Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Tipo de estudo
Intervalo de ano de publicação
1.
Adv Healthc Mater ; : e2304575, 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38436662

RESUMO

The Coronavirus Disease 2019 (COVID-19) pandemic caused by SARS-CoV-2 has a significant impact on global health and the economy. It has underscored the urgent need for a stable, easily produced and effective vaccine. This study presents a novel approach using SARS-CoV-2 spike (S) protein-conjugated nanoparticles (NPs) in combination with cyclic GMP-AMP (cGAMP) (S-NPs-cGAMP) as a subunit vaccine. When mice are immunized, the antiserum of S-NPs-cGAMP group exhibits a 16-fold increase in neutralizing activity against a pseudovirus, compared to S protein group. Additionally, S-NPs-cGAMP induces even higher levels of neutralizing antibodies. Remarkably, the vaccine also triggers a robust humoral immune response, as evidenced by a notable elevation in virus-specific IgG and IgM antibodies. Furthermore, after 42 days of immunization, there is an observed increase in specific immune cell populations in the spleen. CD3+ CD4+ and CD3+ CD8+ T lymphocytes, as well as B220+ CD19+ and CD3- CD49b+ NK lymphocytes, show an upward trend, indicating a positive cellular immune response. Moreover, the S-NPs-cGAMP demonstrates promising results against the Delta strain and exhibits good cross-neutralization potential against other variants. These findings suggest that pDMDAAC NPs is potential adjuvant and could serve as a versatile platform for future vaccine development.

2.
Vaccine ; 42(4): 828-839, 2024 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-38220489

RESUMO

Porcine epidemic diarrhea virus (PEDV) has caused serious economic losses to the pig husbandry worldwide, and the effects of existing commercialized vaccines are suboptimal. Therefore, research to develop an efficacious vaccine for prevention and control of PEDV is essential. In this study, we designed and produced trimerized proteins of full-length PEDV spike (S) protein, S1 subunit, and a tandem of multiple epitopes of S protein using an efficient mammalian expression vector system in HEK 293F cells. The immunogenicity of two commercial adjuvants, M401 and M103, was also evaluated in mice. Enzyme-linked immunosorbent assays demonstrated that all immunized mice generated highly systemic PEDV S-specific IgG and IgA antibodies. Mice in S/M103-immunized group generated the highest neutralizing antibody titer with 1:96. Compared with control group, the subunit vaccines elicited multifunctional CD3+CD4+ and CD3+CD8+ T cells, B220+CD19+ B cells, and CD3-CD49b+ natural killer cells in the spleen. PEDV S/M103 vaccine, which had the best immune effect, was selected for further evaluation in piglets. Immunization with S/M103 vaccine induced high levels of S-specific IgG, IgA, and neutralizing antibodies, and increased the proliferation of peripheral blood mononuclear cells and the expression levels of interferon-γ and interleukin-4 in peripheral blood of piglets. Virus challenge test results showed significantly lower diarrheal index scores and fecal viral loads, and less pathological damage to the intestines in S/M103-immunized piglets than in controls, indicating that S/M103 provides good protection against the virulent virus challenge. Our findings suggest that trimeric PEDV S/M103 has potential as a clinical vaccine candidate.


Assuntos
Infecções por Coronavirus , Vírus da Diarreia Epidêmica Suína , Doenças dos Suínos , Vacinas Virais , Animais , Suínos , Camundongos , Anticorpos Antivirais , Vacinas de Subunidades Proteicas , Linfócitos T CD8-Positivos , Leucócitos Mononucleares , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/veterinária , Vacinas de Subunidades Antigênicas , Imunoglobulina A , Imunoglobulina G , Glicoproteína da Espícula de Coronavírus , Mamíferos
3.
J Virol ; 97(10): e0106323, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37732788

RESUMO

IMPORTANCE: Porcine epidemic diarrhea (PED) caused by PED virus (PEDV) remains a big threat to the swine industry worldwide. Vaccination with live attenuated vaccine is a promising method to prevent and control PED, because it can elicit a more protective immunity than the killed vaccine, subunit vaccine, and so on. In this study, we found two obvious deletions in the genome of a high passage of AH2012/12. We further confirmed the second deletion which contains seven amino acids at the carboxy-terminus of the S2 gene and the start codon of ORF3 can reduce its pathogenicity in vivo. Animal experiments indicated that the recombinant PEDV with deleted carboxy-terminus of S gene showed higher IgG, IgA, neutralization antibodies, and protection effects against virus challenge than the killed vaccine. These data reveal that the engineering of the carboxy-terminus of the S2 gene may be a promising method to develop live attenuated vaccine candidates of PEDV.


Assuntos
Infecções por Coronavirus , Vírus da Diarreia Epidêmica Suína , Doenças dos Suínos , Animais , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/virologia , Diarreia , Vírus da Diarreia Epidêmica Suína/genética , Vírus da Diarreia Epidêmica Suína/patogenicidade , Suínos , Doenças dos Suínos/virologia , Vacinas Atenuadas/genética , Vacinas de Produtos Inativados , Vacinas Virais/genética , Virulência
4.
ACS Appl Mater Interfaces ; 15(25): 29982-29997, 2023 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-37330942

RESUMO

The COVID-19 pandemic continues to spread worldwide. To protect and control the spread of SARS-CoV-2, varieties of subunit vaccines based on spike (S) proteins have been approved for human applications. Here, we report a new subunit vaccine design strategy that functions as both an antigen carrier and an adjuvant in immunization to elicit high-level immune responses. The complex of 2-hydroxypropyl-trimethylammonium chloride chitosan and amylose entangles Au nanoparticles (HTCC/amylose/AuNPs) forming 40 nm nanocarriers with a positive charge. The obtained positively charged nanoparticles reveal many merits, including the larger S protein loading capacity in PBS buffer, higher cellular uptake ability, and lower cell cytotoxicity, supporting their potential as safe vaccine nanocarriers. Two functionalized nanoparticle subunit vaccines are prepared via loading full-length S proteins derived from SARS-CoV-2 variants. In mice, both prepared vaccines elicit high specific IgG antibodies, neutralize antibodies, and immunoglobulin IgG1 and IgG2a. The prepared vaccines also elicit robust T- and B-cell immune responses and increase CD19+ B cells, CD11C+ dendritic cells, and CD11B+ macrophages at the alveoli and bronchi of the immunized mice. Furthermore, the results of skin safety tests and histological observation of organs indicated in vivo safety of HTCC/amylose/AuNP-based vaccines. Summarily, our prepared HTCC/amylose/AuNP have significant potential as general vaccine carriers for the delivery of different antigens with potent immune stimulation.


Assuntos
COVID-19 , Nanopartículas Metálicas , Humanos , Animais , Camundongos , Vacinas contra COVID-19 , Amilose , Ouro , SARS-CoV-2/metabolismo , Pandemias , COVID-19/prevenção & controle , Adjuvantes Imunológicos/farmacologia , Antígenos , Adjuvantes Farmacêuticos , Imunoglobulina G , Vacinas de Subunidades Antigênicas
5.
Vet Microbiol ; 280: 109718, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36871521

RESUMO

The interferon-delta family was first reported in domestic pigs and belongs to the type I interferon (IFN-I) family. The enteric viruses could cause diarrhea in newborn piglets with high morbidity and mortality. We researched the function of the porcine IFN-delta (PoIFN-δ) family in the porcine intestinal epithelial cells (IPEC-J2) cells infected with porcine epidemic diarrhea virus (PEDV). Our study found that all PoIFN-δs shared a typical IFN-I signature and could be divided into five branches in the phylogenic tree. Different strains of PEDV could induce typical IFN transitorily, and the virulent strain AH2012/12 had the strongest induction of porcine IFN-δ and IFN-alpha (PoIFN-α) in the early stage of infection. In addition, it was found that PoIFN-δ5/6/9/11 and PoIFN-δ1/2 were highly expressed in the intestine. PoIFN-δ5 had a better antiviral effect on PEDV compared to PoIFN-δ1 due to its higher induction of ISGs. PoIFN-δ1 and PoIFN-δ5 also activated JAK-STAT and IRS signaling. For other enteric viruses, transmissible gastroenteritis virus (TGEV), porcine deltacoronavirus (PDCoV), and porcine rotavirus (PoRV), PoIFN-δ1 and PoIFN-δ5 both showed an excellent antiviral effect. Transcriptome analyses uncovered the differences in host responses to PoIFN-α and PoIFN-δ5 and revealed thousands of differentially expressed genes were mainly enriched in the inflammatory response, antigen processing and presentation, and other immune-related pathways. PoIFN-δ5 would be a potential antiviral drug, especially against porcine enteric viruses. These studies were the first to report the antiviral function against porcine enteric viruses and broaden the new acquaintances of this type of interferon though not novelly discovered.


Assuntos
Infecções por Coronavirus , Enterovirus Suínos , Vírus da Diarreia Epidêmica Suína , Doenças dos Suínos , Animais , Suínos , Antivirais/farmacologia , Antivirais/uso terapêutico , Transcriptoma , Intestinos , Células Epiteliais , Interferon-alfa/farmacologia , Perfilação da Expressão Gênica/veterinária , Infecções por Coronavirus/veterinária
6.
J Virol ; 96(18): e0102422, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-36037478

RESUMO

Zoonotic coronaviruses represent an ongoing threat to public health. The classical porcine epidemic diarrhea virus (PEDV) first appeared in the early 1970s. Since 2010, outbreaks of highly virulent PEDV variants have caused great economic losses to the swine industry worldwide. However, the strategies by which PEDV variants escape host immune responses are not fully understood. Complement component 3 (C3) is considered a central component of the three complement activation pathways and plays a crucial role in preventing viral infection. In this study, we found that C3 significantly inhibited PEDV replication in vitro, and both variant and classical PEDV strains induced high levels of interleukin-1ß (IL-1ß) in Huh7 cells. However, the PEDV variant strain reduces C3 transcript and protein levels induced by IL-1ß compared with the PEDV classical strain. Examination of key molecules of the C3 transcriptional signaling pathway revealed that variant PEDV reduced C3 by inhibiting CCAAT/enhancer-binding protein ß (C/EBP-ß) phosphorylation. Mechanistically, PEDV nonstructural protein 1 (NSP1) inhibited C/EBP-ß phosphorylation via amino acid residue 50. Finally, we constructed recombinant PEDVs to verify the critical role of amino acid 50 of NSP1 in the regulation of C3 expression. In summary, we identified a novel antiviral role of C3 in inhibiting PEDV replication and the viral immune evasion strategies of PEDV variants. Our study reveals new information on PEDV-host interactions and furthers our understanding of the pathogenic mechanism of this virus. IMPORTANCE The complement system acts as a vital link between the innate and the adaptive immunity and has the ability to recognize and neutralize various pathogens. Activation of the complement system acts as a double-edged sword, as appropriate levels of activation protect against pathogenic infections, but excessive responses can provoke a dramatic inflammatory response and cause tissue damage, leading to pathological processes, which often appear in COVID-19 patients. However, how PEDV, as the most severe coronavirus causing diarrhea in piglets, regulates the complement system has not been previously reported. In this study, for the first time, we identified a novel mechanism of a PEDV variant in the suppression of C3 expression, showing that different coronaviruses and even different subtype strains differ in regulation of C3 expression. In addition, this study provides a deeper understanding of the mechanism of the PEDV variant in immune escape and enhanced virulence.


Assuntos
Complemento C3 , Infecções por Coronavirus , Vírus da Diarreia Epidêmica Suína , Doenças dos Suínos , Proteínas não Estruturais Virais , Replicação Viral , Animais , Antivirais , COVID-19/imunologia , Linhagem Celular Tumoral , Complemento C3/imunologia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Humanos , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/virologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/fisiologia
7.
Vet Microbiol ; 267: 109376, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35217353

RESUMO

Porcine epidemic diarrhea virus (PEDV) is a highly contagious enteropathogenic coronavirus causing severe watery diarrhea and high mortality in piglets. In order to investigate the role of the transcription regulatory sequences (TRSs) in regulation of gene expression and replication of PEDV, the enhanced green fluorescent protein (EGFP) gene, under control of different TRSs of PEDV, were inserted between the N gene and 3' UTR of the PEDV genome using a reverse genetic system. The EGFP expression from different chimeric PEDVs was analyzed for each TRS. TRSs of all the structural and accessory protein genes of PEDV positively regulate EGFP expression at different levels, and the TRS of M protein gene produced the highest level of EGFP. Moreover, this is the first study to show that exogenous gene could be inserted between N gene and 3' UTR of PEDV, and the EGFP insertion had no effect on PEDV replication. Taken together, our study enriched the information of PEDV TRSs on gene expression and replication of PEDV.


Assuntos
Infecções por Coronavirus , Vírus da Diarreia Epidêmica Suína , Doenças dos Suínos , Animais , Infecções por Coronavirus/veterinária , Diarreia/veterinária , Regulação da Expressão Gênica , Vírus da Diarreia Epidêmica Suína/genética , Suínos
8.
Polymers (Basel) ; 12(10)2020 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-33066097

RESUMO

Docetaxel (DTX), as a first-line anti-tumor drug, has been studied for decades for its diverse bioactivities. However, DTX presents poor solubility in water, low bioavailability and serious toxic side effects which has hindered its application in the clinic. To address these problems, docetaxel-sulfobutyl ether-ß-cyclodextrin inclusion complex (DTX-SBE-ß-CD) was prepared successfully by saturated aqueous solution method. Sulfobutyl ether ß-cyclodetrin (SBE-ß-CD) is used as delivery material. For this study, the inclusion complex of docetaxel with sulfobutyl ether ß-cyclodetrin (DTX-SBE-ß-CD) was prepared and optimized its properties to enhance the cytotoxicity of cancer cells. A large number of physical characterization results showed that DTX-SBE-ß-CD inclusion complex was successfully prepared by saturated aqueous solution method. DTX-SBE-ß-CD inclusion complex was optimized by Central Composite Design. DTX-SBE-ß-CD had an inhibitory effect on the in vitro determination of MCF-7 and HepG2 cells by MTT assay. Pharmacokinetic studies were carried out on male Sprague-Dawley rats by tail injection, including the distribution, metabolism and elimination of DTX-SBE-ß-CD in vivo. In the experimental study of inhibition of cancer cells, DTX and DTX-SBE-ß-CD showed apparent concentration-dependent inhibitory actions on tumor cells and the inhibition of DTX-SBE-ß-CD group was more obvious.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA