Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 232
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 54(2): 247-258.e7, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33444549

RESUMEN

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.


Asunto(s)
Virus de la Viruela Vacuna/fisiología , Viruela Vacuna/inmunología , ARN Interferente Pequeño/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Virus Vaccinia/metabolismo , Proteínas Virales/metabolismo , Animales , Evolución Molecular , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Inflamación , Ratones , Ratones Noqueados , Necroptosis/genética , Orthopoxvirus , Filogenia , Proteínas Quinasas/genética , Proteolisis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Análisis de Secuencia de ARN , Proteínas Virales/genética , Replicación Viral
2.
J Virol ; 98(1): e0179123, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38168672

RESUMEN

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.


Asunto(s)
Investigación Biomédica , Contención de Riesgos Biológicos , Virología , Humanos , COVID-19 , Estados Unidos , Virus , Investigación Biomédica/normas
3.
J Gen Virol ; 104(5)2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37195882

RESUMEN

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.


Asunto(s)
Poxviridae , Animales , Humanos , Poxviridae/genética , Peces , Aves , Mamíferos , Reptiles , Genoma Viral , Replicación Viral , Virión
4.
J Virol ; 95(14): e0015121, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-33952639

RESUMEN

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.


Asunto(s)
Gránulos Citoplasmáticos/fisiología , ARN Helicasas DEAD-box/fisiología , Myxoma virus/fisiología , Proteínas de Neoplasias/fisiología , Animales , Antivirales , Línea Celular Tumoral , Gránulos Citoplasmáticos/química , ARN Helicasas DEAD-box/genética , Células HeLa , Humanos , Proteínas de Neoplasias/genética , Biosíntesis de Proteínas , Conejos , Estrés Fisiológico , Proteínas Virales/metabolismo , Replicación Viral
5.
Exp Parasitol ; 239: 108263, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35598646

RESUMEN

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.


Asunto(s)
Antihelmínticos , Myxoma virus , Virus Oncolíticos , Esquistosomiasis mansoni , Animales , Antihelmínticos/farmacología , Femenino , Humanos , Masculino , Mamíferos , Praziquantel/farmacología , Schistosoma mansoni , Esquistosomiasis mansoni/tratamiento farmacológico
6.
J Biol Chem ; 294(21): 8480-8489, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-30940649

RESUMEN

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.


Asunto(s)
Myxoma virus , FN-kappa B/inmunología , Transducción de Señal/inmunología , Proteínas Virales , Secuencias de Aminoácidos , Sustitución de Aminoácidos , Caspasa 1/genética , Caspasa 1/inmunología , Células HeLa , Humanos , Inflamasomas/genética , Mutagénesis Sitio-Dirigida , Mutación Missense , Myxoma virus/química , Myxoma virus/genética , Myxoma virus/inmunología , FN-kappa B/genética , Dominios Proteicos , Transducción de Señal/genética , Células THP-1 , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/inmunología
8.
Proc Natl Acad Sci U S A ; 113(14): 3855-60, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-26903626

RESUMEN

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.


Asunto(s)
Myxoma virus/genética , Proteínas Virales/genética , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/genética , Animales , Australia , Línea Celular Tumoral , Células HeLa , Humanos , Mutación/genética , Myxoma virus/patogenicidad , Conejos , Proteínas Virales/metabolismo , Virulencia/genética , Replicación Viral/genética
9.
Biochemistry ; 57(7): 1096-1107, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29227673

RESUMEN

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.


Asunto(s)
Factores Inmunológicos/química , Myxoma virus/química , Péptidos/química , Serpinas/química , Proteínas Virales/química , Animales , Células CHO , Cricetulus , Cristalografía por Rayos X , Humanos , Factores Inmunológicos/farmacología , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Péptidos/farmacología , Infecciones por Poxviridae/virología , Conformación Proteica , Conejos , Serpinas/farmacología , Proteínas Virales/farmacología
10.
Blood ; 125(24): 3778-88, 2015 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-25904246

RESUMEN

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.


Asunto(s)
Mieloma Múltiple/terapia , Myxoma virus/inmunología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Linfocitos T/inmunología , Linfocitos T/virología , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/terapia , Trasplante de Células Madre Hematopoyéticas , Humanos , Activación de Linfocitos , Infecciones por Poxviridae/inmunología , Linfocitos T/citología , Infecciones Tumorales por Virus/inmunología
11.
J Virol ; 89(3): 1925-31, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25428864

RESUMEN

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.


Asunto(s)
Antivirales/metabolismo , Inmunidad Innata , Poxviridae/inmunología , Proteínas/metabolismo , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular
12.
Cytotherapy ; 18(3): 465-80, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26857235

RESUMEN

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.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Separación Celular/métodos , Neoplasias Hematológicas/patología , Células Madre Hematopoyéticas/citología , Myxoma virus/fisiología , Viroterapia Oncolítica/métodos , Adulto , Antígenos CD34/metabolismo , Autoinjertos/normas , Médula Ósea/patología , Células de la Médula Ósea/patología , Células Cultivadas , Femenino , Trasplante de Células Madre Hematopoyéticas/métodos , Trasplante de Células Madre Hematopoyéticas/normas , Células Madre Hematopoyéticas/fisiología , Humanos , Masculino , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/prevención & control , Acondicionamiento Pretrasplante/métodos
13.
Arch Virol ; 161(11): 2991-3002, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27465567

RESUMEN

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.


Asunto(s)
Endocitosis , Interacciones Huésped-Patógeno , Virus Vaccinia/fisiología , Internalización del Virus , Quinasas p21 Activadas/metabolismo , Animales , Transporte Biológico , Células Cultivadas , Ratones , Ratones Noqueados , Microscopía Confocal , Microscopía Electrónica de Rastreo , Quinasas p21 Activadas/genética
14.
Biochem Biophys Res Commun ; 462(4): 283-7, 2015 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-25843801

RESUMEN

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.


Asunto(s)
Herpesvirus Humano 4/patogenicidad , Linfoma de Células B/prevención & control , Trastornos Linfoproliferativos/complicaciones , Viroterapia Oncolítica , Trasplante/efectos adversos , Animales , Modelos Animales de Enfermedad , Xenoinjertos , Linfoma de Células B/virología , Trastornos Linfoproliferativos/etiología , Ratones
15.
PLoS Pathog ; 9(7): e1003465, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23853588

RESUMEN

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.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Monocitos/inmunología , Myxoma virus/fisiología , Proteínas de Neoplasias/metabolismo , Proteínas Virales/metabolismo , Tropismo Viral , Replicación Viral , eIF-2 Quinasa/metabolismo , Animales , Antivirales/metabolismo , Antivirales/uso terapéutico , Línea Celular , Células Cultivadas , ARN Helicasas DEAD-box/antagonistas & inhibidores , ARN Helicasas DEAD-box/genética , Susceptibilidad a Enfermedades , Femenino , Técnicas de Inactivación de Genes , Humanos , Interferón Tipo I/metabolismo , Interferón Tipo I/uso terapéutico , Monocitos/metabolismo , Monocitos/virología , Mutación , Mixomatosis Infecciosa/prevención & control , Mixomatosis Infecciosa/virología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Conejos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/genética
16.
Nat Rev Immunol ; 2(7): 521-7, 2002 07.
Artículo en Inglés | MEDLINE | ID: mdl-12094226

RESUMEN

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'.


Asunto(s)
Bioterrorismo , Viruela/historia , Virus de la Viruela , Historia del Siglo XV , Historia del Siglo XVI , Historia del Siglo XVII , Historia del Siglo XVIII , Historia del Siglo XIX , Historia del Siglo XX , Historia del Siglo XXI , Historia Antigua , Historia Medieval , Humanos , Viruela/prevención & control , Vacuna contra Viruela/historia , Vacunación/historia , Organización Mundial de la Salud
17.
Mol Cancer ; 13: 82, 2014 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-24725816

RESUMEN

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.


Asunto(s)
Neoplasias de la Vesícula Biliar/genética , Neoplasias de la Vesícula Biliar/terapia , Viroterapia Oncolítica , Animales , Línea Celular Tumoral , Neoplasias de la Vesícula Biliar/patología , Neoplasias de la Vesícula Biliar/virología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ácido Hialurónico/administración & dosificación , Técnicas In Vitro , Ratones , Myxoma virus/genética , Sirolimus/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Clin Immunol ; 153(2): 254-63, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24845791

RESUMEN

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.


Asunto(s)
Artritis Experimental/prevención & control , Inhibidores de Serina Proteinasa/farmacología , Serpinas/farmacología , Proteínas Virales/farmacología , Secuencia de Aminoácidos , Animales , Anticuerpos/sangre , Anticuerpos/inmunología , Artritis Experimental/diagnóstico por imagen , Artritis Experimental/inmunología , Recuento de Células Sanguíneas , Células CHO , Cricetinae , Cricetulus , Ensayo de Inmunoadsorción Enzimática , Femenino , Hipersensibilidad Tardía/inmunología , Hipersensibilidad Tardía/prevención & control , Immunoblotting , Articulaciones/efectos de los fármacos , Articulaciones/inmunología , Articulaciones/patología , Datos de Secuencia Molecular , Myxoma virus/genética , Myxoma virus/metabolismo , Radiografía , Ratas , Ratas Endogámicas , Inhibidores de Serina Proteinasa/genética , Inhibidores de Serina Proteinasa/inmunología , Serpinas/genética , Serpinas/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología
19.
Immunogenetics ; 66(1): 43-52, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24220721

RESUMEN

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.


Asunto(s)
Evolución Molecular , Liebres/genética , ARN Helicasas/inmunología , Conejos/genética , Receptores Virales/inmunología , Virus/inmunología , Animales , Liebres/inmunología , Liebres/virología , Humanos , Modelos Moleculares , Filogenia , Conformación Proteica , ARN Helicasas/genética , ARN Mensajero/genética , Conejos/clasificación , Conejos/inmunología , Conejos/virología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Virales/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
J Virol ; 87(6): 3018-26, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23269801

RESUMEN

Studies on large double-stranded DNA (dsDNA) viruses such as poxviruses have been helpful in identifying a number of viral and cellular growth factors that contribute to our broad understanding of virus-host interaction. Orthopoxviruses and leporipoxviruses are among the most studied viruses in this aspect. However, tanapoxvirus (TPV), a member of the genus Yatapoxvirus, still remains largely unexplored, as the only known hosts for this virus are humans and monkeys. Here, we describe the initial characterization of an epidermal growth factor (EGF)-like growth factor mimicking human neuregulin from TPV, expressed by the TPV-15L gene. Assays using a baculovirus-expressed and tagged TPV-15L protein demonstrated the ability to phosphorylate neuregulin receptors. Neuregulins represent a large family of EGF-like growth factors that play important roles in embryonic endocardium development, Schwann and oligodendrocyte survival and differentiation, localized acetylcholine receptor expression at the neuromuscular junction, and epithelial morphogenesis. Interestingly, certain neuregulin molecules are able to target specific tissues through interactions with heparin sulfate proteoglycans via an immunoglobulin (Ig)-like domain. Analyses of TPV-15L revealed no Ig-like domain, but it retains the ability to bind heparin and phosphorylate neuregulin receptors, providing compelling evidence that TPV-15L is a functional mimetic of neuregulin. TPV-15L knockout virus experiments demonstrate that the virus replicates in human umbilical vein endothelial cells less efficiently than wild-type TPV-Kenya, indicating that this is a nonessential protein for virus viability but can serve a stimulatory role for replication in some cultured cells. However, the precise role of this protein in host-virus interaction still remains to be deduced.


Asunto(s)
Células Endoteliales/virología , Neurregulinas/metabolismo , Proteínas Virales/metabolismo , Factores de Virulencia/metabolismo , Replicación Viral , Yatapoxvirus/patogenicidad , Secuencia de Aminoácidos , Animales , Línea Celular , Técnicas de Inactivación de Genes , Humanos , Datos de Secuencia Molecular , Neurregulinas/genética , Alineación de Secuencia , Proteínas Virales/genética , Factores de Virulencia/genética , Yatapoxvirus/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA