Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20.842
Filtrar
1.
Int J Mol Sci ; 25(17)2024 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-39273370

RESUMO

Zika virus (ZIKV; family, Flaviviridae), which causes congenital Zika syndrome, Guillain-Barré Syndrome, and other severe diseases, is transmitted mainly by mosquitoes; however, the virus can be transmitted through other routes. Among the three structural and seven nonstructural proteins, the surface envelope (E) protein of ZIKV plays a critical role in viral entry and pathogenesis, making it a key target for the development of effective entry inhibitors. This review article describes the life cycle, genome, and encoded proteins of ZIKV, illustrates the structure and function of the ZIKV E protein, summarizes E protein-targeting entry inhibitors (with a focus on those based on natural products and small molecules), and highlights challenges that may potentially hinder the development of effective inhibitors of ZIKV infection. Overall, the article will provide useful guidance for further development of safe and potent ZIKV entry inhibitors targeting the viral E protein.


Assuntos
Antivirais , Proteínas do Envelope Viral , Internalização do Vírus , Infecção por Zika virus , Zika virus , Zika virus/efeitos dos fármacos , Zika virus/fisiologia , Internalização do Vírus/efeitos dos fármacos , Humanos , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/antagonistas & inibidores , Infecção por Zika virus/virologia , Infecção por Zika virus/tratamento farmacológico , Antivirais/farmacologia , Animais
2.
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
3.
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
4.
Proc Natl Acad Sci U S A ; 121(37): e2404250121, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39231203

RESUMO

Human cytomegalovirus (HCMV) glycoprotein B (gB) is a class III membrane fusion protein required for viral entry. HCMV vaccine candidates containing gB have demonstrated moderate clinical efficacy, but no HCMV vaccine has been approved. Here, we used structure-based design to identify and characterize amino acid substitutions that stabilize gB in its metastable prefusion conformation. One variant containing two engineered interprotomer disulfide bonds and two cavity-filling substitutions (gB-C7), displayed increased expression and thermostability. A 2.8 Å resolution cryoelectron microscopy structure shows that gB-C7 adopts a prefusion-like conformation, revealing additional structural elements at the membrane-distal apex. Unlike previous observations for several class I viral fusion proteins, mice immunized with postfusion or prefusion-stabilized forms of soluble gB protein displayed similar neutralizing antibody titers, here specifically against an HCMV laboratory strain on fibroblasts. Collectively, these results identify initial strategies to stabilize class III viral fusion proteins and provide tools to probe gB-directed antibody responses.


Assuntos
Citomegalovirus , Proteínas do Envelope Viral , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Citomegalovirus/imunologia , Humanos , Animais , Camundongos , Microscopia Crioeletrônica , Conformação Proteica , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Internalização do Vírus , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Estabilidade Proteica , Vacinas contra Citomegalovirus/imunologia , Substituição de Aminoácidos , Modelos Moleculares
5.
Nature ; 633(8030): 704-709, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39232163

RESUMO

Fifty-eight million individuals worldwide are affected by chronic hepatitis C virus (HCV) infection, a primary driver of liver cancer for which no vaccine is available1. The HCV envelope proteins E1 and E2 form a heterodimer (E1/E2), which is the target for neutralizing antibodies2. However, the higher-order organization of these E1/E2 heterodimers, as well as that of any Hepacivirus envelope protein complex, remains unknown. Here we determined the cryo-electron microscopy structure of two E1/E2 heterodimers in a homodimeric arrangement. We reveal how the homodimer is established at the molecular level and provide insights into neutralizing antibody evasion and membrane fusion by HCV, as orchestrated by E2 motifs such as hypervariable region 1 and antigenic site 412, as well as the organization of the transmembrane helices, including two internal to E1. This study addresses long-standing questions on the higher-order oligomeric arrangement of Hepacivirus envelope proteins and provides a critical framework in the design of novel HCV vaccine antigens.


Assuntos
Anticorpos Neutralizantes , Microscopia Crioeletrônica , Hepacivirus , Modelos Moleculares , Multimerização Proteica , Proteínas do Envelope Viral , Hepacivirus/química , Hepacivirus/ultraestrutura , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/ultraestrutura , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/química , Humanos , Evasão da Resposta Imune , Fusão de Membrana , Internalização do Vírus , Motivos de Aminoácidos
6.
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
7.
Nat Commun ; 15(1): 8133, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39285186

RESUMO

Limited knowledge exists on the quality of polyclonal antibody responses generated following Marburg virus (MARV) infection and its evolution in survivors. In this study, we evaluate MARV proteome-wide antibody repertoire longitudinally in convalescent phase approximately every six months for five years following MARV infection in ten human survivors. Differential kinetics were observed for IgM vs IgG vs IgA epitope diversity, antibody binding, antibody affinity maturation and Fc-receptor interaction to MARV proteins. Durability of MARV-neutralizing antibodies is low in survivors. MARV infection induces a diverse epitope repertoire with predominance against GP, VP40, VP30 and VP24 that persisted up to 5 years post-exposure. However, the IgM and IgA repertoire declines over time. Within MARV-GP, IgG recognize antigenic sites predominantly in the amino-terminus, wing domain and GP2-heptad repeat. Interestingly, MARV infection generates robust durable FcɣRI, FcɣRIIA and FcɣRIIIA IgG-Fc receptor interactions. Immunization with immunodominant MARV epitopes reveals conserved wing region between GP1 and GP2, induces neutralizing antibodies against MARV. These findings demonstrate that MARV infection generates a diverse, long-lasting, non-neutralizing, IgG antibody repertoire that perturbs disease by FcɣR activity. This information, along with discovery of neutralizing immunogen in wing domain, could aid in development of effective therapeutics and vaccines against Marburg virus.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Doença do Vírus de Marburg , Marburgvirus , Proteoma , Marburgvirus/imunologia , Humanos , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/virologia , Anticorpos Antivirais/imunologia , Anticorpos Neutralizantes/imunologia , Proteoma/imunologia , Feminino , Vacinas Virais/imunologia , Imunoglobulina G/imunologia , Masculino , Epitopos/imunologia , Adulto , Imunoglobulina M/imunologia , Pessoa de Meia-Idade , Estudos Longitudinais , Imunoglobulina A/imunologia , Desenvolvimento de Vacinas , Proteínas do Envelope Viral/imunologia
8.
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
9.
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
10.
Sci Rep ; 14(1): 18975, 2024 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-39152175

RESUMO

Analysis of protein modifications is critical for quality control of therapeutic biologics. However, the identification and quantification of naturally occurring glycation of membrane proteins by mass spectrometry remain technically challenging. We used highly sensitive LC MS/MS analyses combined with multiple enzyme digestions to determine low abundance early-stage lysine glycation products of influenza vaccines derived from embryonated chicken eggs and cultured cells. Straightforward sequencing was enhanced by MS/MS fragmentation of small peptides. As a result, we determined a widespread distribution of lysine modifications attributed by the region-selectivity and site-specificity of glycation toward influenza matrix 1, hemagglutinin and neuraminidase. Topological analysis provides insights into the site-specific lysine glycation, localizing in the distinct structural regions of proteins surrounding the viral envelope membrane. Our finding highlights the proteome-wide discovery of lysine glycation of influenza membrane proteins and potential effects on the structural assembly, stability, receptor binding and enzyme activity, demonstrating that the impacts of accumulated glycation on the quality of products can be directly monitored by mass spectrometry-based structural proteomics analyses.


Assuntos
Espectrometria de Massas em Tandem , Glicosilação , Animais , Vacinas contra Influenza/metabolismo , Neuraminidase/metabolismo , Humanos , Lisina/metabolismo , Embrião de Galinha , Proteínas da Matriz Viral/metabolismo , Proteínas da Matriz Viral/química , Proteômica/métodos , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Cromatografia Líquida
11.
Nat Commun ; 15(1): 6892, 2024 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-39134522

RESUMO

Nipah virus infection, one of the top priority diseases recognized by the World Health Organization, underscores the urgent need to develop effective countermeasures against potential epidemics and pandemics. Here, we identify a fully human single-domain antibody that targets a highly conserved cryptic epitope situated at the dimeric interface of the Nipah virus G protein (receptor binding protein, RBP), as elucidated through structures by high-resolution cryo-electron microscopy (cryo-EM). This unique binding mode disrupts the tetramerization of the G protein, consequently obstructing the activation of the F protein and inhibiting viral membrane fusion. Furthermore, our investigations reveal that this compact antibody displays enhanced permeability across the blood-brain barrier (BBB) and demonstrates superior efficacy in eliminating pseudovirus within the brain in a murine model of Nipah virus infection, particularly compared to the well-characterized antibody m102.4 in an IgG1 format. Consequently, this single-domain antibody holds promise as a therapeutic candidate to prevent Nipah virus infections and has potential implications for vaccine development.


Assuntos
Anticorpos Antivirais , Microscopia Crioeletrônica , Epitopos , Infecções por Henipavirus , Vírus Nipah , Anticorpos de Domínio Único , Vírus Nipah/imunologia , Humanos , Animais , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/prevenção & controle , Infecções por Henipavirus/virologia , Epitopos/imunologia , Camundongos , Anticorpos de Domínio Único/imunologia , Anticorpos de Domínio Único/química , Anticorpos Antivirais/imunologia , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/química , Feminino , Células HEK293
12.
Sci Rep ; 14(1): 19585, 2024 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-39179615

RESUMO

The World Health Organization (WHO) has declared the monkeypox outbreak a public health emergency, as there is no specific therapeutics for monkeypox virus (MPXV) disease. This study focused on docking various commercial drugs and plant-derived compounds against the E8 envelope protein crucial for MPXV attachment and pathogenesis. The target protein structure was modeled based on the vaccinia virus D8L protein. Notably, maraviroc and punicalagin emerged as potential ligands, with punicalagin exhibiting higher binding affinity (- 9.1 kcal/mol) than maraviroc (- 7.8 kcal/mol). Validation through 100 ns molecular dynamics (MD) simulations demonstrated increased stability of the E8-punicalagin complex, with lower RMSD, RMSF, and Rg compared to maraviroc. Enhanced hydrogen bonding, lower solvent accessibility, and compact motions also attributed to higher binding affinity and stability of the complex. MM-PBSA calculations revealed van der Waals, electrostatic, and non-polar solvation as principal stabilizing energies. The binding energy decomposition per residue favored stable interactions between punicalagin and the protein's active site residues (Arg20, Phe56, Glu228, Tyr232) compared to maraviroc. Overall study suggests that punicalagin can act as a potent inhibitor against MPXV. Further research and experimental investigations are warranted to validate its efficacy and safety.


Assuntos
Maraviroc , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Proteínas do Envelope Viral , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/antagonistas & inibidores , Maraviroc/química , Maraviroc/farmacologia , Monkeypox virus/química , Monkeypox virus/metabolismo , Antivirais/química , Antivirais/farmacologia , Ligação de Hidrogênio , Ligação Proteica , Taninos Hidrolisáveis/química , Taninos Hidrolisáveis/farmacologia , Taninos Hidrolisáveis/metabolismo
13.
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
14.
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
15.
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
16.
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
17.
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
18.
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
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...