Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.920
Filtrar
1.
Immun Inflamm Dis ; 12(9): e70021, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39291404

RESUMEN

BACKGROUND: Mycoplasma pneumoniae (MP) is a common respiratory pathogen affecting the longevity of the elderly and the health of children. However, the human vaccine against MP has not been successfully developed till now due to the poor immunogenicity and side effects of MP inactivated or attenuated vaccine. Therefore, it is necessary to develop a MP genetic engineering vaccine with influenza virus strain as vector. METHODS: In this study, the major antigen genes P1a of MP adhesion factor P1(3862-4554 bases) and P30a of P30(49-822 bases) were inserted into the nonstructural protein (NS) gene of Influenza A virus strain A/Puerto Rio/8/34(H1N1), PR8 for short, to construct the recombinant vectors NS-P1a or NS-P30a. The recombinant pHW2000 plasmids containing NS-P1a or NS-P30a were cotransfected with the rest 7 fragments of PR8 into HEK293T cells. After inoculating chicken embryos, the recombinant influenza viruses rFLU-P1a and rFLU-P30a were rescued. RT-PCR and sequencing were used to identify the recombinant viruses. The hemagglutination titers of rFLU-P1a and rFLU-P30a were determined after five successive generations in chicken embryos so as to indicate the genetic stability of the recombinant viruses. The morphology of recombinant influenza viruses was observed under electron microscopy. RESULTS: P1a or P30a was designed to be inserted into the modified NS gene sequence separately and synthesized successfully. RT-PCR identification of the recombinant viruses rFLU-P1a and rFLU-P30a showed that P1a (693 bp), P30a (774 bp), NS-P1a (1992bp) and NS-P30a (2073 bp) bands were found, and the sequencing results were correct. After five successive generations, each virus generation has a certain hemagglutination titer (from 1:32 to 1:64), and the band of P1a or P30a can be seen in the corresponding positions. The virus particles under the electron microscope appeared as spheres or long strips connected by several particles, revealing a complete viral membrane structure composed of virus lipid bilayer, hemagglutinin, neuraminidase, and matrix proteins. CONCLUSION: The recombinant viruses rFLU-P1a and rFLU-P30a which carried the advantaged immune regions of the P1 and P30 genes in MP were successfully constructed and identified. And the genetic stability of rFLU-P1a or rFLU-P30a was relatively high. The typical and complete morphology of influenza virus was observed under the electron microscope. Our research provided a foundation for the further development of MP vaccines for human.


Asunto(s)
Vectores Genéticos , Mycoplasma pneumoniae , Humanos , Mycoplasma pneumoniae/genética , Mycoplasma pneumoniae/inmunología , Animales , Células HEK293 , Vectores Genéticos/genética , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/inmunología , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , Embrión de Pollo , Neumonía por Mycoplasma/inmunología , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/genética
2.
Virus Genes ; 60(5): 475-487, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39102085

RESUMEN

DENV infection outcomes depend on the host's variable expression of immune receptors and mediators, leading to either resolution or exacerbation. While the NS3 protein is known to induce robust immune responses, the specific impact of its protease region epitopes remains unclear. This study investigated the effect of recombinant NS3 protease region proteins from all four DENV serotypes on splenocyte activation in BALB/c mice (n = 5/group). Mice were immunized with each protein, and their splenocytes were subsequently stimulated with homologous antigens. We measured the expression of costimulatory molecules (CD28, CD80, CD86, CD152) by flow cytometry, along with IL-2 production, CD25 expression, and examined the antigen-specific activation of CD4 + and CD8 + T cells. Additionally, the expression of IL-1, IL-10, and TGF-ß1 in splenocytes from immunized animals was assessed. Apoptosis was evaluated using Annexin V/PI staining and DNA fragmentation analysis. Stimulation of splenocytes from immunized mice triggered apoptosis (phosphatidylserine exposure and caspase 3/7 activation) and increased costimulatory molecule expression, particularly CD152. Low IL-2 production and low CD25 expression, as well as sustained expression of the IL-10 gene. These results suggest that these molecules might be involved in mechanisms by which the NS3 protein contributes to viral persistence and disease pathogenesis.


Asunto(s)
Apoptosis , Antígeno CTLA-4 , Virus del Dengue , Ratones Endogámicos BALB C , Bazo , Proteínas no Estructurales Virales , Animales , Ratones , Bazo/inmunología , Bazo/virología , Virus del Dengue/inmunología , Virus del Dengue/genética , Antígeno CTLA-4/genética , Antígeno CTLA-4/inmunología , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/genética , Inmunización , Dengue/inmunología , Dengue/virología , Citocinas/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología
3.
Biologicals ; 87: 101785, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39121525

RESUMEN

Diagnostic assays that are able to detect foot-and-mouth disease (FMD) virus infection in the vaccinated population are essential tools in the progressive control pathway for the FMD. However, testing of serum samples using a single diagnostic assay may not completely substantiate freedom from the virus infection. Therefore, viral non-structural proteins (NSPs)-based various serological assays have been developed for the detection of FMD infection. Nevertheless, the NSPs-based ELISAs have been developed in the indirect-ELISA format, thereby necessitating the use of species-specific conjugated secondary-antibodies for the detection of anti-NSP antibodies in various FMD-susceptible species. Therefore, this study presents a novel recombinant 2B-NSP-based indirect ELISA, employing HRP-conjugated protein-A/G detection system which can detect anti-NSPs antibodies from multiple FMD-susceptible species in a single ELISA platform. Recombinant 2B (r2B) protein was expressed as His-SUMO tagged protein in the E. Coli cells and purified using NI-NTA affinity column chromatography. Using the r2B protein and HRP-conjugated protein A/G, an indirect ELISA was developed and validated for the detection of anti-2B antibodies in serum samples collected from multiple FMD-susceptible animal species with known FMD status. Further, a resampling based statistical technique has been reported for determination of optimal cut-off value for the diagnostic assay. Through this technique, the optimal cut-off of 44 percentage of positivity value was determined for the assay. At this optimal cut-off value, the developed diagnostic assay provided diagnostic sensitivity, specificity, and accuracy, positive and negative predictive values (PPV and NPV) of 92.35 %, 98.41 %, 95.21 %, 98.58 %, and 91.67 %, respectively. The assay was validated further by analyzing random serum samples collected across multi-locations in India. The assay can be used as a single platform for testing serum samples from different species of FMDV-susceptible animals and will be useful for NSP-based serosurveillance of FMDV.


Asunto(s)
Anticuerpos Antivirales , Ensayo de Inmunoadsorción Enzimática , Virus de la Fiebre Aftosa , Fiebre Aftosa , Proteínas no Estructurales Virales , Virus de la Fiebre Aftosa/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Animales , Fiebre Aftosa/diagnóstico , Fiebre Aftosa/inmunología , Fiebre Aftosa/sangre , Fiebre Aftosa/virología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Proteínas no Estructurales Virales/inmunología , Bovinos , Proteínas Recombinantes/inmunología , Porcinos , Especificidad de la Especie
4.
PLoS Negl Trop Dis ; 18(8): e0012146, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39178324

RESUMEN

Mosquito-borne Zika virus (ZIKV) from sub-Saharan Africa has recently gained attention due to its epidemic potential and its capacity to be highly teratogenic. To improve our knowledge on currently circulating strains of African ZIKV, we conducted protein sequence alignment and identified contemporary West Africa NS1 (NS1CWA) protein as a highly conserved viral protein. Comparison of NS1CWA with the NS1 of the historical African ZIKV strain MR766 (NS1MR766), revealed seven amino acid substitutions. The effects of NS1 mutations on protein expression, virus replication, and innate immune activation were assessed in human cells using recombinant NS1 proteins and a chimeric viral clone MR766 with NS1CWA replacing NS1MR766. Our data indicated higher secretion efficiency of NS1CWA compared to NS1MR766 associated with a change in subcellular distribution. A chimeric MR766 virus with NS1CWA instead of authentic protein displayed a greater viral replication efficiency, leading to more pronounced cell death compared to parental virus. Enhanced viral growth was associated with reduced activation of innate immunity. Our data raise questions of the importance of NS1 protein in the pathogenicity of contemporary ZIKV from sub-Saharan Africa and point to differences within viral strains of African lineage.


Asunto(s)
Inmunidad Innata , Proteínas no Estructurales Virales , Replicación Viral , Virus Zika , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/inmunología , Virus Zika/genética , Virus Zika/inmunología , Virus Zika/fisiología , Humanos , Infección por el Virus Zika/virología , Infección por el Virus Zika/inmunología , Animales , Chlorocebus aethiops , Células Vero , África Occidental , Sustitución de Aminoácidos , Línea Celular
5.
Microb Pathog ; 195: 106873, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39173850

RESUMEN

As one of the most important swine enteropathogenic coronavirus, porcine epidemic diarrhea virus (PEDV) is the causative agent of an acute and devastating enteric disease that causes lethal watery diarrhea in suckling piglets. Recent progress in studying PEDV has revealed many intriguing findings on its prevalence and genetic evolution, rapid diagnosis, suppression of host gene expression, and suppression of the host innate immune system. Due to the continuous mutation of the PEDV genome, viral evasions from innate immune defenses and mixed infection with other coronaviruses, the spread of the virus is becoming wider and faster, making it even more necessary to prevent the infections caused by wild-type PEDV variants. It has also been reported that PEDV nsp1 is an essential virulence determinant and is critical for inhibiting host gene expression by structural and biochemical analyses. The inhibition of host protein synthesis employed by PEDV nsp1 may contribute to the regulation of host cell proliferation and immune evasion-related biological functions. In this review, we critically evaluate the recent studies on these aspects of PEDV and assess prospects in understanding the function of PEDV proteins in regulating host innate immune response and viral virulence.


Asunto(s)
Infecciones por Coronavirus , Evasión Inmune , Inmunidad Innata , Virus de la Diarrea Epidémica Porcina , Enfermedades de los Porcinos , Virus de la Diarrea Epidémica Porcina/genética , Virus de la Diarrea Epidémica Porcina/inmunología , Virus de la Diarrea Epidémica Porcina/patogenicidad , Animales , Porcinos , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/virología , Virulencia/genética , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/metabolismo , Interacciones Huésped-Patógeno/inmunología , Factores de Virulencia/genética
6.
Sci Rep ; 14(1): 18883, 2024 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-39143088

RESUMEN

NS1 is an elusive dengue protein, involved in viral replication, assembly, pathogenesis, and immune evasion. Its levels in blood plasm are positively related to disease severity like thrombocytopenia, hemorrhage, and vascular leakage. Despite its pathogenic roles, NS1 is being used in various vaccine formulations due to its sequence conservancy, ability to produce protective antibodies and low risk for inducing antibody-dependent enhancement. In this study, we have used bioinformatics tools and reported literature to develop an NS1 variant (dNS1). Molecular docking studies were performed to evaluate the receptor-binding ability of the NS1 and dNS1 with TLR4. NS1 and dNS1 (153 to 312 amino acid region) genes were cloned, expressed and protein was purified followed by refolding. Docking studies showed the binding of NS1 and dNS1 with the TLR4 receptor which suggests that N and C-terminal sequences of NS1 are not critical for receptor binding. Antibodies against NS1 and dNS1 were raised in rabbits and binding affinity of anti-dNS1 anti-NS1 sera was evaluated against both NS1 and dNS1. Similar results were observed through western blotting which highlight that N and C-terminal deletion of NS1 does not compromise the immunogenic potential of dNS1 hence, supports its use in future vaccine formulations as a substitute for NS1.


Asunto(s)
Vacunas contra el Dengue , Virus del Dengue , Simulación del Acoplamiento Molecular , Proteínas no Estructurales Virales , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Virus del Dengue/inmunología , Virus del Dengue/genética , Animales , Vacunas contra el Dengue/inmunología , Vacunas contra el Dengue/genética , Desarrollo de Vacunas , Conejos , Humanos , Dengue/prevención & control , Dengue/inmunología , Dengue/virología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Unión Proteica , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/inmunología , Eliminación de Secuencia
7.
Emerg Microbes Infect ; 13(1): 2317909, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39133062

RESUMEN

Tick-borne encephalitis virus (TBEV) infection may cause acute central nervous system inflammation varying in clinical manifestations and severity. A possible correlation of TBEV-specific antibody and cell-mediated immune responses, shortly after infection, with clinical manifestations, severity and long-term outcome has been poorly investigated. In a cohort of thirty early tick-borne encephalitis (TBE) patients, we assessed the magnitude, specificity and functional properties of TBEV-specific T-cell and antibody responses. These responses early during disease were assessed in view of clinical manifestations, severity and long-term outcome. TBEV-specific T-cell responses to C, E, NS1, and NS5 proteins were significantly lower in patients with severe acute illness than in patients with mild TBE. Lower T-cell responses to E, NS1, and NS5 proteins also correlated with the development of meningoencephalomyelitis. Virus-specific antibody titres early after infection did not correlate with disease severity, clinical manifestations, or long-term outcome in this study, possibly due to the small number of patients of which matching serum and peripheral blood mononuclear cells were available. The findings suggest that virus-specific T cells afford a certain degree of protection against the development of severe TBEV-induced disease.


Asunto(s)
Anticuerpos Antivirales , Virus de la Encefalitis Transmitidos por Garrapatas , Encefalitis Transmitida por Garrapatas , Linfocitos T , Encefalitis Transmitida por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/virología , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Humanos , Linfocitos T/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Masculino , Femenino , Persona de Mediana Edad , Adulto , Índice de Severidad de la Enfermedad , Anciano , Proteínas no Estructurales Virales/inmunología
8.
Virus Res ; 347: 199431, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38969013

RESUMEN

Usutu virus (USUV) is an emerging flavivirus that can infect birds and mammals. In humans, in severe cases, it may cause neuroinvasive disease. The innate immune system, and in particular the interferon response, functions as the important first line of defense against invading pathogens such as USUV. Many, if not all, viruses have developed mechanisms to suppress and/or evade the interferon response in order to facilitate their replication. The ability of USUV to antagonize the interferon response has so far remained largely unexplored. Using dual-luciferase reporter assays we observed that multiple of the USUV nonstructural (NS) proteins were involved in suppressing IFN-ß production and signaling. In particular NS4A was very effective at suppressing IFN-ß production. We found that NS4A interacted with the mitochondrial antiviral signaling protein (MAVS) and thereby blocked its interaction with melanoma differentiation-associated protein 5 (MDA5), resulting in reduced IFN-ß production. The TM1 domain of NS4A was found to be essential for binding to MAVS. By screening a panel of flavivirus NS4A proteins we found that the interaction of NS4A with MAVS is conserved among flaviviruses. The increased understanding of the role of NS4A in flavivirus immune evasion could aid the development of vaccines and therapeutic strategies.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Flavivirus , Helicasa Inducida por Interferón IFIH1 , Interferón beta , Transducción de Señal , Proteínas no Estructurales Virales , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Flavivirus/inmunología , Flavivirus/genética , Flavivirus/fisiología , Interferón beta/genética , Interferón beta/inmunología , Interferón beta/metabolismo , Helicasa Inducida por Interferón IFIH1/genética , Helicasa Inducida por Interferón IFIH1/metabolismo , Helicasa Inducida por Interferón IFIH1/inmunología , Células HEK293 , Evasión Inmune , Infecciones por Flavivirus/inmunología , Infecciones por Flavivirus/virología , Interacciones Huésped-Patógeno/inmunología , Unión Proteica , Inmunidad Innata , Animales
9.
BMC Infect Dis ; 24(1): 735, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39061013

RESUMEN

BACKGROUND: Dengue infection is a mosquito-borne, endemic viral disease, particularly in developing countries. Here, we report the results of the clinicodemographic, serologic profile and the monthly occurrence of a recent dengue fever outbreak in Puntland State (Somalia). METHODOLOGY: We analyzed the data of 956 dengue-suspected patients who were investigated using the rapid diagnostic testing (RDT) method for detecting NS1 (dengue virus non-structural protein) and IgM antibodies employing the SD Biosensor Dengue Dou NS Ag and IgM test kit (Germany) at the Puntland Public Health Referral Laboratory from November 21, 2022, to May 27, 2023. RESULTS: We found that 118 cases were positive for dengue among the suspected patients enrolled in the present study. Of these cases, 76.2% were dengue NSI positive, 13.6% were dengue IgM positive, and 10.2% were both NSI and IgM positive. The number of females and males in the confirmed cases was equal, and most (48.3%) were aged 20 years or less. 43.1% of them lived in the Nugal region, particularly in Garowe. Clinically, fever was the most frequent symptom (88.9%). The cases peaked in December 2022 but dropped from January to March, with a slight rise in February, and then increased in April and May 2023. CONCLUSION: This study highlights the clinicodemographic characteristics, seroprevalence, and monthly occurrence of dengue in Puntland. We recommend improving vector control measures, enhancing case management, strengthening dengue surveillance, developing an early warning system, and conducting future studies to characterize the circulating strains.


Asunto(s)
Anticuerpos Antivirales , Virus del Dengue , Dengue , Brotes de Enfermedades , Inmunoglobulina M , Humanos , Dengue/epidemiología , Masculino , Femenino , Adulto , Adolescente , Inmunoglobulina M/sangre , Adulto Joven , Anticuerpos Antivirales/sangre , Estudios Retrospectivos , Virus del Dengue/inmunología , Virus del Dengue/aislamiento & purificación , Niño , Persona de Mediana Edad , Preescolar , Proteínas no Estructurales Virales/inmunología
10.
Vet Microbiol ; 296: 110173, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38971119

RESUMEN

Porcine reproductive and respiratory syndrome (PRRS) is the most economically significant disease caused by porcine reproductive and respiratory syndrome virus (PRRSV). Type I interferon (IFN) induces a large number of interferon-stimulated genes (ISGs) expression to inhibit PRRSV infection. To survive in the host, PRRSV has evolved multiple strategies to antagonize host innate immune response. Previous studies have reported that PRRSV N protein decreases the expression of TRIM25 and TRIM25-mediated RIG-I ubiquitination to suppress IFN-ß production. However, whether other PRRSV proteins inhibit the antiviral function of TRIM25 is less well understood. In this study, we first found that PRRSV NSP1α decreased ISGylation of TRIM25. Meanwhile, NSP1α significantly suppressed TRIM25-mediated IFN-ß production to promote PRRSV replication. Further studies demonstrated that PRRSV NSP1α reduced the protein level of TRIM25 in proteasome system but did not regulate the transcription level of TRIM25. In addition, the function of NSP1α in TRIM25 degradation did not rely on its papain-like cysteine protease activity. Taken together, PRRSV NSP1α antagonizes the antiviral response of TRIM25 by mediating TRIM25 degradation to promote PRRSV replication. Our data identify TRIM25 as a natural target of PRRSV NSP1α and reveal a novel mechanism that PRRSV induces TRIM25 degradation and inhibits host antiviral immune response.


Asunto(s)
Inmunidad Innata , Virus del Síndrome Respiratorio y Reproductivo Porcino , Complejo de la Endopetidasa Proteasomal , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Proteínas no Estructurales Virales , Replicación Viral , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Animales , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Porcinos , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/metabolismo , Síndrome Respiratorio y de la Reproducción Porcina/inmunología , Síndrome Respiratorio y de la Reproducción Porcina/virología , Interferón beta/genética , Interferón beta/metabolismo , Interferón beta/inmunología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Línea Celular , Ubiquitinación , Humanos , Células HEK293 , Interacciones Huésped-Patógeno/inmunología
11.
Methods Mol Biol ; 2824: 147-164, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39039412

RESUMEN

Single-domain antibodies, referred to as VHH (variable heavy chains of heavy chain-only antibodies) or in their commercial name as nanobodies, are potent tools for the detection of target proteins in biological samples. They have the advantage of being highly stable, specific, and sensitive, with affinities reaching the nanomolar range. We utilized this tool to develop a rapid detection method that discriminates cells infected with Rift Valley fever virus (RVFV), based on the intracellular detection of the viral nonstructural NSm protein localized on the outer membrane of mitochondria. Here we describe how NSm-specific VHHs have been produced, cloned, and characterized, highlighting their value in RVFV research and diagnosis. This work may also raise interest in other potential applications such as antiviral therapy.


Asunto(s)
Fiebre del Valle del Rift , Virus de la Fiebre del Valle del Rift , Anticuerpos de Dominio Único , Proteínas no Estructurales Virales , Virus de la Fiebre del Valle del Rift/inmunología , Anticuerpos de Dominio Único/inmunología , Humanos , Fiebre del Valle del Rift/inmunología , Fiebre del Valle del Rift/diagnóstico , Fiebre del Valle del Rift/virología , Proteínas no Estructurales Virales/inmunología , Animales , Anticuerpos Antivirales/inmunología
12.
Front Immunol ; 15: 1424307, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39011043

RESUMEN

Introduction: Bluetongue (BT) poses a significant threat to the livestock industry, affecting various animal species and resulting in substantial economic losses. The existence of numerous BT virus (BTV) serotypes has hindered control efforts, highlighting the need for broad-spectrum vaccines. Methodology: In this study, we evaluated the conserved amino acid sequences within key non-structural (NS) proteins of BTV and identified numerous highly conserved murine- and bovine-specific MHC class I-restricted (MHC-I) CD8+ and MHC-II-restricted CD4+ epitopes. We then screened these conserved epitopes for antigenicity, allergenicity, toxicity, and solubility. Using these epitopes, we developed in silico-based broad-spectrum multiepitope vaccines with Toll-like receptor (TLR-4) agonists. The predicted proinflammatory cytokine response was assessed in silico using the C-IMMSIM server. Structural modeling and refinement were achieved using Robetta and GalaxyWEB servers. Finally, we assessed the stability of the docking complexes through extensive 100-nanosecond molecular dynamics simulations before considering the vaccines for codon optimization and in silico cloning. Results: We found many epitopes that meet these criteria within NS1 and NS2 proteins and developed in silico broad-spectrum vaccines. The immune simulation studies revealed that these vaccines induce high levels of IFN-γ and IL-2 in the vaccinated groups. Protein-protein docking analysis demonstrated promising epitopes with strong binding affinities to TLR-4. The docked complexes were stable, with minimal Root Mean Square Deviation and Root Mean Square Fluctuation values. Finally, the in silico-cloned plasmids have high % of GC content with > 0.8 codon adaptation index, suggesting they are suitable for expressing the protein vaccines in prokaryotic system. Discussion: These next-generation vaccine designs are promising and warrant further investigation in wet lab experiments to assess their immunogenicity, safety, and efficacy for practical application in livestock. Our findings offer a robust framework for developing a comprehensive, broad-spectrum vaccine, potentially revolutionizing BT control and prevention strategies in the livestock industry.


Asunto(s)
Virus de la Lengua Azul , Biología Computacional , Epítopos de Linfocito T , Proteínas no Estructurales Virales , Vacunas Virales , Animales , Virus de la Lengua Azul/inmunología , Epítopos de Linfocito T/inmunología , Vacunas Virales/inmunología , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/genética , Ratones , Biología Computacional/métodos , Serogrupo , Bovinos , Lengua Azul/prevención & control , Lengua Azul/inmunología , Lengua Azul/virología , Secuencia Conservada
13.
Viruses ; 16(7)2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39066297

RESUMEN

Rotavirus remains a significant public health threat, especially in low-income countries, where it is the leading cause of severe acute childhood gastroenteritis, contributing to over 128,500 deaths annually. Although the introduction of the Rotarix and RotaTeq vaccines in 2006 marked a milestone in reducing mortality rates, approximately 83,158 preventable deaths persisted, showing ongoing challenges in vaccine accessibility and effectiveness. To address these issues, a novel subcutaneous vaccine formulation targeting multiple rotavirus genotypes has been developed. This vaccine consists of nine VP8* proteins from nine distinct rotavirus genotypes and sub-genotypes (P[4], P[6], P[8]LI, P[8]LIII, P[8]LIV, P[9], P[11], P[14], and P[25]) expressed in E. coli. Two groups of mice were immunized either with a single immunogen, the VP8* from the rotavirus Wa strain (P[8]LI), or with the nonavalent formulation. Preliminary results from mouse immunization studies showed promising outcomes, eliciting antibody responses against six of the nine immunogens. Notably, significantly higher antibody titers against VP8* P[8]LI were observed in the group immunized with the nonavalent vaccine compared to mice specifically immunized against this genotype alone. Overall, the development of parenteral vaccines targeting multiple rotavirus genotypes represents a promising strategy in mitigating the global burden of rotavirus-related morbidity and mortality, offering new avenues for disease prevention and control.


Asunto(s)
Anticuerpos Antivirales , Infecciones por Rotavirus , Vacunas contra Rotavirus , Rotavirus , Vacunas de Subunidad , Animales , Vacunas contra Rotavirus/inmunología , Vacunas contra Rotavirus/administración & dosificación , Ratones , Rotavirus/inmunología , Rotavirus/genética , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/genética , Infecciones por Rotavirus/prevención & control , Infecciones por Rotavirus/inmunología , Infecciones por Rotavirus/virología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Femenino , Ratones Endogámicos BALB C , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/genética , Inmunogenicidad Vacunal , Genotipo , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/genética , Proteínas de Unión al ARN/inmunología , Proteínas de Unión al ARN/genética
14.
Sci Rep ; 14(1): 17645, 2024 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-39085250

RESUMEN

The Middle East has witnessed a greater spread of infectious Dengue viruses, with serotype 2 (DENV-2) being the most prevalent form. Through this work, multi-epitope peptide vaccines against DENV-2 that target E and nonstructural (NS1) proteins were generated through an immunoinformatic approach. MHC class I and II and LBL epitopes among NS1 and envelope E proteins sequences were predicted and their antigenicity, toxicity, and allergenicity were investigated. Studies of the population coverage denoted the high prevalence of NS1 and envelope-E epitopes among different countries where DENV-2 endemic. Further, both the CTL and HTL epitopes retrieved from NS1 epitopes exhibited high conservancies' percentages with other DENV serotypes (1, 3, and 4). Three vaccine constructs were created and the expected immune responses for the constructs were estimated using C-IMMSIM and HADDOCK (against TLR 2,3,4,5, and 7). Molecular dynamics simulation for vaccine construct 2 with TLR4 denoted high binding affinity and stability of the construct with the receptor which might foretell favorable in vivo interaction and immune responses.


Asunto(s)
Vacunas contra el Dengue , Virus del Dengue , Dengue , Serogrupo , Vacunas de Subunidad , Proteínas no Estructurales Virales , Virus del Dengue/inmunología , Vacunas de Subunidad/inmunología , Vacunas contra el Dengue/inmunología , Humanos , Dengue/prevención & control , Dengue/inmunología , Dengue/virología , Proteínas no Estructurales Virales/inmunología , Biología Computacional/métodos , Epítopos de Linfocito T/inmunología , Proteínas del Envoltorio Viral/inmunología , Simulación de Dinámica Molecular , Epítopos/inmunología , Epítopos/química , Vacunas de Subunidades Proteicas
15.
Front Immunol ; 15: 1440407, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39072326

RESUMEN

Introduction: Bluetongue (BT), caused by bluetongue virus (BTV), is an important arthropod-borne livestock disease listed by the World Organization for Animal Health. Live-attenuated and inactivated vaccines have permitted to control BT but they do not simultaneously protect against the myriad of BTV serotypes. Recently, we identified the highly conserved BTV nonstructural protein NS1 and the N-terminal region of NS2 as antigens capable of conferring multiserotype protection against BTV. Methods: Here, we designed Modified Vaccinia Ankara (MVA) viral vectors that expressed BTV-4 proteins VP2 or VP7 along with NS1 and NS2-Nt as well as MVAs that expressed proteins VP2, VP7 or NS1 and NS2-Nt. Results: Immunization of IFNAR(-/-) mice with two doses of MVA-NS1-2A-NS2-Nt protected mice from BTV-4M infection by the induction of an antigen-specific T cell immune response. Despite rMVA expressing VP7 alone were not protective in the IFNAR(-/-) mouse model, inclusion of VP7 in the vaccine formulation amplified the cell-mediated response induced by NS1 and NS2-Nt. Expression of VP2 elicited protective non-cross-reactive neutralizing antibodies (nAbs) in immunized animals and improved the protection observed in the MVA-NS1-2A-NS2-Nt immunized mice when these three BTV antigens were co-expressed. Moreover, vaccines candidates co-expressing VP2 or VP7 along with NS1 and NS2-Nt provided multiserotype protection. We assessed protective efficacy of both vaccine candidates in sheep against virulent challenge with BTV-4M. Discussion: Immunization with MVA-VP7-NS1-2A-NS2-Nt partially dumped viral replication and clinical disease whereas administration of MVA-VP2-NS1-2A-NS2-Nt promoted a complete protection, preventing viraemia and the pathology produced by BTV infection.


Asunto(s)
Virus de la Lengua Azul , Lengua Azul , Proteínas de la Cápside , Vectores Genéticos , Receptor de Interferón alfa y beta , Virus Vaccinia , Proteínas no Estructurales Virales , Vacunas Virales , Animales , Virus de la Lengua Azul/inmunología , Virus de la Lengua Azul/genética , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/genética , Lengua Azul/prevención & control , Lengua Azul/inmunología , Lengua Azul/virología , Ratones , Vacunas Virales/inmunología , Vacunas Virales/genética , Virus Vaccinia/genética , Virus Vaccinia/inmunología , Receptor de Interferón alfa y beta/genética , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/genética , Ratones Noqueados , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Femenino
16.
Mol Diagn Ther ; 28(5): 633-643, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38980575

RESUMEN

BACKGROUND AND OBJECTIVE: Dengue is a major infectious disease with potential for outbreaks and epidemics. A specific and sensitive diagnosis is a prerequisite for clinical management of the disease. We designed our study to identify epitopes on the Dengue virus (DENV) envelope (E) and non-structural protein 1 (NS1) with potential for diagnosis. METHODS: Serology and immunoinformatic approaches were employed. We collected DENV-positive, DENV-negative and Japanese encephalitis virus-positive samples from collaborating hospitals in 2019 and 2022-2023. Seropositive peptides in 15-18 mer peptide arrays of E and NS1 proteins of DENV2 were determined by an indirect enzyme-linked immunosorbent assay. B-cell linear and conformational epitopes were predicted using BepiPred2.0 and ElliPro, respectively. A consensus recombinant peptide was designed, synthesised and evaluated for its diagnostic potential using patient sera. RESULTS: Eight peptides of E protein and six peptides of NS1 protein were identified to be the most frequently recognised by Dengue-positive patients. These peptide sequences were compared with B-cell epitope regions and found to be overlapped with predicted B-cell linear and conformational epitopes. EP11 and NSP15 showed a 100% amino acid sequence overlap with B-cell epitopes. EP1 and NSP15 had 14 whereas EP28, EP31, EP60 16, NSP12 and NSP32 had more than 15 interacting interface residues with a neutralising antibody, suggesting a strength of interaction. Interestingly, potential epitopes identified were localised on the surface of proteins as visualised by PyMOL. Validation with a recombined synthetic peptide yielded 92.3% sensitivity and 91.42% specificity. CONCLUSIONS: Immunodominant regions identified by serology and computationally predicted epitopes overlapped, thereby showing the robustness of the methodology and the peptide designed for diagnosis.


Asunto(s)
Anticuerpos Antivirales , Virus del Dengue , Dengue , Epítopos de Linfocito B , Epítopos Inmunodominantes , Péptidos , Proteínas del Envoltorio Viral , Proteínas no Estructurales Virales , Proteínas no Estructurales Virales/inmunología , Humanos , Virus del Dengue/inmunología , Epítopos Inmunodominantes/inmunología , Dengue/diagnóstico , Dengue/inmunología , Dengue/virología , Dengue/sangre , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/química , Epítopos de Linfocito B/inmunología , Epítopos de Linfocito B/química , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Péptidos/inmunología , Péptidos/química , Ensayo de Inmunoadsorción Enzimática
17.
Vaccine ; 42(22): 126032, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-38964950

RESUMEN

For the rational design of epitope-specific vaccines, identifying epitopes that can be processed and presented is essential. As algorithm-based epitope prediction is frequently discordant with actually recognized CD8+ T-cell epitopes, we developed an in vitro CD8 T-cell priming protocol to enable the identification of truly and functionally expressed HLA class I epitopes. The assay was established and validated to identify epitopes presented by hepatitis C virus (HCV)-infected cells. In vitro priming of naïve CD8 T cells was achieved by culturing unfractionated PBMCs in the presence of a specific cocktail of growth factors and cytokines, and next exposing the cells to hepatic cells expressing the NS3 protein of HCV. After a 10-day co-culture, HCV-specific T-cell responses were identified based on IFN-γ ELISpot analysis. For this, the T cells were restimulated with long synthetic peptides (SLPs) spanning the whole NS3 protein sequence allowing the identification of HCV-specificity. We demonstrated that this protocol resulted in the in vitro priming of naïve precursors to antigen-experienced T-cells specific for 11 out of 98 SLPs tested. These 11 SLPs contain 12 different HLA-A*02:01-restricted epitopes, as predicted by a combination of three epitope prediction algorithms. Furthermore, we identified responses against 3 peptides that were not predicted to contain any immunogenic HLA class I epitopes, yet showed HCV-specific responses in vitro. Separation of CD8+ and CD8- T cells from PBMCs primed in vitro showed responses only upon restimulation with short peptides. We established an in vitro method that enables the identification of HLA class I epitopes resulting from cross-presented antigens and that can cross-prime T cells and allows the effective selection of functional immunogenic epitopes, but also less immunogenic ones, for the design of tailored therapeutic vaccines against persistent viral infections and tumor antigens.


Asunto(s)
Linfocitos T CD8-positivos , Epítopos de Linfocito T , Hepacivirus , Vacunas contra Hepatitis Viral , Proteínas no Estructurales Virales , Linfocitos T CD8-positivos/inmunología , Humanos , Epítopos de Linfocito T/inmunología , Hepacivirus/inmunología , Hepacivirus/genética , Vacunas contra Hepatitis Viral/inmunología , Proteínas no Estructurales Virales/inmunología , Hepatitis C/inmunología , Hepatitis C/prevención & control , Ensayo de Immunospot Ligado a Enzimas/métodos , Antígeno HLA-A2/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Proteasas Virales , Serina Endopeptidasas , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-box
18.
Int J Mol Sci ; 25(14)2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-39062886

RESUMEN

Porcine deltacoronavirus (PDCoV) is an emerging enteric pathogen that causes substantial economic losses in the swine industry worldwide. The PDCoV NS6 protein is an accessory protein that plays a pivotal role in the viral life cycle and immune evasion. However, the functions of NS6 and its role in PDCoV pathogenesis remain largely unknown. In this study, we prepared a monoclonal antibody (mAb) 5-A11 that specifically recognizes the PDCoV NS6 protein. The mAb 5-A11 exhibited high specificity for PDCoV, with no cross-reactivity with several major porcine pathogenic viruses. Furthermore, the epitope recognized by mAb 5-A11 was precisely mapped to residues 70EYGSIYGKDFI80 of the NS6 protein using Western blot analysis. Notably, this epitope is highly conserved among different PDCoV isolates. Substantial variations were observed when comparing this epitope with the corresponding regions in the NS6 proteins of other δ coronaviruses, suggesting potential differences in the structure, function, and antigenicity of their NS6 proteins. Our findings provide valuable tools and insights for further elucidating the functions of the NS6 protein and its role in PDCoV pathogenesis, as well as for developing diagnostic and therapeutic strategies against PDCoV infection.


Asunto(s)
Anticuerpos Monoclonales , Deltacoronavirus , Epítopos , Proteínas no Estructurales Virales , Animales , Anticuerpos Monoclonales/inmunología , Porcinos , Deltacoronavirus/inmunología , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/metabolismo , Epítopos/inmunología , Mapeo Epitopo , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Anticuerpos Antivirales/inmunología , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/inmunología , Ratones , Secuencia de Aminoácidos , Ratones Endogámicos BALB C
19.
Viruses ; 16(7)2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-39066195

RESUMEN

Omsk hemorrhagic fever virus (OHFV) is a member of the tick-borne encephalitis virus (TBEV) complex of the Flaviviridae family. Currently, there are no data on the cross-reactivity of antibodies to the NS1 proteins of OHFV and TBEV. Such data are of major interest for monitoring viral encephalitis of unknown etiology due to the increasing geographical distribution of OHFV. In this study, a recombinant OHFV NS1 protein was produced using the Escherichia coli expression system and purified. The recombinant OHFV NS1 protein was recognized by specific mice immune ascetic fluids to the native OHFV NS1 protein. A Western blot analysis and ELISA of the recombinant NS1 proteins of OHFV and TBEV were used to study the cross-reactivity of antibodies from immune ascites fluid obtained from OHFV-infected mice and mAbs against TBEV NS1. Anti-TBEV NS1 mouse monoclonal antibodies (mAbs) have been shown to not be cross-reactive to the OHFV NS1 protein. Sera from patients with confirmed tick-borne encephalitis (TBE) were examined by ELISA using recombinant OHFV NS1 and TBEV NS1 proteins as antigens. It was shown for the first time that cross-reactive antibodies to the OHFV NS1 protein were not detected in the sera of TBE patients, whereas the sera contained antibodies to the TBEV NS1 protein.


Asunto(s)
Anticuerpos Antivirales , Reacciones Cruzadas , Virus de la Encefalitis Transmitidos por Garrapatas , Encefalitis Transmitida por Garrapatas , Proteínas Recombinantes , Proteínas no Estructurales Virales , Proteínas no Estructurales Virales/inmunología , Encefalitis Transmitida por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/virología , Encefalitis Transmitida por Garrapatas/sangre , Reacciones Cruzadas/inmunología , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Animales , Humanos , Ratones , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/genética , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/genética , Ratones Endogámicos BALB C , Femenino
20.
Vaccine ; 42(25): 126045, 2024 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-38852036

RESUMEN

Yellow fever (YF) is a disease caused by the homonymous flavivirus that can be prevented by a vaccine containing attenuated viruses. Since some individuals cannot receive this vaccine, the development of alternatives is desirable. Here, we developed a recombinant baculovirus (rBV) surface display platform utilizing a chimeric E-NS1 protein as a vaccine candidate. A pBacPAK9 vector containing the baculoviral GP64 signal peptide, the YFV prM, E, NS1 and the ectodomain of VSV-G sequences was synthesized. This transfer plasmid and the bAcGOZA bacmid were cotransfected into Sf9 cells, and an rBV-E-NS1 was obtained, which was characterized by PCR, WB, IFI and FACS analysis. Mice immunized with rBV-E-NS1 elicited a specific humoral and cellular immune response and were protected after YFV infection. In summary, we have developed an rBV that expresses YFV major antigen proteins on its surface, which opens new alternatives that can be tested in a mouse model.


Asunto(s)
Anticuerpos Antivirales , Baculoviridae , Proteínas no Estructurales Virales , Fiebre Amarilla , Virus de la Fiebre Amarilla , Animales , Baculoviridae/genética , Baculoviridae/inmunología , Ratones , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Virus de la Fiebre Amarilla/inmunología , Virus de la Fiebre Amarilla/genética , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/genética , Fiebre Amarilla/prevención & control , Fiebre Amarilla/inmunología , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/genética , Células Sf9 , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Femenino , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/genética , Inmunidad Celular , Ratones Endogámicos BALB C , Inmunidad Humoral , Vectores Genéticos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA