Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
Mol Immunol ; 170: 131-143, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38663254

RESUMO

Mammalian reovirus (MRV) is a non-enveloped, gene segmented double-stranded RNA (dsRNA) virus. It is an important zoonotic pathogen that infects many mammals and vertebrates that act as natural hosts and causes respiratory and digestive tract diseases. Studies have reported that RIG-I and MDA5 in the innate immune cytoplasmic RNA-sensing RIG-like receptor (RLR) signaling pathway can recognize dsRNA from MRV and promote antiviral type I interferon (IFN) responses. However, the mechanism by which many MRV-encoded proteins evade the host innate immune response remains unclear. Here, we show that exogenous µ1 protein promoted the proliferation of MRV in vitro, while knockdown of MRV µ1 protein expression by shRNA could impair MRV proliferation. Specifically, µ1 protein inhibited MRV or poly(I:C)-induced IFN-ß expression, and attenuated RIG-I/MDA5-mediated signaling axis transduction during MRV infection. Importantly, we found that µ1 protein significantly decreased IFN-ß mRNA expression induced by MDA5, RIG-I, MAVS, TBK1, IRF3(5D), and degraded the protein expression of exogenous MDA5, RIG-I, MAVS, TBK1 and IRF3 via the proteasomal and lysosomal pathways. Additionally, we show that µ1 protein can physically interact with MDA5, RIG-I, MAVS, TBK1, and IRF3 and attenuate the RIG-I/MDA5-mediated signaling cascades by blocking the phosphorylation and nuclear translocation of IRF3. In conclusion, our findings reveal that MRV outer capsid protein µ1 is a key factor in antagonizing RLRs signaling cascades and provide new strategies for effective prevention and treatment of MRV infection.


Assuntos
Proteína DEAD-box 58 , Fator Regulador 3 de Interferon , Helicase IFIH1 Induzida por Interferon , Orthoreovirus de Mamíferos , Receptores Imunológicos , Transdução de Sinais , Helicase IFIH1 Induzida por Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Proteína DEAD-box 58/metabolismo , Transdução de Sinais/imunologia , Humanos , Fosforilação , Orthoreovirus de Mamíferos/imunologia , Orthoreovirus de Mamíferos/fisiologia , Células HEK293 , Interferon beta/metabolismo , Interferon beta/imunologia , Animais , Núcleo Celular/metabolismo , Infecções por Reoviridae/imunologia , Proteínas Virais/metabolismo , Transporte Ativo do Núcleo Celular , Imunidade Inata/imunologia , Proteínas Serina-Treonina Quinases
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Virology ; 558: 38-48, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33721728

RESUMO

Mammalian orthoreovirus (MRV) is a safe and effective cancer killing virus that has completed Phase I-III clinical trials against numerous cancer types. While many patients experience benefit from MRV therapy, pre-defined set points necessary for FDA approval have not been reached. Therefore, additional research into MRV biology and the effect of viral therapy on different tumor genetic subtypes and microenvironments is necessary to identify tumors most amenable to MRV virotherapy. In this work we analyzed the stage of viral infection necessary to inhibit HIF-1α, an aggressive cancer activator induced by hypoxia. We demonstrated that two viral capsid proteins were not necessary and that a step parallel with virus core movement across the endosomal membrane was required for this inhibition. Altogether, this work clarifies the mechanisms of MRV-induced HIF-1α inhibition and provides biological relevance for using MRV to inhibit the devastating effects of tumor hypoxia.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Terapia Viral Oncolítica , Orthoreovirus de Mamíferos/fisiologia , Neoplasias da Próstata , Microambiente Tumoral/fisiologia , Linhagem Celular Tumoral , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/análise , Masculino , Hipóxia Tumoral
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.
Viruses ; 13(2)2021 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-33525628

RESUMO

Cells are continually exposed to stressful events, which are overcome by the activation of a number of genetic pathways. The integrated stress response (ISR) is a large component of the overall cellular response to stress, which ultimately functions through the phosphorylation of the alpha subunit of eukaryotic initiation factor-2 (eIF2α) to inhibit the energy-taxing process of translation. This response is instrumental in the inhibition of viral infection and contributes to evolution in viruses. Mammalian orthoreovirus (MRV), an oncolytic virus that has shown promise in over 30 phase I-III clinical trials, has been shown to induce multiple arms within the ISR pathway, but it successfully evades, modulates, or subverts each cellular attempt to inhibit viral translation. MRV has not yet received Food and Drug Administration (FDA) approval for general use in the clinic; therefore, researchers continue to study virus interactions with host cells to identify circumstances where MRV effectiveness in tumor killing can be improved. In this review, we will discuss the ISR, MRV modulation of the ISR, and discuss ways in which MRV interaction with the ISR may increase the effectiveness of cancer therapeutics whose modes of action are altered by the ISR.


Assuntos
Vírus Oncolíticos/fisiologia , Orthoreovirus de Mamíferos/fisiologia , Infecções por Reoviridae/virologia , Estresse Fisiológico , Fator de Iniciação 2 em Eucariotos/metabolismo , Humanos , Terapia Viral Oncolítica , Fosforilação , Biossíntese de Proteínas , Capuzes de RNA/metabolismo , Infecções por Reoviridae/metabolismo
10.
PLoS Pathog ; 16(9): e1008803, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32956403

RESUMO

The Dearing isolate of Mammalian orthoreovirus (T3D) is a prominent model of virus-host relationships and a candidate oncolytic virotherapy. Closely related laboratory strains of T3D, originating from the same ancestral T3D isolate, were recently found to exhibit significantly different oncolytic properties. Specifically, the T3DPL strain had faster replication kinetics in a panel of cancer cells and improved tumor regression in an in vivo melanoma model, relative to T3DTD. In this study, we discover that T3DPL and T3DTD also differentially activate host signalling pathways and downstream gene transcription. At equivalent infectious dose, T3DTD induces higher IRF3 phosphorylation and expression of type I IFNs and IFN-stimulated genes (ISGs) than T3DPL. Using mono-reassortants with intermediate replication kinetics and pharmacological inhibitors of reovirus replication, IFN responses were found to inversely correlate with kinetics of virus replication. In other words, slow-replicating T3D strains induce more IFN signalling than fast-replicating T3D strains. Paradoxically, during co-infections by T3DPL and T3DTD, there was still high IRF3 phosphorylation indicating a phenodominant effect by the slow-replicating T3DTD. Using silencing and knock-out of RIG-I to impede IFN, we found that IFN induction does not affect the first round of reovirus replication but does prevent cell-cell spread in a paracrine fashion. Accordingly, during co-infections, T3DPL continues to replicate robustly despite activation of IFN by T3DTD. Using gene expression analysis, we discovered that reovirus can also induce a subset of genes in a RIG-I and IFN-independent manner; these genes were induced more by T3DPL than T3DTD. Polymorphisms in reovirus σ3 viral protein were found to control activation of RIG-I/ IFN-independent genes. Altogether, the study reveals that single amino acid polymorphisms in reovirus genomes can have large impact on host gene expression, by both changing replication kinetics and by modifying viral protein activity, such that two closely related T3D strains can induce opposite cytokine landscapes.


Assuntos
Proteínas do Capsídeo/metabolismo , Interferons/metabolismo , Polimorfismo Genético , RNA de Cadeia Dupla/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptores do Ácido Retinoico/metabolismo , Infecções por Reoviridae/virologia , Replicação Viral , Proteínas do Capsídeo/genética , Citocinas , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Orthoreovirus de Mamíferos/fisiologia , RNA de Cadeia Dupla/genética , RNA Viral/genética , RNA Viral/metabolismo , Proteínas de Ligação a RNA/genética , Receptores do Ácido Retinoico/genética , Infecções por Reoviridae/genética , Infecções por Reoviridae/metabolismo , Transdução de Sinais
11.
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
12.
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
13.
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
14.
Viruses ; 11(6)2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31216693

RESUMO

Following reovirus infection, cells activate stress responses that repress canonical translation as a mechanism to limit progeny virion production. Work by others suggests that these stress responses, which are part of the integrated stress response (ISR), may benefit rather than repress reovirus replication. Here, we report that compared to untreated cells, treating cells with sodium arsenite (SA) to activate the ISR prior to infection enhanced viral protein expression, percent infectivity, and viral titer. SA-mediated enhancement was not strain-specific, but was cell-type specific. While SA pre-treatment of cells offered the greatest enhancement, treatment within the first 4 h of infection increased the percent of cells infected. SA activates the heme-regulated eIF2α (HRI) kinase, which phosphorylates eukaryotic translation initiation factor 2 alpha (eIF2α) to induce stress granule (SG) formation. Heat shock (HS), another activator of HRI, also induced eIF2α phosphorylation and SGs in cells. However, HS had no effect on percent infectivity or viral yield but did enhance viral protein expression. These data suggest that SA pre-treatment perturbs the cell in a way that is beneficial for reovirus and that this enhancement is independent of SG induction. Understanding how to manipulate the cellular stress responses during infection to enhance replication could help to maximize the oncolytic potential of reovirus.


Assuntos
Arsenitos/toxicidade , Inibidores Enzimáticos/toxicidade , Orthoreovirus de Mamíferos/crescimento & desenvolvimento , Compostos de Sódio/toxicidade , Estresse Fisiológico/efeitos dos fármacos , Replicação Viral , Linhagem Celular , Humanos , Carga Viral
15.
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
16.
Vet Comp Oncol ; 17(2): 184-193, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30761736

RESUMO

Canine histiocytic sarcoma is an aggressive, fatal neoplastic disease with a poor prognosis. Lomustine is generally accepted as the first-line systemic therapy, although this compound does not provide complete regression. Therefore, research into a novel approach against canine histiocytic sarcoma is needed. However, anti-tumour effects of oncolytic therapy using reovirus against histiocytic sarcoma are unknown. Here, we showed that reovirus has oncolytic activity in canine histiocytic sarcoma cell lines in vitro and in vivo. We found that reovirus can replicate and induce caspase-dependent apoptosis in canine histiocytic sarcoma cell lines. A single intra-tumoural injection of reovirus completely suppressed the growth of subcutaneously grafted tumours in NOD/SCID mice. Additionally, we demonstrated that susceptibility to reovirus-induced cell death was attributable to the extent of expression of type I interferons induced by reovirus infection in vitro. In conclusion, oncolytic reovirus appears to be an effective treatment option for histiocytic sarcoma, and therefore warrants further investigation in early clinical trials.


Assuntos
Doenças do Cão/virologia , Sarcoma Histiocítico/veterinária , Terapia Viral Oncolítica/veterinária , Vírus Oncolíticos/patogenicidade , Orthoreovirus de Mamíferos/patogenicidade , Animais , Morte Celular , Linhagem Celular Tumoral/virologia , Cães , Sarcoma Histiocítico/virologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Terapia Viral Oncolítica/métodos , Reação em Cadeia da Polimerase em Tempo Real/veterinária
17.
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
18.
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
19.
J Virol ; 92(10)2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29514905

RESUMO

Several viruses induce intestinal epithelial cell death during enteric infection. However, it is unclear whether proapoptotic capacity promotes or inhibits replication in this tissue. We infected mice with two reovirus strains that infect the intestine but differ in the capacity to alter immunological tolerance to new food antigen. Infection with reovirus strain T1L, which induces an inflammatory immune response to fed antigen, is prolonged in the intestine, whereas T3D-RV, which does not induce this response, is rapidly cleared from the intestine. Compared with T1L, T3D-RV infection triggered apoptosis of intestinal epithelial cells and subsequent sloughing of dead cells into the intestinal lumen. We conclude that the infection advantage of T1L derives from its capacity to subvert host restriction by epithelial cell apoptosis, providing a possible mechanism by which T1L enhances inflammatory signals during antigen feeding. Using a panel of T1L × T3D-RV reassortant viruses, we identified the viral M1 and M2 gene segments as determinants of reovirus-induced apoptosis in the intestine. Expression of the T1L M1 and M2 genes in a T3D-RV background was sufficient to limit epithelial cell apoptosis and enhance viral infection to levels displayed by T1L. These findings define additional reovirus gene segments required for enteric infection of mice and illuminate the antiviral effect of intestinal epithelial cell apoptosis in limiting enteric viral infection. Viral strain-specific differences in the capacity to infect the intestine may be useful in identifying viruses capable of ameliorating tolerance to fed antigen in autoimmune conditions like celiac disease.IMPORTANCE Acute viral infections are thought to be cleared by the host with few lasting consequences. However, there may be much broader and long-lasting effects of viruses on immune homeostasis. Infection with reovirus, a common, nonpathogenic virus, triggers inflammation against innocuous food antigens, implicating this virus in the development of celiac disease, an autoimmune intestinal disorder triggered by exposure to dietary gluten. Using two reovirus strains that differ in the capacity to abrogate oral tolerance, we found that strain-specific differences in the capacity to replicate in the intestine inversely correlate with the capacity to induce apoptotic death of intestinal epithelial cells, providing a host-mediated process to restrict intestinal infection. This work contributes new knowledge about virus-host interactions in the intestine and establishes a foundation for future studies to define mechanisms by which viruses break oral tolerance in celiac disease.


Assuntos
Apoptose/imunologia , Células Epiteliais/imunologia , Mucosa Intestinal/imunologia , Orthoreovirus Mamífero 3/imunologia , Orthoreovirus de Mamíferos/imunologia , Infecções por Reoviridae/imunologia , Animais , Antígenos Virais/imunologia , Linhagem Celular , Cricetinae , Células Epiteliais/patologia , Células Epiteliais/virologia , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Camundongos , Infecções por Reoviridae/patologia
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA