia , Insetos , Masculino , Proteínas do Nucleocapsídeo , Infecções por Orthomyxoviridae/imunologia , Poxviridae/genética , Proteínas de Ligação a RNA/imunologia , Vacinação , Proteínas do Core Viral/imunologia , Proteínas da Matriz Viral/imunologia
19.
Methods Mol Biol ; 1988: 217-248, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31147943

RESUMO

The study of antigen processing and presentation is critical to our understanding of the mechanisms that govern immune surveillance. A typical requirement of assays designed to examine antigen processing and presentation is the de novo biosynthesis of a model antigen. Historically, Vaccinia virus, a poxvirus closely related to Cowpox virus, has enjoyed widespread use for this purpose. Recombinant poxvirus-based expression has a number of advantages over other systems. Poxviruses accommodate the insertion of large pieces of recombinant DNA into their genome, and recombination and selection are relatively efficient. Poxviruses readily infect a variety of cell types, and they drive rapid and high levels of antigen expression. Additionally, they can be utilized in a variety of assays to study both MHC class I restricted and MHC class II restricted antigen processing and presentation. Ultimately, the numerous advantages of poxvirus recombinants have made the Vaccinia expression system a mainstay in the study of processing and presentation over the past two decades. In an attempt to address one shortcoming of Vaccinia virus while simultaneously retaining the benefits inherent to poxviruses, our laboratory has begun to engineer recombinant Ectromelia viruses. Ectromelia virus, or mousepox, is a natural pathogen of murine cells and performing experiments in the context of a natural host-pathogen relationship may elucidate unknown factors that influence epitope generation and host response. This chapter will describe several recombinant poxvirus system protocols used to study both MHC class I and class II antigen processing and presentation, as well as provide insight and troubleshooting techniques to improve the reproducibility and fidelity of these experiments.


Assuntos
Imunoensaio/métodos , Poxviridae/genética , Recombinação Genética/genética , Animais , Citocinas/metabolismo , Citotoxicidade Imunológica , ELISPOT , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Camundongos , Peptídeos/metabolismo , Infecções por Poxviridae/virologia , Coloração e Rotulagem , Linfócitos T/imunologia
20.
J Virol ; 93(13)2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30996093

RESUMO

Vaccinia virus is a promising viral vaccine and gene delivery candidate and has historically been used as a model to study poxvirus-host cell interactions. We employed a genome-wide insertional mutagenesis approach in human haploid cells to identify host factors crucial for vaccinia virus infection. A library of mutagenized HAP1 cells was exposed to modified vaccinia virus Ankara (MVA). Deep-sequencing analysis of virus-resistant cells identified host factors involved in heparan sulfate synthesis, Golgi organization, and vesicular protein trafficking. We validated EXT1, TM9SF2, and TMED10 (TMP21/p23/p24δ) as important host factors for vaccinia virus infection. The critical roles of EXT1 in heparan sulfate synthesis and vaccinia virus infection were confirmed. TM9SF2 was validated as a player mediating heparan sulfate expression, explaining its contribution to vaccinia virus infection. In addition, TMED10 was found to be crucial for virus-induced plasma membrane blebbing and phosphatidylserine-induced macropinocytosis, presumably by regulating the cell surface expression of the TAM receptor Axl.IMPORTANCE Poxviruses are large DNA viruses that can infect a wide range of host species. A number of these viruses are clinically important to humans, including variola virus (smallpox) and vaccinia virus. Since the eradication of smallpox, zoonotic infections with monkeypox virus and cowpox virus are emerging. Additionally, poxviruses can be engineered to specifically target cancer cells and are used as a vaccine vector against tuberculosis, influenza, and coronaviruses. Poxviruses rely on host factors for most stages of their life cycle, including attachment to the cell and entry. These host factors are crucial for virus infectivity and host cell tropism. We used a genome-wide knockout library of host cells to identify host factors necessary for vaccinia virus infection. We confirm a dominant role for heparin sulfate in mediating virus attachment. Additionally, we show that TMED10, previously not implicated in virus infections, facilitates virus uptake by modulating the cellular response to phosphatidylserine.


Assuntos
Haploidia , Heparitina Sulfato/genética , Heparitina Sulfato/isolamento & purificação , Pinocitose/fisiologia , Vaccinia virus/genética , Vaccinia virus/metabolismo , Vacínia/virologia , Proteínas de Transporte Vesicular/metabolismo , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Vírus da Varíola Bovina/genética , Vírus de DNA , Técnicas de Inativação de Genes , Testes Genéticos , Complexo de Golgi , Células HEK293 , Células HeLa , Heparitina Sulfato/metabolismo , Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno , Humanos , Proteínas de Membrana , Monkeypox virus/genética , N-Acetilglucosaminiltransferases , Fosfatidilserinas/metabolismo , Poxviridae/genética , Ligação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA