Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 232
Filter
Add more filters

Publication year range
1.
Immunity ; 54(2): 247-258.e7, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33444549

ABSTRACT

The vaccine strain against smallpox, vaccinia virus (VACV), is highly immunogenic yet causes relatively benign disease. These attributes are believed to be caused by gene loss in VACV. Using a targeted small interfering RNA (siRNA) screen, we identified a viral inhibitor found in cowpox virus (CPXV) and other orthopoxviruses that bound to the host SKP1-Cullin1-F-box (SCF) machinery and the essential necroptosis kinase receptor interacting protein kinase 3 (RIPK3). This "viral inducer of RIPK3 degradation" (vIRD) triggered ubiquitination and proteasome-mediated degradation of RIPK3 and inhibited necroptosis. In contrast to orthopoxviruses, the distantly related leporipoxvirus myxoma virus (MYXV), which infects RIPK3-deficient hosts, lacks a functional vIRD. Introduction of vIRD into VACV, which encodes a truncated and defective vIRD, enhanced viral replication in mice. Deletion of vIRD reduced CPXV-induced inflammation, viral replication, and mortality, which were reversed in RIPK3- and MLKL-deficient mice. Hence, vIRD-RIPK3 drives pathogen-host evolution and regulates virus-induced inflammation and pathogenesis.


Subject(s)
Cowpox virus/physiology , Cowpox/immunology , RNA, Small Interfering/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Vaccinia virus/metabolism , Viral Proteins/metabolism , Animals , Evolution, Molecular , HEK293 Cells , Host-Pathogen Interactions , Humans , Inflammation , Mice , Mice, Knockout , Necroptosis/genetics , Orthopoxvirus , Phylogeny , Protein Kinases/genetics , Proteolysis , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Sequence Analysis, RNA , Viral Proteins/genetics , Virus Replication
2.
J Virol ; 98(1): e0179123, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38168672

ABSTRACT

In the United States (US), biosafety and biosecurity oversight of research on viruses is being reappraised. Safety in virology research is paramount and oversight frameworks should be reviewed periodically. Changes should be made with care, however, to avoid impeding science that is essential for rapidly reducing and responding to pandemic threats as well as addressing more common challenges caused by infectious diseases. Decades of research uniquely positioned the US to be able to respond to the COVID-19 crisis with astounding speed, delivering life-saving vaccines within a year of identifying the virus. We should embolden and empower this strength, which is a vital part of protecting the health, economy, and security of US citizens. Herein, we offer our perspectives on priorities for revised rules governing virology research in the US.


Subject(s)
Biomedical Research , Containment of Biohazards , Virology , Humans , COVID-19 , United States , Viruses , Biomedical Research/standards
3.
J Gen Virol ; 104(5)2023 05.
Article in English | MEDLINE | ID: mdl-37195882

ABSTRACT

Poxviridae is a family of enveloped, brick-shaped or ovoid viruses. The genome is a linear molecule of dsDNA (128-375 kbp) with covalently closed ends. The family includes the sub-families Entomopoxvirinae, whose members have been found in four orders of insects, and Chordopoxvirinae, whose members are found in mammals, birds, reptiles and fish. Poxviruses are important pathogens in various animals, including humans, and typically result in the formation of lesions, skin nodules, or disseminated rash. Infections can be fatal. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Poxviridae, which is available at ictv.global/report/poxviridae.


Subject(s)
Poxviridae , Animals , Humans , Poxviridae/genetics , Fishes , Birds , Mammals , Reptiles , Genome, Viral , Virus Replication , Virion
4.
J Virol ; 95(14): e0015121, 2021 06 24.
Article in English | MEDLINE | ID: mdl-33952639

ABSTRACT

RNA helicase A/DHX9 is required for diverse RNA-related essential cellular functions and antiviral responses and is hijacked by RNA viruses to support their replication. Here, we show that during the late replication stage in human cancer cells of myxoma virus (MYXV), a member of the double-stranded DNA (dsDNA) poxvirus family that is being developed as an oncolytic virus, DHX9, forms unique granular cytoplasmic structures, which we named "DHX9 antiviral granules." These DHX9 antiviral granules are not formed if MYXV DNA replication and/or late protein synthesis is blocked. When formed, DHX9 antiviral granules significantly reduced nascent protein synthesis in the MYXV-infected cancer cells. MYXV late gene transcription and translation were also significantly compromised, particularly in nonpermissive or semipermissive human cancer cells where MYXV replication is partly or completely restricted. Directed knockdown of DHX9 significantly enhanced viral late protein synthesis and progeny virus formation in normally restrictive cancer cells. We further demonstrate that DHX9 is not a component of the canonical cellular stress granules. DHX9 antiviral granules are induced by MYXV, and other poxviruses, in human cells and are associated with other known cellular components of stress granules, dsRNA and virus encoded dsRNA-binding protein M029, a known interactor with DHX9. Thus, DHX9 antiviral granules function by hijacking poxviral elements needed for the cytoplasmic viral replication factories. These results demonstrate a novel antiviral function for DHX9 that is recruited from the nucleus into the cytoplasm, and this step can be exploited to enhance oncolytic virotherapy against the subset of human cancer cells that normally restrict MYXV. IMPORTANCE The cellular DHX9 has both proviral and antiviral roles against diverse RNA and DNA viruses. In this article, we demonstrate that DHX9 can form unique antiviral granules in the cytoplasm during myxoma virus (MYXV) replication in human cancer cells. These antiviral granules sequester viral proteins and reduce viral late protein synthesis and thus regulate MYXV, and other poxviruses, that replicate in the cytoplasm. In addition, we show that in the absence of DHX9, the formation of DHX9 antiviral granules can be inhibited, which significantly enhanced oncolytic MYXV replication in human cancer cell lines where the virus is normally restricted. Our results also show that DHX9 antiviral granules are formed after viral infection but not by common nonviral cellular stress inducers. Thus, our study suggests that DHX9 has antiviral activity in human cancer cells, and this pathway can be targeted for enhanced activity of oncolytic poxviruses against even restrictive cancer cells.


Subject(s)
Cytoplasmic Granules/physiology , DEAD-box RNA Helicases/physiology , Myxoma virus/physiology , Neoplasm Proteins/physiology , Animals , Antiviral Agents , Cell Line, Tumor , Cytoplasmic Granules/chemistry , DEAD-box RNA Helicases/genetics , HeLa Cells , Humans , Neoplasm Proteins/genetics , Protein Biosynthesis , Rabbits , Stress, Physiological , Viral Proteins/metabolism , Virus Replication
5.
Exp Parasitol ; 239: 108263, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35598646

ABSTRACT

Schistosomiasis is a devastating disease caused by parasitic flatworms of the genus Schistosoma. Praziquantel (PZQ), the current treatment of choice, is ineffective against immature worms and cannot prevent reinfection. The continued reliance on a single drug for treatment increases the risk of the development of PZQ-resistant parasites. Reports of PZQ insusceptibility lends urgency to the need for new therapeutics. Here, we report that Myxoma virus (MYXV), an oncolytic pox virus which is non-pathogenic in all mammals except leporids, infects and replicates in S. mansoni schistosomula, juveniles, and adult male and female worms. MYXV infection results in the shredding of the tegument and reduced egg production in vitro, identifying MYXV as the first viral pathogen of schistosomes. MYXV is currently in preclinical studies to manage multiple human cancers, supporting its use in human therapeutics. Our findings raise the exciting possibility that MYXV virus represents a novel and safe class of potential anthelmintic therapeutics.


Subject(s)
Anthelmintics , Myxoma virus , Oncolytic Viruses , Schistosomiasis mansoni , Animals , Anthelmintics/pharmacology , Female , Humans , Male , Mammals , Praziquantel/pharmacology , Schistosoma mansoni , Schistosomiasis mansoni/drug therapy
6.
J Biol Chem ; 294(21): 8480-8489, 2019 05 24.
Article in English | MEDLINE | ID: mdl-30940649

ABSTRACT

Among the repertoire of immunoregulatory proteins encoded by myxoma virus, M013 is a viral homologue of the viral pyrin domain-only protein (vPOP) family. In myeloid cells, M013 protein has been shown to inhibit both the inflammasome and NF-κB signaling pathways by direct binding to ASC1 and NF-κB1, respectively. In this study, a three-dimensional homology model of the M013 pyrin domain (PYD) was built based on similarities to known PYD structures. A distinctive feature of the deduced surface electrostatic map of the M013 PYD is the presence of a negatively region consisting of numerous aspartate and glutamate residues in close proximity. Single-site mutations of aspartate and glutamate residues reveal their role in interactions with ASC-1. The biological significance of charge complementarity in the M013-ASC-1 interaction was further confirmed by functional assays of caspase-1 activation and subsequent secretion of cytokines. M013 also has a unique 33-residue C-terminal tail that follows the N-terminal PYD, and it is enriched in positively charged residues. Deletion of the tail of M013 significantly inhibited the interactions between M013 and NF-κB1, thus compromising the ability of the viral protein to suppress the secretion of pro-inflammatory cytokines. These results demonstrate that vPOP M013 exploits distinct structural motifs to regulate both the inflammasome and NF-κB pathways.


Subject(s)
Myxoma virus , NF-kappa B/immunology , Signal Transduction/immunology , Viral Proteins , Amino Acid Motifs , Amino Acid Substitution , Caspase 1/genetics , Caspase 1/immunology , HeLa Cells , Humans , Inflammasomes/genetics , Mutagenesis, Site-Directed , Mutation, Missense , Myxoma virus/chemistry , Myxoma virus/genetics , Myxoma virus/immunology , NF-kappa B/genetics , Protein Domains , Signal Transduction/genetics , THP-1 Cells , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/immunology
8.
Proc Natl Acad Sci U S A ; 113(14): 3855-60, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26903626

ABSTRACT

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.


Subject(s)
Myxoma virus/genetics , Viral Proteins/genetics , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/genetics , Animals , Australia , Cell Line, Tumor , HeLa Cells , Humans , Mutation/genetics , Myxoma virus/pathogenicity , Rabbits , Viral Proteins/metabolism , Virulence/genetics , Virus Replication/genetics
9.
Biochemistry ; 57(7): 1096-1107, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29227673

ABSTRACT

The Myxomavirus-derived protein Serp-1 has potent anti-inflammatory activity in models of vasculitis, lupus, viral sepsis, and transplant. Serp-1 has also been tested successfully in a Phase IIa clinical trial in unstable angina, representing a "first-in-class" therapeutic. Recently, peptides derived from the reactive center loop (RCL) have been developed as stand-alone therapeutics for reducing vasculitis and improving survival in MHV68-infected mice. However, both Serp-1 and the RCL peptides lose activity in MHV68-infected mice after antibiotic suppression of intestinal microbiota. Here, we utilize a structure-guided approach to design and test a series of next-generation RCL peptides with improved therapeutic potential that is not reduced when the peptides are combined with antibiotic treatments. The crystal structure of cleaved Serp-1 was determined to 2.5 Å resolution and reveals a classical serpin structure with potential for serpin-derived RCL peptides to bind and inhibit mammalian serpins, plasminogen activator inhibitor 1 (PAI-1), anti-thrombin III (ATIII), and α-1 antitrypsin (A1AT), and target proteases. Using in silico modeling of the Serp-1 RCL peptide, S-7, we designed several modified RCL peptides that were predicted to have stronger interactions with human serpins because of the larger number of stabilizing hydrogen bonds. Two of these peptides (MPS7-8 and -9) displayed extended activity, improving survival where activity was previously lost in antibiotic-treated MHV68-infected mice (P < 0.0001). Mass spectrometry and kinetic assays suggest interaction of the peptides with ATIII, A1AT, and target proteases in mouse and human plasma. In summary, we present the next step toward the development of a promising new class of anti-inflammatory serpin-based therapeutics.


Subject(s)
Immunologic Factors/chemistry , Myxoma virus/chemistry , Peptides/chemistry , Serpins/chemistry , Viral Proteins/chemistry , Animals , CHO Cells , Cricetulus , Crystallography, X-Ray , Humans , Immunologic Factors/pharmacology , Mice , Mice, Inbred C57BL , Models, Molecular , Peptides/pharmacology , Poxviridae Infections/virology , Protein Conformation , Rabbits , Serpins/pharmacology , Viral Proteins/pharmacology
10.
Blood ; 125(24): 3778-88, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25904246

ABSTRACT

Allogeneic hematopoietic cell transplant (allo-HCT) can be curative for certain hematologic malignancies, but the risk of graft-versus-host disease (GVHD) is a major limitation for wider application. Ideally, strategies to improve allo-HCT would involve suppression of T lymphocytes that drive GVHD while sparing those that mediate graft-versus-malignancy (GVM). Recently, using a xenograft model, we serendipitously discovered that myxoma virus (MYXV) prevented GVHD while permitting GVM. In this study, we show that MYXV binds to resting, primary human T lymphocytes but will only proceed into active virus infection after the T cells receive activation signals. MYXV-infected T lymphocytes exhibited impaired proliferation after activation with reduced expression of interferon-γ, interleukin-2 (IL-2), and soluble IL-2Rα, but did not affect expression of IL-4 and IL-10. MYXV suppressed T-cell proliferation in 2 patterns (full vs partial) depending on the donor. In terms of GVM, we show that MYXV-infected activated human T lymphocytes effectively deliver live oncolytic virus to human multiple myeloma cells, thus augmenting GVM by transfer of active oncolytic virus to residual cancer cells. Given this dual capacity of reducing GVHD plus increasing the antineoplastic effectiveness of GVM, ex vivo virotherapy with MYXV may be a promising clinical adjunct to allo-HCT regimens.


Subject(s)
Multiple Myeloma/therapy , Myxoma virus/immunology , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Graft vs Host Disease/immunology , Graft vs Host Disease/therapy , Hematopoietic Stem Cell Transplantation , Humans , Lymphocyte Activation , Poxviridae Infections/immunology , T-Lymphocytes/cytology , Tumor Virus Infections/immunology
11.
J Virol ; 89(3): 1925-31, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25428864

ABSTRACT

We show that SAMD9 is an innate host antiviral stress response element that participates in the formation of antiviral granules. Poxviruses, myxoma virus and vaccinia virus specifically, utilize a virus-encoded host range factor(s), such as a member of the C7L superfamily, to antagonize SAMD9 to prevent granule formation in a eukaryotic initiation factor 2α (eIF2α)-independent manner. When SAMD9 is stimulated due to failure of the viral antagonism during infection, the resulting antiviral granules exhibit properties different from those of the canonical stress granules.


Subject(s)
Antiviral Agents/metabolism , Immunity, Innate , Poxviridae/immunology , Proteins/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins
12.
Cytotherapy ; 18(3): 465-80, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26857235

ABSTRACT

BACKGROUND: Relapsing disease is a major challenge after hematopoietic cell transplantation for hematological malignancies. Myxoma virus (MYXV) is an oncolytic virus that can target and eliminate contaminating cancer cells from auto-transplant grafts. The aims of this study were to examine the impact of MYXV on normal hematopoietic stem and progenitor cells and define the optimal treatment conditions for ex vivo virotherapy. METHODS: Bone marrow (BM) and mobilized peripheral blood stem cells (mPBSCs) from patients with hematologic malignancies were treated with MYXV at various time, temperature and incubation media conditions. Treated BM cells from healthy normal donors were evaluated using flow cytometry for MYXV infection, long-term culture-initiating cell (LTC-IC) assay and colony-forming cell (CFC) assay. RESULTS: MYXV initiated infection in up to 45% of antigen-presenting monocytes, B cells and natural killer cells; however, these infections were uniformly aborted in >95% of all cells. Fresh graft sources showed higher levels of MYXV infection initiation than cryopreserved specimens, but in all cases less than 10% of CD34(+) cells could be infected after ex vivo MYXV treatment. MYXV did not impair LTC-IC colony numbers compared with mock treatment. CFC colony types and numbers were also not impaired by MYXV treatment. MYXV incubation time, temperature or culture media did not significantly change the percentage of infected cells, LTC-IC colony formation or CFC colony formation. CONCLUSIONS: Human hematopoietic cells are non-permissive for MYXV. Human hematopoietic stem and progenitor cells were not infected and thus unaffected by MYXV ex vivo treatment.


Subject(s)
Cell Culture Techniques/methods , Cell Separation/methods , Hematologic Neoplasms/pathology , Hematopoietic Stem Cells/cytology , Myxoma virus/physiology , Oncolytic Virotherapy/methods , Adult , Antigens, CD34/metabolism , Autografts/standards , Bone Marrow/pathology , Bone Marrow Cells/pathology , Cells, Cultured , Female , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/standards , Hematopoietic Stem Cells/physiology , Humans , Male , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/prevention & control , Transplantation Conditioning/methods
13.
Arch Virol ; 161(11): 2991-3002, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27465567

ABSTRACT

The orthopoxvirus vaccinia virus (VACV) interacts with both actin and microtubule cytoskeletons in order to generate and spread progeny virions. Here, we present evidence demonstrating the involvement of PAK1 (p21-activated kinase 1) in the dissemination of VACV. Although PAK1 activation has previously been associated with optimal VACV entry via macropinocytosis, its absence does not affect the production of intracellular mature virions (IMVs) and extracellular enveloped virions (EEVs). Our data demonstrate that low-multiplicity infection of PAK1(-/-) MEFs leads to a reduction in plaque size followed by decreased production of both IMVs and EEVs, strongly suggesting that virus spread was impaired in the absence of PAK1. Confocal and scanning electron microscopy showed a substantial reduction in the amount of VACV-induced actin tails in PAK1(-/-) MEFs, but no significant alteration in the total amount of cell-associated enveloped virions (CEVs). Furthermore, the decreased VACV dissemination in PAK1(-/-) cells was correlated with the absence of phosphorylated ARPC1 (Thr21), a downstream target of PAK1 and a key regulatory subunit of the ARP2/3 complex, which is necessary for the formation of actin tails and viral spread. We conclude that PAK1, besides its role in virus entry, also plays a relevant role in VACV dissemination.


Subject(s)
Endocytosis , Host-Pathogen Interactions , Vaccinia virus/physiology , Virus Internalization , p21-Activated Kinases/metabolism , Animals , Biological Transport , Cells, Cultured , Mice , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron, Scanning , p21-Activated Kinases/genetics
14.
Biochem Biophys Res Commun ; 462(4): 283-7, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-25843801

ABSTRACT

Epstein-Barr virus (EBV) has been associated with a variety of epithelial and hematologic malignancies, including B-, T- and NK cell-lymphomas, Hodgkin's disease (HD), post-transplant lymphoproliferative diseases (LPDs), nasopharyngeal and gastric carcinomas, smooth muscle tumors, and HIV-associated lymphomas. Currently, treatment options for EBV-associated malignancies are limited. We have previously shown that myxoma virus specifically targets various human solid tumors and leukemia cells in a variety of animal models, while sparing normal human or murine tissues. Since transplant recipients of bone marrow or solid organs often develop EBV-associated post-transplant LPDs and lymphoma, myxoma virus may be of utility to prevent EBV-associated malignancies in immunocompromised transplant patients where treatment options are frequently limited. In this report, we demonstrate the safety and efficacy of myxoma virus purging as a prophylactic strategy for preventing post-transplant EBV-transformed human lymphomas, using a highly immunosuppressed mouse xenotransplantation model. This provides support for developing myxoma virus as a potential oncolytic therapy for preventing EBV-associated LPDs following transplantation of bone marrow or solid organ allografts.


Subject(s)
Herpesvirus 4, Human/pathogenicity , Lymphoma, B-Cell/prevention & control , Lymphoproliferative Disorders/complications , Oncolytic Virotherapy , Transplantation/adverse effects , Animals , Disease Models, Animal , Heterografts , Lymphoma, B-Cell/virology , Lymphoproliferative Disorders/etiology , Mice
15.
PLoS Pathog ; 9(7): e1003465, 2013.
Article in English | MEDLINE | ID: mdl-23853588

ABSTRACT

Myxoma virus (MYXV)-encoded protein M029 is a member of the poxvirus E3 family of dsRNA-binding proteins that antagonize the cellular interferon signaling pathways. In order to investigate additional functions of M029, we have constructed a series of targeted M029-minus (vMyx-M029KO and vMyx-M029ID) and V5-tagged M029 MYXV. We found that M029 plays a pivotal role in determining the cellular tropism of MYXV in all mammalian cells tested. The M029-minus viruses were able to replicate only in engineered cell lines that stably express a complementing protein, such as vaccinia E3, but underwent abortive or abated infection in all other tested mammalian cell lines. The M029-minus viruses were dramatically attenuated in susceptible host European rabbits and caused no observable signs of myxomatosis. Using V5-tagged M029 virus, we observed that M029 expressed as an early viral protein is localized in both the nuclear and cytosolic compartments in virus-infected cells, and is also incorporated into virions. Using proteomic approaches, we have identified Protein Kinase R (PKR) and RNA helicase A (RHA)/DHX9 as two cellular binding partners of M029 protein. In virus-infected cells, M029 interacts with PKR in a dsRNA-dependent manner, while binding with DHX9 was not dependent on dsRNA. Significantly, PKR knockdown in human cells rescued the replication defect of the M029-knockout viruses. Unexpectedly, this rescue of M029-minus virus replication by PKR depletion could then be reversed by RHA/DHX9 knockdown in human monocytic THP1 cells. This indicates that M029 not only inhibits generic PKR anti-viral pathways, but also binds and conscripts RHA/DHX9 as a pro-viral effector to promote virus replication in THP1 cells. Thus, M029 is a critical host range and virulence factor for MYXV that is required for replication in all mammalian cells by antagonizing PKR-mediated anti-viral functions, and also conscripts pro-viral RHA/DHX9 to promote viral replication specifically in myeloid cells.


Subject(s)
DEAD-box RNA Helicases/metabolism , Monocytes/immunology , Myxoma virus/physiology , Neoplasm Proteins/metabolism , Viral Proteins/metabolism , Viral Tropism , Virus Replication , eIF-2 Kinase/metabolism , Animals , Antiviral Agents/metabolism , Antiviral Agents/therapeutic use , Cell Line , Cells, Cultured , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/genetics , Disease Susceptibility , Female , Gene Knockout Techniques , Humans , Interferon Type I/metabolism , Interferon Type I/therapeutic use , Monocytes/metabolism , Monocytes/virology , Mutation , Myxomatosis, Infectious/prevention & control , Myxomatosis, Infectious/virology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rabbits , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Viral Proteins/antagonists & inhibitors , Viral Proteins/genetics , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/genetics
16.
Nat Rev Immunol ; 2(7): 521-7, 2002 07.
Article in English | MEDLINE | ID: mdl-12094226

ABSTRACT

Smallpox was eradicated in 1977, but it remains a concern owing to the potential use of the causative agent variola virus in bioterrorism. This article provides an overview of the World Health Organization's spectacular success in achieving the eradication of smallpox. It discusses how variola virus could potentially re-emerge and how prepared we are to counter such a re-emergence. Finally, the potential threat from other orthopoxviruses that exist naturally or that have been genetically engineered is considered. In the words of Rep. Christopher Shay, 'Better to be scared by the improbable possibility than to be unprepared for the catastrophic reality'.


Subject(s)
Bioterrorism , Smallpox/history , Variola virus , History, 15th Century , History, 16th Century , History, 17th Century , History, 18th Century , History, 19th Century , History, 20th Century , History, 21st Century , History, Ancient , History, Medieval , Humans , Smallpox/prevention & control , Smallpox Vaccine/history , Vaccination/history , World Health Organization
17.
Mol Cancer ; 13: 82, 2014 Apr 13.
Article in English | MEDLINE | ID: mdl-24725816

ABSTRACT

BACKGROUND: Gallbladder carcinoma (GBC) is highly lethal, and effective treatment will require synergistic anti-tumor management. The study is aimed at investigating the oncolytic value of myxoma virus (MYXV) infection against GBC and optimizing MYXV oncolytic efficiency. METHODS: We examined the permissiveness of GBC cell lines to MYXV infection and compared the effects of MYXV on cell viability among GBC and control permissive glioma cells in vitro and in vivo after MYXV + rapamycin (Rap) treatment, which is known to enhance cell permissiveness to MYXV by upregulating p-Akt levels. We also assessed MYXV + hyaluronan (HA) therapy efficiency by examinating Akt activation status, MMP-9 expression, cell viability, and collagen distribution. We further compared hydraulic conductivity, tumor area, and survival of tumor-bearing mice between the MYXV + Rap and MYXV + HA therapeutic regimens. RESULTS: MYXV + Rap treatment could considerably increase the oncolytic ability of MYXV against GBC cell lines in vitro but not against GBC xenografts in vivo. We found higher levels of collagen IV in GBC tumors than in glioma tumors. Diffusion analysis demonstrated that collagen IV could physically hinder MYXV intratumoral distribution. HA-CD44 interplay was found to activate the Akt signaling pathway, which increases oncolytic rates. HA was also found to enhance the MMP-9 secretion, which contributes to collagen IV degradation. CONCLUSIONS: Unlike MYXV + Rap, MYXV + HA therapy significantly enhanced the anti-tumor effects of MYXV in vivo and prolonged survival of GBC tumor-bearing mice. HA may optimize the oncolytic effects of MYXV on GBC via the HA-CD44 interaction which can promote viral infection and diffusion.


Subject(s)
Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/therapy , Oncolytic Virotherapy , Animals , Cell Line, Tumor , Gallbladder Neoplasms/pathology , Gallbladder Neoplasms/virology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyaluronic Acid/administration & dosage , In Vitro Techniques , Mice , Myxoma virus/genetics , Sirolimus/administration & dosage , Xenograft Model Antitumor Assays
18.
Clin Immunol ; 153(2): 254-63, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24845791

ABSTRACT

Many viruses encode virulence factors to facilitate their own survival by modulating a host's inflammatory response. One of these factors, secreted from cells infected with myxoma virus, is the serine proteinase inhibitor (serpin) Serp-1. Because Serp-1 had demonstrated anti-inflammatory properties in arterial injury models and viral infections, it was cloned and evaluated for therapeutic efficacy in collagen-induced arthritis (CIA). Clinical severity was significantly lower in the Serp-1 protocols (p<0.0001) and blinded radiographs indicated that the Serp-1 group had significantly less erosions than the controls (p<0.01). Delayed-type hypersensitivity was lower in the Serp-1 group but antibody titers to type II collagen were not significantly altered. Recipients had minimal histopathologic synovial changes and did not develop neutralizing antibodies to Serp-1. These results indicate that Serp-1 impedes the pathogenesis of CIA and suggests that the therapeutic potential of serine proteinase inhibitors in inflammatory joint diseases, such as rheumatoid arthritis, should be investigated further.


Subject(s)
Arthritis, Experimental/prevention & control , Serine Proteinase Inhibitors/pharmacology , Serpins/pharmacology , Viral Proteins/pharmacology , Amino Acid Sequence , Animals , Antibodies/blood , Antibodies/immunology , Arthritis, Experimental/diagnostic imaging , Arthritis, Experimental/immunology , Blood Cell Count , CHO Cells , Cricetinae , Cricetulus , Enzyme-Linked Immunosorbent Assay , Female , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/prevention & control , Immunoblotting , Joints/drug effects , Joints/immunology , Joints/pathology , Molecular Sequence Data , Myxoma virus/genetics , Myxoma virus/metabolism , Radiography , Rats , Rats, Inbred Strains , Serine Proteinase Inhibitors/genetics , Serine Proteinase Inhibitors/immunology , Serpins/genetics , Serpins/immunology , Viral Proteins/genetics , Viral Proteins/immunology
19.
Immunogenetics ; 66(1): 43-52, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24220721

ABSTRACT

One of the most severe European rabbit (Oryctolagus cuniculus) pathogens is myxoma virus (MYXV), a rabbit-specific leporipoxvirus that causes the highly lethal disease myxomatosis. Other leporid genera, Sylvilagus and Lepus, encompass species with variable susceptibilities to MYXV, but these do not develop the lethal form of the disease. The protective role of the retinoic acid-inducible gene-I (RIG-I/DDX58) in sensing MYXV in nonpermissive human myeloid cells prompted the study of the RIG-I-like receptor (RLR) family evolution in the three leporid genera. This viral-sensor family also includes the melanoma differentiation-associated factor 5 (MDA5/IFIH1), and the laboratory of genetics and physiology 2 (LGP2/DHX58). Considering specifically the MYXV susceptible host (European rabbit) and one of the virus natural long-term hosts (Sylvilagus bachmani, brush rabbit), the amino acid differences of positively selected sites in RIG-I between the two species were located in the protein region responsible for viral RNA recognition and binding, the repressor domain. Such differences might play a determinant role in how MYXV is sensed. When looking for episodic selection on MDA5 and LGP2 of the eastern cottontail (Sylvilagus floridanus), we also uncovered evidence of selective pressures that might be exerted by a species-specific leporipoxvirus, the Shope fibroma virus. Finally, a putative alternative splicing case was identified in Oryctolagus and Lepus MDA5 isoforms, corresponding to the deletion of one specific exon. This study provided the first insights into the evolution of the leporid RLR gene family that helps illuminate the origins of the species-specific innate responses to pathogens and more specifically to MYXV.


Subject(s)
Evolution, Molecular , Hares/genetics , RNA Helicases/immunology , Rabbits/genetics , Receptors, Virus/immunology , Viruses/immunology , Animals , Hares/immunology , Hares/virology , Humans , Models, Molecular , Phylogeny , Protein Conformation , RNA Helicases/genetics , RNA, Messenger/genetics , Rabbits/classification , Rabbits/immunology , Rabbits/virology , Real-Time Polymerase Chain Reaction , Receptors, Virus/genetics , Reverse Transcriptase Polymerase Chain Reaction
20.
J Virol ; 87(8): 4445-60, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23388707

ABSTRACT

Myxoma virus (MYXV) and vaccinia virus (VACV), two distinct members of the family Poxviridae, are both currently being developed as oncolytic virotherapeutic agents. Recent studies have demonstrated that ex vivo treatment with MYXV can selectively recognize and kill contaminating cancerous cells from autologous bone marrow transplants without perturbing the engraftment of normal CD34(+) hematopoietic stem and progenitor cells. However, the mechanism(s) by which MYXV specifically recognizes and eliminates the cancer cells in the autografts is not understood. While little is known about the cellular attachment factor(s) exploited by MYXV for entry into any target cells, VACV has been shown to utilize cell surface glycosaminoglycans such as heparan sulfate (HS), the extracellular matrix protein laminin, and/or integrin ß1. We have constructed MYXV and VACV virions tagged with the Venus fluorescent protein and compared their characteristics of binding to various human cancer cell lines as well as to primary human leukocytes. We report that the binding of MYXV or VACV to some adherent cell lines could be partially inhibited by heparin, but laminin blocked only VACV binding. In contrast to cultured fibroblasts, the binding of MYXV and VACV to a wide spectrum of primary human leukocytes could not be competed by either HS or laminin. Additionally, MYXV and VACV exhibited very different binding characteristics against certain select human leukocytes, suggesting that the two poxviruses utilize different cell surface determinants for the attachment to these cells. These results indicate that VACV and MYXV can exhibit very different oncolytic tropisms against some cancerous human leukocytes.


Subject(s)
Leukocytes/virology , Myxoma virus/physiology , Vaccinia virus/physiology , Virus Attachment , Cell Line, Tumor , Humans
SELECTION OF CITATIONS
SEARCH DETAIL