Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
mSphere ; 9(6): e0023624, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38757961

RESUMO

Mammalian orthoreovirus (MRV) outer capsid protein σ3 is a multifunctional protein containing a double-stranded RNA-binding domain, which facilitates viral entry and assembly. We reasoned that σ3 has an innate immune evasion function. Here, we show that σ3 protein localizes in the mitochondria and interacts with mitochondrial antiviral signaling protein (MAVS) to activate the intrinsic mitochondria-mediated apoptotic pathway. Consequently, σ3 protein promotes the degradation of MAVS through the intrinsic caspase-9/caspase-3 apoptotic pathway. Moreover, σ3 protein can also inhibit the expression of the components of the RNA-sensing retinoic acid-inducible gene (RIG)-like receptor (RLR) signaling pathway to block antiviral type I interferon responses. Mechanistically, σ3 inhibits RIG-I and melanoma differentiation-associated gene 5 expression is independent of its inhibitory effect on MAVS. Overall, we demonstrate that the MRV σ3 protein plays a vital role in negatively regulating the RLR signaling pathway to inhibit antiviral responses. This enables MRV to evade host defenses to facilitate its own replication providing a target for the development of effective antiviral drugs against MRV. IMPORTANCE: Mammalian orthoreovirus (MRV) is an important zoonotic pathogen, but the regulatory role of its viral proteins in retinoic acid-inducible gene-like receptor (RLR)-mediated antiviral responses is still poorly understood. Herein, we show that MRV σ3 protein co-localizes with mitochondrial antiviral signaling protein (MAVS) in the mitochondria and promotes the mitochondria-mediated intrinsic apoptotic pathway to cleave and consequently degrade MAVS. Furthermore, tryptophan at position 133 of σ3 protein plays a key role in the degradation of MAVS. Importantly, we show that MRV outer capsid protein σ3 is a key factor in antagonizing RLR-mediated antiviral responses, providing evidence to better unravel the infection and transmission mechanisms of MRV.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas do Capsídeo , Orthoreovirus de Mamíferos , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Humanos , Orthoreovirus de Mamíferos/genética , Animais , Apoptose , Proteína DEAD-box 58/metabolismo , Proteína DEAD-box 58/genética , Mitocôndrias/metabolismo , Imunidade Inata , Camundongos , Evasão da Resposta Imune , Células HEK293 , Receptores Imunológicos/metabolismo , Helicase IFIH1 Induzida por Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon/genética , Linhagem Celular , Interações Hospedeiro-Patógeno
2.
J Virol ; 98(6): e0030524, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38771042

RESUMO

Many functions of viral attachment proteins are established, but less is known about the biological importance of viral attachment protein encapsidation efficiency. The mammalian orthoreovirus (reovirus) σ1 attachment protein forms filamentous trimers that incorporate into pentamers of the λ2 capsid protein. Reovirus strains vary in the efficiency of σ1 encapsidation onto progeny virions, which influences viral stability during entry into cells and the efficacy of tumor cell lysis. While the role of σ1 encapsidation has been evaluated in studies using cultured cells, the contribution of attachment protein encapsidation efficiency to viral infection in animals is less clear. Polymorphisms in reovirus σ1 at residues 22 and 249 have been implicated in viral dissemination in mice and susceptibility to proteolysis in the murine intestine, respectively. To determine whether these residues contribute to σ1 encapsidation efficiency, we engineered σ1 mutant viruses with single- and double-residue substitutions at sites 22 and 249. We found that substitutions at these sites alter the encapsidation of σ1 and that reoviruses encapsidating higher amounts of σ1 bind cells more avidly and have a modest replication advantage in a cell-type-specific manner relative to low σ1-encapsidating reoviruses. Furthermore, we found that a high σ1-encapsidating reovirus replicates and disseminates more efficiently in mice relative to a low σ1-encapsidating reovirus. These findings provide evidence of a relationship between viral attachment protein encapsidation efficiency and viral replication in cell culture and animal hosts. IMPORTANCE: Viral attachment proteins can serve multiple functions during viral replication, including attachment to host cells, cell entry and disassembly, and modulation of host immune responses. The relationship between viral attachment protein encapsidation efficiency and viral replication in cells and animals is poorly understood. We engineered and characterized a panel of reoviruses that differ in the capacity to encapsidate the σ1 attachment protein. We found that strains encapsidating σ1 with higher efficiency bind cells more avidly and replicate and spread more efficiently in mice relative to those encapsidating σ1 with lower efficiency. These results highlight a function for σ1 attachment protein capsid abundance in viral replication in cells and animals, which may inform future use of reovirus as an oncolytic therapeutic.


Assuntos
Proteínas do Capsídeo , Infecções por Reoviridae , Replicação Viral , Animais , Camundongos , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Infecções por Reoviridae/virologia , Infecções por Reoviridae/metabolismo , Ligação Viral , Polimorfismo Genético , Orthoreovirus de Mamíferos/genética , Orthoreovirus de Mamíferos/fisiologia , Orthoreovirus de Mamíferos/metabolismo , Montagem de Vírus , Linhagem Celular , Capsídeo/metabolismo , Humanos
3.
Microbiol Spectr ; 12(3): e0176223, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38289932

RESUMO

Mammalian orthoreoviruses (MRVs) infect a wide range of hosts, including humans, livestock, and wildlife. In the present study, we isolated a novel Mammalian orthoreovirus from the intestine of a microbat (Myotis aurascens) and investigated its biological and pathological characteristics. Phylogenetic analysis indicated that the new isolate was serotype 2, sharing the segments with those from different hosts. Our results showed that it can infect a wide range of cell lines from different mammalian species, including human, swine, and non-human primate cell lines. Additionally, media containing trypsin, yeast extract, and tryptose phosphate broth promoted virus propagation in primate cell lines and most human cell lines, but not in A549 and porcine cell lines. Mice infected with this strain via the intranasal route, but not via the oral route, exhibited weight loss and respiratory distress. The virus is distributed in a broad range of organs and causes lung damage. In vitro and in vivo experiments also suggested that the new virus could be a neurotropic infectious strain that can infect a neuroblastoma cell line and replicate in the brains of infected mice. Additionally, it caused a delayed immune response, as indicated by the high expression levels of cytokines and chemokines only at 14 days post-infection (dpi). These data provide an important understanding of the genetics and pathogenicity of mammalian orthoreoviruses in bats at risk of spillover infections.IMPORTANCEMammalian orthoreoviruses (MRVs) have a broad range of hosts and can cause serious respiratory and gastroenteritis diseases in humans and livestock. Some strains infect the central nervous system, causing severe encephalitis. In this study, we identified BatMRV2/SNU1/Korea/2021, a reassortment of MRV serotype 2, isolated from bats with broad tissue tropism, including the neurological system. In addition, it has been shown to cause respiratory syndrome in mouse models. The given data will provide more evidence of the risk of mammalian orthoreovirus transmission from wildlife to various animal species and the sources of spillover infections.


Assuntos
Quirópteros , Orthoreovirus de Mamíferos , Camundongos , Animais , Suínos , Orthoreovirus de Mamíferos/genética , Filogenia , Virulência , Animais Selvagens , República da Coreia , Primatas
4.
Virology ; 587: 109871, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37634292

RESUMO

Mammalian orthoreovirus (MRV) is an oncolytic virus that has been tested in over 30 clinical trials. Increased clinical success has been achieved when MRV is used in combination with other onco-immunotherapies. This has led the field to explore the creation of recombinant MRVs which incorporate immunotherapeutic sequences into the virus genome. This work focuses on creation and characterization of a recombinant MRV, S1/HER2nhd, which encodes a truncated σ1 protein fused in frame with three human epidermal growth factor receptor 2 (HER2) peptides (E75, AE36, and GP2) known to induce HER2 specific CD8+ and CD4+ T cells. We show S1/HER2nhd expresses the σ1 fusion protein containing HER2 peptides in infected cells and on the virion, and infects, replicates in, and reduces survival of HER2+ breast cancer cells. The oncolytic properties of MRV combined with HER2 peptide expression holds potential as a vaccine to prevent recurrences of HER2 expressing cancers.


Assuntos
Neoplasias , Orthoreovirus de Mamíferos , Animais , Humanos , Orthoreovirus de Mamíferos/genética , Proteínas Recombinantes de Fusão/genética , Peptídeos , Mamíferos
5.
DNA Cell Biol ; 42(6): 289-304, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37015068

RESUMO

Orthoreovirus is a nonenveloped double-stranded RNA virus under the Reoviridae family. This group of viruses, especially mammalian orthoreovirus (MRV), are reported with great therapeutic values due to their oncolytic effects. In this review, the life cycle and oncolytic effect of MRV and a few emerging reoviruses were summarized. This article also highlights the challenges and strategies of utilizing MRV and the emerging reoviruses, avian orthoreovirus (ARV) and pteropine orthoreovirus (PRV), as oncolytic viruses (OVs). Besides, the emergence of potential ARV and PRV as OVs were discussed in comparison to MRV. Finally, the risk of reovirus as zoonosis or reverse zoonosis (zooanthroponosis) were debated, and concerns were raised in this article, which warrant continue surveillance of reovirus (MRV, ARV, and PRV) in animals, humans, and the environment.


Assuntos
Vírus Oncolíticos , Orthoreovirus de Mamíferos , Orthoreovirus , Reoviridae , Animais , Humanos , Orthoreovirus/genética , Reoviridae/genética , Orthoreovirus de Mamíferos/genética , Vírus Oncolíticos/genética , Mamíferos
6.
J Virol Methods ; 308: 114574, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35798198

RESUMO

Fluorescence-guided surgery (FGS) is a useful method for removing invasive tumor tissues. For this, near-infrared (NIR) fluorescence probes are suitable for visualizing cancer cells due to their low autofluorescence, and an oncolytic mammalian orthoreovirus (MRV) expressing an NIR fluorescent protein is expected to be a novel tool for FGS. In this study, we identified the optimal insertion site of the NIR fluorescent protein gene iRFP720 (915 nt) in the MRV genome. We constructed genome plasmids for the L1, M1, and S2 segments, where a gene cassette comprising iRFP720 and T2A self-cleaving peptide was inserted in the 5' or 3' region of each segment. Through virus recovery, the recombinant MRV with the gene cassette at the M1 segment's 3' end, T3D-L(M1/3'iRFP720), was capable of replication and passaging with bright NIR fluorescence. However, the replication of T3D-L(M1/3'iRFP720) was approximately 1,000-fold lower than that of the wild-type virus. T3D-L(M1/3'iRFP720) production improved due to the transfection of a fusion-associated small transmembrane protein gene of fusogenic reovirus. Further, fluorescence signals were detected in T3D-L(M1/3'iRFP720)-infected human gastric and pancreatic cancer cells. Thus, the M1 segment's 3' end tolerates the expression of the long iRFP720 gene, which may propel the development of recombinant MRV vectors for FGS.


Assuntos
Orthoreovirus de Mamíferos , Reoviridae , Animais , Humanos , Mamíferos/genética , Orthoreovirus de Mamíferos/genética , Plasmídeos , Reoviridae/genética , Transfecção
7.
Anticancer Res ; 41(5): 2431-2440, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33952468

RESUMO

BACKGROUND/AIM: Oncolytic reovirus, which is a non-enveloped virus possessing a 10-segmented double-stranded RNA genome, has been anticipated as a novel class of antitumor agent. Hepatocellular carcinoma (HCC) is considered to be a target suitable for reovirus-mediated virotherapy. Transforming growth factor (TGF)-ß plays an important role in the pathogenesis of HCC. TGF-ß-signaling inhibitors have proceeded to clinical trials as potential antitumor agents for HCC. On the other hand, TGF-ß is involved in induction of expression of cathepsins B and L, which are important for reovirus infection. It remains to be examined whether TGF-ß signaling inhibitors affect reovirus-mediated lysis of HCC cells. The aim of this study was to evaluate the effects of TGF-ß-signaling inhibitors on tumor cell lysis efficiency of reovirus in human HCC cells. MATERIALS AND METHODS: Reovirus was added to four types of human HCC cell lines pretreated with one of three TGF-ß type I receptor inhibitors: SB431542, A-83-01, or galunisertib (LY2157299). Cell viability, virus genome copy numbers, and virus protein expression were evaluated following reovirus infection. RESULTS: SB431542 significantly inhibited reovirus-mediated killing of human HCC cell lines, while A-83-01 and galunisertib did not inhibit. CONCLUSION: These data indicate that SB431542 inhibited reovirus-mediated lysis of human HCC cells in a TGF-ß signaling-independent manner.


Assuntos
Benzamidas/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Dioxóis/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Fator de Crescimento Transformador beta1/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Sobrevivência Celular/efeitos dos fármacos , Compostos de Epóxi , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/virologia , Orthoreovirus de Mamíferos/efeitos dos fármacos , Orthoreovirus de Mamíferos/genética , Pirazóis/farmacologia , Quinolinas/farmacologia , RNA de Cadeia Dupla/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Tirosina/análogos & derivados , Tirosina/genética
8.
Virus Res ; 296: 198334, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33581186

RESUMO

The intestinal mucosa plays an important role as an immune barrier due to its continual exposure to invading pathogens, including viruses. It is thus highly important to evaluate virus infection profiles in the intestinal mucosa for prevention of virus infection and development of antivirus medicines; however, only a few enterocyte lines are available as in vitro intestinal models for the evaluation of virus infection. In this study, we evaluated profiles of infection and innate immune responses following infection with a mammalian orthoreovirus (hereafter reovirus), which has often been used as a tractable model for studies of viral pathogenesis, in human iPS cell-derived small intestinal epithelial-like cell (hiPS-SIEC) monolayers and cells of a human colon adenocarcinoma cell line, Caco-2. The levels of reovirus infection were similar between hiPS-SIEC and Caco-2 cell monolayers, which are often used as an intestinal model, after apical and basolateral infection. In hiPS-SIEC monolayers, more efficient replication of the virus genome was observed following basolateral infection than apical infection, while apical infection resulted in higher levels of virus protein expression and progeny virus production than basolateral infection. Reovirus significantly induced innate immune responses, including expression of type I and III interferons (IFNs), in hiPS-SIEC monolayers more efficiently than Caco-2 cells. Higher levels of type I and III interferon (IFN) expression were found in hiPS-SIEC monolayers following apical infection than basolateral infection. These results suggested that hiPS-SIECs are a promising in vitro model for the evaluation of virus infection.


Assuntos
Células-Tronco Pluripotentes Induzidas , Orthoreovirus de Mamíferos , Orthoreovirus , Reoviridae , Viroses , Animais , Células CACO-2 , Humanos , Imunidade Inata , Interferons , Mamíferos , Orthoreovirus de Mamíferos/genética
9.
J Virol ; 94(22)2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-32847857

RESUMO

Triple-negative breast cancer (TNBC) constitutes 10 to 15% of all breast cancer and is associated with worse prognosis than other subtypes of breast cancer. Current therapies are limited to cytotoxic chemotherapy, radiation, and surgery, leaving a need for targeted therapeutics to improve outcomes for TNBC patients. Mammalian orthoreovirus (reovirus) is a nonenveloped, segmented, double-stranded RNA virus in the Reoviridae family. Reovirus preferentially kills transformed cells and is in clinical trials to assess its efficacy against several types of cancer. We previously engineered a reassortant reovirus, r2Reovirus, that infects TNBC cells more efficiently and induces cell death with faster kinetics than parental reoviruses. In this study, we sought to understand the mechanisms by which r2Reovirus induces cell death in TNBC cells. We show that r2Reovirus infection of TNBC cells of a mesenchymal stem-like (MSL) lineage downregulates the mitogen-activated protein kinase/extracellular signal-related kinase pathway and induces nonconventional cell death that is caspase-dependent but caspase 3-independent. Infection of different MSL lineage TNBC cells with r2Reovirus results in caspase 3-dependent cell death. We map the enhanced oncolytic properties of r2Reovirus in TNBC to epistatic interactions between the type 3 Dearing M2 gene segment and type 1 Lang genes. These findings suggest that the genetic composition of the host cell impacts the mechanism of reovirus-induced cell death in TNBC. Together, our data show that understanding host and virus determinants of cell death can identify novel properties and interactions between host and viral gene products that can be exploited for the development of improved viral oncolytics.IMPORTANCE TNBC is unresponsive to hormone therapies, leaving patients afflicted with this disease with limited treatment options. We previously engineered an oncolytic reovirus (r2Reovirus) with enhanced infective and cytotoxic properties in TNBC cells. However, how r2Reovirus promotes TNBC cell death is not known. In this study, we show that reassortant r2Reovirus can promote nonconventional caspase-dependent but caspase 3-independent cell death and that the mechanism of cell death depends on the genetic composition of the host cell. We also map the enhanced oncolytic properties of r2Reovirus in TNBC to interactions between a type 3 M2 gene segment and type 1 genes. Our data show that understanding the interplay between the host cell environment and the genetic composition of oncolytic viruses is crucial for the development of efficacious viral oncolytics.


Assuntos
Morte Celular/fisiologia , Reoviridae/fisiologia , Antineoplásicos/farmacologia , Apoptose , Neoplasias da Mama , Caspase 3/metabolismo , Linhagem Celular , Sobrevivência Celular , Humanos , Mitocôndrias/metabolismo , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos , Orthoreovirus de Mamíferos/genética , Reoviridae/genética , Proteínas Virais/metabolismo
10.
Arch Virol ; 165(7): 1541-1550, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32335768

RESUMO

Chinese tree shrews have been used extensively in studies of different types of cancer and for the modeling of viral infections. In the present study, we report the isolation and characterization of two strains of mammalian orthoreovirus (MRV), MRV1/TS/2011 and MRV3/TS/2012, which were isolated from the feces of tree shrews in Yunnan, China. These two strains of MRV were isolated and cultured in both primary tree shrew intestinal epithelial cells (pTIECs) and primary tree shrew alveolar epithelial cells (pTAECs). A neutralization test using immunofluorescence was employed to determine the subtype of each isolate. Viral RNA was extracted and analyzed by polyacrylamide gel electrophoresis (PAGE), and the sequence was determined by next-generation sequencing for construction of a phylogenetic tree and analysis of gene polymorphism. Electron microscopy examination revealed the presence of virus particles with the typical morphological characteristics of MRV. Serotype analysis showed that strain MRV1/TS/2011 was of type I and strain MRV3/TS/2012 was of type III. A sequence comparison showed that the isolates were 25.4% identical in the S1 gene.


Assuntos
Orthoreovirus de Mamíferos/isolamento & purificação , Infecções por Reoviridae/veterinária , Tupaiidae/virologia , Animais , China , Fezes/virologia , Humanos , Orthoreovirus de Mamíferos/classificação , Orthoreovirus de Mamíferos/genética , Filogenia , RNA Viral/genética , Infecções por Reoviridae/virologia , Vírion/classificação , Vírion/genética , Vírion/isolamento & purificação
11.
J Virol ; 94(4)2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31748391

RESUMO

Little is known about how genetic variations in viruses affect their success as therapeutic agents. The type 3 Dearing strain of Mammalian orthoreovirus (T3D) is undergoing clinical trials as an oncolytic virotherapy. Worldwide, studies on reovirus oncolysis use T3D stocks propagated in different laboratories. Here, we report that genetic diversification among T3D stocks from various sources extensively impacts oncolytic activity. The T3D strain from the Patrick Lee laboratory strain (TD3PL) showed significantly stronger oncolytic activities in a murine model of melanoma than the strain from the Terence Dermody laboratory (T3DTD). Overall in vitro replication and cytolytic properties of T3D laboratory strains were assessed by measuring virus plaque size on a panel of human and mouse tumor cells, and results were found to correlate with in vivo oncolytic potency in a melanoma model. T3DPL produced larger plaques than T3DTD and than the T3D strain from the ATCC (T3DATCC) and from the Kevin Coombs laboratory (T3DKC). Reassortant and reverse genetics analyses were used to decipher key genes and polymorphisms that govern enhanced plaque size of T3DPL Five single amino acid changes in the S4, M1, and L3 genome segments of reovirus were each partially correlated with plaque size and when combined were able to fully account for differences between T3DPL and T3DTD Moreover, polymorphisms were discovered in T3DTD that promoted virus replication and spread in tumors, and a new T3DPL/T3DTD hybrid was generated with enhanced plaque size compared to that of T3DPL Altogether, single amino acid changes acquired during laboratory virus propagation can have a large impact on reovirus therapeutic potency and warrant consideration as possible confounding variables between studies.IMPORTANCE The reovirus serotype 3 Dearing (T3D) strain is in clinical trials for cancer therapy. We find that closely related laboratory strains of T3D exhibit large differences in their abilities to replicate in cancer cells in vitro, which correlates with oncolytic activity in a in a murine model of melanoma. The study reveals that five single amino acid changes among three reovirus genes strongly impact reovirus therapeutic potency. In general, the findings suggest that attention should be given to genomic divergence of virus strains during research and optimization for cancer therapy.


Assuntos
Orthoreovirus Mamífero 3/genética , Terapia Viral Oncolítica/métodos , Replicação Viral/genética , Aminoácidos/genética , Animais , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Variação Genética/genética , Humanos , Orthoreovirus Mamífero 3/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Orthoreovirus de Mamíferos/genética , Orthoreovirus de Mamíferos/metabolismo , Filogenia , Reoviridae/genética , Proteínas Virais/metabolismo
12.
J Virol ; 93(14)2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31068423

RESUMO

Wild-type mammalian reoviruses (MRVs) have been evaluated as oncolytic agents against various cancers; however, genetic modification methods for improving MRV agents have not been exploited fully. In the present study, using MRV strain T1L, we generated a reporter MRV that expresses a NanoLuc luciferase (NLuc) gene and used it for noninvasive imaging of MRV infection in tumor xenograft mice. NLuc and a P2A self-cleaving peptide gene cassette were placed upstream of the L1 gene open reading frame to enable bicistronic expression of NLuc and the L1 gene product. BALB/c nude mice intranasally infected with MRV expressing NLuc (rsT1L-NLuc) displayed bioluminescent signals in the chest area at 4 days postinfection (dpi), which is consistent with natural MRV infection in the lung. Furthermore, to monitor tumor-selective infection by MRV, nude mice bearing human cancer xenografts were infected intravenously with rsT1L-NLuc. Bioluminescent signals were detected in tumors as early as 3 dpi and persisted for 2 months. The results demonstrate the utility of an autonomous replicating reporter MRV for noninvasive live imaging of replicating oncolytic MRV agents.IMPORTANCE Engineering of recombinant MRV for improved oncolytic activity has not yet been achieved due to difficulty in generating autonomous replicating MRV harboring transgenes. Here, we constructed a reporter MRV that can be used to monitor cancer-selective infection by oncolytic MRV in a mouse model. Among the numerous oncolytic viruses, MRV has an advantage in that the wild-type virus shows marked oncolytic activity in patients without any notable adverse effects. The reporter MRV developed herein will open avenues to the development of recombinant MRV vectors armed with anticancer transgenes.


Assuntos
Regulação Viral da Expressão Gênica , Luciferases/biossíntese , Medições Luminescentes , Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos/metabolismo , Orthoreovirus de Mamíferos/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Luciferases/genética , Camundongos , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Neoplasias/virologia , Vírus Oncolíticos/genética , Orthoreovirus de Mamíferos/genética , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cancer Gene Ther ; 26(9-10): 268-281, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30467340

RESUMO

Oncolytic reovirus administration has been well tolerated by cancer patients in clinical trials. However, its anti-cancer efficacy as a monotherapy remains to be augmented. We and others have previously demonstrated the feasibility of producing replication-competent reoviruses expressing a heterologous transgene. Here, we describe the production of recombinant reoviruses expressing murine (mm) or human (hs) GM-CSF (rS1-mmGMCSF and rS1-hsGMCSF, respectively). The viruses could be propagated up to 10 passages while deletion mutants occurred only occasionally. In infected cell cultures, the secretion of GM-CSF protein (up to 481 ng/106 cells per day) was demonstrated by ELISA. The secreted mmGM-CSF protein was functional in cell culture, as demonstrated by the capacity to stimulate the survival and proliferation of the GM-CSF-dependent dendritic cell (DC) line D1, and by its ability to generate DCs from murine bone marrow cells. Importantly, in a murine model of pancreatic cancer we found a systemic increase in DC and T-cell activation upon intratumoral administration of rS1-mmGMCSF. These data demonstrate that reoviruses expressing functional GM-CSF can be generated and have the potential to enhance anti-tumor immune responses. The GM-CSF reoviruses represent a promising new agent for use in oncolytic virotherapy strategies.


Assuntos
Vetores Genéticos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Imunidade/genética , Imunomodulação/genética , Vírus Oncolíticos/genética , Orthoreovirus de Mamíferos/genética , Animais , Linhagem Celular , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Ordem dos Genes , Engenharia Genética , Terapia Genética , Vetores Genéticos/administração & dosagem , Humanos , Imunoterapia/métodos , Camundongos , Terapia Viral Oncolítica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Transgenes
14.
Nat Microbiol ; 3(4): 481-493, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29531365

RESUMO

Viruses are molecular machines sustained through a life cycle that requires replication within host cells. Throughout the infectious cycle, viral and cellular components interact to advance the multistep process required to produce progeny virions. Despite progress made in understanding the virus-host protein interactome, much remains to be discovered about the cellular factors that function during infection, especially those operating at terminal steps in replication. In an RNA interference screen, we identified the eukaryotic chaperonin T-complex protein-1 (TCP-1) ring complex (TRiC; also called CCT for chaperonin containing TCP-1) as a cellular factor required for late events in the replication of mammalian reovirus. We discovered that TRiC functions in reovirus replication through a mechanism that involves folding the viral σ3 major outer-capsid protein into a form capable of assembling onto virus particles. TRiC also complexes with homologous capsid proteins of closely related viruses. Our data define a critical function for TRiC in the viral assembly process and raise the possibility that this mechanism is conserved in related non-enveloped viruses. These results also provide insight into TRiC protein substrates and establish a rationale for the development of small-molecule inhibitors of TRiC as potential antiviral therapeutics.


Assuntos
Proteínas do Capsídeo/genética , Capsídeo/metabolismo , Chaperonina com TCP-1/genética , Orthoreovirus de Mamíferos/genética , Montagem de Vírus/genética , Animais , Células CACO-2 , Proteínas do Capsídeo/metabolismo , Linhagem Celular Tumoral , Células Endoteliais/virologia , Células HEK293 , Ensaios de Triagem em Larga Escala , Humanos , Camundongos , Orthoreovirus de Mamíferos/crescimento & desenvolvimento , Dobramento de Proteína , Interferência de RNA , RNA Interferente Pequeno/genética
15.
Virus Res ; 247: 40-46, 2018 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-29382551

RESUMO

In the course of previous works, it was observed that the virus laboratory stock (T3DS) differs in sequence from the virus encoded by the ten plasmids currently in use in many laboratories (T3DK), and derived from a different original virus stock. Seven proteins are affected by these sequence differences. In the present study, replication of T3DK was shown to be more sensitive to the antiviral effect of interferon. Infection by the T3DK virus was also shown to induce the production of higher amount of ß and α-interferons compared to T3DS. Two proteins, the µ2 and λ2 proteins, were found to be responsible for increased sensitivity to interferon while both µ2 and λ1 are responsible for increased interferon secretion. Altogether this supports the idea that multiple reovirus proteins are involved in the control of induction of interferon and virus sensitivity to the interferon-induced response. While interrelated, interferon induction and sensitivity can be separated by defined gene combinations. While both µ2 and λ2 were previously suspected of a role in the control of the interferon response, other proteins are also likely involved, as first shown here for λ1. This also further stresses that due caution should be exerted when comparing different virus isolates with different genetic background.


Assuntos
Proteínas do Capsídeo/genética , Proteínas de Ligação a DNA/genética , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Nucleotidiltransferases/genética , Orthoreovirus de Mamíferos/efeitos dos fármacos , Proteínas de Ligação a RNA/genética , Vírus Reordenados/efeitos dos fármacos , Proteínas do Core Viral/genética , Proteínas Virais/genética , Animais , Antivirais/farmacologia , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Cricetulus , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/virologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/imunologia , Fibroblastos/virologia , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Patógeno , Interferon-alfa/biossíntese , Interferon beta/biossíntese , Camundongos , Modelos Moleculares , Nucleotidiltransferases/química , Nucleotidiltransferases/metabolismo , Orthoreovirus de Mamíferos/genética , Orthoreovirus de Mamíferos/imunologia , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Vírus Reordenados/genética , Vírus Reordenados/imunologia , Genética Reversa , Proteínas do Core Viral/química , Proteínas do Core Viral/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo
16.
BMC Infect Dis ; 18(1): 33, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29325543

RESUMO

BACKGROUND: We report a rare case of Mammalian orthoreovirus (MRV) infection in a child with a primary immunodeficiency (PID). Infections with Mammalian orthoreovirus are very rare and probably of zoonotic origin. Only a few cases have been described so far, including one with similar pathogenesis as in our case. CASE PRESENTATION: The patient, age 11, presented with flu-like symptoms and persistent severe diarrhea. Enterovirus has been detected over several months, however, exact typing of a positive cell culture remained inconclusive. Unbiased metagenomic sequencing then detected MRV in stool samples from several time points. The sequencing approach further revealed co-infection with a recombinant Coxsackievirus and Adenovirus. MRV-specific antibodies detected by immunofluorescence proved that the patient seroconverted. CONCLUSION: This case highlights the potential of unbiased metagenomic sequencing in supplementing routine diagnostic methods, especially in situations of chronic infection with multiple viruses as seen here in an immunocompromised host. The origin, transmission routes and implications of MRV infection in humans merit further investigation.


Assuntos
Infecções por Adenoviridae/virologia , Infecções por Coxsackievirus/virologia , Síndromes de Imunodeficiência/complicações , Metagenômica/métodos , Infecções por Reoviridae/virologia , Infecções por Adenoviridae/etiologia , Criança , Coinfecção , Infecções por Coxsackievirus/etiologia , Diarreia/virologia , Enterovirus/genética , Enterovirus/patogenicidade , Infecções por Enterovirus/virologia , Feminino , Humanos , Síndromes de Imunodeficiência/virologia , Orthoreovirus de Mamíferos/genética , Orthoreovirus de Mamíferos/patogenicidade , Infecções por Reoviridae/etiologia
17.
J Virol ; 91(11)2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28298603

RESUMO

Reoviruses, like many eukaryotic viruses, contain an inverted 7-methylguanosine (m7G) cap linked to the 5' nucleotide of mRNA. The traditional functions of capping are to promote mRNA stability, protein translation, and concealment from cellular proteins that recognize foreign RNA. To address the role of mRNA capping during reovirus replication, we assessed the benefits of adding the African swine fever virus NP868R capping enzyme during reovirus rescue. C3P3, a fusion protein containing T7 RNA polymerase and NP868R, was found to increase protein expression 5- to 10-fold compared to T7 RNA polymerase alone while enhancing reovirus rescue from the current reverse genetics system by 100-fold. Surprisingly, RNA stability was not increased by C3P3, suggesting a direct effect on protein translation. A time course analysis revealed that C3P3 increased protein synthesis within the first 2 days of a reverse genetics transfection. This analysis also revealed that C3P3 enhanced processing of outer capsid µ1 protein to µ1C, a previously described hallmark of reovirus assembly. Finally, to determine the rate of infectious-RNA incorporation into new virions, we developed a new recombinant reovirus S1 gene that expressed the fluorescent protein UnaG. Following transfection of cells with UnaG and infection with wild-type virus, passage of UnaG through progeny was significantly enhanced by C3P3. These data suggest that capping provides nontraditional functions to reovirus, such as promoting assembly and infectious-RNA incorporation.IMPORTANCE Our findings expand our understanding of how viruses utilize capping, suggesting that capping provides nontraditional functions to reovirus, such as promoting assembly and infectious-RNA incorporation, in addition to enhancing protein translation. Beyond providing mechanistic insight into reovirus replication, our findings also show that reovirus reverse genetics rescue is enhanced 100-fold by the NP868R capping enzyme. Since reovirus shows promise as a cancer therapy, efficient reovirus reverse genetics rescue will accelerate production of recombinant reoviruses as candidates to enhance therapeutic potency. NP868R-assisted reovirus rescue will also expedite production of recombinant reovirus for mechanistic insights into reovirus protein function and structure.


Assuntos
Vírus da Febre Suína Africana/enzimologia , Nucleotidiltransferases/metabolismo , Orthoreovirus de Mamíferos/genética , Orthoreovirus de Mamíferos/fisiologia , RNA Viral/metabolismo , Vírion/fisiologia , Montagem de Vírus , Vírus da Febre Suína Africana/genética , Linhagem Celular , Proteínas Recombinantes de Fusão/metabolismo , Genética Reversa , Vírion/genética , Replicação Viral
18.
Proc Natl Acad Sci U S A ; 114(9): 2349-2354, 2017 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-28137864

RESUMO

Rotaviruses (RVs) are highly important pathogens that cause severe diarrhea among infants and young children worldwide. The understanding of the molecular mechanisms underlying RV replication and pathogenesis has been hampered by the lack of an entirely plasmid-based reverse genetics system. In this study, we describe the recovery of recombinant RVs entirely from cloned cDNAs. The strategy requires coexpression of a small transmembrane protein that accelerates cell-to-cell fusion and vaccinia virus capping enzyme. We used this system to obtain insights into the process by which RV nonstructural protein NSP1 subverts host innate immune responses. By insertion into the NSP1 gene segment, we recovered recombinant viruses that encode split-green fluorescent protein-tagged NSP1 and NanoLuc luciferase. This technology will provide opportunities for studying RV biology and foster development of RV vaccines and therapeutics.


Assuntos
Metiltransferases/genética , Complexos Multienzimáticos/genética , Nucleotidiltransferases/genética , Orthoreovirus de Mamíferos/genética , Orthoreovirus/genética , Monoéster Fosfórico Hidrolases/genética , Plasmídeos/metabolismo , Genética Reversa/métodos , Proteínas não Estruturais Virais/genética , Proteínas Virais/genética , Animais , Sequência de Bases , Linhagem Celular , Linhagem Celular Tumoral , Cricetulus , DNA Complementar/genética , DNA Complementar/metabolismo , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Células Epiteliais/virologia , Regulação da Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Luciferases/genética , Luciferases/metabolismo , Metiltransferases/metabolismo , Complexos Multienzimáticos/metabolismo , Nucleotidiltransferases/metabolismo , Orthoreovirus/metabolismo , Orthoreovirus de Mamíferos/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Plasmídeos/química , Transdução Genética , Vaccinia virus/genética , Vaccinia virus/metabolismo , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/metabolismo
20.
Hum Gene Ther ; 27(2): 127-33, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26603492

RESUMO

Unlike for other digestive cancer entities, chemotherapy, radiotherapy, and targeted therapies have, so far, largely failed to improve patient survival in pancreatic adenocarcinoma (PDAC), which remains the fourth leading cause of cancer-related death in Europe and the United States. In this context, gene therapy may offer a new avenue for patients with PDAC. In this review, we explore the research currently ongoing in French laboratories aimed at defeating PDAC using nonviral therapeutic gene delivery, targeted transgene expression, or oncolytic virotherapy that recently or will soon bridge the gap between experimental models of cancer and clinical trials. These studies are likely to change clinical practice or thinking about PDAC management, as they represent a major advance not only for PDAC but may also significantly influence the field of gene-based molecular treatment of cancer.


Assuntos
Adenocarcinoma/terapia , Regulação Neoplásica da Expressão Gênica , Terapia Genética/métodos , Terapia Viral Oncolítica/métodos , Neoplasias Pancreáticas/terapia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenovírus Humanos/genética , Adenovírus Humanos/metabolismo , Ensaios Clínicos como Assunto , Desoxicitidina Quinase/genética , Desoxicitidina Quinase/metabolismo , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Núcleosídeo-Fosfato Quinase/genética , Núcleosídeo-Fosfato Quinase/metabolismo , Orthoreovirus de Mamíferos/genética , Orthoreovirus de Mamíferos/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA