Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Viruses ; 16(3)2024 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-38543776

RESUMEN

Rotaviruses are a significant cause of severe, potentially life-threatening gastroenteritis in infants and the young of many economically important animals. Although vaccines against porcine rotavirus exist, both live oral and inactivated, their effectiveness in preventing gastroenteritis is less than ideal. Thus, there is a need for the development of new generations of porcine rotavirus vaccines. The Ohio State University (OSU) rotavirus strain represents a Rotavirus A species with a G5P[7] genotype, the genotype most frequently associated with rotavirus disease in piglets. Using complete genome sequences that were determined via Nanopore sequencing, we developed a robust reverse genetics system enabling the recovery of recombinant (r)OSU rotavirus. Although rOSU grew to high titers (~107 plaque-forming units/mL), its growth kinetics were modestly decreased in comparison to the laboratory-adapted OSU virus. The reverse genetics system was used to generate the rOSU rotavirus, which served as an expression vector for a foreign protein. Specifically, by engineering a fused NSP3-2A-UnaG open reading frame into the segment 7 RNA, we produced a genetically stable rOSU virus that expressed the fluorescent UnaG protein as a functional separate product. Together, these findings raise the possibility of producing improved live oral porcine rotavirus vaccines through reverse-genetics-based modification or combination porcine rotavirus vaccines that can express neutralizing antigens for other porcine enteric diseases.


Asunto(s)
Gastroenteritis , Infecciones por Rotavirus , Vacunas contra Rotavirus , Rotavirus , Humanos , Animales , Porcinos , Genética Inversa , Ohio , Universidades , Infecciones por Rotavirus/prevención & control , Infecciones por Rotavirus/veterinaria , Gastroenteritis/prevención & control , Gastroenteritis/veterinaria
2.
PLoS Pathog ; 18(3): e1010398, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35320319

RESUMEN

Successful initiation of infection by many different viruses requires their uptake into the endosomal compartment. While some viruses exit this compartment early, others must reach the degradative, acidic environment of the late endosome. Mammalian orthoreovirus (reovirus) is one such late penetrating virus. To identify host factors that are important for reovirus infection, we performed a CRISPR-Cas9 knockout (KO) screen that targets over 20,000 genes in fibroblasts derived from the embryos of C57/BL6 mice. We identified seven genes (WDR81, WDR91, RAB7, CCZ1, CTSL, GNPTAB, and SLC35A1) that were required for the induction of cell death by reovirus. Notably, CRISPR-mediated KO of WD repeat-containing protein 81 (WDR81) rendered cells resistant to reovirus infection. Susceptibility to reovirus infection was restored by complementing KO cells with human WDR81. Although the absence of WDR81 did not affect viral attachment efficiency or uptake into the endosomal compartments for initial disassembly, it reduced viral gene expression and diminished infectious virus production. Consistent with the role of WDR81 in impacting the maturation of endosomes, WDR81-deficiency led to the accumulation of reovirus particles in dead-end compartments. Though WDR81 was dispensable for infection by VSV (vesicular stomatitis virus), which exits the endosomal system at an early stage, it was required for VSV-EBO GP (VSV that expresses the Ebolavirus glycoprotein), which must reach the late endosome to initiate infection. These results reveal a previously unappreciated role for WDR81 in promoting the replication of viruses that transit through late endosomes.


Asunto(s)
Infecciones por Reoviridae , Reoviridae , Animales , Sistemas CRISPR-Cas , Endosomas/metabolismo , Mamíferos , Ratones , Reoviridae/genética , Infecciones por Reoviridae/metabolismo , Repeticiones WD40
3.
J Virol ; 96(2): e0187921, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-34757847

RESUMEN

Although a broad range of viruses cause myocarditis, the mechanisms that underlie viral myocarditis are poorly understood. Here, we report that the M2 gene is a determinant of reovirus myocarditis. The M2 gene encodes outer capsid protein µ1, which mediates host membrane penetration during reovirus entry. We infected newborn C57BL/6 mice with reovirus strain type 1 Lang (T1L) or a reassortant reovirus in which the M2 gene from strain type 3 Dearing (T3D) was substituted into the T1L genetic background (T1L/T3DM2). T1L was nonlethal in wild-type mice, whereas more than 90% of mice succumbed to T1L/T3DM2 infection. T1L/T3DM2 produced higher viral loads than T1L at the site of inoculation. In secondary organs, T1L/T3DM2 was detected with more rapid kinetics and reached higher peak titers than T1L. We found that hearts from T1L/T3DM2-infected mice were grossly abnormal, with large lesions indicative of substantial inflammatory infiltrate. Lesions in T1L/T3DM2-infected mice contained necrotic cardiomyocytes with pyknotic debris, as well as extensive lymphocyte and histiocyte infiltration. In contrast, T1L induced the formation of small purulent lesions in a small subset of animals, consistent with T1L being mildly myocarditic. Finally, more activated caspase-3-positive cells were observed in hearts from animals infected with T1L/T3DM2 than T1L. Together, our findings indicate that substitution of the T3D M2 allele into an otherwise T1L genetic background is sufficient to change a nonlethal infection into a lethal infection. Our results further indicate that T3D M2 enhances T1L replication and dissemination in vivo, which potentiates the capacity of reovirus to cause myocarditis. IMPORTANCE Reovirus is a nonenveloped virus with a segmented double-stranded RNA genome that serves as a model for studying viral myocarditis. The mechanisms by which reovirus drives myocarditis development are not fully elucidated. We found that substituting the M2 gene from strain type 3 Dearing (T3D) into an otherwise type 1 Lang (T1L) genetic background (T1L/T3DM2) was sufficient to convert the nonlethal T1L strain into a lethal infection in neonatal C57BL/6 mice. T1L/T3DM2 disseminated more efficiently and reached higher maximum titers than T1L in all organs tested, including the heart. T1L is mildly myocarditic and induced small areas of cardiac inflammation in a subset of mice. In contrast, hearts from mice infected with T1L/T3DM2 contained extensive cardiac inflammatory infiltration and more activated caspase-3-positive cells, which is indicative of apoptosis. Together, our findings identify the reovirus M2 gene as a new determinant of reovirus-induced myocarditis.


Asunto(s)
Proteínas de la Cápside/metabolismo , Orthoreovirus Mamífero 3/patogenicidad , Miocarditis/virología , Infecciones por Reoviridae/virología , Animales , Animales Recién Nacidos , Proteínas de la Cápside/genética , Inflamación , Orthoreovirus Mamífero 3/genética , Orthoreovirus Mamífero 3/metabolismo , Ratones , Ratones Endogámicos C57BL , Miocarditis/mortalidad , Miocarditis/patología , Orthoreovirus de los Mamíferos/genética , Orthoreovirus de los Mamíferos/metabolismo , Orthoreovirus de los Mamíferos/patogenicidad , Infecciones por Reoviridae/mortalidad , Infecciones por Reoviridae/patología , Carga Viral , Virulencia , Replicación Viral
4.
Viruses ; 13(2)2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33494426

RESUMEN

Mammalian orthoreovirus (reovirus), a dsRNA virus with a multilayered capsid, serves as a model system for studying the entry of similar viruses. The outermost layer of this capsid undergoes processing to generate a metastable intermediate. The metastable particle undergoes further remodeling to generate an entry-capable form that delivers the genome-containing inner capsid, or core, into the cytoplasm. In this review, we highlight capsid proteins and the intricacies of their interactions that control the stability of the capsid and consequently impact capsid structural changes that are prerequisites for entry. We also discuss a novel proviral role of host membranes in promoting capsid conformational transitions. Current knowledge gaps in the field that are ripe for future investigation are also outlined.


Asunto(s)
Proteínas de la Cápside/metabolismo , Orthoreovirus de los Mamíferos/fisiología , Proteolisis , Infecciones por Reoviridae/virología , Virión/fisiología , Internalización del Virus , Animales , Proteínas de la Cápside/genética , Línea Celular , Ratones , Orthoreovirus de los Mamíferos/genética , Dominios Proteicos , Infecciones por Reoviridae/genética , Virión/genética
5.
J Virol ; 93(9)2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30787157

RESUMEN

The environment represents a significant barrier to infection. Physical stressors (heat) or chemical agents (ethanol) can render virions noninfectious. As such, discrete proteins are necessary to stabilize the dual-layered structure of mammalian orthoreovirus (reovirus). The outer capsid participates in cell entry: (i) σ3 is degraded to generate the infectious subviral particle, and (ii) µ1 facilitates membrane penetration and subsequent core delivery. µ1-σ3 interactions also prevent inactivation; however, this activity is not fully characterized. Using forward and reverse genetic approaches, we identified two mutations (µ1 M258I and σ3 S344P) within heat-resistant strains. σ3 S344P was sufficient to enhance capsid integrity and to reduce protease sensitivity. Moreover, these changes impaired replicative fitness in a reassortant background. This work reveals new details regarding the determinants of reovirus stability.IMPORTANCE Nonenveloped viruses rely on protein-protein interactions to shield their genomes from the environment. The capsid, or protective shell, must also disassemble during cell entry. In this work, we identified a determinant within mammalian orthoreovirus that regulates heat resistance, disassembly kinetics, and replicative fitness. Together, these findings show capsid function is balanced for optimal replication and for spread to a new host.


Asunto(s)
Proteínas de la Cápside , Cápside/metabolismo , Calor , Orthoreovirus Mamífero 3 , Mutación , Animales , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular , Orthoreovirus Mamífero 3/genética , Orthoreovirus Mamífero 3/metabolismo , Ratones , Estabilidad Proteica
6.
J Virol ; 93(2)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30381491

RESUMEN

The mammalian orthoreovirus (reovirus) outer capsid is composed of 200 µ1-σ3 heterohexamers and a maximum of 12 σ1 trimers. During cell entry, σ3 is degraded by luminal or intracellular proteases to generate the infectious subviral particle (ISVP). When ISVP formation is prevented, reovirus fails to establish a productive infection, suggesting proteolytic priming is required for entry. ISVPs are then converted to ISVP*s, which is accompanied by µ1 rearrangements. The µ1 and σ3 proteins confer resistance to inactivating agents; however, neither the impact on capsid properties nor the mechanism (or basis) of inactivation is fully understood. Here, we utilized T1L/T3D M2 and T3D/T1L S4 to investigate the determinants of reovirus stability. Both reassortants encode mismatched subunits. When µ1-σ3 were derived from different strains, virions resembled wild-type particles in structure and protease sensitivity. T1L/T3D M2 and T3D/T1L S4 ISVPs were less thermostable than wild-type ISVPs. In contrast, virions were equally susceptible to heating. Virion associated µ1 adopted an ISVP*-like conformation concurrent with inactivation; σ3 preserves infectivity by preventing µ1 rearrangements. Moreover, thermostability was enhanced by a hyperstable variant of µ1. Unlike the outer capsid, the inner capsid (core) was highly resistant to elevated temperatures. The dual layered architecture allowed for differential sensitivity to inactivating agents.IMPORTANCE Nonenveloped and enveloped viruses are exposed to the environment during transmission to a new host. Protein-protein and/or protein-lipid interactions stabilize the particle and protect the viral genome. Mammalian orthoreovirus (reovirus) is composed of two concentric, protein shells. The µ1 and σ3 proteins form the outer capsid; contacts between neighboring subunits are thought to confer resistance to inactivating agents. We further investigated the determinants of reovirus stability. The outer capsid was disrupted concurrent with the loss of infectivity; virion associated µ1 rearranged into an altered conformation. Heat sensitivity was controlled by σ3; however, particle integrity was enhanced by a single µ1 mutation. In contrast, the inner capsid (core) displayed superior resistance to heating. These findings reveal structural components that differentially contribute to reovirus stability.


Asunto(s)
Proteínas de la Cápside/química , Cápside/metabolismo , Reoviridae/fisiología , Animales , Cápside/química , Línea Celular , Microscopía por Crioelectrón , Ratones , Modelos Moleculares , Conformación Proteica , Estabilidad Proteica , Reoviridae/metabolismo , Termodinámica , Internalización del Virus
7.
Bio Protoc ; 8(2)2018 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-29552594

RESUMEN

Mammalian orthoreovirus (reovirus) utilizes pore forming peptides to penetrate host cell membranes. This step is essential for delivering its genome containing core particle during viral entry. This protocol describes an in vitro assay for measuring reovirus-induced pore formation.

8.
Bio Protoc ; 8(2)2018 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-29399600

RESUMEN

The mammalian orthoreovirus (reovirus) outer capsid undergoes a series of conformational changes prior to or during viral entry. These transitions are necessary for delivering the genome-containing core across host cell membranes. This protocol describes an in vitro assay for monitoring the transition into a membrane penetration-active form (i.e., ISVP*).

9.
J Virol ; 92(6)2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29298891

RESUMEN

The mammalian orthoreovirus (reovirus) outer capsid, which is composed of 200 µ1/σ3 heterohexamers and a maximum of 12 σ1 trimers, contains all of the proteins that are necessary for attaching to and entering host cells. Following attachment, reovirus is internalized by receptor-mediated endocytosis and acid-dependent cathepsin proteases degrade the σ3 protein. This process generates a metastable intermediate, called infectious subviral particle (ISVP), in which the µ1 membrane penetration protein is exposed. ISVPs undergo a second structural rearrangement to deposit the genome-containing core into the host cytoplasm. The conformationally altered particle is called ISVP*. ISVP-to-ISVP* conversion culminates in the release of µ1 N- and C-terminal fragments, µ1N and Φ, respectively. Released µ1N is thought to facilitate core delivery by generating size-selective pores within the endosomal membrane, whereas the precise role of Φ, particularly in the context of viral entry, is undefined. In this report, we characterize a recombinant reovirus that fails to cleave Φ from µ1 in vitro Φ cleavage, which is not required for ISVP-to-ISVP* conversion, enhances the disruption of liposomal membranes and facilitates the recruitment of ISVP*s to the site of pore formation. Moreover, the Φ cleavage-deficient strain initiates infection of host cells less efficiently than the parental strain. These results indicate that µ1N and Φ contribute to reovirus pore forming activity.IMPORTANCE Host membranes represent a physical barrier that prevents infection. To overcome this barrier, viruses utilize diverse strategies, such as membrane fusion or membrane disruption, to access internal components of the cell. These strategies are characterized by discrete protein-protein and protein-lipid interactions. The mammalian orthoreovirus (reovirus) outer capsid undergoes a series of well-defined conformational changes, which conclude with pore formation and delivery of the viral genetic material. In this report, we characterize the role of the small, reovirus-derived Φ peptide in pore formation. Φ cleavage from the outer capsid enhances membrane disruption and facilitates the recruitment of virions to membrane-associated pores. Moreover, Φ cleavage promotes the initiation of infection. Together, these results reveal an additional component of the reovirus pore forming apparatus and highlight a strategy for penetrating host membranes.


Asunto(s)
Proteínas de la Cápside/metabolismo , Orthoreovirus de los Mamíferos/metabolismo , Proteolisis , Infecciones por Reoviridae/metabolismo , Virión/metabolismo , Animales , Proteínas de la Cápside/genética , Línea Celular , Ratones , Orthoreovirus de los Mamíferos/genética , Dominios Proteicos , Infecciones por Reoviridae/genética , Infecciones por Reoviridae/patología , Virión/genética
10.
J Virol ; 91(20)2017 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-28794028

RESUMEN

Reovirus particles are covered with 200 µ1/σ3 heterohexamers. Following attachment to cell surface receptors, reovirus is internalized by receptor-mediated endocytosis. Within the endosome, particles undergo a series of stepwise disassembly events. First, the σ3 protector protein is degraded by cellular proteases to generate infectious subviral particles (ISVPs). Second, the µ1 protein rearranges into a protease-sensitive conformation to generate ISVP*s and releases two virus-encoded peptides, µ1N and Φ. The released peptides promote delivery of the genome-containing core by perforating the endosomal membrane. Thus, to establish a productive infection, virions must be stable in the environment but flexible to disassemble in response to the appropriate cellular cue. The reovirus outer capsid is stabilized by µ1 intratrimer, intertrimer, and trimer-core interactions. As a consequence of ISVP-to-ISVP* conversion, neighboring µ1 trimers unwind and separate. Located within the µ1 jelly roll ß barrel domain, which is a known regulator of ISVP* formation, residues 340 to 343 form a loop and have been proposed to facilitate viral entry. To test this idea, we generated recombinant reoviruses that encoded deletions within this loop (Δ341 and Δ342). Both deletions destabilized the outer capsid. Notably, Δ342 impaired the viral life cycle; however, replicative fitness was restored by an additional change (V403A) within the µ1 jelly roll ß barrel domain. In the Δ341 and Δ342 backgrounds, V403A also rescued defects in ISVP-to-ISVP* conversion. Together, these findings reveal a new region that regulates reovirus disassembly and how perturbing a metastable capsid can compromise replicative fitness.IMPORTANCE Capsids of nonenveloped viruses are composed of protein complexes that encapsulate, or form a shell around, nucleic acid. The protein-protein interactions that form this shell must be stable to protect the viral genome but also sufficiently flexible to disassemble during cell entry. Thus, capsids adopt conformations that undergo rapid disassembly in response to a specific cellular cue. In this work, we identify a new region within the mammalian orthoreovirus outer capsid that regulates particle stability. Amino acid deletions that destabilize this region impair the viral replication cycle. Nonetheless, replicative fitness is restored by a compensatory mutation that restores particle stability. Together, this work demonstrates the critical balance between assembling virions that are stable and maintaining conformational flexibility. Any factor that perturbs this balance has the potential to block a productive infection.

11.
J Virol ; 91(6)2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28077640

RESUMEN

Necroptosis, a regulated form of necrotic cell death, requires the activation of the RIP3 kinase. Here, we identify that infection of host cells with reovirus can result in necroptosis. We find that necroptosis requires sensing of the genomic RNA within incoming virus particles via cytoplasmic RNA sensors to produce type I interferon (IFN). While these events that occur prior to the de novo synthesis of viral RNA are required for the induction of necroptosis, they are not sufficient. The induction of necroptosis also requires late stages of reovirus infection. Specifically, efficient synthesis of double-stranded RNA (dsRNA) within infected cells is required for necroptosis. These data indicate that viral RNA interfaces with host components at two different stages of infection to induce necroptosis. This work provides new molecular details about events in the viral replication cycle that contribute to the induction of necroptosis following infection with an RNA virus.IMPORTANCE An appreciation of how cell death pathways are regulated following viral infection may reveal strategies to limit tissue destruction and prevent the onset of disease. Cell death following virus infection can occur by apoptosis or a regulated form of necrosis known as necroptosis. Apoptotic cells are typically disposed of without activating the immune system. In contrast, necroptotic cells alert the immune system, resulting in inflammation and tissue damage. While apoptosis following virus infection has been extensively investigated, how necroptosis is unleashed following virus infection is understood for only a small group of viruses. Here, using mammalian reovirus, we highlight the molecular mechanism by which infection with a dsRNA virus results in necroptosis.


Asunto(s)
Muerte Celular , Interacciones Huésped-Patógeno , Inmunidad Innata , ARN Viral/metabolismo , Reoviridae/inmunología , Reoviridae/fisiología , Animales , Línea Celular , Fibroblastos/inmunología , Fibroblastos/fisiología , Fibroblastos/virología , Ratones
12.
J Biol Chem ; 291(52): 26773-26785, 2016 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-27875299

RESUMEN

Virus-host interactions play a role in many stages of the viral lifecycle, including entry. Reovirus, a model system for studying the entry mechanisms of nonenveloped viruses, undergoes a series of regulated structural transitions that culminate in delivery of the viral genetic material. Lipids can trigger one of these conformational changes, infectious subviral particle (ISVP)-to-ISVP* conversion. ISVP* formation releases two virally encoded peptides, myristoylated µ1N (myr-µ1N) and Φ. Among these, myr-µ1N is sufficient to form pores within membranes. Released myr-µ1N can also promote ISVP* formation in trans Using thermal inactivation as a readout for ISVP-to-ISVP* conversion, we demonstrate that lipids render ISVPs less thermostable in a virus concentration-dependent manner. Under conditions in which neither lipids alone nor myr-µ1N alone promotes ISVP-to-ISVP* conversion, myr-µ1N induces particle uncoating when lipids are present. These data suggest that the pore-forming activity and the ISVP*-promoting activity of myr-µ1N are linked. Lipid-associated myr-µ1N interacts with ISVPs and triggers efficient ISVP* formation. The cooperativity between a reovirus component and lipids reveals a distinct virus-host interaction in which membranes can facilitate nonenveloped virus entry.


Asunto(s)
Proteínas de la Cápside/metabolismo , Membrana Celular/metabolismo , Péptidos de Penetración Celular/metabolismo , Lípidos de la Membrana/metabolismo , Infecciones por Reoviridae/metabolismo , Reoviridae/fisiología , Virión/fisiología , Secuencia de Aminoácidos , Animales , Proteínas de la Cápside/química , Membrana Celular/química , Membrana Celular/virología , Permeabilidad de la Membrana Celular , Células Cultivadas , Liposomas/química , Ratones , Modelos Biológicos , Ácidos Mirísticos/metabolismo , Conformación Proteica , Procesamiento Proteico-Postraduccional , Infecciones por Reoviridae/virología , Homología de Secuencia de Aminoácido , Internalización del Virus
13.
J Virol ; 90(23): 10951-10962, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27681135

RESUMEN

Proteins that form the reovirus outer capsid play an active role in the entry of reovirus into host cells. Among these, the σ1 protein mediates attachment of reovirus particles to host cells via interaction with cell surface glycans or the proteinaceous receptor junctional adhesion molecule A (JAM-A). The µ1 protein functions to penetrate the host cell membrane to allow delivery of the genome-containing viral core particle into the cytoplasm to initiate viral replication. We demonstrate that a reassortant virus that expresses the M2 gene-encoded µ1 protein derived from prototype strain T3D in an otherwise prototype T1L background (T1L/T3DM2) infects cells more efficiently than parental T1L. Unexpectedly, the enhancement in infectivity of T1L/T3DM2 is due to its capacity to attach to cells more efficiently. We present genetic data implicating the central region of µ1 in altering the cell attachment property of reovirus. Our data indicate that the T3D µ1-mediated enhancement in infectivity of T1L is dependent on the function of σ1 and requires the expression of JAM-A. We also demonstrate that T1L/T3DM2 utilizes JAM-A more efficiently than T1L. These studies revealed a previously unknown relationship between two nonadjacent reovirus outer capsid proteins, σ1 and µ1. IMPORTANCE: How reovirus attaches to host cells has been extensively characterized. Attachment of reovirus to host cells is mediated by the σ1 protein, and properties of σ1 influence the capacity of reovirus to target specific host tissues and produce disease. Here, we present new evidence indicating that the cell attachment properties of σ1 are influenced by the nature of µ1, a capsid protein that does not physically interact with σ1. These studies could explain the previously described role for µ1 in influencing reovirus pathogenesis. These studies are also of broader significance because they highlight an example of how genetic reassortment between virus strains could produce phenotypes that are distinct from those of either parent.


Asunto(s)
Proteínas de la Cápside/fisiología , Orthoreovirus Mamífero 3/fisiología , Orthoreovirus Mamífero 3/patogenicidad , Animales , Proteínas de la Cápside/genética , Moléculas de Adhesión Celular/fisiología , Línea Celular , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/fisiología , Orthoreovirus Mamífero 3/genética , Ratones , Orthoreovirus de los Mamíferos/genética , Orthoreovirus de los Mamíferos/patogenicidad , Orthoreovirus de los Mamíferos/fisiología , Receptores de Superficie Celular/fisiología , Receptores Virales/fisiología , Infecciones por Reoviridae/etiología , Infecciones por Reoviridae/virología , Virulencia/genética , Virulencia/fisiología , Acoplamiento Viral
14.
J Virol ; 90(5): 2628-38, 2015 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-26699639

RESUMEN

UNLABELLED: Cellular entry of nonenveloped and enveloped viruses is often accompanied by dramatic conformational changes within viral structural proteins. These rearrangements are triggered by a variety of mechanisms, such as low pH, virus-receptor interactions, and virus-host chaperone interactions. Reoviruses, a model system for entry of nonenveloped viruses, undergo a series of disassembly steps within the host endosome. One of these steps, infectious subviral particle (ISVP)-to-ISVP* conversion, is necessary for delivering the genome-containing viral core into host cells, but the physiological trigger that mediates ISVP-to-ISVP* conversion during cell entry is unknown. Structural studies of the reovirus membrane penetration protein, µ1, predict that interactions between µ1 and negatively charged lipid head groups may promote ISVP* formation; however, experimental evidence for this idea is lacking. Here, we show that the presence of polyanions (SO4(2-) and HPO4(2-)) or lipids in the form of liposomes facilitates ISVP-to-ISVP* conversion. The requirement for charged lipids appears to be selective, since phosphatidylcholine and phosphatidylethanolamine promoted ISVP* formation, whereas other lipids, such as sphingomyelin and sulfatide, either did not affect ISVP* formation or prevented ISVP* formation. Thus, our work provides evidence that interactions with membranes can function as a trigger for a nonenveloped virus to gain entry into host cells. IMPORTANCE: Cell entry, a critical stage in the virus life cycle, concludes with the delivery of the viral genetic material across host membranes. Regulated structural transitions within nonenveloped and enveloped viruses are necessary for accomplishing this step; these conformational changes are predominantly triggered by low pH and/or interactions with host proteins. In this work, we describe a previously unknown trigger, interactions with lipid membranes, which can induce the structural rearrangements required for cell entry. This mechanism operates during entry of mammalian orthoreoviruses. We show that interactions between reovirus entry intermediates and lipid membranes devoid of host proteins promote conformational changes within the viral outer capsid that lead to membrane penetration. Thus, this work illustrates a novel strategy that nonenveloped viruses can use to gain access into cells and how viruses usurp disparate host factors to initiate infection.


Asunto(s)
Proteínas de la Cápside/metabolismo , Lípidos de la Membrana/metabolismo , Fosfolípidos/metabolismo , Reoviridae/fisiología , Internalización del Virus , Animales , Proteínas de la Cápside/química , Línea Celular , Liposomas/metabolismo , Ratones , Modelos Moleculares , Conformación Proteica/efectos de los fármacos , Reoviridae/química
15.
Appl Environ Microbiol ; 80(10): 3219-32, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24632256

RESUMEN

Cellular processes, such as the digestion of macromolecules, phosphate acquisition, and cell motility, require bacterial secretion systems. In Bacillus subtilis, the predominant protein export pathways are Sec (generalized secretory pathway) and Tat (twin-arginine translocase). Unlike Sec, which secretes unfolded proteins, the Tat machinery secretes fully folded proteins across the plasma membrane and into the medium. Proteins are directed for Tat-dependent export by N-terminal signal peptides that contain a conserved twin-arginine motif. Thus, utilizing the Tat secretion system by fusing a Tat signal peptide is an attractive strategy for the production and export of heterologous proteins. As a proof of concept, we expressed green fluorescent protein (GFP) fused to the PhoD Tat signal peptide in the laboratory and ancestral strains of B. subtilis. Secretion of the Tat-GFP construct, as well as secretion of proteins in general, was substantially increased in the ancestral strain. Furthermore, our results show that secreted, fluorescent GFP could be purified directly from the extracellular medium. Nonetheless, export was not dependent on the known Tat secretion components or the signal peptide twin-arginine motif. We propose that the ancestral strain contains additional Tat components and/or secretion regulators that were abrogated following domestication.


Asunto(s)
Bacillus subtilis/enzimología , Proteínas Bacterianas/metabolismo , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/metabolismo , Transferasas/metabolismo , Arginina/metabolismo , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sistemas de Secreción Bacterianos , Proteínas Fluorescentes Verdes/genética , Pliegue de Proteína , Señales de Clasificación de Proteína , Transporte de Proteínas , Transferasas/química , Transferasas/genética
16.
J Virol ; 87(22): 12216-26, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24006438

RESUMEN

The genus Alphavirus consists of a group of enveloped, single-stranded RNA viruses, many of which are transmitted by arthropods to a wide range of vertebrate host species. Here we report that Sindbis virus (SINV) produced from a representative mammalian cell line consists of at least two unique particle subpopulations, separable on the basis of virion density. In contrast, mosquito-derived SINV consists of a homogeneous population of particles. Our findings indicate that the denser particle subpopulation, SINV(Heavy), is more infectious on a per-particle basis than SINV(Light). SINV produced in mosquito cell lines (SINV(C6/36)) exhibited particle-to-PFU ratios similar to those observed for SINV(Heavy). In mammalian cells, viral RNA was synthesized and accumulated more rapidly following infection with SINV(Heavy) or SINV(C6/36) than following infection with SINV(Light), due partly to enhanced translation of viral genomic RNA early in infection. Analysis of the individual particle subpopulations indicated that SINV(Heavy) and SINV(C6/36) contain host-derived factors whose presence correlates with the enhanced translation, RNA synthesis, and infectivity observed for these particles.


Asunto(s)
Infecciones por Alphavirus/transmisión , Culicidae/virología , Fibroblastos/virología , Interacciones Huésped-Patógeno , Riñón/virología , Virus Sindbis/patogenicidad , Infecciones por Alphavirus/virología , Animales , Células Cultivadas , Cricetinae , Reactivos de Enlaces Cruzados , Fibroblastos/patología , Células HEK293 , Humanos , Inmunoprecipitación , Riñón/patología , Ratones , Reacción en Cadena de la Polimerasa , ARN Viral/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Internalización del Virus , Replicación Viral
17.
J Virol ; 86(24): 13609-20, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23035234

RESUMEN

The 80 trimeric, glycoprotein spikes that cover the surface of alphavirus particles are required for mediating viral entry into a host cell. Spike assembly is a regulated process that requires interactions between five structural proteins, E3, E2, 6K and its translational frameshift product TF, and E1. E3 is a small, ∼65-amino-acid glycoprotein that has two known functions: E3 serves as the signal sequence for translocation of the E3-E2-6K-E1 polyprotein into the endoplasmic reticulum (ER), and cleavage of E3 from E2 is essential for virus maturation. Nonetheless, when E3 is replaced with an ER signal sequence, spikes do not form and infectious particles are not assembled, suggesting an additional role(s) for E3 in the viral life cycle. To further investigate the role of E3 in spike assembly, we made chimeric viruses in which E3 from one alphavirus species is replaced with E3 from another species. Our results demonstrate that when E3 is interchanged between alphavirus species that belong to the same virus clade, viral titers and particle morphologies and compositions are similar to what are observed for the parental virus. In contrast, for chimeras in which E3 is derived from a different clade than the parental virus, we observed reduced titers and the formation of particles with atypical morphologies and protein compositions. We further characterized the E3 chimeras using a combination of structure-function and revertant analyses. This work revealed two specific interactions between E3 and its cognate E2 glycoprotein that are important for regulating spike assembly.


Asunto(s)
Alphavirus/fisiología , Glicoproteínas/fisiología , Proteínas Virales/fisiología , Alphavirus/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Cricetinae , Cartilla de ADN , Glicoproteínas/química , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Proteínas Virales/química
18.
J Virol ; 86(6): 3100-11, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22238319

RESUMEN

There are 80 trimeric, glycoprotein spikes that cover the surface of an alphavirus particle. The spikes, which are composed of three E2 and E1 glycoprotein heterodimers, are responsible for receptor binding and mediating fusion between the viral and host-cell membranes during entry. In addition, the cytoplasmic domain of E2 interacts with the nucleocapsid core during the last stages of particle assembly, possibly to aid in particle stability. During assembly, the spikes are nonfusogenic until the E3 glycoprotein is cleaved from E2 in the trans-Golgi network. Thus, a mutation in E2 potentially has effects on virus entry, spike assembly, or spike maturation. E2 is a highly conserved, cysteine-rich transmembrane glycoprotein. We made single cysteine-to-serine mutations within two distinct regions of the E2 ectodomain in both Sindbis virus and Ross River virus. Each of the E2 Cys mutants produced fewer infectious particles than wild-type virus. Further characterization of the mutant viruses revealed differences in particle morphology, fusion activity, and polyprotein cleavage between Sindbis and Ross River virus mutants, despite the mutations being made at corresponding positions in E2. The nonconserved assembly defects suggest that E2 folding and function is species dependent, possibly due to interactions with a virus-specific chaperone.


Asunto(s)
Infecciones por Alphavirus/virología , Virus Chikungunya/fisiología , Mutación , Virus Sindbis/fisiología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Ensamble de Virus , Aedes , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Línea Celular , Virus Chikungunya/química , Virus Chikungunya/genética , Secuencia Conservada , Cricetinae , Cisteína/genética , Cisteína/metabolismo , Humanos , Datos de Secuencia Molecular , Alineación de Secuencia , Virus Sindbis/química , Virus Sindbis/genética , Proteínas del Envoltorio Viral/metabolismo
19.
Cancer ; 116(17): 4043-53, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20564122

RESUMEN

BACKGROUND: Aberrant DNA methylation is a hallmark of cancer, and DNA methyltransferase inhibitors have demonstrated clinical efficacy in hematologic malignancies. On the basis of preclinical studies indicating that hypomethylating agents can reverse platinum resistance in ovarian cancer cells, the authors conducted a phase 1 trial of low-dose decitabine combined with carboplatin in patients with recurrent, platinum-resistant ovarian cancer. METHODS: Decitabine was administered intravenously daily for 5 days, before carboplatin (area under the curve, 5) on Day 8 of a 28-day cycle. By using a standard 3 + 3 dose escalation, decitabine was tested at 2 dose levels: 10 mg/m(2) (7 patients) or 20 mg/m(2) (3 patients). Peripheral blood mononuclear cells (PBMCs) and plasma collected on Days 1 (pretreatment), 5, 8, and 15 were used to assess global (LINE-1 repetitive element) and gene-specific DNA methylation. RESULTS: Dose-limiting toxicity (DLT) at the 20-mg/m(2) dose was grade 4 neutropenia (2 patients), and no DLTs were observed at 10 mg/m(2). The most common toxicities were nausea, allergic reactions, neutropenia, fatigue, anorexia, vomiting, and abdominal pain, the majority being grades 1-2. One complete response was observed, and 3 additional patients had stable disease for >/=6 months. LINE-1 hypomethylation on Days 8 and 15 was detected in DNA from PBMCs. Of 5 ovarian cancer-associated methylated genes, HOXA11 and BRCA1 were demethylated in plasma on Days 8 and 15. CONCLUSIONS: Repetitive low-dose decitabine is tolerated when combined with carboplatin in ovarian cancer patients, and demonstrates biological (ie, DNA-hypomethylating) activity, justifying further testing for clinical efficacy. Cancer 2010. (c) 2010 American Cancer Society.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Azacitidina/análogos & derivados , Carboplatino/administración & dosificación , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Compuestos de Platino/uso terapéutico , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Azacitidina/administración & dosificación , Azacitidina/farmacología , Metilación de ADN , Decitabina , Esquema de Medicación , Resistencia a Antineoplásicos , Femenino , Humanos , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...