Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(20): e2115354119, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35549551

RESUMO

Myxoma virus (MYXV) causes localized cutaneous fibromas in its natural hosts, tapeti and brush rabbits; however, in the European rabbit, MYXV causes the lethal disease myxomatosis. Currently, the molecular mechanisms underlying this increased virulence after cross-species transmission are poorly understood. In this study, we investigated the interaction between MYXV M156 and the host protein kinase R (PKR) to determine their crosstalk with the proinflammatory nuclear factor kappa B (NF-κB) pathway. Our results demonstrated that MYXV M156 inhibits brush rabbit PKR (bPKR) more strongly than European rabbit PKR (ePKR). This moderate ePKR inhibition could be improved by hyperactive M156 mutants. We hypothesized that the moderate inhibition of ePKR by M156 might incompletely suppress the signal transduction pathways modulated by PKR, such as the NF-κB pathway. Therefore, we analyzed NF-κB pathway activation with a luciferase-based promoter assay. The moderate inhibition of ePKR resulted in significantly higher NF-κB­dependent reporter activity than complete inhibition of bPKR. We also found a stronger induction of the NF-κB target genes TNFα and IL-6 in ePKR-expressing cells than in bPKR-expressing cells in response to M156 in both transfection and infections assays. Furthermore, a hyperactive M156 mutant did not cause ePKR-dependent NF-κB activation. These observations indicate that M156 is maladapted for ePKR inhibition, only incompletely blocking translation in these hosts, resulting in preferential depletion of short­half-life proteins, such as the NF-κB inhibitor IκBα. We speculate that this functional activation of NF-κB induced by the intermediate inhibition of ePKR by M156 may contribute to the increased virulence of MYXV in European rabbits.


Assuntos
Interações Hospedeiro-Patógeno , Myxoma virus , Mixomatose Infecciosa , NF-kappa B , Coelhos , eIF-2 Quinase , Animais , Redes e Vias Metabólicas , Myxoma virus/genética , Myxoma virus/patogenicidade , Mixomatose Infecciosa/metabolismo , Mixomatose Infecciosa/virologia , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/metabolismo , Coelhos/virologia , eIF-2 Quinase/metabolismo
2.
Methods Mol Biol ; 2225: 1-23, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33108654

RESUMO

Viral diseases, whether of animals or humans, are normally considered as problems to be managed. However, in Australia, two viruses have been used as landscape-scale therapeutics to control European rabbits (Oryctolagus cuniculus), the preeminent invasive vertebrate pest species. Rabbits have caused major environmental and agricultural losses and contributed to extinction of native species. It was not until the introduction of Myxoma virus that effective control of this pest was obtained at a continental scale. Subsequent coevolution of rabbit and virus saw a gradual reduction in the effectiveness of biological control that was partially ameliorated by the introduction of the European rabbit flea to act as an additional vector for the virus. In 1995, a completely different virus, Rabbit hemorrhagic disease virus (RHDV), escaped from testing and spread through the Australian rabbit population and again significantly reduced rabbit numbers and environmental impacts. The evolutionary pressures on this virus appear to be producing quite different outcomes to those that occurred with myxoma virus and the emergence and invasion of a novel genotype of RHDV in 2014 have further augmented control. Molecular studies on myxoma virus have demonstrated multiple proteins that manipulate the host innate and adaptive immune response; however the molecular basis of virus attenuation and reversion to virulence are not yet understood.


Assuntos
Agentes de Controle Biológico , Infecções por Caliciviridae/veterinária , Vírus da Doença Hemorrágica de Coelhos/patogenicidade , Myxoma virus/patogenicidade , Mixomatose Infecciosa/virologia , Reprodução , Animais , Austrália , Coevolução Biológica , Infecções por Caliciviridae/mortalidade , Infecções por Caliciviridae/virologia , Feminino , Expressão Gênica , Genótipo , Vírus da Doença Hemorrágica de Coelhos/genética , Interações Hospedeiro-Patógeno/genética , Insetos Vetores/virologia , Espécies Introduzidas , Masculino , Myxoma virus/genética , Mixomatose Infecciosa/mortalidade , Mixomatose Infecciosa/patologia , Coelhos , Sifonápteros/virologia , Proteínas Virais/genética , Proteínas Virais/metabolismo
3.
J Immunother Cancer ; 8(1)2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32581062

RESUMO

BACKGROUND: Oncolytic therapy uses live-replicating viruses to improve the immunological status of treated tumors. Critically, while these viruses are known to self-amplify in vivo, clinical oncolytic therapies still appear to display a strong dose dependence and the mechanisms mediating this dose dependence are not well understood. METHODS: To explore this apparent contradiction, we investigated how the initial dose of oncolytic myxoma virus affected the subsequent ability of treatment to alter the immunological status of tumors as well as synergize with programmed cell death protein 1 (PD1) blockade. RESULTS: Our results indicate that, due to viral self-amplification in vivo, the overall load of myxoma virus rapidly normalizes within treated tumors despite up to 3-log differences in inoculating dose. Because of this, therapeutic efficacy in the absence of checkpoint blockade is largely dose independent. Despite this rapid normalization, however, treatment with high or low doses of myxoma virus induces distinct immunological changes within treated tumors. Critically, these changes appear to be durably programmed based on the initial oncolytic dose with low-dose treatment failing to induce immunological improvements despite rapidly achieving equivalent viral burdens. Finally, due to the distinct immunological profiles induced by high and low myxoma virus doses, oncolytic efficacy resulting from combination with PD1 blockade therapy displays a strong dose dependence. CONCLUSIONS: Taken together, these data suggest that the ability of oncolytic myxoma virus to immunologically reprogram treated tumors is dependent on initial viral dose. Additionally, this work could provide a possible mechanistic explanation for clinical results observed with other oncolytic viruses.


Assuntos
Myxoma virus/patogenicidade , Vírus Oncolíticos/patogenicidade , Replicação Viral/genética , Animais , Humanos , Camundongos
5.
Bioessays ; 40(8): e1800050, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29869436

RESUMO

Cancer cells seem to exploit mechanisms that evolve as part of physiological tolerance, which is a complementary and often beneficial form of defense. The study of physiological systems of tolerance can therefore provide insights into the development of a state of host tolerance of cancer, and how to break it. Analysis of these models has the potential to improve our understanding of existing immunological therapeutic targets, and help to identify future targets and rational therapeutic combinations. The treatment of cancer with immune checkpoint inhibitors aims to reverse the progression to tolerance of cancer, and achieve an immunogenic, rather than tolerogenic, homeostasis. Broadening the efficacy and durability of checkpoint inhibitors focuses on reversing tolerance and stimulating immunogenicity in the cancer, host, and environment. Two examples of important physiological states of tolerance that may inform tolerance of cancer are microbial infection and placental reproduction. These states of tolerance result from bilateral shaping of host and non-self, akin to immunoediting in cancer, and offer reliable models to study the immune tolerance paradigm.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Tolerância Imunológica/fisiologia , Infecções/imunologia , Neoplasias/imunologia , Placenta/fisiologia , Aloenxertos/imunologia , Animais , Feminino , Humanos , Microbiota , Myxoma virus/patogenicidade , Infecções por Poxviridae/mortalidade , Gravidez , Microambiente Tumoral/imunologia
6.
J Virol ; 92(7)2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29343569

RESUMO

Poxviruses are large, DNA viruses whose protein capsid is surrounded by one or more lipid envelopes. Embedded into these lipid envelopes are three conserved viral proteins which are thought to mediate binding of virions to target cells. While the function of these proteins has been studied in vitro, their specific roles during the pathogenesis of poxviral disease remain largely unclear. Here we present data demonstrating that the putative chondroitin binding protein M083 from the leporipoxvirus myxoma virus is a significant virulence factor during infection of susceptible Oryctolagus rabbits. Removal of M083 results in a reduced capacity of virus to spread beyond the regional lymph nodes and completely eliminates infection-mediated mortality. In vitro, removal of M083 results in only minor intracellular replication defects but causes a significant reduction in the ability of myxoma virus to spread from infected epithelial cells onto primary lymphocytes. We hypothesize that the physiological role of M083 is therefore to mediate the spread of myxoma virus onto rabbit lymphocytes, allowing these cells to disseminate virus throughout infected rabbits.IMPORTANCE Poxviruses represent both a class of human pathogens and potential therapeutic agents for the treatment of human malignancy. Understanding the basic biology of these agents is therefore significant to human health in a variety of ways. While the mechanisms mediating poxviral binding have been well studied in vitro, how these mechanisms impact poxviral pathogenesis in vivo remains unclear. The current study advances our understanding of how poxviral binding impacts viral pathogenesis by demonstrating that the putative chondroitin binding protein M083 plays a critical role during the pathogenesis of myxoma virus in susceptible Oryctolagus rabbits by impacting viral dissemination through changes in the transfer of virions onto primary splenocytes.


Assuntos
Linfócitos/virologia , Myxoma virus , Proteínas Virais , Células A549 , Animais , Humanos , Linfócitos/metabolismo , Linfócitos/patologia , Myxoma virus/genética , Myxoma virus/metabolismo , Myxoma virus/patogenicidade , Coelhos , Proteínas Virais/genética , Proteínas Virais/metabolismo
7.
J Virol ; 91(20)2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28768866

RESUMO

The coevolution of myxoma virus (MYXV) and wild European rabbits in Australia and Europe is a paradigm for the evolution of a pathogen in a new host species. Genomic analyses have identified the mutations that have characterized this evolutionary process, but defining causal mutations in the pathways from virulence to attenuation and back to virulence has not been possible. Using reverse genetics, we examined the roles of six selected mutations found in Australian field isolates of MYXV that fall in known or potential virulence genes. Several of these mutations occurred in genes previously identified as virulence genes in whole-gene knockout studies. Strikingly, no single or double mutation among the mutations tested had an appreciable impact on virulence. This suggests either that virulence evolution was defined by amino acid changes other than those analyzed here or that combinations of multiple mutations, possibly involving epistatic interactions or noncoding sequences, have been critical in the ongoing evolution of MYXV virulence. In sum, our results show that single-gene knockout studies of a progenitor virus can have little power to predict the impact of individual mutations seen in the field. The genetic determinants responsible for this canonical case of virulence evolution remain to be determined.IMPORTANCE The species jump of myxoma virus (MYXV) from the South American tapeti to the European rabbit populations of Australia and Europe is a canonical example of host-pathogen coevolution. Detailed molecular studies have identified multiple genes in MYXV that are critical for virulence, and genome sequencing has revealed the evolutionary history of MYXV in Australia and Europe. However, it has not been possible to categorically identify the key mutations responsible for the attenuation of or reversion to virulence during this evolutionary process. Here we use reverse genetics to examine the role of mutations in viruses isolated early and late in the Australian radiation of MYXV. Surprisingly, none of the candidate mutations that we identified as likely having roles in attenuation proved to be important for virulence. This indicates that considerable caution is warranted when interpreting the possible role of individual mutations during virulence evolution.


Assuntos
Genoma Viral , Mutação , Myxoma virus/genética , Myxoma virus/patogenicidade , Genética Reversa , Fatores de Virulência/genética , Animais , Austrália , Evolução Molecular , Técnicas de Inativação de Genes , Genômica , Myxoma virus/classificação , Myxoma virus/isolamento & purificação , Filogenia , Coelhos , Virulência
8.
Proc Natl Acad Sci U S A ; 114(35): 9397-9402, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28808019

RESUMO

In host-pathogen arms races, increases in host resistance prompt counteradaptation by pathogens, but the nature of that counteradaptation is seldom directly observed outside of laboratory models. The best-documented field example is the coevolution of myxoma virus (MYXV) in European rabbits. To understand how MYXV in Australia has continued to evolve in wild rabbits under intense selection for genetic resistance to myxomatosis, we compared the phenotypes of the progenitor MYXV and viral isolates from the 1950s and the 1990s in laboratory rabbits with no resistance. Strikingly, and unlike their 1950s counterparts, most virus isolates from the 1990s induced a highly lethal immune collapse syndrome similar to septic shock. Thus, the next step in this canonical case of coevolution after a species jump has been further escalation by the virus in the face of widespread host resistance.


Assuntos
Myxoma virus/genética , Infecções por Poxviridae/veterinária , Coelhos/virologia , Infecções Tumorais por Vírus/veterinária , Animais , Austrália/epidemiologia , Evolução Biológica , Myxoma virus/patogenicidade , Infecções por Poxviridae/epidemiologia , Infecções por Poxviridae/patologia , Fatores de Tempo , Infecções Tumorais por Vírus/epidemiologia , Infecções Tumorais por Vírus/patologia , Virulência
9.
PLoS Pathog ; 13(3): e1006252, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28253375

RESUMO

The co-evolution of myxoma virus (MYXV) and the European rabbit occurred independently in Australia and Europe from different progenitor viruses. Although this is the canonical study of the evolution of virulence, whether the genomic and phenotypic outcomes of MYXV evolution in Europe mirror those observed in Australia is unknown. We addressed this question using viruses isolated in the United Kingdom early in the MYXV epizootic (1954-1955) and between 2008-2013. The later UK viruses fell into three distinct lineages indicative of a long period of separation and independent evolution. Although rates of evolutionary change were almost identical to those previously described for MYXV in Australia and strongly clock-like, genome evolution in the UK and Australia showed little convergence. The phenotypes of eight UK viruses from three lineages were characterized in laboratory rabbits and compared to the progenitor (release) Lausanne strain. Inferred virulence ranged from highly virulent (grade 1) to highly attenuated (grade 5). Two broad disease types were seen: cutaneous nodular myxomatosis characterized by multiple raised secondary cutaneous lesions, or an amyxomatous phenotype with few or no secondary lesions. A novel clinical outcome was acute death with pulmonary oedema and haemorrhage, often associated with bacteria in many tissues but an absence of inflammatory cells. Notably, reading frame disruptions in genes defined as essential for virulence in the progenitor Lausanne strain were compatible with the acquisition of high virulence. Combined, these data support a model of ongoing host-pathogen co-evolution in which multiple genetic pathways can produce successful outcomes in the field that involve both different virulence grades and disease phenotypes, with alterations in tissue tropism and disease mechanisms.


Assuntos
Evolução Molecular , Myxoma virus/genética , Myxoma virus/patogenicidade , Mixomatose Infecciosa/genética , Virulência/genética , Animais , Austrália , Genes Virais/genética , Genótipo , Fenótipo , Filogenia , Reação em Cadeia da Polimerase , Coelhos , Reino Unido
10.
Viruses ; 9(2)2017 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-28157174

RESUMO

Myxoma virus (MYXV) is Leporipoxvirus that possesses a specific rabbit-restricted host tropism but exhibits a much broader  cellular host range in cultured cells. MYXV is able to efficiently  block all aspects of the type I interferon (IFN)-induced  antiviral  state  in rabbit cells, partially in  human  cells  and  very  poorly  in  mouse  cells.  The mechanism(s) of this species-specific inhibition of  type I IFN-induced antiviral state is not well understood. Here we demonstrate that MYXV encoded  protein  M029, a truncated relative of the vaccinia virus (VACV) E3 double-stranded RNA (dsRNA)  binding  protein  that  inhibits  protein  kinase  R (PKR),  can  also  antagonize the type I IFN-induced  antiviral state in a highly species-specific manner. In cells pre-treated with type I IFN prior to  infection,  MYXV  exploits  M029  to  overcome  the  induced  antiviral  state completely in rabbit cells,  partially  in  human  cells,  but  not at all in mouse cells. However, in cells pre-infected with MYXV,  IFN-induced  signaling  is fully  inhibited  even  in the  absence  of M029 in cells from all three species,  suggesting  that  other  MYXV  protein(s)  apart  from  M029  block  IFN  signaling  in  a  speciesindependent  manner.  We  also  show  that  the  antiviral  state  induced in rabbit, human or mouse cells  by  type  I IFN  can  inhibit M029-knockout MYXV even when PKR is genetically knocked-out, suggesting  that  M029  targets  other  host  proteins  for  this  antiviral state inhibition. Thus, the MYXV  dsRNA  binding  protein  M029  not  only  antagonizes  PKR  from  multiple  species  but  also blocks the  type I IFN antiviral state independently of PKR in a highly species-specific fashion.


Assuntos
Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Interferon Tipo I/antagonistas & inibidores , Myxoma virus/patogenicidade , Proteínas de Ligação a RNA/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Humanos , Camundongos , Coelhos
11.
Proc Natl Acad Sci U S A ; 113(14): 3855-60, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26903626

RESUMO

Myxoma virus (MYXV) is a rabbit-specific poxvirus, which is highly virulent in European rabbits. The attenuation of MYXV and the increased resistance of rabbits following the release of MYXV in Australia is one of the best-documented examples of host-pathogen coevolution. To elucidate the molecular mechanisms that contribute to the restriction of MYXV infection to rabbits and MYXV attenuation in the field, we have studied the interaction of the MYXV protein M156 with the host antiviral protein kinase R (PKR). In yeast and cell-culture transfection assays, M156 only inhibited rabbit PKR but not PKR from other tested mammalian species. Infection assays with human HeLa PKR knock-down cells, which were stably transfected with human or rabbit PKR, revealed that only human but not rabbit PKR was able to restrict MYXV infection, whereas both PKRs were able to restrict replication of a vaccinia virus (VACV) strain that lacks the PKR inhibitors E3 and K3. Inactivation of M156R led to MYXV virus attenuation in rabbit cells, which was rescued by the ectopic expression of VACV E3 and K3. We further show that a mutation in the M156 encoding gene that was identified in more than 50% of MYXV field isolates from Australia resulted in an M156 variant that lost its ability to inhibit rabbit PKR and led to virus attenuation. The species-specific inhibition of rabbit PKR by M156 and the M156 loss-of-function in Australian MYXV field isolates might thus contribute to the species specificity of MYXV and to the attenuation in the field, respectively.


Assuntos
Myxoma virus/genética , Proteínas Virais/genética , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/genética , Animais , Austrália , Linhagem Celular Tumoral , Células HeLa , Humanos , Mutação/genética , Myxoma virus/patogenicidade , Coelhos , Proteínas Virais/metabolismo , Virulência/genética , Replicação Viral/genética
12.
PLoS One ; 10(2): e0118806, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25705900

RESUMO

Myxoma virus (MYXV) induces a lethal disease called Myxomatosis in European rabbits. MYXV is one of the rare viruses that encodes an α2,3-sialyltransferase through its M138L gene. In this study, we showed that although the absence of the enzyme was not associated with any in vitro deficit, the M138L deficient strains are highly attenuated in vivo. Indeed, while all rabbits infected with the parental and the revertant strains died within 9 days post-infection from severe myxomatosis, all but one rabbit inoculated with the M138L deficient strains survived the infection. In primary lesions, this resistance to the infection was associated with an increased ability of innate immune cells, mostly neutrophils, to migrate to the site of virus replication at 4 days post-infection. This was followed by the development of a better specific immune response against MYXV. Indeed, at day 9 post-infection, we observed an important proliferation of lymphocytes and an intense congestion of blood vessels in lymph nodes after M138L knockouts infection. Accordingly, in these rabbits, we observed an intense mononuclear cell infiltration throughout the dermis in primary lesions and higher titers of neutralizing antibodies. Finally, this adaptive immune response provided protection to these surviving rabbits against a challenge with the MYXV WT strain. Altogether, these results show that expression of the M138L gene contributes directly or indirectly to immune evasion by MYXV. In the future, these results could help us to better understand the pathogenesis of myxomatosis but also the importance of glycans in regulation of immune responses.


Assuntos
Tolerância Imunológica/imunologia , Myxoma virus/imunologia , Mixomatose Infecciosa/imunologia , Sialiltransferases/imunologia , Proteínas Virais/imunologia , Imunidade Adaptativa/imunologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , DNA Viral/sangue , DNA Viral/genética , DNA Viral/imunologia , Técnicas de Inativação de Genes , Interações Hospedeiro-Patógeno/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Masculino , Myxoma virus/patogenicidade , Myxoma virus/fisiologia , Mixomatose Infecciosa/sangue , Mixomatose Infecciosa/virologia , Coelhos , Sialiltransferases/genética , Sialiltransferases/metabolismo , Análise de Sobrevida , Fatores de Tempo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Virulência/genética , Virulência/imunologia , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Fatores de Virulência/metabolismo
13.
Trends Microbiol ; 23(2): 83-90, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25455418

RESUMO

Although viral emergence is commonly associated with cross-species transmission, the processes and determinants of viral evolution in a novel host environment are poorly understood. We address key questions in virus emergence and evolution using data generated from two unique natural experiments: the deliberate release of myxoma virus (MYXV) and rabbit hemorrhagic disease virus (RHDV) as biological control (biocontrol) agents against the European rabbit in Australia, and which have been of enormous benefit to Australia's ecosystem and agricultural industries. Notably, although virulence evolution in MYXV and RHDV followed different trajectories, a strongly parallel evolutionary process was observed in Australia and Europe. These biocontrol agents were also characterized by a lack of transmission to nontarget host species, suggesting that there are major barriers to successful emergence.


Assuntos
Agentes de Controle Biológico , Doenças Transmissíveis Emergentes/virologia , Evolução Molecular , Vírus da Doença Hemorrágica de Coelhos , Myxoma virus , Coelhos , Agricultura , Animais , Austrália , Infecções por Caliciviridae/microbiologia , Infecções por Caliciviridae/transmissão , Infecções por Caliciviridae/veterinária , Doenças Transmissíveis Emergentes/transmissão , Ecossistema , Europa (Continente) , Vírus da Doença Hemorrágica de Coelhos/genética , Vírus da Doença Hemorrágica de Coelhos/patogenicidade , Myxoma virus/genética , Myxoma virus/patogenicidade , Mixomatose Infecciosa/transmissão , Mixomatose Infecciosa/virologia , Filogenia , Virulência/genética
14.
Virology ; 464-465: 134-145, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25068401

RESUMO

Although the production of single gene knockout viruses is a useful strategy to study viral gene functions, the redundancy of many host interactive genes within a complex viral genome can obscure their collective functions. In this study, a rabbit-specific poxvirus, myxoma virus (MYXV), was genetically altered to disrupt multiple members of the poxviral ankyrin-repeat (ANK-R) protein superfamily, M-T5, M148, M149 and M150. A particularly robust activation of the NF-κB pathway was observed in A549 cells following infection with the complete ANK-R knockout (vMyx-ANKsKO). Also, an increased release of IL-6 was only observed upon infection with vMyx-ANKsKO. In virus-infected rabbit studies, vMyx-ANKsKO was the most extensively attenuated and produced the smallest primary lesion of all ANK-R mutant constructs. This study provides the first insights into the shared functions of the poxviral ANK-R protein superfamily in vitro and in vivo.


Assuntos
Myxoma virus/patogenicidade , Infecções por Poxviridae/veterinária , Coelhos/imunologia , Coelhos/virologia , Proteínas Virais/metabolismo , Animais , Repetição de Anquirina , Deleção de Genes , Interleucina-6/genética , Interleucina-6/imunologia , Myxoma virus/genética , Myxoma virus/metabolismo , NF-kappa B/genética , NF-kappa B/imunologia , Infecções por Poxviridae/imunologia , Infecções por Poxviridae/virologia , Coelhos/genética , Proteínas Virais/química , Proteínas Virais/genética , Virulência
15.
Chin Med J (Engl) ; 127(12): 2350-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24931255

RESUMO

BACKGROUND: Gallbladder carcinoma (GBC) has a high mortality rate, requiring synergistic anti-tumor management for effective treatment. The myxoma virus (MYXV) exhibits a modest clinical value through its oncolytic potential and narrow host tropism. METHODS: We performed viral replication assays, cell viability assays, migration assays, and xenograft tumor models to demonstrate that bone marrow-derived stem cells (BMSCs) may enhance efficiency of intravenous MYXV delivery. RESULTS: We examined the permissiveness of various GBC cell lines towards MYXV infection and found two supported single and multiple rounds of MYXV replication, leading to an oncolytic effect. Furthermore, we found that BMSCs exhibited tropism for GBC cells within a Matrigel migration system. BMSCs failed to affect the growth of GBC cells, in terms of tumor volume and survival time. Finally, we demonstrated in vivo that intravenous injection of MYXV-infected BMSCs significantly improves the oncolytic effect of MYXV alone, almost to the same extent as intratumoral injection of MYXV. CONCLUSION: This study indicates that BMSCs are a promising novel vehicle for MYXV to clinically address gallbladder tumors.


Assuntos
Células da Medula Óssea/citologia , Neoplasias da Vesícula Biliar/terapia , Myxoma virus/patogenicidade , Células-Tronco/citologia , Animais , Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , Feminino , Neoplasias da Vesícula Biliar/virologia , Humanos , Imuno-Histoquímica , Camundongos , Células-Tronco/fisiologia , Replicação Viral/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
J Virol ; 87(23): 12900-15, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24067966

RESUMO

The evolutionary interplay between myxoma virus (MYXV) and the European rabbit (Oryctolagus cuniculus) following release of the virus in Australia in 1950 as a biological control is a classic example of host-pathogen coevolution. We present a detailed genomic and phylogeographic analysis of 30 strains of MYXV, including the Australian progenitor strain Standard Laboratory Strain (SLS), 24 Australian viruses isolated from 1951 to 1999, and three isolates from the early radiation in Britain from 1954 and 1955. We show that in Australia MYXV has spread rapidly on a spatial scale, with multiple lineages cocirculating within individual localities, and that both highly virulent and attenuated viruses were still present in the field through the 1990s. In addition, the detection of closely related virus lineages at sites 1,000 km apart suggests that MYXV moves freely in geographic space, with mosquitoes, fleas, and rabbit migration all providing means of transport. Strikingly, despite multiple introductions, all modern viruses appear to be ultimately derived from the original introductions of SLS. The rapidity of MYXV evolution was also apparent at the genomic scale, with gene duplications documented in a number of viruses. Duplication of potential virulence genes may be important in increasing the expression of virulence proteins and provides the basis for the evolution of novel functions. Mutations leading to loss of open reading frames were surprisingly frequent and in some cases may explain attenuation, but no common mutations that correlated with virulence or attenuation were identified.


Assuntos
Evolução Molecular , Genoma Viral , Interações Hospedeiro-Patógeno , Myxoma virus/genética , Infecções por Poxviridae/veterinária , Coelhos/virologia , Adaptação Fisiológica , Animais , Dados de Sequência Molecular , Myxoma virus/isolamento & purificação , Myxoma virus/patogenicidade , Myxoma virus/fisiologia , Filogenia , Filogeografia , Infecções por Poxviridae/transmissão , Infecções por Poxviridae/virologia , Virulência
17.
J Virol ; 87(22): 12080-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23986601

RESUMO

Myxomatosis is a rapidly lethal disease of European rabbits that is caused by myxoma virus (MYXV). The introduction of a South American strain of MYXV into the European rabbit population of Australia is the classic case of host-pathogen coevolution following cross-species transmission. The most virulent strains of MYXV for European rabbits are the Californian viruses, found in the Pacific states of the United States and the Baja Peninsula, Mexico. The natural host of Californian MYXV is the brush rabbit, Sylvilagus bachmani. We determined the complete sequence of the MSW strain of Californian MYXV and performed a comparative analysis with other MYXV genomes. The MSW genome is larger than that of the South American Lausanne (type) strain of MYXV due to an expansion of the terminal inverted repeats (TIRs) of the genome, with duplication of the M156R, M154L, M153R, M152R, and M151R genes and part of the M150R gene from the right-hand (RH) end of the genome at the left-hand (LH) TIR. Despite the extreme virulence of MSW, no novel genes were identified; five genes were disrupted by multiple indels or mutations to the ATG start codon, including two genes, M008.1L/R and M152R, with major virulence functions in European rabbits, and a sixth gene, M000.5L/R, was absent. The loss of these gene functions suggests that S. bachmani is a relatively recent host for MYXV and that duplication of virulence genes in the TIRs, gene loss, or sequence variation in other genes can compensate for the loss of M008.1L/R and M152R in infections of European rabbits.


Assuntos
Adaptação Fisiológica/genética , Genoma Viral , Myxoma virus/genética , Mixomatose Infecciosa/virologia , Infecções Tumorais por Vírus/virologia , Proteínas Virais/genética , Virulência/genética , Animais , Sequência de Bases , Evolução Biológica , California , Europa (Continente) , México , Dados de Sequência Molecular , Myxoma virus/classificação , Myxoma virus/patogenicidade , Mixomatose Infecciosa/genética , Filogenia , Coelhos , Homologia de Sequência do Ácido Nucleico , Sequências Repetidas Terminais/genética , Infecções Tumorais por Vírus/genética , Replicação Viral
18.
Vaccine ; 31(39): 4252-8, 2013 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-23726825

RESUMO

Many common neoplasms are still noncurative with current standards of cancer therapy. More therapeutic modalities need to be developed to significantly prolong the lives of patients and eventually cure a wider spectrum of cancers. Oncolytic virotherapy is one of the promising new additions to clinical cancer therapeutics. Successful oncolytic virotherapy in the clinic will be those strategies that best combine tumor cell oncolysis with enhanced immune responses against tumor antigens. The current candidate oncolytic viruses all share the common property that they are relatively nonpathogenic to humans, yet they have the ability to replicate selectively in human cancer cells and induce cancer regression by direct oncolysis and/or induction of improved anti-tumor immune responses. Many candidate oncolytic viruses are in various stages of clinical and preclinical development. One such preclinical candidate is myxoma virus (MYXV), a member of the Poxviridae family that, in its natural setting, exhibits a very restricted host range and is only pathogenic to European rabbits. Despite its narrow host range in nature, MYXV has been shown to productively infect various classes of human cancer cells. Several preclinical in vivo modeling studies have demonstrated that MYXV is an attractive and safe candidate oncolytic virus, and hence, MYXV is currently being developed as a potential therapeutic for several cancers, such as pancreatic cancer, glioblastoma, ovarian cancer, melanoma, and hematologic malignancies. This review highlights the preclinical cancer models that have shown the most promise for translation of MYXV into human clinical trials.


Assuntos
Myxoma virus/patogenicidade , Terapia Viral Oncolítica , Vírus Oncolíticos/patogenicidade , Animais , Ensaios Clínicos como Assunto , Humanos , Modelos Animais , Myxoma virus/imunologia , Neoplasias/terapia , Vírus Oncolíticos/fisiologia , Coelhos/virologia
19.
PLoS Pathog ; 8(10): e1002950, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23055928

RESUMO

The attenuation of myxoma virus (MYXV) following its introduction as a biological control into the European rabbit populations of Australia and Europe is the canonical study of the evolution of virulence. However, the evolutionary genetics of this profound change in host-pathogen relationship is unknown. We describe the genome-scale evolution of MYXV covering a range of virulence grades sampled over 49 years from the parallel Australian and European epidemics, including the high-virulence progenitor strains released in the early 1950s. MYXV evolved rapidly over the sampling period, exhibiting one of the highest nucleotide substitution rates ever reported for a double-stranded DNA virus, and indicative of a relatively high mutation rate and/or a continually changing selective environment. Our comparative sequence data reveal that changes in virulence involved multiple genes, likely losses of gene function due to insertion-deletion events, and no mutations common to specific virulence grades. Hence, despite the similarity in selection pressures there are multiple genetic routes to attain either highly virulent or attenuated phenotypes in MYXV, resulting in convergence for phenotype but not genotype.


Assuntos
Evolução Molecular , Genoma Viral , Myxoma virus/genética , Myxoma virus/patogenicidade , Mixomatose Infecciosa/virologia , Animais , Austrália , Sequência de Bases , Evolução Biológica , DNA Viral/genética , Europa (Continente) , Dados de Sequência Molecular , Taxa de Mutação , Filogenia , Coelhos , Análise de Sequência de DNA
20.
PLoS One ; 7(5): e36823, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22606294

RESUMO

Plasmacytoid dendritic cells (pDCs) play important roles in antiviral innate immunity by producing type I interferon (IFN). In this study, we assess the immune responses of primary human pDCs to two poxviruses, vaccinia and myxoma virus. Vaccinia, an orthopoxvirus, was used for immunization against smallpox, a contagious human disease with high mortality. Myxoma virus, a Leporipoxvirus, causes lethal disease in rabbits, but is non-pathogenic in humans. We report that myxoma virus infection of human pDCs induces IFN-α and TNF production, whereas vaccinia infection does not. Co-infection of pDCs with myxoma virus plus vaccinia blocks myxoma induction effects. We find that heat-inactivated vaccinia (Heat-VAC; by incubating the virus at 55°C for 1 h) gains the ability to induce IFN-α and TNF in primary human pDCs. Induction of IFN-α in pDCs by myxoma virus or Heat-VAC is blocked by chloroquine, which inhibits endosomal acidification required for TLR7/9 signaling, and by inhibitors of cellular kinases PI3K and Akt. Using purified pDCs from genetic knockout mice, we demonstrate that Heat-VAC-induced type I IFN production in pDCs requires the endosomal RNA sensor TLR7 and its adaptor MyD88, transcription factor IRF7 and the type I IFN feedback loop mediated by IFNAR1. These results indicate that (i) vaccinia virus, but not myxoma virus, expresses inhibitor(s) of the poxvirus sensing pathway(s) in pDCs; and (ii) Heat-VAC infection fails to produce inhibitor(s) but rather produces novel activator(s), likely viral RNA transcripts that are sensed by the TLR7/MyD88 pathway. Using vaccinia gene deletion mutants, we show that the Z-DNA/RNA binding domain at the N-terminus of the vaccinia immunomodulatory E3 protein is an antagonist of the innate immune response of human pDCs to poxvirus infection and TLR agonists. The myxoma virus ortholog of vaccinia E3 (M029) lacks the N-terminal Z-DNA/RNA binding domain, which might contribute to the immunostimulating properties of myxoma virus.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Imunidade Inata , Proteínas de Ligação a RNA/imunologia , Vírus Vaccinia/imunologia , Proteínas Virais/imunologia , Animais , Cloroquina/farmacologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Regulação para Baixo , Humanos , Interferon-alfa/biossíntese , Glicoproteínas de Membrana/metabolismo , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , Myxoma virus/genética , Myxoma virus/imunologia , Myxoma virus/patogenicidade , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Coelhos , Receptor 7 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Vírus Vaccinia/genética , Vírus Vaccinia/patogenicidade , Proteínas Virais/química , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...