Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 624
Filtrar
1.
J Virol ; 98(2): e0197523, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38294249

RESUMO

The highly pathogenic arenavirus, Junín virus (JUNV), expresses three truncated alternative isoforms of its nucleoprotein (NP), i.e., NP53kD, NP47kD, and NP40kD. While both NP47kD and NP40kD have been previously shown to be products of caspase cleavage, here, we show that expression of the third isoform NP53kD is due to alternative in-frame translation from M80. Based on this information, we were able to generate recombinant JUNVs lacking each of these isoforms. Infection with these mutants revealed that, while all three isoforms contribute to the efficient control of caspase activation, NP40kD plays the predominant role. In contrast to full-length NP (i.e., NP65kD), which is localized to inclusion bodies, where viral RNA synthesis takes place, the loss of portions of the N-terminal coiled-coil region in these isoforms leads to a diffuse cytoplasmic distribution and a loss of function in viral RNA synthesis. Nonetheless, NP53kD, NP47kD, and NP40kD all retain robust interferon antagonistic and 3'-5' exonuclease activities. We suggest that the altered localization of these NP isoforms allows them to be more efficiently targeted by activated caspases for cleavage as decoy substrates, and to be better positioned to degrade viral double-stranded (ds)RNA species that accumulate in the cytoplasm during virus infection and/or interact with cytosolic RNA sensors, thereby limiting dsRNA-mediated innate immune responses. Taken together, this work provides insight into the mechanism by which JUNV leverages apoptosis during infection to generate biologically distinct pools of NP and contributes to our understanding of the expression and biological relevance of alternative protein isoforms during virus infection.IMPORTANCEA limited coding capacity means that RNA viruses need strategies to diversify their proteome. The nucleoprotein (NP) of the highly pathogenic arenavirus Junín virus (JUNV) produces three N-terminally truncated isoforms: two (NP47kD and NP40kD) are known to be produced by caspase cleavage, while, here, we show that NP53kD is produced by alternative translation initiation. Recombinant JUNVs lacking individual NP isoforms revealed that all three isoforms contribute to inhibiting caspase activation during infection, but cleavage to generate NP40kD makes the biggest contribution. Importantly, all three isoforms retain their ability to digest double-stranded (ds)RNA and inhibit interferon promoter activation but have a diffuse cytoplasmic distribution. Given the cytoplasmic localization of both aberrant viral dsRNAs, as well as dsRNA sensors and many other cellular components of innate immune activation pathways, we suggest that the generation of NP isoforms not only contributes to evasion of apoptosis but also robust control of the antiviral response.


Assuntos
Caspases , Citoplasma , Febre Hemorrágica Americana , Interações Hospedeiro-Patógeno , Imunidade Inata , Vírus Junin , Nucleoproteínas , Biossíntese de Proteínas , Humanos , Apoptose , Inibidores de Caspase/metabolismo , Caspases/metabolismo , Citoplasma/metabolismo , Citoplasma/virologia , Ativação Enzimática , Febre Hemorrágica Americana/imunologia , Febre Hemorrágica Americana/virologia , Interferons/genética , Interferons/imunologia , Vírus Junin/genética , Vírus Junin/metabolismo , Vírus Junin/patogenicidade , Nucleoproteínas/biossíntese , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/metabolismo , RNA Viral/biossíntese , RNA Viral/genética , Replicação Viral
2.
PLoS Pathog ; 16(6): e1008610, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32603377

RESUMO

Newcastle disease virus (NDV), a member of the Paramyxoviridae family, can activate PKR/eIF2α signaling cascade to shutoff host and facilitate viral mRNA translation during infection, however, the mechanism remains unclear. In this study, we revealed that NDV infection up-regulated host cap-dependent translation machinery by activating PI3K/Akt/mTOR and p38 MAPK/Mnk1 pathways. In addition, NDV infection induced p38 MAPK/Mnk1 signaling participated 4E-BP1 hyperphosphorylation for efficient viral protein synthesis when mTOR signaling is inhibited. Furthermore, NDV NP protein was found to be important for selective cap-dependent translation of viral mRNAs through binding to eIF4E during NDV infection. Taken together, NDV infection activated multiple signaling pathways for selective viral protein synthesis in infected cells, via interaction between viral NP protein and host translation machinery. Our results may help to design novel targets for therapeutic intervention against NDV infection and to understand the NDV anti-oncolytic mechanism.


Assuntos
Proteínas Aviárias , Fator de Iniciação 4E em Eucariotos , Sistema de Sinalização das MAP Quinases , Vírus da Doença de Newcastle , Nucleoproteínas , RNA Mensageiro , RNA Viral , Proteínas Virais , Animais , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Embrião de Galinha , Galinhas , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Células HEK293 , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Vírus da Doença de Newcastle/genética , Vírus da Doença de Newcastle/metabolismo , Proteínas do Nucleocapsídeo , Nucleoproteínas/biossíntese , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Biossíntese de Proteínas , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Proteínas Virais/biossíntese , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
J Virol ; 89(22): 11734-8, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26355095

RESUMO

Recombinant glycoprotein-deficient lymphocytic choriomeningitis virus-based vaccine vectors (rLCMV/ΔGP) are potent CD8(+) T cell inducers. To investigate the underlying molecular requirements, we generated a nucleoprotein-deficient vector counterpart (rLCMV/ΔNP). NP but not GP is a minimal trans-acting factor for viral transcription and genome replication. We found that, unlike rLCMV/ΔGP, rLCMV/ΔNP failed to elicit detectable CD8(+) T cell responses unless NP was trans complemented in a transgenic host. Hence, NP-dependent intracellular gene expression is essential for LCMV vector immunogenicity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Nucleoproteínas/biossíntese , Vacinas Virais/imunologia , Regiões 3' não Traduzidas/genética , Animais , Linhagem Celular , Cricetinae , Expressão Gênica/imunologia , Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Transgênicos , Nucleoproteínas/genética , Nucleoproteínas/imunologia
4.
J Virol ; 89(2): 1119-28, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25378485

RESUMO

UNLABELLED: Boid inclusion body disease (BIDB) is a fatal disease of boid snakes, the etiology of which has only recently been revealed following the identification of several novel arenaviruses in diseased snakes. BIBD-associated arenaviruses (BIBDAV) are genetically divergent from the classical Old and New World arenaviruses and also differ substantially from each other. Even though there is convincing evidence that BIBDAV are indeed the etiological agent of BIBD, the BIBDAV reservoir hosts--if any exist besides boid snakes themselves--are not yet known. In this report, we use University of Helsinki virus (UHV; a virus that we isolated from a Boa constrictor with BIBD) to show that BIBDAV can also replicate effectively in mammalian cells, including human cells, provided they are cultured at 30°C. The infection induces the formation of cytoplasmic inclusion bodies (IB), comprised mainly of viral nucleoprotein (NP), similar to those observed in BIBD and in boid cell cultures. Transferring infected cells from 30°C to 37°C ambient temperature resulted in progressive declines in IB formation and in the amounts of viral NP and RNA, suggesting that BIBDAV growth is limited at 37°C. These observations indirectly indicate that IB formation is linked to viral replication. In addition to mammalian and reptilian cells, UHV infected arthropod (tick) cells when grown at 30°C. Even though our findings suggest that BIBDAV have a high potential to cross the species barrier, their inefficient growth at mammalian body temperatures indicates that the reservoir hosts of BIBDAV are likely species with a lower body temperature, such as snakes. IMPORTANCE: The newly discovered boid inclusion body disease-associated arenaviruses (BIBDAV) of reptiles have drastically altered the phylogeny of the family Arenavirus. Prior to their discovery, known arenaviruses were considered mainly rodent-borne viruses, with each arenavirus species having its own reservoir host. BIBDAV have so far been demonstrated in captive boid snakes, but their possible reservoir host(s) have not yet been identified. Here we show, using University of Helsinki virus as a model, that these viruses are able to infect mammalian (including human) and arthropod cells. Our results provide in vitro proof of the considerable ability of arenaviruses to cross species barriers. However, our data indicate that BIBDAV growth occurs at 30°C but is inhibited at 37°C, implying that crossing of the species barrier would be hindered by the body temperature of mammalian species.


Assuntos
Infecções por Arenaviridae/veterinária , Arenaviridae/fisiologia , Arenaviridae/efeitos da radiação , Boidae , Replicação Viral/efeitos da radiação , Animais , Arenaviridae/isolamento & purificação , Infecções por Arenaviridae/virologia , Linhagem Celular , Especificidade de Hospedeiro , Humanos , Corpos de Inclusão Viral , Mamíferos , Nucleoproteínas/biossíntese , RNA Viral/biossíntese , Temperatura , Carrapatos , Proteínas Virais/biossíntese
5.
J Comp Pathol ; 149(1): 82-93, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23369809

RESUMO

The chicken and duck are important hosts of Newcastle disease virus (NDV) with distinctive responses to infection. NDV infection in ducks is often subclinical and chronic, while in chickens the infection is clinically apparent and transient. These differences may be due to in part to the host response to NDV infection. This study compares the histopathological changes in the spleen, thymus and bursa of Fabricius following infection with NDV in chickens and ducks. The 9a5b isolate of NDV was inoculated intranasally into specific pathogen-free chickens and Japanese commercial ducks. NDV nucleoprotein (NDV-NP) and interferon (IFN)-ß were detected in tissues by immunohistochemistry (IHC), apoptosis was detected by haematoxylin and eosin staining, caspase-3 IHC and the TUNEL assay. Labelling of NDV-NP and lymphoid depletion were most marked in chicken tissues. The pattern of apoptosis in the spleen differed between chickens and ducks. In chickens there were numerous apoptotic cells in the peri-ellipsoidal white pulp and the peri-ellipsoidal, peri-arteriolar and peri-venous lymphoid sheaths, while apoptosis in duck spleens was mainly within the germinal centres. Lymphoid depletion was the main feature in the bursal and thymic tissues of chickens, but apoptosis was marked in these organs in ducks. Expression of IFN-ß appeared earlier and was more intense in the tissues from ducks compared with those from chickens. The differences in IFN-ß and NDV-NP expression may reflect the relative clinical severity of the infection in the two avian species.


Assuntos
Doença de Newcastle/imunologia , Doença de Newcastle/patologia , Animais , Bolsa de Fabricius/patologia , Galinhas , Patos , Imuno-Histoquímica , Interferon beta/biossíntese , Masculino , Doença de Newcastle/virologia , Vírus da Doença de Newcastle , Proteínas do Nucleocapsídeo , Nucleoproteínas/biossíntese , Especificidade da Espécie , Baço/patologia , Timo/patologia , Proteínas Virais/biossíntese
6.
J Biol Chem ; 288(12): 8702-8711, 2013 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-23382389

RESUMO

The arenavirus nucleoprotein (NP) can suppress induction of type I interferon (IFN). This anti-IFN activity is thought to be shared by all arenaviruses with the exception of Tacaribe virus (TCRV). To identify the TCRV NP amino acid residues that prevent its IFN-countering ability, we created a series of NP chimeras between residues of TCRV NP and Pichinde virus (PICV) NP, an arenavirus NP with potent anti-IFN function. Chimera NP analysis revealed that a minimal four amino acid stretch derived from PICV NP could impart efficient anti-IFN activity to TCRV NP. Strikingly, the TCRV NP gene cloned and sequenced from viral stocks obtained through National Institutes of Health Biodefense and Emerging Infections (BEI) resources deviated from the reference sequence at this particular four-amino acid region, GPPT (GenBank KC329849) versus DLQL (GenBank NC004293), respectively at residues 389-392. When efficiently expressed in cells through codon-optimization, TCRV NP containing the GPPT residues rescued the antagonistic IFN function. Consistent with cell expression results, TCRV infection did not stimulate an IFNß response early in infection in multiple cells types (e.g. A549, P388D1), and IRF-3 was not translocated to the nucleus in TCRV-infected A549 cells. Collectively, these data suggest that certain TCRV strain variants contain the important NP amino acids necessary for anti-IFN activity.


Assuntos
Arenavirus do Novo Mundo/fisiologia , Interferon beta/metabolismo , Nucleoproteínas/química , Proteínas Recombinantes de Fusão/química , Proteínas Virais/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Arenavirus do Novo Mundo/imunologia , Núcleo Celular/metabolismo , Chlorocebus aethiops , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/genética , Camundongos , Dados de Sequência Molecular , Nucleoproteínas/biossíntese , Nucleoproteínas/imunologia , Regiões Promotoras Genéticas , Transporte Proteico , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/imunologia , Ativação Transcricional , Células Vero , Proteínas Virais/biossíntese , Proteínas Virais/imunologia
7.
Int J Mol Med ; 31(3): 525-32, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23337979

RESUMO

Newcastle disease virus (NDV) AF2240 Malaysian strain is a very virulent avian virus. NDV strain AF2240 was previously demonstrated to induce apoptosis in human breast carcinoma MCF-7 cells. However, at which stage of the NDV life cycle apoptosis is induced and whether NDV replication and protein synthesis are involved in apoptosis induction have yet to be determined. In the present study, we investigated the time course of NDV strain AF2240 nucleoprotein (NP) gene expression and the early apoptotic signs in the form of activation of caspase-8 and mitochondrial transition pore opening. In addition, the induction of apoptosis by both ultraviolet-inactivated and cycloheximide-treated NDV-infected MCF-7 cells were examined. Our findings showed that NDV strain AF2240 induced apoptosis at 1 h post-infection (pi) through activation of mitochondrial transition pore opening and at 2 h through activation of caspase-8, while the NP gene was expressed at 6 h pi. The induced apoptosis was independent of both virus replication and protein synthesis. In conclusion, NDV strain AF2240 induces apoptosis at an early stage of its life cycle, possibly during virus binding or fusion with the cell membrane. The mitochondrial-related pathway may be the central activator in NDV strain AF2240-induced apoptosis.


Assuntos
Apoptose , Vírus da Doença de Newcastle/crescimento & desenvolvimento , Vírus da Doença de Newcastle/metabolismo , Ligação Viral , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/virologia , Caspase 8/metabolismo , Linhagem Celular Tumoral , Chlorocebus aethiops , Cicloeximida/farmacologia , Ativação Enzimática , Feminino , Humanos , Estágios do Ciclo de Vida , Células MCF-7 , Mitocôndrias , Proteínas do Nucleocapsídeo , Nucleoproteínas/biossíntese , Nucleoproteínas/genética , Raios Ultravioleta , Células Vero , Proteínas Virais/biossíntese , Proteínas Virais/genética , Replicação Viral
8.
J Biotechnol ; 163(3): 333-8, 2013 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-23178898

RESUMO

Rabies is one of the most fatal zoonotic diseases in developing countries, where a safe, cheap and effective vaccine against the disease remains unaffordable. In this paper, we describe a new silkworm-baculovirus expression system to express the nucleoprotein (N) gene of rabies virus and evaluation of the immune response in BALB/c mice. A recombinant baculovirus -rBmNPV(RV-N) carrying the N gene of rabies virus Evelyn Rokitniki Abelseth (ERA) strain was constructed and the N protein expression was evaluated in Bombyx mori (BmN) cells and silkworm pupae by immunofluorescence staining, Western blots and enzyme-linked immunosorbent assay (ELISA). The immune response to vaccines was evaluated based on serum IgG antibody titers and challenge experiments. The study revealed that N protein of rabies virus can be highly expressed in silkworm baculovirus expression system and the vaccine of N antigen presents a promising approach for the prevention of rabies virus.


Assuntos
Bombyx/genética , Nucleoproteínas/genética , Nucleoproteínas/imunologia , Vírus da Raiva/genética , Vírus da Raiva/imunologia , Animais , Anticorpos Antivirais/imunologia , Baculoviridae/genética , Baculoviridae/imunologia , Bombyx/imunologia , Linhagem Celular , Feminino , Imunofluorescência/métodos , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Nucleoproteínas/biossíntese , Pupa/imunologia , Raiva/imunologia , Raiva/prevenção & controle , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia
9.
PLoS One ; 7(5): e37337, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22615984

RESUMO

Lymphocytic choriomeningitis virus (LCMV), a natural murine pathogen, is a member of the Arenavirus family, may cause atypical meningitis in humans, and has been utilized extensively as a model pathogen for the study of virus-induced disease and immune responses. Historically, viral titers have been quantified by a standard plaque assay, but for non-cytopathic viruses including LCMV this requires lengthy incubation, so results cannot be obtained rapidly. Additionally, due to specific technical constraints of the plaque assay including the visual detection format, it has an element of subjectivity along with limited sensitivity. In this study, we describe the development of a FACS-based assay that utilizes detection of LCMV nucleoprotein (NP) expression in infected cells to determine viral titers, and that exhibits several advantages over the standard plaque assay. We show that the LCMV-NP FACS assay is an objective and reproducible detection method that requires smaller sample volumes, exhibits a ∼20-fold increase in sensitivity to and produces results three times faster than the plaque assay. Importantly, when applied to models of acute and chronic LCMV infection, the LCMV-NP FACS assay revealed the presence of infectious virus in samples that were determined to be negative by plaque assay. Therefore, this technique represents an accelerated, enhanced and objective alternative method for detection of infectious LCMV that is amenable to adaptation for other viral infections as well as high throughput diagnostic platforms.


Assuntos
Citometria de Fluxo/métodos , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/crescimento & desenvolvimento , Nucleoproteínas/biossíntese , Carga Viral/métodos , Animais , Anticorpos Antivirais/biossíntese , Coriomeningite Linfocítica/sangue , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/metabolismo , Camundongos , Nucleoproteínas/imunologia , Sensibilidade e Especificidade , Ensaio de Placa Viral , Viremia/diagnóstico
10.
J Virol Methods ; 179(1): 70-6, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22001274

RESUMO

Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne viral zoonosis distributed widely in Africa, Asia, Russia and the Balkans. The emergence and re-emergence of CCHFV emphasize the importance of increasing both human and veterinary surveillance and developing diagnostic capacity. Recombinant CCHFV nucleocapsid protein (NP) has been expressed using insect cells and mammalian cells and used as a diagnostic tool but bacterial expression has not been described previously. The S gene of CCHFV was codon optimized and the NP expressed in Escherichia coli from the synthetic gene. The protein was reacted against serum samples collected from confirmed CCHFV patients at varying intervals after the onset of illness from acute to convalescent stages using both an ELISA and a Western blot. To confirm that the protein was able to induce a humoral antibody response that could be detected using CCHFV antigen derived from live virus, mice were immunized and serum samples were tested using IF slides prepared from CCHFV infected Vero cells. The recombinant antigen was able to detect IgG antibody in acute and convalescent sera. In addition, a detectable IgG antibody response was induced in mice immunized using NP. The results suggest that proteins expressed in a bacterial system lacking post-translational modifications can be used in ELISA to detect IgG antibody against CCHFV in human sera which may be used for routine diagnosis and seroepidemiology.


Assuntos
Anticorpos Antivirais/sangue , Antígenos Virais , Ensaio de Imunoadsorção Enzimática/métodos , Expressão Gênica , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Nucleoproteínas , Animais , Antígenos Virais/biossíntese , Antígenos Virais/genética , Western Blotting , Criança , Pré-Escolar , Chlorocebus aethiops , Escherichia coli/genética , Humanos , Imunoglobulina G/sangue , Lactente , Recém-Nascido , Camundongos , Nucleoproteínas/biossíntese , Nucleoproteínas/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Células Vero
11.
J Virol Methods ; 158(1-2): 93-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19428575

RESUMO

Human respiratory syncytial virus (HRSV) is the major pathogen leading to respiratory disease in infants and neonates worldwide. An effective vaccine has not yet been developed against this virus, despite considerable efforts in basic and clinical research. HRSV replication is independent of the nuclear RNA processing constraints, since the virus genes are adapted to the cytoplasmic transcription, a process performed by the viral RNA-dependent RNA polymerase. This study shows that meaningful nuclear RNA polymerase II dependent expression of the HRSV nucleoprotein (N) and phosphoprotein (P) proteins can only be achieved with the optimization of their genes, and that the intracellular localization of N and P proteins changes when they are expressed out of the virus replication context. Immunization tests performed in mice resulted in the induction of humoral immunity using the optimized genes. This result was not observed for the non-optimized genes. In conclusion, optimization is a valuable tool for improving expression of HRSV genes in DNA vaccines.


Assuntos
Anticorpos Antivirais/sangue , Nucleoproteínas/imunologia , Fosfoproteínas/imunologia , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Proteínas Virais/imunologia , Animais , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Nucleoproteínas/biossíntese , Nucleoproteínas/genética , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Vacinas contra Vírus Sincicial Respiratório/biossíntese , Vacinas contra Vírus Sincicial Respiratório/genética , Proteínas Virais/biossíntese , Proteínas Virais/genética
12.
Artigo em Inglês | MEDLINE | ID: mdl-19194011

RESUMO

Ebola VP35 is a multifunctional protein that is important for host immune suppression and pathogenesis. VP35 contains an N-terminal oligomerization domain and a C-terminal interferon inhibitory domain (IID). Mutations within the VP35 IID result in loss of host immune suppression. Here, efforts to crystallize recombinantly overexpressed VP35 IID that was purified from Escherichia coli are described. Native and selenomethionine-labeled crystals belonging to the orthorhombic space group P2(1)2(1)2(1) were obtained by the hanging-drop vapor-diffusion method and diffraction data were collected at the ALS synchrotron.


Assuntos
Ebolavirus/química , Ebolavirus/patogenicidade , Regulação Viral da Expressão Gênica/fisiologia , Interferons/antagonistas & inibidores , Nucleoproteínas/química , Nucleoproteínas/fisiologia , Proteínas do Core Viral/química , Proteínas do Core Viral/fisiologia , Difração de Raios X , Cristalização , Interferons/química , Proteínas do Nucleocapsídeo , Nucleoproteínas/biossíntese , Estrutura Terciária de Proteína/fisiologia , Proteínas do Core Viral/biossíntese , Proteínas Virais Reguladoras e Acessórias/biossíntese , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/isolamento & purificação , Virulência
13.
J Reprod Dev ; 55(2): 214-8, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19194063

RESUMO

The dead end gene, coding an RNA binding protein, is predominantly expressed in the germ cells of vertebrates. Recently, we cloned chicken dead end homologue (CDH) and showed that expression of CDH mRNA is highly specific to primordial germ cells (PGCs) at early embryonic stages. To date, the subcelluler localization of Dead end protein in germ cell has been largely unknown due to lack of an antibody. Here, we raised a polyclonal antibody against chicken dead end homologue (CDH) to elucidate its subcellular localization in the germ cells. For comparative studies with CDH, a polyclonal antibody against chicken vasa homologue (CVH), a well-known germ cell marker, was also raised. Immunoblotting analysis for CDH protein showed a single band with a molecular size of approximately 60 kDa in the ovarian and testicular proteins. Immunofluorescence studies revealed that CDH protein was exclusively localized in the nuclei of primordial germ cells (PGCs) and germ cells at later stages, while CVH was localized in the cytoplasm. Interestingly, the germ cells distributed at the basal sides of seminiferous epithelia, such as spermatogonia, were strongly positive to CDH protein. The current study provides novel evidence that CDH is a nucleoprotein of germ cells, including PGCs.


Assuntos
Galinhas/metabolismo , Células Germinativas/metabolismo , Nucleoproteínas/biossíntese , Ovário/metabolismo , Proteínas de Ligação a RNA/biossíntese , Testículo/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Galinhas/genética , Feminino , Masculino , Dados de Sequência Molecular , Nucleoproteínas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária
14.
Dev Biol (Basel) ; 131: 477-82, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18634510

RESUMO

The objective of this study was to obtain transgenic maize expressing the rabies virus glycoprotein (G) of the Vnukovo strain and to evaluate its immunogenicity in mice, by the oral route. The ubiquitin maize promoter fused to the whole coding region of the rabies virus G gene, and a constitutive promoter from cauliflowermosaic virus (CaMV)were used. Maize embryogenic callus were transformed with the above construct by biolistics. Regenerated maize plants were recovered and grown in a greenhouse. The presence of the G gene and its product was detected by PCR and western blot, respectively. The amount of G protein detected in the grains was approximately 1% of the total soluble plant protein. Transformed kernels containing 50 microg of G protein were given once by the oral route in adult mice (BALB-C strain). Challenge was undertaken at 90-days post-vaccination using a lethal dose of a vampire bat rabies virus (100 LD 50% in mice); vampire bats are one of the main reservoirs in Latin America. The edible vaccine induced viral neutralizing antibodies (VNA) which, protected mice 100% against challenge. The control group did not survive. The G protein of the Vnukovo strain expressed in transgenic maize may be considered as an oral immunogen against rabies, conferring cross-protection.


Assuntos
Glicoproteínas/imunologia , Nucleoproteínas/imunologia , Vacina Antirrábica/administração & dosagem , Proteínas Virais/imunologia , Zea mays , Administração Oral , Animais , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/imunologia , Glicoproteínas/genética , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Nucleoproteínas/biossíntese , Nucleoproteínas/genética , Plantas Geneticamente Modificadas , Vírus da Raiva/genética , Vírus da Raiva/imunologia , Proteínas Virais/biossíntese , Proteínas Virais/genética , Zea mays/genética , Zea mays/virologia
15.
Plant Cell Rep ; 27(4): 677-85, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18270708

RESUMO

Transgenic plants have been employed successfully as a low-cost system for the production of therapeutically valuable proteins including antibodies, antigens and hormones. Here, we report expression of a full-length nucleoprotein gene of rabies virus in transgenic tomato plants. The nucleoprotein was also transiently expressed in Nicotiana benthamiana plants by agroinfiltration. In both cases, the nucleoprotein was expressed at high levels, 1-5% of total soluble protein in tomato and 45% in N. benthamiana. Previously, only epitopes of the nucleoprotein had been expressed in plants. The presence and expression of the transgene was verified by PCR, Southern, northern and western blots. Mice were immunized both intraperitoneally (i.p.) and orally with tomato protein extracts containing the N protein induced the production of antibodies. The antibody titer of mice immunized i.p., was at least four times higher than that of mice immunized orally. These results were reflected in the challenge experiments where i.p.-immunized mice were partially protected against a peripheral virus challenge whereas orally immunized mice were not. This protection was comparable to that obtained in previous experiments employing different expression systems. Work is in progress to express both G and N proteins in transgenic plants and evaluate protection in mice.


Assuntos
/metabolismo , Nucleoproteínas/biossíntese , Nucleoproteínas/imunologia , Vírus da Raiva/fisiologia , Solanum lycopersicum/metabolismo , Proteínas Virais/biossíntese , Proteínas Virais/imunologia , Animais , Formação de Anticorpos , Antígenos Virais/biossíntese , Antígenos Virais/genética , Antígenos Virais/imunologia , Imunização , Camundongos , Nucleoproteínas/genética , Plantas Geneticamente Modificadas/metabolismo , Raiva/imunologia , Raiva/prevenção & controle , Vírus da Raiva/imunologia , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Virais/genética
16.
Med Microbiol Immunol ; 197(1): 21-7, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17611773

RESUMO

A cell line of Madin-Darby canine kidney (MDCK) cells persistently infected with human influenza A virus has been established and designated as MDCK-IVpi cells. Production of progeny virus in MDCK-IVpi cells was suppressed when the cells were incubated in the presence of 10% fetal calf serum (FCS). FCS impaired virus mRNA synthesis in MDCK-IVpi cells, which resulted in a scarcity of virus proteins for virion formation. However, MDCK-IVpi cells well supported the growth of superinfecting heterologous influenza viruses, even in the presence of FCS. A certain fetuin-like substance in FCS might be responsible for the observed inhibition of virus replication.


Assuntos
Regulação Viral da Expressão Gênica/efeitos dos fármacos , Genoma Viral , Vírus da Influenza A/fisiologia , Rim/virologia , Replicação Viral , Animais , Antivirais/farmacologia , Bovinos , Células Cultivadas , Cães , Sangue Fetal/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/biossíntese , Microscopia de Fluorescência , Proteínas do Nucleocapsídeo , Nucleoproteínas/biossíntese , RNA Mensageiro/biossíntese , Proteínas de Ligação a RNA/biossíntese , Proteínas do Core Viral/biossíntese , Proteínas da Matriz Viral/biossíntese , Ensaio de Placa Viral , alfa-Fetoproteínas/farmacologia
17.
Antiviral Res ; 76(2): 186-93, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17719657

RESUMO

RNA interference (RNAi) is a powerful tool to silence gene expression. Small interfering RNA (siRNA)-induced RNA degradation has been recently used as an antivirus agent to inhibit specific virus replication. Here, we showed that several siRNAs specific for conserved regions of influenza virus matrix (M2) and nucleocapsid protein (NP) genes could effectively inhibit expression of the corresponding viral protein. We also evaluated the antiviral potential of these siRNAs targeting M2 and NP of H5N1 avian influenza virus (AIV), which are essential to viral replication. We investigated the inhibitory effect of M2-specific siRNAs and NP-specific siRNAs on influenza A virus (H5N1, H1N1 and H9N2) replication in Madin-Darby canine kidney (MDCK) cells and BALB/c mice. The results showed that treatment with these siRNAs could specifically inhibit influenza A virus replication in MDCK cells (0.51-1.63 TCID(50) reduction in virus titers), and delivery of pS-M48 and pS-NP1383 significantly reduced lung virus titers in the infected mice (16-50-fold reduction in lung virus titers) and partially protected the mice from lethal influenza virus challenge (a survival rate of 4/8 for H1N1 virus-infected mice and 2/8 for H5N1 virus infected mice). Moreover, the treatment of pS-M48 and pS-NP1383 could suppress replication of different subtypes of influenza A viruses, including a H5N1 highly pathogenic avian isolate strain. The results provided a basis for further development of siRNA for prophylaxis and therapy of influenza virus infection in humans and animals.


Assuntos
Vírus da Influenza A Subtipo H1N1/genética , Virus da Influenza A Subtipo H5N1/genética , Vírus da Influenza A Subtipo H9N2/genética , Infecções por Orthomyxoviridae/terapia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico , Animais , Peso Corporal , Linhagem Celular , Cães , Feminino , Terapia Genética/métodos , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Nucleocapsídeo , Nucleoproteínas/biossíntese , Nucleoproteínas/genética , Infecções por Orthomyxoviridae/genética , Proteínas de Ligação a RNA/biossíntese , Proteínas de Ligação a RNA/genética , Análise de Sobrevida , Proteínas do Core Viral/biossíntese , Proteínas do Core Viral/genética , Proteínas da Matriz Viral/biossíntese , Proteínas da Matriz Viral/genética
18.
Virus Genes ; 35(3): 521-9, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17619134

RESUMO

In Europe, three genotypes of the genus Lyssavirus, family Rhabdoviridae, are present, classical rabies virus (RABV, genotype 1), European bat lyssavirus type 1 (EBLV-1, genotype 5) and European bat lyssavirus type 2 (EBLV-2, genotype 6). The entire authentic nucleoprotein (N protein) encoding sequences of RABV (challenge virus standard, CVS, strain), EBLV-1 and EBLV-2 were expressed in yeast Saccharomyces cerevisiae at high level. Purification of recombinant N proteins by caesium chloride gradient centrifugation resulted in yields between 14-17, 25-29 and 18-20 mg/l of induced yeast culture for RABV-CVS, EBLV-1 and EBLV-2, respectively. The purified N proteins were evaluated by negative staining electron microscopy, which revealed the formation of nucleocapsid-like structures. The antigenic conformation of the N proteins was investigated for their reactivity with monoclonal antibodies (mAbs) directed against different lyssaviruses. The reactivity pattern of each mAb was virtually identical between immunofluorescence assay with virus-infected cells, and ELISA and dot blot assay using the corresponding recombinant N proteins. These observations lead us to conclude that yeast-expressed lyssavirus N proteins share antigenic properties with naturally expressed virus protein. These recombinant proteins have the potential for use as components of serological assays for lyssaviruses.


Assuntos
Antígenos Virais/imunologia , Lyssavirus/genética , Lyssavirus/imunologia , Nucleoproteínas/imunologia , Saccharomyces cerevisiae/metabolismo , Proteínas Virais/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/metabolismo , Anticorpos Antivirais/isolamento & purificação , Anticorpos Antivirais/metabolismo , Antígenos Virais/biossíntese , Antígenos Virais/isolamento & purificação , Clonagem Molecular , Ensaio de Imunoadsorção Enzimática , Técnica Direta de Fluorescência para Anticorpo , Expressão Gênica , Immunoblotting , Camundongos , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Nucleoproteínas/biossíntese , Nucleoproteínas/isolamento & purificação , Ligação Proteica , Saccharomyces cerevisiae/genética , Alinhamento de Sequência , Proteínas Virais/biossíntese , Proteínas Virais/isolamento & purificação , Virossomos/ultraestrutura
19.
Clin Vaccine Immunol ; 14(9): 1182-9, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17634509

RESUMO

Diagnostic systems for Lassa fever (LF), a viral hemorrhagic fever caused by Lassa virus (LASV), such as enzyme immunoassays for the detection of LASV antibodies and LASV antigens, were developed using the recombinant nucleoprotein (rNP) of LASV (LASV-rNP). The LASV-rNP was expressed in a recombinant baculovirus system. LASV-rNP was used as an antigen in the detection of LASV-antibodies and as an immunogen for the production of monoclonal antibodies. The LASV-rNP was also expressed in HeLa cells by transfection with the expression vector encoding cDNA of the LASV-NP gene. An immunoglobulin G enzyme-linked immunosorbent assay (ELISA) using LASV-rNP and an indirect immunofluorescence assay using LASV-rNP-expressing HeLa cells were confirmed to have high sensitivity and specificity in the detection of LASV-antibodies. A novel monoclonal antibody to LASV-rNP, monoclonal antibody 4A5, was established. A sandwich antigen capture (Ag-capture) ELISA using the monoclonal antibody and an anti-LASV-rNP rabbit serum as capture and detection antibodies, respectively, was then developed. Authentic LASV nucleoprotein in serum samples collected from hamsters experimentally infected with LASV was detected by the Ag-capture ELISA. The Ag-capture ELISA specifically detected LASV-rNP but not the rNPs of lymphocytic choriomeningitis virus or Junin virus. The sensitivity of the Ag-capture ELISA in detecting LASV antigens was comparable to that of reverse transcription-PCR in detecting LASV RNA. These LASV rNP-based diagnostics were confirmed to be useful in the diagnosis of LF even in institutes without a high containment laboratory, since the antigens can be prepared without manipulation of the infectious viruses.


Assuntos
Febre Lassa/diagnóstico , Nucleoproteínas/imunologia , Proteínas Virais/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/genética , Anticorpos Antivirais/imunologia , Antígenos Virais/biossíntese , Antígenos Virais/genética , Antígenos Virais/imunologia , Baculoviridae/genética , Cricetinae , Ensaio de Imunoadsorção Enzimática/métodos , Epitopos de Linfócito B/imunologia , Técnica Indireta de Fluorescência para Anticorpo/métodos , Haplorrinos , Células HeLa , Humanos , Imunoglobulina G/imunologia , Insetos , Febre Lassa/genética , Febre Lassa/imunologia , Febre Lassa/virologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Nucleoproteínas/biossíntese , Nucleoproteínas/genética , Coelhos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Sensibilidade e Especificidade , Proteínas Virais/biossíntese , Proteínas Virais/genética
20.
J Gen Virol ; 88(Pt 8): 2280-2290, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17622633

RESUMO

The matrix (M1) protein of influenza A virus is a multifunctional protein that plays essential structural and functional roles in the virus life cycle. It drives virus budding and is the major protein component of the virion, where it forms an intermediate layer between the viral envelope and integral membrane proteins and the genomic ribonucleoproteins (RNPs). It also helps to control the intracellular trafficking of RNPs. These roles are mediated primarily via protein-protein interactions with viral and possibly cellular proteins. Here, the regions of M1 involved in binding the viral RNPs and in mediating homo-oligomerization are identified. In vitro, by using recombinant proteins, it was found that the middle domain of M1 was responsible for binding NP and that this interaction did not require RNA. Similarly, only M1 polypeptides containing the middle domain were able to bind to RNP-M1 complexes isolated from purified virus. When M1 self-association was examined, all three domains of the protein participated in homo-oligomerization although, again, the middle domain was dominant and self-associated efficiently in the absence of the N- and C-terminal domains. However, when the individual fragments of M1 were tagged with green fluorescent protein and expressed in virus-infected cells, microscopy of filamentous particles showed that only full-length M1 was incorporated into budding virions. It is concluded that the middle domain of M1 is primarily responsible for binding NP and self-association, but that additional interactions are required for efficient incorporation of M1 into virus particles.


Assuntos
Vírus da Influenza A/fisiologia , Nucleoproteínas/metabolismo , Estrutura Terciária de Proteína/fisiologia , Proteínas da Matriz Viral/metabolismo , Vírion/metabolismo , Animais , Linhagem Celular , Dimerização , Cães , Escherichia coli/metabolismo , Proteínas de Membrana/metabolismo , Nucleoproteínas/biossíntese , Nucleoproteínas/genética , Peptídeos/metabolismo , Ligação Proteica , Proteínas Recombinantes/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas Virais de Fusão/biossíntese , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo , Proteínas da Matriz Viral/biossíntese , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/genética , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...