Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.126
Filtrar
1.
Sci Rep ; 14(1): 20924, 2024 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-39251790

RESUMO

Human herpes viruses (HHV) are ubiquitous and have been implicated in numerous long-term health conditions. Since the association between viral exposure and long-term health impacts is partially influenced by variation in human leukocyte antigen (HLA) genes, we evaluated in silico the binding affinities of 9 HHV envelope glycoproteins with 127 common HLA Class I and Class II molecules. The findings show substantial variability in HHV binding affinity across viruses, HLA Class, HLA genes, and HLA alleles. Specific findings were as follows: (1) the predicted binding affinities of HHVs were characterized by four distinct groupings-[HHV1, HHV2], [HHV3, HHV4, HHV5], [HHV6A], [HHV6B, HHV7, HHV8]-with relatively lower binding affinities for HHV1, HHV2, and HHV6a compared to other HHVs; (2) significantly higher binding affinity was found for HLA Class I relative to Class II; (3) analyses within each class demonstrated that alleles of the C gene (for Class I) and DRB1 gene (for Class II) had the highest binding affinities; and (4) for each virus, predicted binding affinity to specific alleles varied, with HHV6a having the lowest affinity for HHV-HLA complexes, and HHV3, HHV4, and HHV5 having the highest. Since HLA-antigen binding is the first step in initiating an immune response to foreign antigens, these relative differences in HHV binding affinities are likely to influence long-term health impacts such that the cells infected with viruses associated with higher binding affinities across common HLA alleles may be more reduced in numbers, thereby lowering the potential for long-term sequelae of their infections.


Assuntos
Alelos , Proteínas do Envelope Viral , Humanos , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/metabolismo , Herpesviridae/imunologia , Herpesviridae/genética , Antígenos HLA/genética , Antígenos HLA/imunologia , Ligação Proteica , Imunogenética , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia
2.
J Infect Dev Ctries ; 18(8): 1281-1290, 2024 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-39288397

RESUMO

INTRODUCTION: Rabies is a fatal infectious disease, that poses a major public health threat in developing countries. With an annual death toll of approximately 59,000, more than half of which are children, an urgent need exists for a safe, affordable, and effective preventive measure against rabies virus infection. METHODOLOGY: A recombinant rabies vaccine called Ad5-dRVG was constructed by introducing two copies of the rabies virus glycoprotein into a human adenoviral vector. Virus-neutralizing assays and virus challenge experiments were employed to evaluate the Ad5-dRVG vaccine. RESULTS: Our findings demonstrate that a single dose of Ad5-dRVG, administered either intramuscularly or orally, elicited significantly stronger immune responses than Ad5-RVG. Moreover, both vaccines provided complete protection in mice. Notably, the vaccine exhibited remarkable efficacy even at low doses, suggesting potential cost reduction in production. CONCLUSIONS: The development of the Ad5-dRVG recombinant rabies vaccine represents a significant advancement in rabies prevention. Its enhanced immunogenicity, demonstrated efficacy and potential cost savings make it a promising candidate for widespread use.


Assuntos
Vetores Genéticos , Glicoproteínas , Vacina Antirrábica , Vírus da Raiva , Raiva , Vacinas Sintéticas , Animais , Vacina Antirrábica/imunologia , Vacina Antirrábica/genética , Vacina Antirrábica/administração & dosagem , Raiva/prevenção & controle , Raiva/imunologia , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/administração & dosagem , Glicoproteínas/imunologia , Glicoproteínas/genética , Camundongos , Vírus da Raiva/imunologia , Vírus da Raiva/genética , Feminino , Anticorpos Antivirais/sangue , Adenoviridae/genética , Camundongos Endogâmicos BALB C , Injeções Intramusculares , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Humanos , Modelos Animais de Doenças , Administração Oral , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , Eficácia de Vacinas
3.
Nature ; 633(8030): 695-703, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39232167

RESUMO

Viral glycoproteins drive membrane fusion in enveloped viruses and determine host range, tissue tropism and pathogenesis1. Despite their importance, there is a fragmentary understanding of glycoproteins within the Flaviviridae2, a large virus family that include pathogens such as hepatitis C, dengue and Zika viruses, and numerous other human, animal and emergent viruses. For many flaviviruses the glycoproteins have not yet been identified, for others, such as the hepaciviruses, the molecular mechanisms of membrane fusion remain uncharacterized3. Here we combine phylogenetic analyses with protein structure prediction to survey glycoproteins across the entire Flaviviridae. We find class II fusion systems, homologous to the Orthoflavivirus E glycoprotein in most species, including highly divergent jingmenviruses and large genome flaviviruses. However, the E1E2 glycoproteins of the hepaciviruses, pegiviruses and pestiviruses are structurally distinct, may represent a novel class of fusion mechanism, and are strictly associated with infection of vertebrate hosts. By mapping glycoprotein distribution onto the underlying phylogeny, we reveal a complex evolutionary history marked by the capture of bacterial genes and potentially inter-genus recombination. These insights, made possible through protein structure prediction, refine our understanding of viral fusion mechanisms and reveal the events that have shaped the diverse virology and ecology of the Flaviviridae.


Assuntos
Evolução Molecular , Flaviviridae , Glicoproteínas , Modelos Moleculares , Filogenia , Proteínas do Envelope Viral , Flaviviridae/genética , Flaviviridae/classificação , Glicoproteínas/química , Glicoproteínas/metabolismo , Glicoproteínas/genética , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Animais , Humanos
4.
Proc Natl Acad Sci U S A ; 121(39): e2408078121, 2024 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-39292744

RESUMO

The Pentamer complex of Human Cytomegalovirus (HCMV) consists of the viral glycoproteins gH, gL, UL128, UL130, and UL131 and is incorporated into infectious virions. HCMV strains propagated extensively in vitro in fibroblasts carry UL128, UL130, or UL131 alleles that do not make a functional complex and thus lack Pentamer function. Adding functional Pentamer to such strains decreases virus growth in fibroblasts. Here, we show that the Pentamer inhibits productive HCMV replication in fibroblasts by repressing viral Immediate Early (IE) transcription. We show that ectopic expression of the viral IE1 protein, a target of Pentamer-mediated transcriptional repression, complements the growth defect of a Pentamer-positive virus. Furthermore, we show that the Pentamer also represses viral IE transcription in cell types where HCMV in vitro latency is studied. Finally, we identify UL130 as a functional subunit of the Pentamer for IE transcriptional repression and demonstrate that cyclic AMP Response Element (CRE) and NFkB sites within the Major Immediate Early Promoter that drives IE1 transcription contribute to this repression. We conclude that the HCMV Pentamer represses viral IE transcription.


Assuntos
Infecções por Citomegalovirus , Citomegalovirus , Fibroblastos , Proteínas Imediatamente Precoces , Transcrição Gênica , Proteínas do Envelope Viral , Humanos , Citomegalovirus/genética , Citomegalovirus/fisiologia , Citomegalovirus/metabolismo , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Proteínas Imediatamente Precoces/genética , Fibroblastos/virologia , Fibroblastos/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/genética , Regulação Viral da Expressão Gênica , Replicação Viral/genética , Glicoproteínas/metabolismo , Glicoproteínas/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Proteínas Virais/metabolismo , Proteínas Virais/genética , Genes Precoces , Regiões Promotoras Genéticas
5.
Front Cell Infect Microbiol ; 14: 1445115, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39220282

RESUMO

The human respiratory syncytial virus (RSV) is a significant health concern, particularly for infants, young children, and the elderly. This virus is known to evolve continuously due to environmental factors and herd immunity. In light of this, our study aimed to analyze the genetic variability of the G protein in RSV-A and RSV-B genotypes in Kuwait from 2020 to 2022. Between January 2020 and September 2022, we collected 490 respiratory samples from hospitalized patients with acute respiratory tract infections. These samples were tested and confirmed positive for RSV using multiplex Real-Time PCR. Subsequently, the samples underwent nucleic acid sequencing using the advanced Nanopore sequencing technology to analyze the full-length G gene. Sequence analysis showed that 64 isolates (76%) were RSV-A, and 20 isolates (24%) were RSV-B. The G genes of RSV-A belonged to genotype GA2.3.5, while all the RSV-B genotypes belonged to GB5.0.5a. New lineages and sub-lineages of RSV-A and RSV-B were detected, indicating the circulation of new strains in Kuwait. Many unique and new amino acid changes, including insertions, were found in the G proteins of Kuwaiti isolates, with the highest variability in the second hypervariable region. An increased number of N and O-linked glycosylation sites were also identified in the G protein, which could speculate to alter the antigenicity of RSV. The identified changes in the G protein of RSV-A and RSV-B genotypes might result from immune pressure and could affect the antigenic characteristics of circulating strains in Kuwait. This could potentially lead to new RSV variants that can evade the immune response. Our in-depth analysis of the G proteins of both RSV-A and RSV-B could aid in the development of more potent treatments and vaccines.


Assuntos
Variação Genética , Genótipo , Filogenia , Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano , Humanos , Kuweit , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/classificação , Vírus Sincicial Respiratório Humano/isolamento & purificação , Vírus Sincicial Respiratório Humano/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Glicosilação , Lactente , Pré-Escolar , Feminino , Masculino , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia , Adulto , Criança , Pessoa de Meia-Idade , Idoso , Adulto Jovem , Adolescente , Infecções Respiratórias/virologia , Proteínas do Envelope Viral/genética
6.
PLoS Pathog ; 20(8): e1012468, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39146367

RESUMO

Genetic editing of the germline using CRISPR/Cas9 technology has made it possible to alter livestock traits, including the creation of resistance to viral diseases. However, virus adaptability could present a major obstacle in this effort. Recently, chickens resistant to avian leukosis virus subgroup J (ALV-J) were developed by deleting a single amino acid, W38, within the ALV-J receptor NHE1 using CRISPR/Cas9 genome editing. This resistance was confirmed both in vitro and in vivo. In vitro resistance of W38-/- chicken embryonic fibroblasts to all tested ALV-J strains was shown. To investigate the capacity of ALV-J for further adaptation, we used a retrovirus reporter-based assay to select adapted ALV-J variants. We assumed that adaptive mutations overcoming the cellular resistance would occur within the envelope protein. In accordance with this assumption, we isolated and sequenced numerous adapted virus variants and found within their envelope genes eight independent single nucleotide substitutions. To confirm the adaptive capacity of these substitutions, we introduced them into the original retrovirus reporter. All eight variants replicated effectively in W38-/- chicken embryonic fibroblasts in vitro while in vivo, W38-/- chickens were sensitive to tumor induction by two of the variants. Importantly, receptor alleles with more extensive modifications have remained resistant to the virus. These results demonstrate an important strategy in livestock genome engineering towards antivirus resistance and illustrate that cellular resistance induced by minor receptor modifications can be overcome by adapted virus variants. We conclude that more complex editing will be necessary to attain robust resistance.


Assuntos
Vírus da Leucose Aviária , Leucose Aviária , Galinhas , Animais , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/fisiologia , Galinhas/virologia , Leucose Aviária/virologia , Leucose Aviária/genética , Doenças das Aves Domésticas/virologia , Doenças das Aves Domésticas/genética , Resistência à Doença/genética , Sistemas CRISPR-Cas , Edição de Genes , Embrião de Galinha , Evolução Molecular , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Fibroblastos/virologia , Fibroblastos/metabolismo
7.
Emerg Microbes Infect ; 13(1): 2392651, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39155772

RESUMO

Ebola disease is a lethal viral hemorrhagic fever caused by ebolaviruses within the Filoviridae family with mortality rates of up to 90%. Monoclonal antibody (mAb) based therapies have shown great potential for the treatment of EVD. However, the potential emerging ebolavirus isolates and the negative effect of decoy protein on the therapeutic efficacy of antibodies highlight the necessity of developing novel antibodies to counter the threat of Ebola. Here, 11 fully human mAbs were isolated from transgenic mice immunized with GP protein and recombinant vesicular stomatitis virus-bearing GP (rVSV-EBOV GP). These mAbs were divided into five groups according to their germline genes and exhibited differential binding activities and neutralization capabilities. In particular, mAbs 8G6, 2A4, and 5H4 were cross-reactive and bound at least three ebolavirus glycoproteins. mAb 4C1 not only exhibited neutralizing activity but no cross-reaction with sGP. mAb 7D8 exhibited the strongest neutralizing capacity. Further analysis on the critical residues for the bindings of 4C1 and 8G6 to GPs was conducted using antibodies complementarity-determining regions (CDRs) alanine scanning. It has been shown that light chain CDR3 played a crucial role in binding and neutralization and that any mutation in CDRs could not improve the binding of 4C1 to sGP. Importantly, mAbs 7D8, 8G6, and 4C1 provided complete protections against EBOV infection in a hamster lethal challenge model when administered 12 h post-infection. These results support mAbs 7D8, 8G6, and 4C1 as potent antibody candidates for further investigations and pave the way for further developments of therapies and vaccines.


Assuntos
Anticorpos Monoclonais , Anticorpos Neutralizantes , Anticorpos Antivirais , Modelos Animais de Doenças , Ebolavirus , Doença pelo Vírus Ebola , Animais , Ebolavirus/imunologia , Ebolavirus/genética , Anticorpos Monoclonais/imunologia , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Doença pelo Vírus Ebola/virologia , Anticorpos Antivirais/imunologia , Cricetinae , Camundongos , Anticorpos Neutralizantes/imunologia , Humanos , Camundongos Transgênicos , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , Reações Cruzadas
8.
BMC Biotechnol ; 24(1): 59, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39192233

RESUMO

BACKGROUND: Crimean-Congo hemorrhagic fever (CCHF) is a tick-borne zoonotic disease that presents with severe hemorrhagic manifestations and is associated with significant fatality rates. The causative agent, Crimean-Congo Hemorrhagic Fever Virus (CCHFV), is a high-priority pathogen identified by the World Health Organization with no approved vaccine or specific treatment available. In addition, there is a critical need for enhanced diagnostic tools to improve public health awareness, prevention measures, and disease control strategies. METHODS: We designed plasmids to enable the purification of soluble CCHFV glycoprotein Gc expressed in mammalian 293 F cells, followed by purification using affinity and size exclusion chromatography. The purified antigen was analyzed by SDS-PAGE and Western blotting to confirm its reactivity to antibodies from CCHF survivors. Additionally, an in-house indirect ELISA was developed using the purified Gc as a coating antigen. RESULTS: The optimized expression system successfully produced soluble and pure Gc antigen after affinity chromatography. The protein showed specific reactivity with CCHFV-positive serum antibodies in Western blot analysis. The indirect ELISA assay demonstrated high efficacy in distinguishing between CCHFV-positive and -negative serum samples, indicating its potential as a valuable diagnostic tool. Size exclusion chromatography further confirmed the presence of aggregates in our protein preparation. CONCLUSIONS: The purified Gc antigen shows promise for developing direct diagnostic assays for CCHFV. The antigen's suitability for subunit vaccine development and its application as bait for monoclonal antibody isolation from survivors could be investigated further. This work lays the foundation for future research into the development of rapid diagnostic tests for field deployment.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo , Proteínas Recombinantes , Humanos , Vírus da Febre Hemorrágica da Crimeia-Congo/imunologia , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação , Células HEK293 , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/isolamento & purificação , Febre Hemorrágica da Crimeia/diagnóstico , Febre Hemorrágica da Crimeia/virologia , Ensaio de Imunoadsorção Enzimática , Animais , Cromatografia de Afinidade/métodos , Cromatografia em Gel , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue
9.
Viruses ; 16(8)2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-39205190

RESUMO

Transduction of producer cells during lentiviral vector (LVV) production causes the loss of 70-90% of viable particles. This process is called retro-transduction and it is a consequence of the interaction between the LVV envelope protein, VSV-G, and the LDL receptor located on the producer cell membrane, allowing lentiviral vector transduction. Avoiding retro-transduction in LVV manufacturing is crucial to improve net production and, therefore, the efficiency of the production process. Here, we describe a method for quantifying the transduction of producer cells and three different strategies that, focused on the interaction between VSV-G and the LDLR, aim to reduce retro-transduction.


Assuntos
Vetores Genéticos , Lentivirus , Receptores de LDL , Transdução Genética , Vetores Genéticos/genética , Lentivirus/genética , Humanos , Receptores de LDL/metabolismo , Receptores de LDL/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Células HEK293 , Glicoproteínas de Membrana
10.
Viruses ; 16(8)2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39205211

RESUMO

The West Nile virus (WNV) subtype Kunjin virus (WNVKUN) is endemic to Australia. Here, we characterized the classical WNVKUN strain, OR393. The original OR393 strain contained two types of viruses: small plaque-forming virus (SP) and large plaque-forming virus (LP). The amino acid residues at positions 156 and 332 in the E protein (E156 and E332) of SP were Ser and Lys (E156S/332K), respectively, whereas those in LP were Phe and Thr (E156F/332T). SP grew slightly faster than LP in vitro. The E protein of SP was N-glycosylated, whereas that of LP was not. Analysis using two recombinant single-mutant LP viruses, rKUNV-LP-EF156S and rKUNV-LP-ET332K, indicated that E156S enlarged plaques formed by LP, but E332K potently reduced them, regardless of the amino acid at E156. rKUNV-LP-EF156S showed significantly higher neuroinvasive ability than LP, SP, and rKUNV-LP-ET332K. Our results indicate that the low-pathogenic classical WNVKUN can easily change its pathogenicity through only a few amino acid substitutions in the E protein. It was also found that Phe at E156 of the rKUNV-LP-ET332K was easily changed to Ser during replication in vitro and in vivo, suggesting that E156S is advantageous for the propagation of WNVKUN in mammalian cells.


Assuntos
Proteínas do Envelope Viral , Ensaio de Placa Viral , Febre do Nilo Ocidental , Vírus do Nilo Ocidental , Animais , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/patogenicidade , Vírus do Nilo Ocidental/fisiologia , Camundongos , Febre do Nilo Ocidental/virologia , Virulência , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/química , Aminoácidos/metabolismo , Aminoácidos/genética , Replicação Viral , Chlorocebus aethiops , Substituição de Aminoácidos , Células Vero , Feminino , Humanos , Austrália , Linhagem Celular
11.
Int J Mol Sci ; 25(16)2024 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-39201420

RESUMO

Multivalent live-attenuated or inactivated vaccines are often used to control the bovine viral diarrhea disease (BVD). Still, they retain inherent disadvantages and do not provide the expected protection. This study developed a new vaccine prototype, including the external segment of the E2 viral protein from five different subgenotypes selected after a massive screening. The E2 proteins of every subgenotype (1aE2, 1bE2, 1cE2, 1dE2, and 1eE2) were produced in mammalian cells and purified by IMAC. An equimolar mixture of E2 proteins formulated in an oil-in-water adjuvant made up the vaccine candidate, inducing a high humoral response at 50, 100, and 150 µg doses in sheep. A similar immune response was observed in bovines at 50 µg. The cellular response showed a significant increase in the transcript levels of relevant Th1 cytokines, while those corresponding to the Th2 cytokine IL-4 and the negative control were similar. High levels of neutralizing antibodies against the subgenotype BVDV1a demonstrated the effectiveness of our vaccine candidate, similar to that observed in the sera of animals vaccinated with the commercial vaccine. These results suggest that our vaccine prototype could become an effective recombinant vaccine against the BVD.


Assuntos
Anticorpos Antivirais , Doença das Mucosas por Vírus da Diarreia Viral Bovina , Vacinas de Subunidades Antigênicas , Vacinas Sintéticas , Vacinas Virais , Animais , Bovinos , Vacinas Virais/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Vacinas Sintéticas/imunologia , Doença das Mucosas por Vírus da Diarreia Viral Bovina/prevenção & controle , Doença das Mucosas por Vírus da Diarreia Viral Bovina/imunologia , Doença das Mucosas por Vírus da Diarreia Viral Bovina/virologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Ovinos , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , Citocinas/metabolismo , Vírus da Diarreia Viral Bovina/imunologia , Vírus da Diarreia Viral Bovina/genética , Vírus da Diarreia Viral Bovina Tipo 1/imunologia , Vírus da Diarreia Viral Bovina Tipo 1/genética
12.
Int J Mol Sci ; 25(16)2024 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-39201685

RESUMO

An efficient gene transfer and expression tool is lacking for shrimps and shrimp cells. To solve this, this study has developed a shrimp DNA virus-mediated gene transfer and expression system, consisting of insect Sf9 cells for viral packaging, the shrimp viral vector of pUC19-IHHNV-PH-GUS and the baculoviral vector of Bacmid or Bacmid-VP28 encoding the shrimp WSSV envelope protein VP28. The pUC19-IHHNV-PH-GUS vector was constructed by assembling the genomic DNA of shrimp infectious hypodermal and hematopoietic necrosis virus (IHHNV), which has shortened inverted terminal repeats, into a pUC19 backbone, and then an expression cassette of baculoviral polyhedron (PH) promoter-driven GUS (ß-glucuronidase) reporter gene was inserted immediately downstream of IHHNV for proof-of-concept. It was found that the viral vector of pUC19-IHHNV-PH-GUS could be successfully packaged into IHHNV-like infective virions in the Sf9 cells, and the gene transfer efficiency of this system was evaluated and verified in three systems of Sf9 cells, shrimp hemolymph cells and tissues of infected shrimps, but the GUS expression could only be detected in cases where the viral vector was co-transfected or co-infected with a baculovirus of Bacmid or Bacmid-VP28 due to the Bacmid-dependence of the PH promoter. Moreover, the packaging and infection efficiencies could be significantly improved when Bacmid-VP28 was used instead of Bacmid.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos , Penaeidae , Animais , Penaeidae/virologia , Penaeidae/genética , Células Sf9 , Vetores Genéticos/genética , Baculoviridae/genética , Regiões Promotoras Genéticas , Spodoptera/virologia , Densovirinae/genética , Expressão Gênica , Vírus da Síndrome da Mancha Branca 1/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Glucuronidase/genética , Glucuronidase/metabolismo
13.
Viruses ; 16(8)2024 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-39205163

RESUMO

Pseudorabies virus (PRV) is one of the herpes viruses that can infect a wide range of animals including pigs, cattle, sheep, mice, and wild animals. PRV is a neurotropic alphaherpesvirus capable of infecting a variety of mammals. There is a rising interest in the targeted application of probiotic bacteria to prevent viral diseases, including PRV. In this study, the surface expression of enhanced green fluorescent protein (EGFP) on recombinant Lactiplantibacillus plantarum NC8 (rNC8) through the LP3065 LPxTG motif of Lactobacillus plantarum WCFS1 was generated. The surface expression was observed through confocal microscopy. Dendritic cell targeting peptides (DCpep) were also fused with LPxTG that help to bind with mouse DCs. The PRV-gD was cloned in LP3065 LPxTG, resulting in the generation of rNC8-LP3065-gD. Inactivated rNC8-LP3065-gD was administered intravenously in mice on days 1 and 7 at a dose of 200 µL (109 CFU/mouse) for monitoring immunogenicity. Subsequently, a challenge dose of PRV TJ (104 TCID50) was administered intramuscularly at 14 days post-immunization. The survival rate of the immunized mice reached 80% (4/5) with no significant signs of illness. A significant rise in anti-gD antibodies was detected in the immunized mice by ELISA. Quantitative PCR (qPCR) results showed decreased viral loading in different body tissues. Flow cytometry of lymphocytes derived from mice spleen indicated an increase in CD3+CD4+ T cells, but CD3+CD8+ T cells were not detected. Moreover, it offers a model to delineate immune correlates with rNC8-induced immunity against swine viral diseases.


Assuntos
Herpesvirus Suídeo 1 , Pseudorraiva , Animais , Herpesvirus Suídeo 1/imunologia , Herpesvirus Suídeo 1/genética , Camundongos , Pseudorraiva/prevenção & controle , Pseudorraiva/imunologia , Pseudorraiva/virologia , Feminino , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Lactobacillus plantarum/genética , Lactobacillus plantarum/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , Camundongos Endogâmicos BALB C , Vacinas contra Pseudorraiva/imunologia , Suínos , Proteínas de Fluorescência Verde/genética , Técnicas de Visualização da Superfície Celular
14.
Cell Stem Cell ; 31(9): 1298-1314.e8, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39146934

RESUMO

Endogenous retroviruses (ERVs) occupy a significant part of the human genome, with some encoding proteins that influence the immune system or regulate cell-cell fusion in early extra-embryonic development. However, whether ERV-derived proteins regulate somatic development is unknown. Here, we report a somatic developmental function for the primate-specific ERVH48-1 (SUPYN/Suppressyn). ERVH48-1 encodes a fragment of a viral envelope that is expressed during early embryonic development. Loss of ERVH48-1 led to impaired mesoderm and cardiomyocyte commitment and diverted cells to an ectoderm-like fate. Mechanistically, ERVH48-1 is localized to sub-cellular membrane compartments through a functional N-terminal signal peptide and binds to the WNT antagonist SFRP2 to promote its polyubiquitination and degradation, thus limiting SFRP2 secretion and blocking repression of WNT/ß-catenin signaling. Knockdown of SFRP2 or expression of a chimeric SFRP2 with the ERVH48-1 signal peptide rescued cardiomyocyte differentiation. This study demonstrates how ERVH48-1 modulates WNT/ß-catenin signaling and cell type commitment in somatic development.


Assuntos
Diferenciação Celular , Retrovirus Endógenos , Proteínas de Membrana , Miócitos Cardíacos , Via de Sinalização Wnt , Humanos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/citologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Retrovirus Endógenos/metabolismo , Retrovirus Endógenos/genética , Animais , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/genética , Primatas , Células HEK293 , Mesoderma/metabolismo
15.
Emerg Microbes Infect ; 13(1): 2392661, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39137287

RESUMO

mRNA platform holds promise for next-generation Varicella-zoster Virus (VZV) vaccine development due to its high potency at inducing strong T-cell response. Built upon the design of our 1st-generation VZV mRNA vaccine that encodes for full-length gE antigen, in this study we reported on a novel combinatorial strategy to further optimize the gE-encoding mRNA sequence through signal peptide replacement, C-terminal modification, and insertion of mRNA-stabilizing motif, which collectively contributed to significantly improved vaccine immunogenicity. In adult mice, aged mice, and immunocompromised mice, this optimized VZV mRNA vaccine showed strong superiority in multiple aspects including the induction of gE-specific antibodies, specific memory B-cell response, as well as Th1-type T-cell response.


Assuntos
Anticorpos Antivirais , Herpesvirus Humano 3 , Proteínas do Envelope Viral , Animais , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , Herpesvirus Humano 3/imunologia , Herpesvirus Humano 3/genética , Camundongos , Anticorpos Antivirais/imunologia , Humanos , Vacinas de mRNA , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Feminino , Desenvolvimento de Vacinas , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Imunogenicidade da Vacina , Vacina contra Varicela/imunologia , Vacina contra Varicela/administração & dosagem , Vacina contra Varicela/genética , Linfócitos B/imunologia , Camundongos Endogâmicos BALB C , Células Th1/imunologia
16.
J Virol ; 98(9): e0054024, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39162433

RESUMO

Systemic viral infection of insects typically begins with the primary infection of midgut epithelial cells (enterocytes) and subsequent transit of the progeny virus in an apical-to-basal orientation into the hemocoel. For insect-vectored viruses, an oppositely oriented process (basal-to-apical transit) occurs upon secondary infection of salivary glands and is necessary for virus transmission to non-insect hosts. To examine this inversely oriented virus transit in these polarized tissues, we assessed the intracellular trafficking of two model viral envelope proteins (baculovirus GP64 and vesicular stomatitis virus G) in the midgut and salivary gland cells of the model insect, Drosophila melanogaster. Using fly lines that inducibly express either GP64 or VSV G, we found that each protein, expressed alone, was trafficked basally in midgut enterocytes. In salivary gland cells, VSV G was trafficked apically in most but not all cells, whereas GP64 was consistently trafficked basally. We demonstrated that a YxxØ motif present in both proteins was critical for basal trafficking in midgut enterocytes but dispensable for trafficking in salivary gland cells. Using RNAi, we found that clathrin adaptor protein complexes AP-1 and AP-3, as well as seven Rab GTPases, were involved in polarized VSV G trafficking in midgut enterocytes. Our results indicate that these viral envelope proteins encode the requisite information and require no other viral factors for appropriately polarized trafficking. In addition, they exploit tissue-specific differences in protein trafficking pathways to facilitate virus egress in the appropriate orientation for establishing systemic infections and vectoring infection to other hosts. IMPORTANCE: Viruses that use insects as hosts must navigate specific routes through different insect tissues to complete their life cycles. The routes may differ substantially depending on the life cycle of the virus. Both insect pathogenic viruses and insect-vectored viruses must navigate through the polarized cells of the midgut epithelium to establish a systemic infection. In addition, insect-vectored viruses must also navigate through the polarized salivary gland epithelium for transmission. Thus, insect-vectored viruses appear to traffic in opposite directions in these two tissues. In this study, we asked whether two viral envelope proteins (VSV G and baculovirus GP64) alone encode the signals necessary for the polarized trafficking associated with their respective life cycles. Using Drosophila as a model to examine tissue-specific polarized trafficking of these viral envelope proteins, we identified one of the virus-encoded signals and several host proteins associated with regulating the polarized trafficking in the midgut epithelium.


Assuntos
Drosophila melanogaster , Transporte Proteico , Glândulas Salivares , Proteínas do Envelope Viral , Animais , Glândulas Salivares/virologia , Glândulas Salivares/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/genética , Drosophila melanogaster/virologia , Drosophila melanogaster/metabolismo , Insetos Vetores/virologia , Insetos Vetores/metabolismo , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Enterócitos/virologia , Enterócitos/metabolismo , Trato Gastrointestinal/virologia , Trato Gastrointestinal/metabolismo
17.
Emerg Microbes Infect ; 13(1): 2389115, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39129566

RESUMO

Rabies is a lethal zoonotic disease that threatens human health. As the only viral surface protein, the rabies virus (RABV) glycoprotein (G) induces main neutralizing antibody (Nab) responses; however, Nab titre is closely correlated with the conformation of G. Virus-like particles (VLP) formed by the co-expression of RABV G and matrix protein (M) improve retention and antigen presentation, inducing broad, durable immune responses. RABV nucleoprotein (N) can elicit humoral and cellular immune responses. Hence, we developed a series of nucleoside-modified RABV mRNA vaccines encoding wild-type G, soluble trimeric RABV G formed by an artificial trimer motif (tG-MTQ), membrane-anchored prefusion-stabilized G (preG). Furthermore, we also developed RABV VLP mRNA vaccine co-expressing preG and M to generate VLPs, and VLP/N mRNA vaccine co-expressing preG, M, and N. The RABV mRNA vaccines induced higher humoral and cellular responses than inactivated rabies vaccine, and completely protected mice against intracerebral challenge. Additionally, the IgG and Nab titres in RABV preG, VLP and VLP/N mRNA groups were significantly higher than those in G and tG-MTQ groups. A single administration of VLP or VLP/N mRNA vaccines elicited protective Nab responses, the Nab titres were significantly higher than that in inactivated rabies vaccine group at day 7. Moreover, RABV VLP and VLP/N mRNA vaccines showed superior capacities to elicit potent germinal centre, long-lived plasma cell and memory B cell responses, which linked to high titre and durable Nab responses. In summary, our data demonstrated that RABV VLP and VLP/N mRNA vaccines could be promising candidates against rabies.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Imunidade Celular , Imunidade Humoral , Vacina Antirrábica , Vírus da Raiva , Raiva , Vacinas de Partículas Semelhantes a Vírus , Animais , Vacina Antirrábica/imunologia , Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/genética , Raiva/prevenção & controle , Raiva/imunologia , Vírus da Raiva/imunologia , Vírus da Raiva/genética , Camundongos , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/genética , Feminino , Vacinas de mRNA/imunologia , Camundongos Endogâmicos BALB C , Nucleosídeos/imunologia , Glicoproteínas/imunologia , Glicoproteínas/genética , Humanos , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Proteínas da Matriz Viral/imunologia , Proteínas da Matriz Viral/genética , Antígenos Virais/imunologia , Antígenos Virais/genética , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , RNA Mensageiro/genética , RNA Mensageiro/imunologia
18.
Arch Virol ; 169(8): 172, 2024 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-39096433

RESUMO

Goatpox and sheeppox are highly contagious and economically important viral diseases of small ruminants. Due to the risk they pose to animal health, livestock production, and international trade, capripoxviruses are a considerable threat to the livestock economy. In this study, we expressed two core proteins (A4L and A12L) and one extracellular enveloped virion protein (A33R) of goatpox virus in a baculovirus expression vector system and evaluated their use as diagnostic antigens in ELISA. Full-length A4L, A12L, and A33R genes of the GTPV Uttarkashi strain were amplified, cloned into the pFastBac HT A donor vector, and introduced into DH10Bac cells containing a baculovirus shuttle vector plasmid to generate recombinant bacmids. The recombinant baculoviruses were produced in Sf-21 cells by transfection, and proteins were expressed in TN5 insect cells. The recombinant proteins were analysed by SDS-PAGE and confirmed by western blot, with expected sizes of ~30 kDa, ~31 kDa, and ~32 kDa for A4L, A12L, and A33R, respectively. The recombinant proteins were purified, and the immunoreactivity of the purified proteins was confirmed by western blot using anti-GTPV serum. The antigenic specificity of the expressed proteins as diagnostic antigens was evaluated by testing their reactivity with infected, vaccinated, and negative GTPV/SPPV serum in indirect ELISA, and the A33R-based indirect ELISA was optimized. The diagnostic sensitivity and specificity of the A33R-based indirect ELISA were found to be of 89% and 94% for goats and 98% and 91%, for sheep, respectively. No cross-reactivity was observed with other related viruses. The recombinant-A33R-based indirect ELISA developed in the present study shows that it has potential for the detection of antibodies in GTPV and SPPV infected/vaccinated animals.


Assuntos
Baculoviridae , Capripoxvirus , Ensaio de Imunoadsorção Enzimática , Doenças das Cabras , Cabras , Proteínas do Envelope Viral , Capripoxvirus/genética , Capripoxvirus/isolamento & purificação , Baculoviridae/genética , Animais , Doenças das Cabras/virologia , Doenças das Cabras/diagnóstico , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Cabras/virologia , Ensaio de Imunoadsorção Enzimática/métodos , Infecções por Poxviridae/diagnóstico , Infecções por Poxviridae/veterinária , Infecções por Poxviridae/virologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/imunologia , Vírion/genética , Proteínas do Core Viral/genética , Proteínas do Core Viral/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Células Sf9 , Antígenos Virais/genética , Antígenos Virais/imunologia , Linhagem Celular , Expressão Gênica
19.
Genes (Basel) ; 15(8)2024 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-39202352

RESUMO

Atypical porcine pestivirus (APPV) can cause congenital tremor type A-II in neonatal piglets, posing a significant threat to swine herd health globally. Our previous study demonstrated that the Mut domains, comprising 112 amino acids at the N-terminus, are the primary functional regions of the E2 protein of APPV. This study identified 14 host cellular proteins that exhibit potential interactions with the Mut domains of the E2 protein using yeast two-hybrid screening. Using bioinformatics analysis, we discovered that the Mut domains of the E2 protein might exert regulatory effects on apoptosis by modulating energy metabolism within the mitochondria. We also conducted co-immunoprecipitation, glutathione S-transferase pull-down, and immunofluorescence assays to confirm the interaction between the Mut domains of the E2 protein and cathepsin H and signal sequence receptor subunit 4 (SSR4). Ultimately, SSR4 enhanced APPV replication in vitro. In summary, our study successfully elucidated the interactions between the Mut domains of the E2 protein and host cell protein, predicted the potential pathways implicated in these interactions, and demonstrated SSR4 involvement in APPV infection. These significant findings contribute valuable knowledge toward a deeper understanding of APPV pathogenesis and the role of the Mut domains of the E2 protein in this intricate process.


Assuntos
Infecções por Pestivirus , Pestivirus , Animais , Pestivirus/genética , Pestivirus/metabolismo , Suínos , Infecções por Pestivirus/veterinária , Infecções por Pestivirus/virologia , Infecções por Pestivirus/genética , Doenças dos Suínos/virologia , Doenças dos Suínos/genética , Doenças dos Suínos/metabolismo , Interações Hospedeiro-Patógeno/genética , Domínios Proteicos , Replicação Viral/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Humanos , Mapas de Interação de Proteínas/genética
20.
Methods Mol Biol ; 2824: 121-133, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39039410

RESUMO

The Rift Valley fever virus (RVFV), transmitted through mosquito bites, leads to severe illness in humans and livestock throughout Africa and the Arabian Peninsula, causing significant morbidity and mortality. As of now, there are no verified and efficacious drugs or licensed vaccines accessible for the prevention or treatment of RVFV infections in both humans and livestock. The mature RVFV virion has two envelope proteins on its surface: glycoprotein N (GN) and glycoprotein C (GC). These proteins play a significant role in facilitating the virus's entry into the host cell, making them prominent targets for entry mechanism research as well as targets for drugs and vaccine development. The initial stage in obtaining atomic-resolution structural and mechanistic information on viral entry as well as developing biochemical and biophysical research tools involves recombinant protein production. In this chapter, we describe a simplified and scalable protocol facilitating the generation of high-quality, high-titer baculovirus virus for expression and purification of RVFV GC, utilizing the baculovirus-mediated expression system in insect cells.


Assuntos
Baculoviridae , Proteínas Recombinantes , Vírus da Febre do Vale do Rift , Proteínas do Envelope Viral , Baculoviridae/genética , Animais , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/isolamento & purificação , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Vírus da Febre do Vale do Rift/genética , Células Sf9 , Expressão Gênica , Humanos , Vetores Genéticos/genética , Clonagem Molecular/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...