Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Vaccines (Basel) ; 12(8)2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39203972

RESUMEN

The recent worldwide incidence of mpox infection and concerns about future emerging variants of mpox viruses highlight the need for the development of a new generation of mpox vaccines. To achieve this goal, we utilized our norovirus S nanoparticle vaccine platform to produce and evaluate two pseudovirus nanoparticles (PVNPs), S-L1 and S-J1. These PVNPs displayed the L1 neutralizing antigen target of the vaccinia virus and a yet-untested J1 antigen of the mpox virus, respectively, with the aim of creating an effective nanoparticle-based mpox vaccine. Each self-assembled PVNP consists of an inner shell resembling the interior layer of the norovirus capsid and multiple L1 or J1 antigens on the surface. The PVNPs improved the antibody responses toward the displayed L1 or J1 antigens in mice, resulting in significantly greater L1/J1-specific IgG and IgA titers than those elicited by the corresponding free L1 or J1 antigens. After immunization with the S-L1 PVNPs, the mouse sera exhibited high neutralizing antibody titers against the vaccinia virus, and the S-L1 PVNPs provided mice with 100% protection against mortality caused by vaccinia virus challenge. In contrast, the S-J1 PVNPs induced low neutralizing antibody titers and conferred mice weak protective immunity. These data confirm that the L1 protein is an excellent vaccine target and that the readily available S-L1 PVNPs are a promising mpox vaccine candidate worthy of further development.

2.
Sci Rep ; 14(1): 19537, 2024 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-39174674

RESUMEN

Porcine epidemic diarrhea virus (PEDV) is a serious disease that poses a significant threat to the pig industry. This study focused on analyzing the Spike protein of PEDV, which harbors crucial antigenic determinants, in identifying dominant epitopes. Immunoinformatics tools were used to screen for B-cell, CD4+ and CD8+ predominance epitopes. These epitopes were then connected to the N-terminal of ferritin to form a self-assembled nanoparticle vaccine. Various physical and chemical properties of the candidate vaccine were analyzed, including secondary structure prediction, tertiary structure modeling, molecular docking, immune response simulation and computer cloning. The results demonstrated that the candidate vaccine was antigenic, soluble, stable, non-allergic, and formed a stable complex with the target receptor TLR-3. Immune simulation analysis showed that the candidate vaccine effectively stimulated both cellular and humoral reactions, leading to increased related cytokines production. Furthermore, efficient and stable expression of the candidate vaccine was achieved through reverse translation in the Escherichia coli K12 expression system following codon optimization and in silico cloning. The developed nanoparticle candidate vaccine in this study holds promise as an effective PEDV vaccine candidate, offering a new approach for the research, development and improvement of vaccines targeting porcine enteric diarrhea coronavirus.


Asunto(s)
Infecciones por Coronavirus , Inmunoinformática , Virus de la Diarrea Epidémica Porcina , Enfermedades de los Porcinos , Vacunas Virales , Animales , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/virología , Epítopos/inmunología , Epítopos de Linfocito B/inmunología , Epítopos de Linfocito T/inmunología , Inmunoinformática/métodos , Simulación del Acoplamiento Molecular , Virus de la Diarrea Epidémica Porcina/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/química , Porcinos , Enfermedades de los Porcinos/prevención & control , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/virología , Vacunas Virales/inmunología
3.
J Funct Biomater ; 15(8)2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39194667

RESUMEN

With the rapid development of tumor immunotherapy, nanoparticle vaccines have attracted much attention as potential therapeutic strategies. A systematic review and analysis must be carried out to investigate the effect of mannose modification on the immune response to nanoparticles in regulating the tumor microenvironment, as well as to explore its potential clinical application in tumor therapy. Despite the potential advantages of nanoparticle vaccines in immunotherapy, achieving an effective immune response in the tumor microenvironment remains a challenge. Tumor immune escape and the overexpression of immunosuppressive factors limit its clinical application. Therefore, our review explored how to intervene in the immunosuppressive mechanism in the tumor microenvironment through the use of mannan-decorated lipid calcium phosphate nanoparticle vaccines to improve the efficacy of immunotherapy in patients with tumors and to provide new ideas and strategies for the field of tumor therapy.

4.
Int J Mol Sci ; 25(13)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-39000366

RESUMEN

As a highly pathogenic avian virus, H5 influenza poses a serious threat to livestock, the poultry industry, and public health security. Hemagglutinin (HA) is both the dominant epitope and the main target of influenza-neutralizing antibodies. Here, we designed a nanoparticle hemagglutinin influenza vaccine to improve the immunogenicity of the influenza vaccine. In this study, HA5 subtype influenza virus was used as the candidate antigen and was combined with the artificially designed double-branch scaffold protein I53_dn5 A and B. A structurally correct and bioactive trimer HA5-I53_dn5B/Y98F was obtained through secretion and purification using an insect baculovirus expression system; I53_dn5A was obtained by purification using a prokaryotic expression system. HA5-I53_dn5B/Y98F and I53_dn5A self-assembled into spherical nanoparticles (HA5-I53_dn5) in vitro with a diameter of about 45 nm. Immunization and serum test results showed that both HA5-I53_dn5B/Y98F and HA5-I53_dn5 could induce HA5-specific antibodies; however, the immunogenicity of HA5-I53_dn5 was better than that of HA5-I53_dn5B/Y98F. Groups treated with HA5-I53_dn5B and HA5-I53_dn5 nanoparticles produced IgG antibody titers that were not statistically different from those of the nanoparticle-containing adjuvant group. This production of trimerized HA5-I53_dn5B and HA5-I53_dn5 nanoparticles using baculovirus expression provides a reference for the development of novel, safe, and efficient influenza vaccines.


Asunto(s)
Anticuerpos Antivirales , Glicoproteínas Hemaglutininas del Virus de la Influenza , Vacunas contra la Influenza , Nanopartículas , Vacunas contra la Influenza/inmunología , Animales , Nanopartículas/química , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Ratones , Ratones Endogámicos BALB C , Formación de Anticuerpos/inmunología , Femenino , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Humanos
5.
Vet Microbiol ; 296: 110198, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39067145

RESUMEN

Senecavirus A (SVA) is a causative agent that can cause vesicular disease in swine, which causes a great threat to the swine husbandry in the world. Therefore, it is necessary to develop a vaccine that can effectively prevent the spread of SVA. In this study, we developed a 24-polymeric nano-scaffold using ß-annulus peptide from tomato bushy effect virus (TBSV) by coupling this antigen to SVA B cell epitope VP121-26 and VP2 proteins via linkers, respectively. The SVA-based nanoparticle protein of the VP1(B)-ß-VP2 was expressed and purified by low-cost prokaryotic system to prepare a SVA nanoparticle vaccine. The immunological protective effect of SVA nanoparticle vaccine was evaluated in mouse and swine models, respectively. The results suggested that both mice and swine could induce high levels SVA neutralizing antibodies and IgG antibodies after two doses immunization. In addition, the swine challenge protection experiment showed that the protection rate of immune SVA nanoparticle vaccine and SVA inactivated vaccine both were 80 %, while the negative control had no protection effect. It demonstrated that SVA nanoparticle vaccine effectively prevented SVA infection in swine. In summary, the preparation of SVA vaccine by using ß-annulus peptide is a promising candidate vaccine for prevent SVA transmission, and provides a new idea for the development of novel SVA vaccines.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Nanovacunas , Infecciones por Picornaviridae , Picornaviridae , Enfermedades de los Porcinos , Vacunas Virales , Animales , Femenino , Ratones , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Proteínas de la Cápside/inmunología , Ratones Endogámicos BALB C , Nanovacunas/administración & dosificación , Nanovacunas/inmunología , Picornaviridae/inmunología , Infecciones por Picornaviridae/veterinaria , Infecciones por Picornaviridae/prevención & control , Infecciones por Picornaviridae/inmunología , Infecciones por Picornaviridae/virología , Porcinos , Enfermedades de los Porcinos/prevención & control , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/inmunología , Proteínas Estructurales Virales/inmunología , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación
6.
Front Mol Biosci ; 11: 1403635, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38933369

RESUMEN

Since the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) outbreak, several solutions have been proposed to manage the disease. The most viable option for controlling this virus is to produce effective vaccines. Most of the current SARS-CoV-2 vaccines have focused on the infusion spike protein. Spike exists as a trimer and plays a vital role in infecting host cells by binding to the Angiotensin-Converting Enzyme 2 (ACE2) receptor through its Receptor Binding Domain (RBD). Ferritin protein, a naturally occurring iron-storage protein, has gained attention for vaccine production due to its self-assembling property, non-toxic nature, and biocompatibility. Ferritin nanocages have recently been employed in the development of a SARS-CoV-2 vaccination eliciting not only long-term protective memory cells but also a sustained antibody response. In this study, a combination of in silico investigations including molecular docking, molecular dynamics simulations, and immune simulations were carried out to computationally model the monomeric spike protein on the ferritin nanocage as well as to evaluate its stability and interactions for the first time. The structural dynamics of the modeled complex demonstrated noticeable stability. In particular, the Receptor Binding Domain (RBD) and ferritin within the monomeric spike-ferritin complex illustrated significant stability. The lack of alterations in the secondary structure further supported the overall steadiness of the complex. The decline in the distance between ferritin and spike suggests a strong interaction over time. The cross-correlation matrices revealed that the monomeric spike and ferritin move towards each other supporting the stable interaction between spike and ferritin. Further, the orientation of monomeric spike protein within the ferritin unit facilitated the exposure of critical epitopes, specifically upward active Receptor Binding Domain (RBD), enabling effective interactions with the ACE2 receptor. The immune simulations of the model indicated high-level stimulations of both cellular and humoral immunity in the human body. It was also found that the employed model is effective regardless of the mutated spikes in different variants. These findings shed light on the current status of the SARS-CoV-2-ferritin nanoparticle vaccines and could be used as a framework for other similar vaccine designs.

7.
Pharmaceutics ; 16(6)2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38931864

RESUMEN

We describe the current Good Manufacturing Practice (cGMP) production and subsequent characterization of eOD-GT8 60mer, a glycosylated self-assembling nanoparticle HIV-1 vaccine candidate and germline targeting priming immunogen. Production was carried out via transient expression in the human embryonic kidney 293 (HEK293) cell line followed by a combination of purification techniques. A large-scale cGMP (200 L) production run yielded 354 mg of the purified eOD-GT8 60mer drug product material, which was formulated at 1 mg/mL in 10% sucrose in phosphate-buffered saline (PBS) at pH 7.2. The clinical trial material was comprehensively characterized for purity, antigenicity, glycan composition, amino acid sequence, and aggregation and by several safety-related tests during cGMP lot release. A comparison of the purified products produced at the 1 L scale and 200 L cGMP scale demonstrated the consistency and robustness of the transient transfection upstream process and the downstream purification strategies. The cGMP clinical trial material was tested in a Phase 1 clinical trial (NCT03547245), is currently being stored at -80 °C, and is on a stability testing program as per regulatory guidelines. The methods described here illustrate the utility of transient transfection for cGMP production of complex products such as glycosylated self-assembling nanoparticles.

8.
Front Immunol ; 15: 1387811, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38911870

RESUMEN

The Nipah virus (NiV), a highly deadly bat-borne paramyxovirus, poses a substantial threat due to recurrent outbreaks in specific regions, causing severe respiratory and neurological diseases with high morbidity. Two distinct strains, NiV-Malaysia (NiV-M) and NiV-Bangladesh (NiV-B), contribute to outbreaks in different geographical areas. Currently, there are no commercially licensed vaccines or drugs available for prevention or treatment. In response to this urgent need for protection against NiV and related henipaviruses infections, we developed a novel homotypic virus-like nanoparticle (VLP) vaccine co-displaying NiV attachment glycoproteins (G) from both strains, utilizing the self-assembling properties of ferritin protein. In comparison to the NiV G subunit vaccine, our nanoparticle vaccine elicited significantly higher levels of neutralizing antibodies and provided complete protection against a lethal challenge with NiV infection in Syrian hamsters. Remarkably, the nanoparticle vaccine stimulated the production of antibodies that exhibited superior cross-reactivity to homologous or heterologous henipavirus. These findings underscore the potential utility of ferritin-based nanoparticle vaccines in providing both broad-spectrum and long-term protection against NiV and emerging zoonotic henipaviruses challenges.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Ferritinas , Infecciones por Henipavirus , Mesocricetus , Nanopartículas , Virus Nipah , Vacunas Virales , Animales , Virus Nipah/inmunología , Infecciones por Henipavirus/prevención & control , Infecciones por Henipavirus/inmunología , Ferritinas/inmunología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Cricetinae , Vacunas de Partículas Similares a Virus/inmunología , Vacunas de Partículas Similares a Virus/administración & dosificación , Femenino , Humanos , Nanovacunas
9.
Front Immunol ; 15: 1361323, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38835763

RESUMEN

Introduction: Swine influenza viruses (SIVs) pose significant economic losses to the pig industry and are a burden on global public health systems. The increasing complexity of the distribution and evolution of different serotypes of influenza strains in swine herds escalates the potential for the emergence of novel pandemic viruses, so it is essential to develop new vaccines based on swine influenza. Methods: Here, we constructed a self-assembling ferritin nanoparticle vaccine based on the hemagglutinin (HA) extracellular domain of swine influenza A (H1N1) virus using insect baculovirus expression vector system (IBEVS), and after two immunizations, the immunogenicities and protective efficacies of the HA-Ferritin nanoparticle vaccine against the swine influenza virus H1N1 strain in mice and piglets were evaluated. Results: Our results demonstrated that HA-Ferritin nanoparticle vaccine induced more efficient immunity than traditional swine influenza vaccines. Vaccination with the HA-Ferritin nanoparticle vaccine elicited robust hemagglutinin inhibition titers and antigen-specific IgG antibodies and increased cytokine levels in serum. MF59 adjuvant can significantly promote the humoral immunity of HA-Ferritin nanoparticle vaccine. Furthermore, challenge tests showed that HA-Ferritin nanoparticle vaccine conferred full protection against lethal challenge with H1N1 virus and significantly decreased the severity of virus-associated lung lesions after challenge in both BALB/c mice and piglets. Conclusion: Taken together, these results indicate that the hemagglutinin extracellular-based ferritin nanoparticle vaccine may be a promising vaccine candidate against SIVs infection.


Asunto(s)
Anticuerpos Antivirales , Ferritinas , Glicoproteínas Hemaglutininas del Virus de la Influenza , Subtipo H1N1 del Virus de la Influenza A , Vacunas contra la Influenza , Ratones Endogámicos BALB C , Nanopartículas , Infecciones por Orthomyxoviridae , Animales , Subtipo H1N1 del Virus de la Influenza A/inmunología , Ferritinas/inmunología , Vacunas contra la Influenza/inmunología , Porcinos , Ratones , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Enfermedades de los Porcinos/prevención & control , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/virología , Femenino , Nanovacunas
10.
Cell Rep Med ; 5(6): 101614, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38897173

RESUMEN

Proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to the low-density lipoprotein receptor (LDLR) and mediates its internalization and degradation, resulting in an increase in LDL cholesterol levels. Recently, PCSK9 emerged as a therapeutic target for hypercholesterolemia and atherosclerosis. In this study, we develop a PCSK9 nanoparticle (NP) vaccine by covalently conjugating the catalytic domain (aa 153-aa 454, D374Y) of PCSK9 to self-assembled 24-mer ferritin NPs. We demonstrate that the PCSK9 NP vaccine effectively induces interfering antibodies against PCSK9 and reduces serum lipids levels in both a high-fat diet-induced hypercholesterolemia model and an adeno-associated virus-hPCSK9D374Y-induced hypercholesterolemia model. Additionally, the vaccine significantly reduces plaque lesion areas in the aorta and macrophages infiltration in an atherosclerosis mouse model. Furthermore, we discover that the vaccine's efficacy relied on T follicular help cells and LDLR. Overall, these findings suggest that the PCSK9 NP vaccine holds promise as an effective treatment for hypercholesterolemia and atherosclerosis.


Asunto(s)
Aterosclerosis , Modelos Animales de Enfermedad , Hipercolesterolemia , Nanopartículas , Proproteína Convertasa 9 , Receptores de LDL , Vacunas , Proproteína Convertasa 9/inmunología , Proproteína Convertasa 9/metabolismo , Animales , Hipercolesterolemia/patología , Nanopartículas/química , Vacunas/inmunología , Ratones , Receptores de LDL/metabolismo , Aterosclerosis/prevención & control , Aterosclerosis/inmunología , Aterosclerosis/patología , Ratones Endogámicos C57BL , Humanos , Dieta Alta en Grasa , Masculino , Nanovacunas
11.
Front Immunol ; 15: 1372349, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38698863

RESUMEN

Pseudomonas aeruginosa (Pa) is an opportunistic bacterial pathogen responsible for severe hospital acquired infections in immunocompromised and elderly individuals. Emergence of increasingly drug resistant strains and the absence of a broad-spectrum prophylactic vaccine against both T3SA+ (type III secretion apparatus) and ExlA+/T3SA- Pa strains worsen the situation in a post-pandemic world. Thus, we formulated a candidate subunit vaccine (called ExlA/L-PaF/BECC/ME) against both Pa types. This bivalent vaccine was generated by combining the C-terminal active moiety of exolysin A (ExlA) produced by non-T3SA Pa strains with our T3SA-based vaccine platform, L-PaF, in an oil-in-water emulsion. The ExlA/L-PaF in ME (MedImmune emulsion) was then mixed with BECC438b, an engineered lipid A analogue and a TLR4 agonist. This formulation was administered intranasally (IN) to young and elderly mice to determine its potency across a diverse age-range. The elderly mice were used to mimic the infection seen in elderly humans, who are more susceptible to serious Pa disease compared to their young adult counterparts. After Pa infection, mice immunized with ExlA/L-PaF/BECC/ME displayed a T cell-mediated adaptive response while PBS-vaccinated mice experienced a rapid onset inflammatory response. Important genes and pathways were observed, which give rise to an anti-Pa immune response. Thus, this vaccine has the potential to protect aged individuals in our population from serious Pa infection.


Asunto(s)
Emulsiones , Infecciones por Pseudomonas , Vacunas contra la Infección por Pseudomonas , Pseudomonas aeruginosa , Vacunas de Subunidad , Animales , Pseudomonas aeruginosa/inmunología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Ratones , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/prevención & control , Vacunas contra la Infección por Pseudomonas/inmunología , Vacunas contra la Infección por Pseudomonas/administración & dosificación , Femenino , Desarrollo de Vacunas , Humanos , Anticuerpos Antibacterianos/inmunología , Anticuerpos Antibacterianos/sangre , Modelos Animales de Enfermedad , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/genética
12.
Vaccines (Basel) ; 12(5)2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38793797

RESUMEN

Malaria is caused by eukaryotic protozoan parasites of the genus Plasmodium. There are 249 million new cases and 608,000 deaths annually, and new interventions are desperately needed. Malaria vaccines can be divided into three categories: liver stage, blood stage, or transmission-blocking vaccines. Transmission-blocking vaccines prevent the transmission of disease by the mosquito vector from one human to another. Pfs230 is one of the leading transmission-blocking vaccine antigens for malaria. Here, we describe the development of a 24-copy self-assembling nanoparticle vaccine comprising domain 1 of Pfs230 genetically fused to H. pylori ferritin. The single-component Pfs230D1-ferritin construct forms a stable and homogenous 24-copy nanoparticle with good production yields. The nanoparticle is highly immunogenic, as two low-dose vaccinations of New Zealand White rabbits elicited a potent and durable antibody response with high transmission-reducing activity when formulated in two distinct adjuvants suitable for translation to human use. This single-component 24-copy Pfs230D1-ferritin nanoparticle vaccine has the potential to improve production pipelines and the cost of manufacturing a potent and durable transmission-blocking vaccine for malaria control.

13.
ACS Nano ; 18(19): 12235-12260, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38696217

RESUMEN

Variants of coronavirus porcine epidemic diarrhea virus (PEDV) frequently emerge, causing an incomplete match between the vaccine and variant strains, which affects vaccine efficacy. Designing vaccines with rapidly replaceable antigens and high efficacy is a promising strategy for the prevention of infection with PEDV variant strains. In our study, three different types of self-assembled nanoparticles (nps) targeting receptor-binding N-terminal domain (NTD) and C-terminal domain (CTD) of S1 protein, named NTDnps, CTDnps, and NTD/CTDnps, were constructed and evaluated as vaccine candidates against PEDV. NTDnps and CTDnps vaccines mediated significantly higher neutralizing antibody (NAb) titers than NTD and CTD recombinant proteins in mice. The NTD/CTDnps in varying ratios elicited significantly higher NAb titers when compared with NTDnps and CTDnps alone. The NTD/CTDnps (3:1) elicited NAb with titers up to 92.92% of those induced by the commercial vaccine. Piglets immunized with NTD/CTDnps (3:1) achieved a passive immune protection rate of 83.33% of that induced by the commercial vaccine. NTD/CTDnps (3:1) enhanced the capacity of mononuclear macrophages and dendritic cells to take up and present antigens by activating major histocompatibility complex I and II molecules to stimulate humoral and cellular immunity. These data reveal that a combination of S1-NTD and S1-CTD antigens targeting double receptor-binding domains strengthens the protective immunity of nanoparticle vaccines against PEDV. Our findings will provide a promising vaccine candidate against PEDV.


Asunto(s)
Nanopartículas , Virus de la Diarrea Epidémica Porcina , Vacunas Virales , Virus de la Diarrea Epidémica Porcina/inmunología , Animales , Nanopartículas/química , Porcinos , Ratones , Vacunas Virales/inmunología , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/inmunología , Ratones Endogámicos BALB C , Antígenos Virales/inmunología , Antígenos Virales/química , Anticuerpos Neutralizantes/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/química , Dominios Proteicos/inmunología , Femenino , Nanovacunas
14.
Front Immunol ; 15: 1328266, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38550592

RESUMEN

Background: Porcine deltacoronavirus (PDCoV), a novel swine enteropathogenic coronavirus, challenges the global swine industry. Currently, there are no approaches preventing swine from PDCoV infection. Methods: A new PDCoV strain named JS2211 was isolated. Next, the dimer receptor binding domain of PDCoV spike protein (RBD-dimer) was expressed using the prokaryotic expression system, and a novel nanoparticle containing RBD-dimer and ferritin (SC-Fe) was constructed using the SpyTag/SpyCatcher system. Finally, the immunoprotection of RBD-Fe nanoparticles was evaluated in mice. Results: The novel PDCoV strain was located in the clade of the late Chinese isolate strains and close to the United States strains. The RBD-Fe nanoparticles were successfully established. Immune responses of the homologous prime-boost regime showed that RBD-Fe nanoparticles efficiently elicited specific humoral and cellular immune responses in mice. Notably, high level PDCoV RBD-specific IgG and neutralizing antibody (NA) could be detected, and the histopathological results showed that PDCoV infection was dramatically reduced in mice immunized with RBD-Fe nanoparticles. Conclusion: This study effectively developed a candidate nanoparticle with receptor binding domain of PDCoV spike protein that offers protection against PDCoV infection in mice.


Asunto(s)
Nanovacunas , Glicoproteína de la Espiga del Coronavirus , Porcinos , Animales , Ratones , Deltacoronavirus , Inmunidad , SARS-CoV-2
15.
Int J Biol Macromol ; 264(Pt 2): 130820, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38484812

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and its variants has resulted in global economic losses and posed a threat to human health. The pandemic highlights the urgent need for an efficient, easily producible, and broad-spectrum vaccine. Here, we present a potentially universal strategy for the rapid and general design of vaccines, focusing on the design and testing of omicron BA.5 RBD-conjugated self-assembling ferritin nanoparticles (NPs). The covalent bonding of RBD-Fc to protein A-ferritin was easily accomplished through incubation, resulting in fully multivalent RBD-conjugated NPs that exhibited high structural uniformity, stability, and efficient assembly. The ferritin nanoparticle vaccine synergistically stimulated the innate immune response, Tfh-GCB-plasma cell-mediated activation of humoral immunity and IFN-γ-driven cellular immunity. This nanoparticle vaccine induced a high level of cross-neutralizing responses and protected golden hamsters challenged with multiple mutant strains from infection-induced clinical disease, providing a promising strategy for broad-spectrum vaccine development for SARS-CoV-2 prophylaxis. In conclusion, the nanoparticle conjugation platform holds promise for its potential universality and competitive immunization efficacy and is expected to facilitate the rapid manufacturing and broad application of next-generation vaccines.


Asunto(s)
COVID-19 , Nanopartículas , Animales , Cricetinae , Humanos , SARS-CoV-2 , COVID-19/prevención & control , Vacunas contra la COVID-19 , Inmunidad Innata , Ferritinas/genética , Nanovacunas , Anticuerpos Neutralizantes , Anticuerpos Antivirales
16.
J Virol ; 98(3): e0120623, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38305154

RESUMEN

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with greater transmissibility or immune evasion properties has jeopardized the existing vaccine and antibody-based countermeasures. Here, we evaluated the efficacy of boosting pre-immune hamsters with protein nanoparticle vaccines (Novavax, Inc.) containing recombinant Prototype (Wuhan-1) or BA.5 S proteins against a challenge with the Omicron BA.5 variant of SARS-CoV-2. Serum antibody binding and neutralization titers were quantified before challenge, and viral loads were measured 3 days after challenge. Boosting with Prototype or BA.5 vaccine induced similar antibody binding responses against ancestral Wuhan-1 or BA.5 S proteins, and neutralizing activity of Omicron BA.1 and BA.5 variants. One and three months after vaccine boosting, hamsters were challenged with the Omicron BA.5 variant. Prototype and BA.5 vaccine-boosted hamsters had reduced viral infection in the nasal washes, nasal turbinates, and lungs compared to unvaccinated animals. Although no significant differences in virus load were detected between the Prototype and BA.5 vaccine-boosted animals, fewer breakthrough infections were detected in the BA.5-vaccinated hamsters. Thus, immunity induced by Prototype or BA.5 S protein nanoparticle vaccine boosting can protect against the Omicron BA.5 variant in the Syrian hamster model. IMPORTANCE: As SARS-CoV-2 continues to evolve, there may be a need to update the vaccines to match the newly emerging variants. Here, we compared the protective efficacy of the updated BA.5 and the original Wuhan-1 COVID-19 vaccine against a challenge with the BA.5 Omicron variant of SARS-CoV-2 in hamsters. Both vaccines induced similar levels of neutralizing antibodies against multiple variants of SARS-CoV-2. One and three months after the final immunization, hamsters were challenged with BA.5. No differences in protection against the BA.5 variant virus were observed between the two vaccines, although fewer breakthrough infections were detected in the BA.5-vaccinated hamsters. Together, our data show that both protein nanoparticle vaccines are effective against the BA.5 variant of SARS-CoV-2 but given the increased number of breakthrough infections and continued evolution, it is important to update the COVID-19 vaccine for long-term protection.


Asunto(s)
Vacunas contra la COVID-19 , Nanovacunas , SARS-CoV-2 , Animales , Cricetinae , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Infección Irruptiva/inmunología , Infección Irruptiva/prevención & control , Infección Irruptiva/virología , COVID-19/inmunología , COVID-19/prevención & control , COVID-19/virología , Vacunas contra la COVID-19/inmunología , Mesocricetus/inmunología , Mesocricetus/virología , Nanovacunas/inmunología , SARS-CoV-2/inmunología , Inmunización Secundaria , Carga Viral
17.
Adv Sci (Weinh) ; 11(9): e2303366, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38105421

RESUMEN

To combat SARS-CoV-2 variants and MERS-CoV, as well as the potential re-emergence of SARS-CoV and spillovers of sarbecoviruses, which pose a significant threat to global public health, vaccines that can confer broad-spectrum protection against betacoronaviruses (ß-CoVs) are urgently needed. A mosaic ferritin nanoparticle vaccine is developed that co-displays the spike receptor-binding domains of SARS-CoV, MERS-CoV, and SARS-CoV-2 Wild-type (WT) strain and evaluated its immunogenicity and protective efficacy in mice and nonhuman primates. A low dose of 10 µg administered at a 21-day interval induced a Th1-biased immune response in mice and elicited robust cross-reactive neutralizing antibody responses against a variety of ß-CoVs, including a series of SARS-CoV-2 variants. It is also able to effectively protect against challenges of SARS-CoV, MERS-CoV, and SARS-CoV-2 variants in not only young mice but also the more vulnerable mice through induction of long-lived immunity. Together, these results suggest that this mosaic 3-RBD nanoparticle has the potential to be developed as a pan-ß-CoV vaccine.


Asunto(s)
Infecciones por Coronavirus , Coronavirus del Síndrome Respiratorio de Oriente Medio , Nanopartículas , Vacunas Virales , Humanos , Animales , Ratones , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Infecciones por Coronavirus/prevención & control , SARS-CoV-2 , Coronavirus del Síndrome Respiratorio de Oriente Medio/química , Modelos Animales
18.
Vaccine ; 42(2): 111-119, 2024 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-38097456

RESUMEN

The first influenza virus infection (imprinting) can lead to long-term immune memory and influence subsequent vaccinations and infections. Previously, we reported a polyethyleneimine (PEI)-Aichi hemagglutinin (HA)/CpG (PHC) nanoparticle with cross-protective potential against homologous and heterologous influenza strains. Here we studied how influenza immune imprinting influences the antibody responses to the PHC vaccination and the protection against heterosubtypic virus challenges. We found that pre-existing virus immunity can maintain or synergize the vaccine-induced antibody titers, depending on the imprinting virus HA phylogenetic group. The HA group 1 virus (PR8, H1N1)-imprinted mice displayed comparable antigen-specific antibody responses to those without imprinting post-PHC vaccination. In contrast, the group 2 virus (Phi, H3N2)-imprinted mice showed significantly more robust and balanced antibodies post-vaccination, conferring complete protection against body weight loss and lung inflammation upon heterosubtypic reassortant A/Shanghai/2/2013 (rSH, H7N9) virus challenge. Our findings suggest that influenza imprinting from the same HA phylogenetic group can synergize subsequent vaccination, conferring heterosubtypic protection.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Subtipo H7N9 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Infecciones por Orthomyxoviridae , Animales , Ratones , Humanos , Gripe Humana/prevención & control , Hemaglutininas , Nanovacunas , Polietileneimina , Subtipo H3N2 del Virus de la Influenza A , Filogenia , Anticuerpos Antivirales , China , Glicoproteínas Hemaglutininas del Virus de la Influenza , Ratones Endogámicos BALB C
19.
J Nanobiotechnology ; 21(1): 479, 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-38093320

RESUMEN

Vaccination is still the most promising strategy for combating influenza virus pandemics. However, the highly variable characteristics of influenza virus make it difficult to develop antibody-based universal vaccines, until now. Lung tissue-resident memory T cells (TRM), which actively survey tissues for signs of infection and react rapidly to eliminate infected cells without the need for a systemic immune reaction, have recently drawn increasing attention towards the development of a universal influenza vaccine. We previously designed a sequential immunization strategy based on orally administered Salmonella vectored vaccine candidates. To further improve our vaccine design, in this study, we used two different dendritic cell (DC)-targeting strategies, including a single chain variable fragment (scFv) targeting the surface marker DC-CD11c and DC targeting peptide 3 (DCpep3). Oral immunization with Salmonella harboring plasmid pYL230 (S230), which displayed scFv-CD11c on the bacterial surface, induced dramatic production of spleen effector memory T cells (TEM). On the other hand, intranasal boost immunization using purified DCpep3-decorated 3M2e-ferritin nanoparticles in mice orally immunized twice with S230 (S230inDC) significantly stimulated the differentiation of lung CD11b+ DCs, increased intracellular IL-17 production in lung CD4+ T cells and elevated chemokine production in lung sections, such as CXCL13 and CXCL15, as determined by RNAseq and qRT‒PCR assays, resulting in significantly increased percentages of lung TRMs, which could provide efficient protection against influenza virus challenge. The dual DC targeting strategy, together with the sequential immunization approach described in this study, provides us with a novel "prime and pull" strategy for addressing the production of protective TRM cells in vaccine design.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Virus de la Influenza A , Vacunas contra la Influenza , Infecciones por Orthomyxoviridae , Ratones , Animales , Células T de Memoria , Pulmón , Células Dendríticas , Infecciones por Orthomyxoviridae/prevención & control
20.
Int J Biol Macromol ; 253(Pt 5): 127159, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-37778577

RESUMEN

SARS-CoV-2 is a particularly transmissible virus that renders the worldwide COVID-19 pandemic and global severe respiratory distress syndrome. Protein-based vaccines hold great advantages to build the herd immunity for their specificity, effectiveness, and safety. Receptor-binding domain (RBD) of SARS-CoV-2 is an appealing antigen for vaccine development. However, adjuvants and delivery system are necessitated to enhance the immunogenicity of RBD. In the present study, RBD was chemically conjugated with loxoribine and SpyCatcher/SpyTag, followed by assembly to form a nanoparticle vaccine. Loxoribine (a TLR7/8 agonist) acted as an adjuvant, and nanoparticles functioned as delivery system for the antigen and the adjuvant. The nanoparticle vaccine elicited high RBD-specific antibody titers, high neutralizing antibody titer, and strong ACE2-blocking activity. It stimulated high splenic levels of Th1-type cytokines (IFN-γ and IL-2) and Th2-type cytokines (IL-4 and IL-5) in BALB/c mice. It promoted the splenocyte proliferation, enhanced the CD4+ and CD8+ T cell percentage and stimulated the maturation of dendritic cells. The vaccine did not render apparent toxicity to the organs of mice. Thus, the nanoparticle vaccine was of potential to act as a preliminarily safe and effective candidate against SARS-CoV-2.


Asunto(s)
COVID-19 , Nanopartículas , Animales , Humanos , Ratones , Vacunas contra la COVID-19 , SARS-CoV-2 , COVID-19/prevención & control , Pandemias , Adyuvantes Inmunológicos , Adyuvantes Farmacéuticos , Citocinas , Ratones Endogámicos BALB C , Anticuerpos Neutralizantes , Anticuerpos Antivirales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA