Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 300
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(4): e2305745121, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38236731

RESUMEN

The development of vaccines, which induce effective immune responses while ensuring safety and affordability, remains a substantial challenge. In this study, we proposed a vaccine model of a restructured "head-to-tail" dimer to efficiently stimulate B cell response. We also demonstrate the feasibility of using this model to develop a paramyxovirus vaccine through a low-cost rice endosperm expression system. Crystal structure and small-angle X-ray scattering data showed that the restructured hemagglutinin-neuraminidase (HN) formed tetramers with fully exposed quadruple receptor binding domains and neutralizing epitopes. In comparison with the original HN antigen and three traditional commercial whole virus vaccines, the restructured HN facilitated critical epitope exposure and initiated a faster and more potent immune response. Two-dose immunization with 0.5 µg of the restructured antigen (equivalent to one-127th of a rice grain) and one-dose with 5 µg completely protected chickens against a lethal challenge of the virus. These results demonstrate that the restructured HN from transgenic rice seeds is safe, effective, low-dose useful, and inexpensive. We provide a plant platform and a simple restructured model for highly effective vaccine development.


Asunto(s)
Oryza , Paramyxovirinae , Vacunas Virales , Animales , Pollos , Virus de la Enfermedad de Newcastle , Oryza/genética , Diseño Universal , Epítopos , Anticuerpos Antivirales
2.
J Virol ; 98(1): e0165423, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38169290

RESUMEN

Jeilongviruses are emerging single-stranded negative-sense RNA viruses in the Paramyxoviridae family. Tailam paramyxovirus (TlmPV) is a Jeilongvirus that was identified in 2011. Very little is known about the mechanisms that regulate viral replication in these newly emerging viruses. Among the non-structural viral proteins of TlmPV, the C protein is predicted to be translated from an open reading frame within the phosphoprotein gene through alternative translation initiation. Though the regulatory roles of C proteins in virus replication of other paramyxoviruses have been reported before, the function of the TlmPV C protein and the relevant molecular mechanisms have not been reported. Here, we show that the C protein is expressed in TlmPV-infected cells and negatively modulates viral RNA replication. The TlmPV C protein interacts with the P protein, negatively impacting the interaction between N and P, resulting in inhibition of viral RNA replication. Deletion mutagenesis studies indicate that the 50 amino-terminal amino acid residues of the C protein are dispensable for its inhibition of virus RNA replication and interaction with the P protein.IMPORTANCETailam paramyxovirus (TlmPV) is a newly identified paramyxovirus belonging to the Jeilongvirus genus, of which little is known. In this work, we confirmed the expression of the C protein in TlmPV-infected cells, assessed its function, and defined a potential mechanism of action. This is the first time that the existence of a Jeilongvirus C protein has been confirmed and its role in viral replication has been reported.


Asunto(s)
Paramyxovirinae , Proteínas Virales , Replicación Viral , Paramyxovirinae/genética , Paramyxovirinae/fisiología , ARN Viral/genética , Proteínas Virales/genética , Animales , Cricetinae , Línea Celular
3.
J Virol ; 97(1): e0180222, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36521070

RESUMEN

Tailam paramyxovirus (TlmPV) was identified in Sikkim Rats in Hong Kong, China in 2011. Its negative sense RNA genome is similar to J paramyxovirus (JPV) and Beilong paramyxovirus (BeiPV), the prototypes of the recently established genus Jeilongvirus. TlmPV genome is predicted to have eight genes in the order 3'-N-P/V/C-M-F-SH-TM-G/X-L-5'. The predicted size of the TlmPV G protein is 1,052 amino acid (aa) residues and much larger than G proteins of typical paramyxoviruses, which are often less than 800 aa. In addition to G open reading frame (ORF) in the G gene, another ORF, termed ORF-X exists in the G gene transcript. Similar ORF-X exists in JPV and BeiPV G gene, but their expression in virus-infected cells has not been confirmed. In this study, we generated infectious TlmPV using a newly developed reverse genetics system. We have found that the G protein of TlmPV is truncated in cultured cells: stop codons emerged in the G open reading frame, resulting in deletions of amino acid residues beyond residue 732. We have obtained infectious TlmPV lacking the C-terminal 307 aa (rTlmPV-G745) and TlmPV lacking the C-terminal 306 aa and the ORF-X (rTlmPV-GΔ746-X). The recombinant TlmPVs lacking the C-terminal 300 aa reach a higher peak viral titer and have improved genome stability in tissue cultured cells. The work indicates that the C-terminal of the G protein of TlmPV and ORF-X are not required for replication in tissue culture cells, and the deletion of the C-terminal confers a growth advantage in tissue culture cells. IMPORTANCE TlmPV is a member of the recently established genus Jeilongvirus. TlmPV encodes a large G protein and its G gene contains ORF-X. In this work, infectious TlmPV was recovered using reverse genetics. Using this system, we have demonstrated that 300 aa of C-terminal of G and the ORF-X are not required for viral replication in tissue culture cells.


Asunto(s)
Proteínas de Unión al GTP , Sistemas de Lectura Abierta , Paramyxovirinae , Replicación Viral , Animales , Ratas , Células Cultivadas , Proteínas de Unión al GTP/genética , Paramyxovirinae/genética , Paramyxovirinae/fisiología
4.
Virol J ; 21(1): 146, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918816

RESUMEN

The genus Jeilongvirus comprises non-segmented negative-stranded RNA viruses that are classified within the Paramyxoviridae family by phylogeny. Jeilongviruses are found in various reservoirs, including rodents and bats. Rodents are typical viral reservoirs with diverse spectra and zoonotic potential. Little is currently known about jeilongviruses in rodents from central China. The study utilized high-throughput and Sanger sequencing to obtain jeilongvirus genomes, including those of two novel strains (HBJZ120/CHN/2021 (17,468 nt) and HBJZ157/CHN/2021 (19,143 nt)) and three known viruses (HBXN18/CHN/2021 (19,212 nt), HBJZ10/CHN/2021 (19,700 nt), HBJM106/CHN/2021 (18,871 nt)), which were characterized by genome structure, identity matrix, and phylogenetic analysis. Jeilongviruses were classified into three subclades based on their topology, phylogeny, and hosts. Based on the amino acid sequence identities and phylogenetic analysis of the L protein, HBJZ120/CHN/2021 and HBJZ157/CHN/2021 were found to be strains rather than novel species. Additionally, according to specific polymerase chain reaction screening, the positive percentage of Beilong virus in Hubei was 6.38%, suggesting that Beilong virus, belonging to the Jeilongvirus genus, is likely to be widespread in wild rodents. The identification of novel strains further elucidated the genomic diversity of jeilongviruses. Additionally, the prevalence of jeilongviruses in Hubei, China, was profiled, establishing a foundation for the surveillance and early warning of emerging paramyxoviruses.


Asunto(s)
Genoma Viral , Filogenia , Roedores , Animales , China , Roedores/virología , Animales Salvajes/virología , Paramyxovirinae/genética , Paramyxovirinae/clasificación , Paramyxovirinae/aislamiento & purificación , ARN Viral/genética , Infecciones por Paramyxoviridae/veterinaria , Infecciones por Paramyxoviridae/virología , Infecciones por Paramyxoviridae/epidemiología , Secuenciación de Nucleótidos de Alto Rendimiento , Reservorios de Enfermedades/virología , Análisis de Secuencia de ADN
5.
J Gen Virol ; 104(11)2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37962188

RESUMEN

Parainfluenza virus type 5 (PIV5) can either have a persistent or a lytic phenotype in cultured cells. We have previously shown that the phenotype is determined by the phosphorylation status of the phosphoprotein (P). Single amino acid substitutions at critical residues, including a serine-to-phenylalanine substitution at position 157 on P, result in a switch between persistent and lytic phenotypes. Here, using PIV5 vectors expressing either mCherry or GFP with persistent or lytic phenotypes, we show that in co-infections the persistent phenotype is dominant. Thus, in contrast to the cell death observed with cells infected solely with the lytic variant, in co-infected cells persistence is immediately established and both lytic and persistent genotypes persist. Furthermore, 10-20 % of virus released from dually infected cells contains both genotypes, indicating that PIV5 particles can package more than one genome. Co-infected cells continue to maintain both genotypes/phenotypes during cell passage, as do individual colonies of cells derived from a culture of persistently infected cells. A refinement of our model on how the dynamics of virus selection may occur in vivo is presented.


Asunto(s)
Coinfección , Virus de la Parainfluenza 5 , Paramyxovirinae , Infecciones por Respirovirus , Humanos , Virus de la Parainfluenza 5/genética , Fenotipo
6.
Proc Natl Acad Sci U S A ; 117(9): 4931-4941, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32075920

RESUMEN

Paramyxoviruses are enveloped, nonsegmented, negative-strand RNA viruses that cause a wide spectrum of human and animal diseases. The viral genome, packaged by the nucleoprotein (N), serves as a template for the polymerase complex, composed of the large protein (L) and the homo-tetrameric phosphoprotein (P). The ∼250-kDa L possesses all enzymatic activities necessary for its function but requires P in vivo. Structural information is available for individual P domains from different paramyxoviruses, but how P interacts with L and how that affects the activity of L is largely unknown due to the lack of high-resolution structures of this complex in this viral family. In this study we determined the structure of the L-P complex from parainfluenza virus 5 (PIV5) at 4.3-Šresolution using cryoelectron microscopy, as well as the oligomerization domain (OD) of P at 1.4-Šresolution using X-ray crystallography. P-OD associates with the RNA-dependent RNA polymerase domain of L and protrudes away from it, while the X domain of one chain of P is bound near the L nucleotide entry site. The methyltransferase (MTase) domain and the C-terminal domain (CTD) of L adopt a unique conformation, positioning the MTase active site immediately above the poly-ribonucleotidyltransferase domain and near the likely exit site for the product RNA 5' end. Our study reveals a potential mechanism that mononegavirus polymerases may employ to switch between transcription and genome replication. This knowledge will assist in the design and development of antivirals against paramyxoviruses.


Asunto(s)
Metiltransferasas/química , Metiltransferasas/metabolismo , Paramyxovirinae/enzimología , Proteínas Virales/química , Proteínas Virales/metabolismo , Dominio Catalítico , Microscopía por Crioelectrón , Cristalografía por Rayos X , Genoma Viral , Metiltransferasas/genética , Modelos Moleculares , Nucleoproteínas/química , Virus de la Parainfluenza 5/química , Paramyxovirinae/genética , Fosfoproteínas/química , Unión Proteica , Conformación Proteica , Dominios Proteicos
7.
J Virol ; 95(22): e0132121, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34469242

RESUMEN

H5N1, an avian influenza virus, is known to circulate in many Asian countries, such as Bangladesh, China, Cambodia, Indonesia, and Vietnam. The current FDA-approved H5N1 vaccine has a moderate level of efficacy. A safe and effective vaccine is needed to prevent outbreaks of highly pathogenic avian influenza (HPAI) H5N1 in humans. Nonsegmented negative-sense single-stranded viruses (NNSVs) are widely used as a vector to develop vaccines for humans, animals, and poultry. NNSVs stably express foreign genes without integrating with the host genome. J paramyxovirus (JPV) is a nonsegmented negative-strand RNA virus and a member of the proposed genus Jeilongvirus in the family Paramyxoviridae. JPV-specific antibodies have been detected in rodents, bats, humans, and pigs, but the virus is not associated with disease in any species other than mice. JPV replicates in the respiratory tract of mice and efficiently expresses the virus-vectored foreign genes in tissue culture cells. In this work, we explored JPV as a vector for developing an H5N1 vaccine using intranasal delivery. We incorporated hemagglutinin (HA) of H5N1 into the JPV genome by replacing the small hydrophobic (SH) gene to generate a recombinant JPV expressing HA (rJPV-ΔSH-H5). A single intranasal administration of rJPV-ΔSH-H5 protected mice from a lethal HPAI H5N1 challenge. Intranasal vaccination of rJPV-ΔSH-H5 in rhesus macaques elicited antigen-specific humoral and cell-mediated immune responses. This work demonstrates that JPV is a promising vaccine vector. IMPORTANCE A highly pathogenic avian influenza (HPAI) H5N1 outbreak in Southeast Asia destroyed millions of birds. Transmission of H5N1 into humans resulted in deaths in many countries. In this work, we developed a novel H5N1 vaccine candidate using J paramyxovirus (JPV) as a vector and demonstrated that JPV is an efficacious vaccine vector in animals. Nonsegmented negative-sense single-stranded viruses (NNSVs) stably express foreign genes without integrating into the host genome. JPV, an NNSV, replicates efficiently in the respiratory tract and induces robust immune responses.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Paramyxovirinae/inmunología , Animales , Línea Celular , Chlorocebus aethiops , Cricetinae , Perros , Femenino , Humanos , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos BALB C , Desarrollo de Vacunas
8.
PLoS Pathog ; 16(11): e1008972, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33152032

RESUMEN

Paramyxo- and filovirus nucleocapsids (NCs) have bipartite promoters at their 3' ends to initiate RNA synthesis. The 2 elements, promoter element 1 (PE1) and promoter element 2 (PE2), are separated by a spacer region that must be exactly a multiple of 6 nucleotides (nt) long. Paramyxovirus NCs have 13 nucleoprotein (NP) subunits/turn, such that PE1 and PE2 are juxtaposed on the same face of the NC helix, for concerted recognition by the viral polymerase. Ebola virus (EBOV) NCs, in contrast, have 25 to 28 subunits/turn, meaning that PE1 and PE2 cannot be juxtaposed. However, there is evidence that the number of subunits/turn at the 3' end of the EBOV NC is variable. We propose a paramyxovirus-like model for EBOV explaining why there are 8 contiguous copies of the PE2 repeat when 3 are sufficient, why expanding this run to 13 further improves minigenome performance, and why there is a limit to the number of hexa-nt that can be inserted in the spacer region.


Asunto(s)
Ebolavirus/genética , Genoma Viral/genética , Fiebre Hemorrágica Ebola/virología , Nucleoproteínas/genética , Paramyxovirinae/genética , Regiones Promotoras Genéticas/genética , Humanos , Nucleocápside/genética , Alineación de Secuencia
9.
J Virol ; 94(6)2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31896588

RESUMEN

Influenza viruses are highly infectious and are the leading cause of human respiratory diseases and may trigger severe epidemics and occasional pandemics. Although antiviral drugs against influenza viruses have been developed, there is an urgent need to design new strategies to develop influenza virus inhibitors due to the increasing resistance of viruses toward currently available drugs. In this study, we examined the antiviral activity of natural compounds against the following influenza virus strains: A/WSN/33 (H1N1), A/Udorn/72 (H3N2), and B/Lee/40. Papaverine (a nonnarcotic alkaloid that has been used for the treatment of heart disease, impotency, and psychosis) was found to be an effective inhibitor of multiple strains of influenza virus. Kinetic studies demonstrated that papaverine inhibited influenza virus infection at a late stage in the virus life cycle. An alteration in influenza virus morphology and viral ribonucleoprotein (vRNP) localization was observed as an effect of papaverine treatment. Papaverine is a well-known phosphodiesterase inhibitor and also modifies the mitogen-activated protein kinase (MAPK) pathway by downregulating the phosphorylation of MEK and extracellular signal-regulated kinase (ERK). Thus, the modulation of host cell signaling pathways by papaverine may be associated with the nuclear retention of vRNPs and the reduction of influenza virus titers. Interestingly, papaverine also inhibited paramyxoviruses parainfluenza virus 5 (PIV5), human parainfluenza virus 3 (HPIV3), and respiratory syncytial virus (RSV) infections. We propose that papaverine can be a potential candidate to be used as an antiviral agent against a broad range of influenza viruses and paramyxoviruses.IMPORTANCE Influenza viruses are important human pathogens that are the causative agents of epidemics and pandemics. Despite the availability of an annual vaccine, a large number of cases occur every year globally. Here, we report that papaverine, a vasodilator, shows inhibitory action against various strains of influenza virus as well as the paramyxoviruses PIV5, HPIV3, and RSV. A significant effect of papaverine on the influenza virus morphology was observed. Papaverine treatment of influenza-virus-infected cells resulted in the inhibition of virus at a later time in the virus life cycle through the suppression of nuclear export of vRNP and also interfered with the host cellular cAMP and MEK/ERK cascade pathways. This study explores the use of papaverine as an effective inhibitor of both influenza viruses as well as paramyxoviruses.


Asunto(s)
Antivirales/farmacología , Reposicionamiento de Medicamentos , Infecciones por Orthomyxoviridae , Orthomyxoviridae/metabolismo , Papaverina/farmacología , Infecciones por Paramyxoviridae , Paramyxovirinae/metabolismo , Animales , Perros , Evaluación Preclínica de Medicamentos , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células de Riñón Canino Madin Darby , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/patología , Infecciones por Paramyxoviridae/metabolismo , Infecciones por Paramyxoviridae/patología
10.
J Virol ; 94(19)2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32669342

RESUMEN

Medically important paramyxoviruses, such as measles, mumps, parainfluenza, Nipah, and Hendra viruses, infect host cells by directing fusion of the viral and cellular plasma membranes. Upon infection, paramyxoviruses cause a second type of membrane fusion, cell-cell fusion (syncytium formation), which is linked to pathogenicity. Host cell receptor binding causes conformational changes in the attachment glycoprotein (HN, H, or G) that trigger a conformational cascade in the fusion (F) glycoprotein that mediates membrane fusion. F, a class I fusion protein, contains the archetypal heptad repeat regions 1 (HR1) and 2 (HR2). It is well established that binding of HR1 and HR2 is key to fusing viral and cellular membranes. In this study, we uncovered a novel fusion-modulatory role of a third structurally conserved helical region (HR3) in F. Based on its location within the F structure, and structural differences between its prefusion and postfusion conformations, we hypothesized that the HR3 modulates triggering of the F conformational cascade (still requiring G). We used the deadly Nipah virus (NiV) as an important paramyxoviral model to perform alanine scan mutagenesis and a series of multidisciplinary structural/functional analyses that dissect the various states of the membrane fusion cascade. Remarkably, we found that specific residues within the HR3 modulate not only early F-triggering but also late extensive fusion pore expansion steps in the membrane fusion cascade. Our results characterize these novel fusion-modulatory roles of the F HR3, improving our understanding of the membrane fusion process for NiV and likely for the related Henipavirus genus and possibly Paramyxoviridae family members.IMPORTANCE The Paramyxoviridae family includes important human and animal pathogens, such as measles, mumps, and parainfluenza viruses and the deadly henipaviruses Nipah (NiV) and Hendra (HeV) viruses. Paramyxoviruses infect the respiratory tract and the central nervous system (CNS) and can be highly infectious. Most paramyxoviruses have a limited host range. However, the biosafety level 4 NiV and HeV are highly pathogenic and have a wide mammalian host range. Nipah viral infections result in acute respiratory syndrome and severe encephalitis in humans, leading to 40 to 100% mortality rates. The lack of licensed vaccines or therapeutic approaches against NiV and other important paramyxoviruses underscores the need to understand viral entry mechanisms. In this study, we uncovered a novel role of a third helical region (HR3) of the NiV fusion glycoprotein in the membrane fusion process that leads to viral entry. This discovery sets HR3 as a new candidate target for antiviral strategies for NiV and likely for related viruses.


Asunto(s)
Fusión de Membrana/fisiología , Virus Nipah/metabolismo , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/metabolismo , Animales , Chlorocebus aethiops , Encefalitis/virología , Células HEK293 , Infecciones por Henipavirus/virología , Especificidad del Huésped , Humanos , Modelos Moleculares , Virus Nipah/genética , Paramyxovirinae , Conformación Proteica , Dominios Proteicos , Alineación de Secuencia , Homología Estructural de Proteína , Células Vero , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales de Fusión/genética , Internalización del Virus
11.
J Gen Virol ; 101(5): 467-472, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32100701

RESUMEN

The paramyxoviruses Hendra virus (HeV) and parainfluenza virus 5 (PIV5) require the fusion (F) protein to efficiently infect cells. For fusion to occur, F undergoes dramatic, essentially irreversible conformational changes to merge the viral and cell membranes into a continuous bilayer. Recently, a transmembrane (TM) domain leucine/isoleucine (L/I) zipper was shown to be critical in maintaining the expression, stability and pre-fusion conformation of HeV F, allowing for fine-tuned timing of membrane fusion. To analyse the effect of the TM domain L/I zipper in another paramyxovirus, we created alanine mutations to the TM domain of PIV5 F, a paramyxovirus model system. Our data show that while the PIV5 F TM L/I zipper does not significantly affect total expression and only modestly affects surface expression and pre-fusion stability, it is critical for fusogenic activity. These results suggest that the roles of TM L/I zipper motifs differ among members of the family Paramyxoviridae.


Asunto(s)
Membrana Celular/genética , Isoleucina/genética , Leucina/genética , Mutación/genética , Virus de la Parainfluenza 5/genética , Dominios Proteicos/genética , Proteínas Virales de Fusión/genética , Secuencia de Aminoácidos , Animales , Línea Celular , Chlorocebus aethiops , Fusión de Membrana/genética , Paramyxovirinae/genética , Células Vero
12.
J Gen Virol ; 101(1): 44-58, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31793855

RESUMEN

Although most non-segmented negative-strand RNA viruses (NNSVs) replicate in the cytoplasm, NNSV proteins often exert host manipulatory functions in the nucleus. Matrix (M) proteins of henipaviruses and other paramyxoviruses shuttle through the nucleus, where host factors may bind for M modification or host-cell manipulation. Acidic leucine-rich nuclear phosphoprotein 32 family member B (ANP32B) is an interactor of Hendra and Nipah virus M. Both accumulate in the nucleus in an ANP32B-dependent manner. Here we demonstrate that the nuclear localization signal (NLS) of ANP32B is dispensable for HeV M binding. Specific purification of M-ANP32B but not of M-ANP32A complexes revealed that neither the negatively charged acidic nor the leucine-rich regions of ANP32 proteins per se mediate interactions with henipavirus M proteins. Whereas pneumovirus M did not interact with ANP32B, Newcastle disease virus (NDV, genus Avulavirus), Sendai virus (SeV, genus Respirovirus), Measles virus (MeV, genus Morbillivirus) and Canine distemper virus (CDV, genus Morbillivirus) M were able to form complexes with ANP32B. However, in contrast to NDV M and SeV M, which accumulated in the nucleus ANP32B dependently, both morbillivirus Ms did not accumulate in the nucleus, neither at ANP32B overexpression nor after nuclear protein export inhibition. These results indicate that intracellular compartmentalization of cytoplasmic morbillivirus M and nuclear ANP32B prevented an intracellular interaction. Overall, we provide evidence for a general ability of paramyxovirus M proteins to interact with ANP32B. This suggests a conserved, yet to be clarified mechanism might play a role in host manipulation and immune regulation in infected hosts.


Asunto(s)
Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Paramyxovirinae/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Línea Celular , Núcleo Celular/metabolismo , Núcleo Celular/virología , Citoplasma/metabolismo , Células HEK293 , Humanos , Señales de Localización Nuclear/metabolismo
13.
J Virol ; 93(17)2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31189700

RESUMEN

We have developed a high-throughput sequencing (HTS) workflow for investigating paramyxovirus transcription and replication. We show that sequencing of oligo(dT)-selected polyadenylated mRNAs, without considering the orientation of the RNAs from which they had been generated, cannot accurately be used to analyze the abundance of viral mRNAs because genomic RNA copurifies with the viral mRNAs. The best method is directional sequencing of infected cell RNA that has physically been depleted of ribosomal and mitochondrial RNA followed by bioinformatic steps to differentiate data originating from genomes from viral mRNAs and antigenomes. This approach has the advantage that the abundance of viral mRNA (and antigenomes) and genomes can be analyzed and quantified from the same data. We investigated the kinetics of viral transcription and replication during infection of A549 cells with parainfluenza virus type 2 (PIV2), PIV3, PIV5, or mumps virus and determined the abundances of individual viral mRNAs and readthrough mRNAs. We found that the mRNA abundance gradients differed significantly between all four viruses but that for each virus the pattern remained relatively stable throughout infection. We suggest that rapid degradation of non-poly(A) mRNAs may be primarily responsible for the shape of the mRNA abundance gradient in parainfluenza virus 3, whereas a combination of this factor and disengagement of RNA polymerase at intergenic sequences, particularly those at the NP:P and P:M gene boundaries, may be responsible in the other viruses.IMPORTANCE High-throughput sequencing (HTS) of virus-infected cells can be used to study in great detail the patterns of virus transcription and replication. For paramyxoviruses, and by analogy for all other negative-strand RNA viruses, we show that directional sequencing must be used to distinguish between genomic RNA and mRNA/antigenomic RNA because significant amounts of genomic RNA copurify with poly(A)-selected mRNA. We found that the best method is directional sequencing of total cell RNA, after the physical removal of rRNA (and mitochondrial RNA), because quantitative information on the abundance of both genomic RNA and mRNA/antigenomes can be simultaneously derived. Using this approach, we revealed new details of the kinetics of virus transcription and replication for parainfluenza virus (PIV) type 2, PIV3, PIV5, and mumps virus, as well as on the relative abundance of the individual viral mRNAs.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Infecciones por Paramyxoviridae/virología , Paramyxovirinae/fisiología , ARN Mensajero/genética , Secuenciación Completa del Genoma/métodos , Células A549 , Regulación Viral de la Expresión Génica , Tamaño del Genoma , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Paramyxovirinae/clasificación , Paramyxovirinae/patogenicidad , ARN Viral/genética , Especificidad de la Especie , Replicación Viral
14.
J Virol ; 93(17)2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31217248

RESUMEN

Enveloped viruses utilize surface glycoproteins to bind and fuse with a target cell membrane. The zoonotic Hendra virus (HeV), a member of the family Paramyxoviridae, utilizes the attachment protein (G) and the fusion protein (F) to perform these critical functions. Upon triggering, the trimeric F protein undergoes a large, irreversible conformation change to drive membrane fusion. Previously, we have shown that the transmembrane (TM) domain of the F protein, separate from the rest of the protein, is present in a monomer-trimer equilibrium. This TM-TM association contributes to the stability of the prefusion form of the protein, supporting a role for TM-TM interactions in the control of F protein conformational changes. To determine the impact of disrupting TM-TM interactions, constructs expressing the HeV F TM with limited flanking sequences were synthesized. Coexpression of these constructs with HeV F resulted in dramatic reductions in the stability of F protein expression and fusion activity. In contrast, no effects were observed when the HeV F TM constructs were coexpressed with the nonhomologous parainfluenza virus 5 (PIV5) fusion protein, indicating a requirement for specific interactions. To further examine this, a TM peptide homologous to the PIV5 F TM domain was synthesized. Addition of the peptide prior to infection inhibited infection with PIV5 but did not significantly affect infection with human metapneumovirus, a related virus. These results indicate that targeted disruption of TM-TM interactions significantly impact viral fusion protein stability and function, presenting these interactions as a novel target for antiviral development.IMPORTANCE Enveloped viruses require virus-cell membrane fusion to release the viral genome and replicate. The viral fusion protein triggers from the pre- to the postfusion conformation, an essentially irreversible change, to drive membrane fusion. We found that small proteins containing the TM and a limited flanking region homologous to the fusion protein of the zoonotic Hendra virus reduced protein expression and fusion activity. The introduction of exogenous TM peptides may displace a TM domain, disrupting native TM-TM interactions and globally destabilizing the fusion protein. Supporting this hypothesis, we showed that a sequence-specific transmembrane peptide dramatically reduced viral infection in another enveloped virus model, suggesting a broader inhibitory mechanism. Viral fusion protein TM-TM interactions are important for protein function, and disruption of these interactions dramatically reduces protein stability.


Asunto(s)
Paramyxovirinae/metabolismo , Péptidos/farmacología , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética , Animales , Sitios de Unión/efectos de los fármacos , Chlorocebus aethiops , Virus Hendra/química , Virus Hendra/genética , Virus Hendra/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas/efectos de los fármacos , Virus de la Parainfluenza 5/química , Virus de la Parainfluenza 5/genética , Virus de la Parainfluenza 5/metabolismo , Paramyxovirinae/química , Paramyxovirinae/genética , Conformación Proteica/efectos de los fármacos , Dominios Proteicos/efectos de los fármacos , Estabilidad Proteica , Células Vero , Proteínas Virales de Fusión/efectos de los fármacos
15.
J Virol ; 93(22)2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31462574

RESUMEN

Hendra virus (HeV) is a zoonotic paramyxovirus that utilizes a trimeric fusion (F) protein within its lipid bilayer to mediate membrane merger with a cell membrane for entry. Previous HeV F studies showed that transmembrane domain (TMD) interactions are important for stabilizing the prefusion conformation of the protein prior to triggering. Thus, the current model for HeV F fusion suggests that modulation of TMD interactions is critical for initiation and completion of conformational changes that drive membrane fusion. HeV F constructs (T483C/V484C, V484C/N485C, and N485C/P486C) were generated with double cysteine substitutions near the N-terminal region of the TMD to study the effect of altered flexibility in this region. Oligomeric analysis showed that the double cysteine substitutions successfully promoted intersubunit disulfide bond formation in HeV F. Subsequent fusion assays indicated that the introduction of disulfide bonds in the mutants prohibited fusion events. Further testing confirmed that T483C/V484C and V484C/N485C were expressed at the cell surface at levels that would allow for fusion. Attempts to restore fusion with a reducing agent were unsuccessful, suggesting that the introduced disulfide bonds were likely buried in the membrane. Conformational analysis showed that T483C/V484C and V484C/N485C were able to bind a prefusion conformation-specific antibody prior to cell disruption, indicating that the introduced disulfide bonds did not significantly affect protein folding. This study is the first to report that TMD dissociation is required for HeV F fusogenic activity and strengthens our model for HeV fusion.IMPORTANCE The paramyxovirus Hendra virus (HeV) causes severe respiratory illness and encephalitis in humans. To develop therapeutics for HeV and related viral infections, further studies are needed to understand the mechanisms underlying paramyxovirus fusion events. Knowledge gained in studies of the HeV fusion (F) protein may be applicable to a broad span of enveloped viruses. In this study, we demonstrate that disulfide bonds introduced between the HeV F transmembrane domains (TMDs) block fusion. Depending on the location of these disulfide bonds, HeV F can still fold properly and bind a prefusion conformation-specific antibody prior to cell disruption. These findings support our current model for HeV membrane fusion and expand our knowledge of the TMD and its role in HeV F stability and fusion promotion.


Asunto(s)
Virus Hendra/metabolismo , Infecciones por Henipavirus/metabolismo , Proteínas Virales de Fusión/metabolismo , Secuencia de Aminoácidos/genética , Animales , Línea Celular , Chlorocebus aethiops , Virus Hendra/genética , Humanos , Fusión de Membrana/fisiología , Paramyxovirinae/metabolismo , Dominios Proteicos/genética , Pliegue de Proteína , Células Vero , Proteínas Virales de Fusión/genética , Internalización del Virus
16.
PLoS Pathog ; 14(2): e1006889, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29425244

RESUMEN

Paramyxoviruses represent a family of RNA viruses causing significant human diseases. These include measles virus, the most infectious virus ever reported, in addition to parainfluenza virus, and other emerging viruses. Paramyxoviruses likely share common replication machinery but their mechanisms of RNA biosynthesis activities and details of their complex polymerase structures are unknown. Mechanistic and functional details of a paramyxovirus polymerase would have sweeping implications for understanding RNA virus replication and for the development of new antiviral medicines. To study paramyxovirus polymerase structure and function, we expressed an active recombinant Nipah virus (NiV) polymerase complex assembled from the multifunctional NiV L protein bound to its phosphoprotein cofactor. NiV is an emerging highly pathogenic virus that causes severe encephalitis and has been declared a global public health concern due to its high mortality rate. Using negative-stain electron microscopy, we demonstrated NiV polymerase forms ring-like particles resembling related RNA polymerases. We identified conserved sequence elements driving recognition of the 3'-terminal genomic promoter by NiV polymerase, and leading to initiation of RNA synthesis, primer extension, and transition to elongation mode. Polyadenylation resulting from NiV polymerase stuttering provides a mechanistic basis for transcription termination. It also suggests a divergent adaptation in promoter recognition between pneumo- and paramyxoviruses. The lack of available antiviral therapy for NiV prompted us to identify the triphosphate forms of R1479 and GS-5734, two clinically relevant nucleotide analogs, as substrates and inhibitors of NiV polymerase activity by delayed chain termination. Overall, these findings provide low-resolution structural details and the mechanism of an RNA polymerase from a previously uncharacterized virus family. This work illustrates important functional differences yet remarkable similarities between the polymerases of nonsegmented negative-strand RNA viruses.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/metabolismo , Virus Nipah/genética , Fosfoproteínas/metabolismo , Elongación de la Transcripción Genética , Iniciación de la Transcripción Genética , Terminación de la Transcripción Genética , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/genética , Virus Nipah/enzimología , Paramyxovirinae/enzimología , Paramyxovirinae/genética , Paramyxovirinae/metabolismo , Fosfoproteínas/química , Fosfoproteínas/genética , ARN Viral/genética , ARN Viral/metabolismo , Proteínas Virales/química , Proteínas Virales/genética , Replicación Viral
17.
PLoS Pathog ; 14(1): e1006877, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29381763

RESUMEN

The interferon (IFN) system represents the first line of defense against a wide range of viruses. Virus infection rapidly triggers the transcriptional induction of IFN-ß and IFN Stimulated Genes (ISGs), whose protein products act as viral restriction factors by interfering with specific stages of virus life cycle, such as entry, transcription, translation, genome replication, assembly and egress. Here, we report a new mode of action of an ISG, IFN-induced TDRD7 (tudor domain containing 7) inhibited paramyxovirus replication by inhibiting autophagy. TDRD7 was identified as an antiviral gene by a high throughput screen of an ISG shRNA library for blocking IFN's protective effect against Sendai virus (SeV) replication. The antiviral activity of TDRD7 against SeV, human parainfluenza virus 3 and respiratory syncytial virus was confirmed by its genetic ablation or ectopic expression in several types of mouse and human cells. TDRD7's antiviral action was mediated by its ability to inhibit autophagy, a cellular catabolic process which was robustly induced by SeV infection and required for its replication. Mechanistic investigation revealed that TDRD7 interfered with the activation of AMP-dependent kinase (AMPK), an enzyme required for initiating autophagy. AMPK activity was required for efficient replication of several paramyxoviruses, as demonstrated by its genetic ablation or inhibition of its activity by TDRD7 or chemical inhibitors. Therefore, our study has identified a new antiviral ISG with a new mode of action.


Asunto(s)
Antivirales/farmacología , Autofagia , Interferones/farmacología , Paramyxovirinae/fisiología , Ribonucleoproteínas/fisiología , Replicación Viral/efectos de los fármacos , Animales , Autofagia/genética , Autofagia/inmunología , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/genética , Ratones , Ratones Endogámicos C57BL , Ribonucleoproteínas/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Replicación Viral/genética
18.
Virus Genes ; 56(2): 150-167, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32076918

RESUMEN

The viruses historically implicated or currently considered as candidates for misuse in bioterrorist events are poxviruses, filoviruses, bunyaviruses, orthomyxoviruses, paramyxoviruses and a number of arboviruses causing encephalitis, including alpha- and flaviviruses. All these viruses are of concern for public health services when they occur in natural outbreaks or emerge in unvaccinated populations. Recent events and intelligence reports point to a growing risk of dangerous biological agents being used for nefarious purposes. Public health responses effective in natural outbreaks of infectious disease may not be sufficient to deal with the severe consequences of a deliberate release of such agents. One important aspect of countermeasures against viral biothreat agents are the antiviral treatment options available for use in post-exposure prophylaxis. These issues were adressed by the organizers of the 16th Medical Biodefense Conference, held in Munich in 2018, in a special session on the development of drugs to treat infections with viruses currently perceived as a threat to societies or associated with a potential for misuse as biothreat agents. This review will outline the state-of-the-art methods in antivirals research discussed and provide an overview of antiviral compounds in the pipeline that are already approved for use or still under development.


Asunto(s)
Antivirales/uso terapéutico , Arbovirus/efectos de los fármacos , Bioterrorismo/prevención & control , Virosis/tratamiento farmacológico , Arbovirus/patogenicidad , Filoviridae/efectos de los fármacos , Filoviridae/patogenicidad , Humanos , Orthobunyavirus/efectos de los fármacos , Orthobunyavirus/patogenicidad , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/patogenicidad , Paramyxovirinae/efectos de los fármacos , Paramyxovirinae/patogenicidad , Poxviridae/efectos de los fármacos , Poxviridae/patogenicidad , Virosis/virología
19.
J Gen Virol ; 100(3): 403-413, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30688635

RESUMEN

Bats are the reservoir hosts for multiple viruses with zoonotic potential, including coronaviruses, paramyxoviruses and filoviruses. Urine collected from Australian pteropid bats was assessed for the presence of paramyxoviruses. One of the viruses isolated was Teviot virus (TevPV), a novel rubulavirus previously isolated from pteropid bat urine throughout the east coast of Australia. Here, we further characterize TevPV through analysis of whole-genome sequencing, growth kinetics, antigenic relatedness and the experimental infection of ferrets and mice. TevPV is phylogenetically and antigenically most closely related to Tioman virus (TioPV). Unlike many other rubulaviruses, cell receptor attachment by TevPV does not appear to be sialic acid-dependent, with the receptor for host cell entry being unknown. The infection of ferrets and mice suggested that TevPV has a low pathogenic potential in mammals. Infected ferrets seroconverted by 10 days post-infection without clinical signs of disease. Furthermore, infected ferrets did not shed virus in any respiratory secretions, suggesting a low risk of onward transmission of TevPV. No productive infection was observed in the mouse infection study.


Asunto(s)
Quirópteros/virología , Infecciones por Paramyxoviridae/veterinaria , Paramyxovirinae/aislamiento & purificación , Animales , Australia , Hurones , Genoma Viral , Ratones , Infecciones por Paramyxoviridae/virología , Paramyxovirinae/genética , Paramyxovirinae/patogenicidad , Paramyxovirinae/fisiología , Filogenia , Virulencia
20.
J Virol ; 92(5)2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29237838

RESUMEN

One of the first defenses against infecting pathogens is the innate immune system activated by cellular recognition of pathogen-associated molecular patterns (PAMPs). Although virus-derived RNA species, especially copyback (cb)-type defective interfering (DI) genomes, have been shown to serve as real PAMPs, which strongly induce interferon-beta (IFN-ß) during mononegavirus infection, the mechanisms underlying DI generation remain unclear. Here, for the first time, we identified a single amino acid substitution causing production of cbDI genomes by successful isolation of two distinct types of viral clones with cbDI-producing and cbDI-nonproducing phenotypes from the stock Sendai virus (SeV) strain Cantell, which has been widely used in a number of studies on antiviral innate immunity as a representative IFN-ß-inducing virus. IFN-ß induction was totally dependent on the presence of a significant amount of cbDI genome-containing viral particles (DI particles) in the viral stock, but not on deficiency of the IFN-antagonistic viral accessory proteins C and V. Comparison of the isolates indicated that a single amino acid substitution found within the N protein of the cbDI-producing clone was enough to cause the emergence of DI genomes. The mutated N protein of the cbDI-producing clone resulted in a lower density of nucleocapsids than that of the DI-nonproducing clone, probably causing both production of the DI genomes and their formation of a stem-loop structure, which serves as an ideal ligand for RIG-I. These results suggested that the integrity of mononegaviral nucleocapsids might be a critical factor in avoiding the undesirable recognition of infection by host cells.IMPORTANCE The type I interferon (IFN) system is a pivotal defense against infecting RNA viruses that is activated by sensing viral RNA species. RIG-I is a major sensor for infection with most mononegaviruses, and copyback (cb)-type defective interfering (DI) genomes have been shown to serve as strong RIG-I ligands in real infections. However, the mechanism underlying production of cbDI genomes remains unclear, although DI genomes emerge as the result of an error during viral replication with high doses of viruses. Sendai virus has been extensively studied and is unique in that its interaction with innate immunity reveals opposing characteristics, such as high-level IFN-ß induction and strong inhibition of type I IFN pathways. Our findings provide novel insights into the mechanism of production of mononegaviral cbDI genomes, as well as virus-host interactions during innate immunity.


Asunto(s)
Sustitución de Aminoácidos/inmunología , Virus Defectuosos/genética , Interferón beta/metabolismo , Nucleoproteínas/inmunología , Paramyxovirinae/genética , Paramyxovirinae/inmunología , Virus Sendai/genética , Sustitución de Aminoácidos/genética , Animales , Línea Celular , Proteína 58 DEAD Box , Virus Defectuosos/inmunología , Femenino , Regulación de la Expresión Génica , Genoma Viral , Células HeLa , Interacciones Huésped-Parásitos/genética , Interacciones Huésped-Parásitos/inmunología , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/análisis , Interferón Tipo I/inmunología , Ratones , Ratones Endogámicos C57BL , Mutación , Nucleocápside/metabolismo , Nucleoproteínas/genética , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/virología , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , ARN Viral/genética , Receptores Inmunológicos , Virus Sendai/inmunología , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA