Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.126
Filter
Add more filters

Complementary Medicines
Publication year range
1.
ACS Biomater Sci Eng ; 10(5): 3387-3400, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38656158

ABSTRACT

Given the worldwide problem posed by enteric pathogens, the discovery of safe and efficient intestinal adjuvants combined with novel antigen delivery techniques is essential to the design of mucosal vaccines. In this work, we designed poly (lactic-co-glycolic acid) (PLGA)-based nanoparticles (NPs) to codeliver all-trans retinoic acid (atRA), novel antigens, and CpG. To address the insolubility of the intestinal adjuvant atRA, we utilized PLGA to encapsulate atRA and form a "nanocapsid" with polydopamine. By leveraging polydopamine, we adsorbed the water-soluble antigens and the TLR9 agonist CpG onto the NPs' surface, resulting in the pathogen-mimicking PLPCa NPs. In this study, the novel fusion protein (HBf), consisting of the Mycobacterium avium subspecies paratuberculosis antigens HBHA, Ag85B, and Bfra, was coloaded onto the NPs. In vitro, PLPCa NPs were shown to promote the activation and maturation of bone marrow-derived dendritic cells. Additionally, we found that PLPCa NPs created an immune-rich microenvironment at the injection site following intramuscular administration. From the results, the PLPCa NPs induced strong IgA levels in the gut in addition to enhancing powerful systemic immune responses. Consequently, significant declines in the bacterial burden and inflammatory score were noted in PLPCa NPs-treated mice. In summary, PLPCa can serve as a novel and safe vaccine delivery platform against gut pathogens, such as paratuberculosis, capable of activating both systemic and intestinal immunity.


Subject(s)
Nanoparticles , Paratuberculosis , Animals , Nanoparticles/chemistry , Paratuberculosis/immunology , Paratuberculosis/prevention & control , Mice , Tretinoin/chemistry , Tretinoin/pharmacology , Mycobacterium avium subsp. paratuberculosis/immunology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Antigens, Bacterial/immunology , Antigens, Bacterial/chemistry , Dendritic Cells/immunology , Dendritic Cells/drug effects , Intestines/immunology , Intestines/microbiology , Mice, Inbred C57BL , Female , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/administration & dosage , Bacterial Vaccines/immunology , Mice, Inbred BALB C
2.
ACS Infect Dis ; 10(5): 1552-1560, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38623820

ABSTRACT

Tyrosine cross-linking has recently been used to produce nanoclusters (NCs) from peptides to enhance their immunogenicity. In this study, NCs were generated using the ectodomain of the ion channel Matrix 2 (M2e) protein, a conserved influenza surface antigen. The NCs were administered via intranasal (IN) or intramuscular (IM) routes in a mouse model in a prime-boost regimen in the presence of the adjuvant CpG. After boost, a significant increase in anti-M2e IgG and its subtypes was observed in the serum and lungs of mice vaccinated through the IM and IN routes; however, significant enhancement in anti-M2e IgA in lungs was observed only in the IN group. Analysis of cytokine concentrations in stimulated splenocyte cultures indicated a Th1/Th17-biased response. Mice were challenged with a lethal dose of A/California/07/2009 (H1N1pdm), A/Puerto Rico/08/1934 (H1N1), or A/Hong Kong/08/1968 (H3N2) strains. Mice that received M2e NCs + CpG were significantly protected against these strains and showed decreased lung viral titers compared with the naive mice and M2e NC-alone groups. The IN-vaccinated group showed superior protection against the H3N2 strain as compared to the IM group. This research extends our earlier efforts involving the tyrosine-based cross-linking method and highlights the potential of this technology in enhancing the immunogenicity of short peptide immunogens.


Subject(s)
Antibodies, Viral , Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Orthomyxoviridae Infections , Tyrosine , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Mice , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Tyrosine/chemistry , Tyrosine/pharmacology , Influenza A Virus, H1N1 Subtype/immunology , Female , Antibodies, Viral/blood , Antibodies, Viral/immunology , Viral Matrix Proteins/immunology , Viral Matrix Proteins/genetics , Mice, Inbred BALB C , Influenza A Virus, H3N2 Subtype/immunology , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Lung/virology , Lung/immunology , Administration, Intranasal , Injections, Intramuscular , Cytokines , Cross Protection , Viroporin Proteins
3.
Sci Adv ; 10(14): eadk8093, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38578989

ABSTRACT

Trained immunity is one of the mechanisms by which BCG vaccination confers persistent nonspecific protection against diverse diseases. Genomic differences between the different BCG vaccine strains that are in global use could result in variable protection against tuberculosis and therapeutic effects on bladder cancer. In this study, we found that four representative BCG strains (BCG-Russia, BCG-Sweden, BCG-China, and BCG-Pasteur) covering all four genetic clusters differed in their ability to induce trained immunity and nonspecific protection. The trained immunity induced by BCG was associated with the Akt-mTOR-HIF1α axis, glycolysis, and NOD-like receptor signaling pathway. Multi-omics analysis (epigenomics, transcriptomics, and metabolomics) showed that linoleic acid metabolism was correlated with the trained immunity-inducing capacity of different BCG strains. Linoleic acid participated in the induction of trained immunity and could act as adjuvants to enhance BCG-induced trained immunity, revealing a trained immunity-inducing signaling pathway that could be used in the adjuvant development.


Subject(s)
BCG Vaccine , Tuberculosis , Humans , Linoleic Acid , Trained Immunity , Multiomics , Adjuvants, Immunologic/pharmacology
4.
Sci Rep ; 14(1): 7994, 2024 04 05.
Article in English | MEDLINE | ID: mdl-38580687

ABSTRACT

Cordyceps militaris (L.) Link (C. militaris) contains various beneficial substances, including polysaccharides (galactomannan), nucleotides (adenosine and cordycepin), cordycepic acid, amino acids, and sterols (ergosterol and beta-sitosterol). It also contains other essential nutrients, such as protein, vitamins (E, K, B1, B2, and B12), and minerals (potassium, sodium, calcium, magnesium, iron, zinc, and selenium). Due to the numerous health benefits of supplements and products containing C. militaris extract, their popularity has increased. However, the immunostimulant effect of C. militaris remains unclear. Therefore, this study developed a functional beverage from the submerged fermentation of C. militaris (FCM) and aimed to investigate the potential of FCM in healthy male and female volunteers in Phayao Province, Thailand. This study provides essential information for the development of healthy drink products. Healthy men and women were provided either FCM containing 2.85 mg of cordycepin or placebo for 8 weeks (n = 10 for each gender). The immune cell markers, immunoglobulins, and safety parameters were assessed initially at baseline and at 4 and 8 weeks. The NK cell activity markedly increased in the male FCM group from baseline (p = 0.049) to 4 weeks after receiving FCM. Compared with those in the placebo group, the NK activity in women who received FCM for 8 weeks significantly increased (p = 0.023) from baseline. Within-group analysis revealed that the IL-1ß levels were markedly reduced in the male FCM group (p = 0.049). Furthermore, the IL-6 levels decreased from baseline in the female FCM group (p = 0.047). The blood sugar, lipid, and safety indices were not different between the experimental groups. FCM can potentially be developed as an immune-boosting supplement without liver, kidney, or blood component toxicity.


Subject(s)
Cordyceps , Adult , Humans , Male , Female , Cordyceps/chemistry , Deoxyadenosines/pharmacology , Adenosine/metabolism , Adjuvants, Immunologic/pharmacology , Liver , Immunity
5.
PLoS One ; 19(4): e0298117, 2024.
Article in English | MEDLINE | ID: mdl-38573916

ABSTRACT

Selection of adjuvant to be combined with the antigen is an extremely important point for formulating effective vaccines. The aim of this study was to evaluate reactogenicity, levels of IgM, IgG and subclasses (IgG1, IgG2b and IgG3), and protection elicited by vaccine formulations with association of chitosan coated alginate or Montanide ISA 61 with γ-irradiated Brucella ovis. The alginate/chitosan biopolymers as well as the Montanide ISA 61 emulsion elicited intense and long-lasting local response, especially when associated with the antigen. However, Montanide ISA 61 induced less intense reactogenicity when compared to alginate/chitosan. Furthermore, γ-irradiated B. ovis with Montanide ISA 61 induced higher levels of IgG2b an important marker of cellular immune response. In conclusion, Montanide ISA 61 resulted in milder reactogenicity when compared to the alginate/chitosan, while it induced a high IgG2b/IgG1 ratio compatible with a Th1 profile response.


Subject(s)
Chitosan , Mineral Oil , Vaccines , Animals , Mice , Sheep , Adjuvants, Vaccine , Capsules , Adjuvants, Immunologic/pharmacology , Immunoglobulin G , Mice, Inbred BALB C
6.
Biomater Sci ; 12(9): 2292-2301, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38498328

ABSTRACT

Colorectal cancer (CRC) ranks among the most prevalent cancers globally, demanding innovative therapeutic strategies. Immunotherapy, a promising avenue, employs cancer vaccines to activate the immune system against tumors. However, conventional approaches fall short of eliciting robust responses within the gastrointestinal (GI) tract, where CRC originates. Harnessing the potential of all-trans retinoic acid (ATRA) and cytosine-phosphorothioate-guanine (CpG), we developed layered nanoparticles using a layer-by-layer assembly method to co-deliver these agents. ATRA, crucial for gut immunity, was efficiently encapsulated alongside CpG within these nanoparticles. Administering these ATRA@CpG-NPs, combined with ovalbumin peptide (OVA), effectively inhibited orthotopic CRC growth in mice. Our approach leveraged the inherent benefits of ATRA and CpG, demonstrating superior efficacy in activating dendritic cells, imprinting T cells with gut-homing receptors, and inhibiting tumor growth. This mucosal adjuvant presents a promising strategy for CRC immunotherapy, showcasing the potential for targeting gut-associated immune responses in combating colorectal malignancies.


Subject(s)
Colorectal Neoplasms , Dinucleoside Phosphates , Nanoparticles , Tretinoin , Tretinoin/chemistry , Tretinoin/administration & dosage , Tretinoin/pharmacology , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/immunology , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Mice , Humans , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Mice, Inbred C57BL , Female , Immunotherapy/methods , Ovalbumin/administration & dosage , Ovalbumin/immunology , Ovalbumin/chemistry , Cell Line, Tumor , Mice, Inbred BALB C , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Layer-by-Layer Nanoparticles
7.
J Vis Exp ; (204)2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38465928

ABSTRACT

Cationic nanostructures have emerged as an adjuvant and antigen delivery system that enhances dendritic cell maturation, ROS generation, and antigen uptake and then promotes antigen-specific immune responses. In recent years, retinoic acid (RA) has received increasing attention due to its effect in activating the mucosal immune response; however, in order to use RA as a mucosal adjuvant, it is necessary to solve the problem of its dissolution, loading, and delivery. Here, we describe a cationic nanoemulsion-encapsulated retinoic acid (CNE-RA) delivery system composed of the cationic lipid 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOTAP), retinoic acid, squalene as the oil phase, polysorbate 80 as surfactant, and sorbitan trioleate 85 as co-surfactant. Its physical and chemical properties were characterized using dynamic light scattering and a spectrophotometer. Immunization of mice with the mixture of antigen (ovalbumin, OVA) and CNE-RA significantly elevated the levels of anti-OVA secretory immunoglobulin A (sIgA) in vaginal lavage fluid and the small intestinal lavage fluid of mice compared with OVA alone. This protocol describes a detailed method for the preparation, characterization, and evaluation of the adjuvant effect of CNE-RA.


Subject(s)
Adjuvants, Immunologic , Immunization , Female , Animals , Mice , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemistry , Mucous Membrane , Vaccination , Antigens , Immunity, Mucosal , Surface-Active Agents/pharmacology , Ovalbumin , Mice, Inbred BALB C
8.
J Med Chem ; 67(7): 5603-5616, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38513080

ABSTRACT

Vaccines are one of the greatest achievements of modern medicine. Due to their safer profile, the latest investigations usually focus on subunit vaccines. However, the active component often needs to be coupled with an adjuvant to be effective and properly trigger an immune response. We are developing a new synthetic monosaccharide-based TLR4 agonist, such as glucosamine-derived compounds FP18 and FP20, as a potential vaccine adjuvant. In this study, we present a new FP20 derivative, FP20Hmp, with a hydroxylated ester linked to the glucosamine core. We show that the modification introduced improves the activity of the adjuvant and its solubility. This study presents the synthesis of FP20Hmp, its in vitro characterization, and in vivo activity while coupled with the ovalbumin antigen or in formulation with an enterococcal antigen. We show that FP20Hmp enables increased production of antigen-specific antibodies that bind to the whole bacterium.


Subject(s)
Adjuvants, Vaccine , Enterococcus faecium , Toll-Like Receptor 4 , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemistry , Vaccines, Subunit , Glucosamine
9.
Poult Sci ; 103(4): 103496, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38330890

ABSTRACT

The avian influenza virus is infected through the mucosal route, thus mucosal barrier defense is very important. While the inactivated H9N2 vaccine cannot achieve sufficient mucosal immunity, adjuvants are needed to induce mucosal and systemic immunity to prevent poultry from H9N2 influenza virus infection. Our previous study found that polysaccharide from Atractylodes macrocephala Koidz binding with zinc oxide nanoparticles (AMP-ZnONPs) had immune-enhancing effects in vitro. This study aimed to evaluate the mucosal immune responses of oral whole-inactivated H9N2 virus (WIV)+AMP-ZnONPs and its impact on the animal challenge protection, and the corresponding changes of pulmonary metabolomics after the second immunization. The results showed that compared to the WIV, the combined treatment of WIV and AMP-ZnONPs significantly enhanced the HI titer, IgG and specific sIgA levels, the number of goblet cells and intestinal epithelial lymphocytes (iIELs) as well as the expression of J-chain, polymeric immunoglobulin receptor (pIgR), interleukin-10 (IL-10), tumor necrosis factor-α (TNF-α) and transforming growth factor-ß (TGF-ß). In viral attack experiments, WIV combing with AMP-ZnONPs effectively reduced lung damage and viral titers in throat swabs. Interestingly, significant changes of both the IgA intestinal immune network and PPAR pathway could also be found in the WIV+AMP-ZnONPs group compared to the non-infected group. Taken together, these findings suggest that AMP-ZnONPs can serve as a potential mucosal vaccine adjuvant, thereby avoiding adverse stress and corresponding costs caused by vaccine injection.


Subject(s)
Influenza A Virus, H9N2 Subtype , Influenza Vaccines , Influenza in Birds , Vaccines , Animals , Immunity, Mucosal , Chickens , Antibodies, Viral , Adjuvants, Immunologic/pharmacology , Administration, Oral , Vaccines, Inactivated , Influenza in Birds/prevention & control
10.
Int J Biol Macromol ; 262(Pt 2): 129982, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38354941

ABSTRACT

Oral vaccines are a safe and convenient alternative to injected vaccines and have great potential to prevent major infectious diseases. However, the harsh gastrointestinal (GI) environment, mucus barriers, low immunogenicity, and lack of effective and safe mucosal adjuvants are the major challenges for oral vaccine delivery. In recent years, nanoparticle-based strategies have become attractive for improving oral vaccine delivery. Here, the dendritic fibrous nano-silica (DFNS) grafted with Cistanche deserticola polysaccharide (CDP) nanoparticles (CDP-DFNS) were prepared and investigated how to impact the immune responses. CDP-DFNS facilitated the antigen uptake in mouse bone marrow-derived dendritic cells (BMDCs), and induce the activation of DCs in vitro. Furthermore, in vivo experiments, the result showed that the uptake efficiency by Peyer's patches (PPs) of CDP-DFNS/BSA was the best. And CDP-DFNS/BSA then significantly activated the DCs in lamina propria (LP), and T/B cells in PPs and mesenteric lymph nodes (MLNs). Moreover, the memory T cell responses in later period of vaccination was stronger than other groups. In addition, CDP-DFNS/BSA enhanced BSA-specific antibody IgG, IgA production, and SIgA secretion, was effective at inducing a strong mixed Th1/Th2 response and mucosal antibody responses. These results indicated that CDP-DFNS deserves further consideration as an oral vaccine adjuvant delivery system.


Subject(s)
Cistanche , Vaccines , Animals , Mice , Adjuvants, Vaccine , Silicon Dioxide , Mucous Membrane , Adjuvants, Immunologic/pharmacology , Adjuvants, Pharmaceutic , Polysaccharides/pharmacology , Immunity, Mucosal
11.
Biomater Sci ; 12(7): 1771-1787, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38385306

ABSTRACT

In the development of cancer vaccines, antigens are delivered to elicit potent and specific T-cell responses to eradicate tumour cells. Nonetheless, successful vaccines are often hampered by the poor immunogenicity of tumour antigens, rapid clearance by the innate immunity, and limited cross-presentation on MHC-I to activate CD8+ T-cells arm. To address these issues, we developed dextran-based nanogels to promote antigen uptake, storage, and cross-presentation on MHC-I, while directing immunogenic maturation of the antigen-presenting cells (APCs). To promote the nanocarriers interaction with cells, we modified DX with L-arginine (Arg), whose immunomodulatory activities have been well documented. The ArgDX nanogel performance was compared with the nanogel modified with L-histidine (His) and L-glutamate (Glut). Moreover, we introduced pH-sensitive hydrazone crosslinking during the nanogel formation for the conjugation and controlled release of antigen ovalbumin (OVA). The OVA-laden nanogels have an average size of 325 nm. We demonstrated that the nanogels could rapidly release cargoes upon a pH change from 7 to 5 within 8 days, indicating the controlled release of antigens in the acidic cellular compartments upon internalization. Our results revealed that the ArgDX nanogel could promote greater antigen uptake and storage in DCs in vitro and promoted a stronger immunogenic maturation of DCs and M1 polarization of the macrophages. The OVA signals were co-localized with lysosomal compartments up till 96 hours post-treatment and washing, suggesting the nanogels could facilitate prolonged antigen storage and supply from endo-lysosomal compartments. Furthermore, all the tested nanogel formulations retained antigens at the skin injection sites until day 21. Such delayed clearance could be due to the formation of micron-sized aggregates of OVA-laden nanogels, extending the interactions with the resident DCs. Amongst the amino acid modifications, ArgDX nanogels promoted the highest level of lymph node homing signal CCR7 on DCs. The nanogels also showed higher antigen presentation on both MHC-I and II than DX in vitro. In the in vivo immune studies, ArgDX nanogels were more superior in inducing cellular and humoral immunity than the other treatment groups on day 21 post-treatment. These results suggested that ArgDX nanogel is a promising self-adjuvanted nanocarrier for vaccine delivery.


Subject(s)
Cancer Vaccines , Immunity, Humoral , Polyethylene Glycols , Polyethyleneimine , Animals , Mice , Nanogels , Dextrans , CD8-Positive T-Lymphocytes , Delayed-Action Preparations , Dendritic Cells , Antigens , Adjuvants, Immunologic/pharmacology , Ovalbumin/chemistry , Mice, Inbred C57BL
12.
Anal Chem ; 96(8): 3362-3372, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38348659

ABSTRACT

Recently, we described synthetic sulfolipids named Sulfavants as a novel class of molecular adjuvants based on the sulfoquinovosyl-diacylglycerol skeleton. The members of this family, Sulfavant A (1), Sulfavant R (2), and Sulfavant S (3), showed important effects on triggering receptor expressed on myeloid cells 2 (TREM2)-induced differentiation and maturation of human dendritic cells (hDC), through a novel cell mechanism underlying the regulation of the immune response. As these molecules are involved in biological TREM2-mediated processes crucial for cell survival, here, we report the synthesis and application of a fluorescent analogue of Sulfavant A bearing the 4,4-difluoro-1,3,5,7-tetramethyl-4-bora-3a,4a-diaza-s-indacene moiety (Me4-BODIPY). The fluorescent derivative, named PB-SULF A (4), preserving the biological activity of Sulfavants, opens the way to chemical biology and cell biology experiments to better understand the interactions with cellular and in vivo organ targets and to improve our comprehension of complex molecular mechanisms underlying the not fully understood ligand-induced TREM2 activity.


Subject(s)
Boron Compounds , Fluorescent Dyes , Humans , Fluorescent Dyes/chemistry , Boron Compounds/pharmacology , Boron Compounds/chemistry , Adjuvants, Immunologic/pharmacology , Membrane Glycoproteins , Receptors, Immunologic
13.
Int J Mol Sci ; 25(4)2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38396809

ABSTRACT

H9N2 avian influenza poses a significant public health risk, necessitating effective vaccines for mass immunization. Oral inactivated vaccines offer advantages like the ease of administration, but their efficacy often requires enhancement through mucosal adjuvants. In a previous study, we established a novel complex of polysaccharide from Atractylodes macrocephala Koidz binding with zinc oxide nanoparticles (AMP-ZnONPs) and preliminarily demonstrated its immune-enhancing function. This work aimed to evaluate the efficacy of AMP-ZnONPs as adjuvants in an oral H9N2-inactivated vaccine and the vaccine's impact on intestinal mucosal immunity. In this study, mice were orally vaccinated on days 0 and 14 after adapting to the environment. AMP-ZnONPs significantly improved HI titers, the levels of specific IgG, IgG1 and IgG2a in serum and sIgA in intestinal lavage fluid; increased the number of B-1 and B-2 cells and dendritic cell populations; and enhanced the mRNA expression of intestinal homing factors and immune-related cytokines. Interestingly, AMP-ZnONPs were more likely to affect B-1 cells than B-2 cells. AMP-ZnONPs showed mucosal immune enhancement that was comparable to positive control (cholera toxin, CT), but not to the side effect of weight loss caused by CT. Compared to the whole-inactivated H9N2 virus (WIV) group, the WIV + AMP-ZnONP and WIV + CT groups exhibited opposite shifts in gut microbial abundance. AMP-ZnONPs serve as an effective and safe mucosal adjuvant for oral WIV, improving cellular, humoral and mucosal immunity and microbiota in the gastrointestinal tract, avoiding the related undesired effects of CT.


Subject(s)
Atractylodes , Influenza A Virus, H9N2 Subtype , Influenza Vaccines , Zinc Oxide , Animals , Mice , Adjuvants, Immunologic/pharmacology , Immunity, Mucosal , Vaccines, Inactivated , Polysaccharides/pharmacology , Antibodies, Viral
14.
Pharm Dev Technol ; 29(2): 75-85, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38217108

ABSTRACT

To develop a novel water-in-oil-in-water (W/O/W) adjuvant and evaluate the effect on foot-and-mouth disease (FMD) inactivated vaccine, in this study, we prepared the novel nano-emulsion adjuvant based on QS-21 (BEA) which is composed of the mixture of mineral oil Marcol52, surfactant Tween80, oleate polyoxyethylene ether ester, polyoxyethylene palmitic acid ester and span80, cosurfactant polyethylene glycol and QS-21. The two-step emulsification method formed the W/O/W nano-emulsion with two films and three-phase structures. The effective particle diameter of the BEA was about 184 nm, and it has good thermal stability. Then, BEA was emulsified as an adjuvant to prepare for the inactivated FMDV vaccine, and BALB/c mice and pigs were immunized to evaluate its safety and immunization effect. The results showed that the inactivated BEA-FMDV vaccine significantly increased BALB/c mice and pigs' antibodies and cytokine IFN-γ in serum. Meanwhile, the pig-neutralizing antibodies were higher than control group. Safety tests found no symptoms of FMD or significant toxic reactions. After 28 days of immunization, the protection rate can reach 93.3%. The BEA vaccine had good stability at 4 °C, no stratification after 180 days, and the content of 146S in the vaccine did not decrease. In conclusion, the BEA prepared in this study is suitable for FMDV inactivated vaccine and is an effective adjuvant.


Subject(s)
Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Viral Vaccines , Mice , Animals , Swine , Foot-and-Mouth Disease/prevention & control , Vaccines, Inactivated , Water , Antibodies, Viral , Adjuvants, Immunologic/pharmacology , Polyethylene Glycols , Esters
15.
Sci China Life Sci ; 67(4): 680-697, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38206438

ABSTRACT

The study of tumor nanovaccines (NVs) has gained interest because they specifically recognize and eliminate tumor cells. However, the poor recognition and internalization by dendritic cells (DCs) and insufficient immunogenicity restricted the vaccine efficacy. Herein, we extracted two molecular-weight Astragalus polysaccharides (APS, 12.19 kD; APSHMw, 135.67 kD) from Radix Astragali and made them self-assemble with OVA257-264 directly forming OVA/APS integrated nanocomplexes through the microfluidic method. The nanocomplexes were wrapped with a sheddable calcium phosphate layer to improve stability. APS in the formed nanocomplexes served as drug carriers and immune adjuvants for potent tumor immunotherapy. The optimal APS-NVs were approximately 160 nm with uniform size distribution and could remain stable in physiological saline solution. The FITC-OVA in APS-NVs could be effectively taken up by DCs, and APS-NVs could stimulate the maturation of DCs, improving the antigen cross-presentation efficiency in vitro. The possible mechanism was that APS can induce DC activation via multiple receptors such as dectin-1 and Toll-like receptors 2 and 4. Enhanced accumulation of APS-NVs both in draining and distal lymph nodes were observed following s.c. injection. Smaller APS-NVs could easily access the lymph nodes. Furthermore, APS-NVs could markedly promote antigen delivery efficiency to DCs and activate cytotoxic T cells. In addition, APS-NVs achieve a better antitumor effect in established B16-OVA melanoma tumors compared with the OVA+Alum treatment group. The antitumor mechanism correlated with the increase in cytotoxic T cells in the tumor region. Subsequently, the poor tumor inhibitory effect of APS-NVs on the nude mouse model of melanoma also confirmed the participation of antitumor adaptive immune response induced by NVs. Therefore, this study developed a promising APS-based tumor NV that is an efficient tumor immunotherapy without systemic side effects.


Subject(s)
Cancer Vaccines , Melanoma , Mice , Animals , Nanovaccines , Melanoma/pathology , Dendritic Cells , Adjuvants, Immunologic/pharmacology , Immunotherapy , Antigens , Polysaccharides/chemistry , Mice, Inbred C57BL
16.
Int J Pharm ; 652: 123802, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38218508

ABSTRACT

Adjuvants are necessary for protein vaccines and have been used for nearly 100 years. However, developing safe and effective adjuvants is still urgently needed. Polysaccharides isolated from traditional Chinese medicine are considered novel vaccine adjuvant sources. This study aimed to investigate the adjuvant activity and immune-enhancing mechanisms of the microparticulated Polygonatum sibiricum polysaccharide (MP-PSP) modified by calcium carbonate. PSP demonstrated adjuvant activity, and MP-PSP further showed a higher humoral response compared to PSP. Subsequently, MP-PSP was elucidated to improving the immunity by slowing the rate of antigen release and activating dendritic cells along with interleukin-6 secretion through toll-like receptor 4 signaling, followed by T follicular helper cell and B cell interactions. Moreover, MP-PSP had a good safety profile in vaccinated mice. Thus, MP-PSP may be a promising vaccine adjuvant and warrants further investigation.


Subject(s)
Adjuvants, Vaccine , Polygonatum , Mice , Animals , Signal Transduction , Adjuvants, Immunologic/pharmacology , Polysaccharides/pharmacology
17.
Fish Shellfish Immunol ; 146: 109369, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38220122

ABSTRACT

Damiana (Turnera diffusa Willd) was evaluated in vitro for antioxidant and antibacterial activities against Staphylococcus aureus and Streptococcus pyogenes (as a preliminary screening assessment) by high-performance thin-layer chromatography (HPTLC)-Direct bioautography. A study was performed in vivo to evaluate the effects of Damiana enriched diets at 0.5 % on immune parameters in mucus and serum and gene expression in Almaco Jack (Seriola rivoliana) intestine after two and four weeks; an infection with Aeromonas hydrophila at 1x107 colony forming units (CFU) followed and an ex vivo study was carried out using head-kidney leukocytes. Ferric reducing ability of plasma (FRAP) and 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid (ABTS) assays showed high antioxidant activities in Damiana leaves; even in the ABTS assay, Damiana at 300 µg/mL showed similar activity to ascorbic acid - the standard control. Damiana exhibited strong in vitro antimicrobial activity against S. aureus and S. pyogenes. In vivo studies showed a strong enhancement of myeloperoxidase, nitric oxide, superoxide dismutase, and catalase activities in mucus and serum of S. rivoliana supplemented with Damiana; their immunological response enhanced after infection with A. hydrophila. IL-1ß, TNF-α, and IL-10 gene expressions upregulated in the fish intestine challenged with the bacterium. Piscidin and macrophage (MARCO) receptor gene expression up-regulated at week 4 and down-regulated after infection. Intestinal histology results confirm that Damiana not cause inflammation or damage. Finally, the ex vivo study confirmed the immunostimulant and protective effects of Damiana through increased phagocytic, respiratory burst, myeloperoxidase activities and nitric oxide generation before and upon the bacterial encounter. These results support the idea that Damiana has the potential as an immunostimulant additive for diets in aquaculture by enhancing immune parameters and protecting Almaco Jack against A. hydrophila infections upon four weeks of supplementation.


Subject(s)
Benzothiazoles , Fish Diseases , Gram-Negative Bacterial Infections , Sulfonic Acids , Turnera , Animals , Turnera/chemistry , Antioxidants/metabolism , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/metabolism , Nitric Oxide/metabolism , Staphylococcus aureus/metabolism , Dietary Supplements/analysis , Diet , Peroxidase/metabolism , Aeromonas hydrophila , Gram-Negative Bacterial Infections/veterinary , Animal Feed/analysis
18.
Phytomedicine ; 124: 155302, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38176273

ABSTRACT

BACKGROUND: Extensive investigation has been undertaken about the utilization of saponin adjuvants in vaccines intended for veterinary and human applications. AB4 is the main constituent of the traditional Chinese medicine, Pulsatilla chinensis (Bunge) Regel, and has immunomodulatory activity. However, there is a paucity of reports on AB4 as a potential adjuvant. PURPOSE: The objective of this work was to clarify the adjuvant role of AB4 and the molecular mechanisms that underlie its immunomodulatory actions. STUDY DESIGN AND METHODS: The immunomodulatory effects of AB4 were investigated using network pharmacological analyses. These effects were validated by evaluating the developmental status of the immune organs and by using the following techniques: ELISA for the quantification of serum-specific antibodies to determine immune-related cytokine levels; the MTS method for the assessment of proliferative activity of splenic lymphocytes; flow cytometry to analyze lymphocyte and dendritic cell activation status; and western blotting for mechanistic analysis at the protein level. RESULTS: The network pharmacological analysis predicted a total of 52 targets and 12 pathways for AB4 to exert immunomodulatory effects. In a mouse model with immunity to OVA, the introduction of AB4 resulted in the enhancement of immunological organ growth and maturation, elevation of blood antibodies targeting OVA, and amplification of the production of cytokines associated with Th1 and Th2 immune responses. Additionally, the administration of AB4 resulted in a notable augmentation of lymphocyte proliferation and an elevation in the CD4+/CD8+ T lymphocyte ratios. Furthermore, the administration of AB4 enhanced the maturation process of DCs in the draining LNs and increased the production of co-stimulatory factors and MHC II molecules. AB4 induces the upregulation of TLR4 and IKK proteins, as well as the phosphorylation of NF-κB p65 protein within the TLR4/NF-κB signaling cascade, while concurrently suppressing the expression of IκBα protein. CONCLUSION: The specific immunoadjuvant effects of AB4 have been demonstrated to modulate the growth and maturation of immune organs and enhance the secretion and cellular activity of pertinent immune molecules. The utilization of network pharmacology, combined within and in vivo vitro assays, clarified the adjuvant function of AB4, which potentially involves the regulation of the TLR4/NF-κB signaling pathway.


Subject(s)
NF-kappa B , Saponins , Animals , Mice , Humans , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Network Pharmacology , Adjuvants, Immunologic/pharmacology , Cytokines/metabolism , Saponins/pharmacology , Saponins/metabolism , Adjuvants, Pharmaceutic , Dendritic Cells
19.
Angew Chem Int Ed Engl ; 63(10): e202318530, 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38196070

ABSTRACT

Dendritic cell (DC) maturation and antigen presentation are key factors for successful vaccine-based cancer immunotherapy. This study developed manganese-based layered double hydroxide (Mn-LDH) nanoparticles as a self-adjuvanted vaccine carrier that not only promoted DC maturation through synergistically depleting endogenous glutathione (GSH) and activating STING signaling pathway, but also facilitated the delivery of model antigen ovalbumin (OVA) into lymph nodes and subsequent antigen presentation in DCs. Significant therapeutic-prophylactic efficacy of the OVA-loaded Mn-LDH (OVA/Mn-LDH) nanovaccine was determined by the tumor growth inhibition in the mice bearing B16-OVA tumor. Our results showed that the OVA/Mn-LDH nanoparticles could be a potent delivery system for cancer vaccine development without the need of adjuvant. Therefore, the combination of GSH exhaustion and STING pathway activation might be an advisable approach for promoting DC maturation and antigen presentation, finally improving cancer vaccine efficacy.


Subject(s)
Cancer Vaccines , Nanoparticles , Neoplasms , Mice , Animals , Vaccine Efficacy , Neoplasms/pathology , Immunotherapy/methods , Adjuvants, Immunologic/pharmacology , Glutathione , Dendritic Cells , Mice, Inbred C57BL , Ovalbumin
20.
Microb Pathog ; 186: 106489, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38061666

ABSTRACT

Trichinellosis caused by Trichinella spiralis (T. spiralis) is a zoonotic disease that poses a substantial risk to human health. At present, vaccines used to prevent trichinellosis are effective, but the production of antibody levels and immunogenicity are low. Adjuvants can increase antibody levels and vaccine immunogenicity. As a result, it is critical to develop an effective adjuvant for the T. spiralis vaccine. Recent research has shown that traditional Chinese medicine polysaccharides with low-toxicity and biodegradability can act as adjuvants in vaccines. In this study, BALB/c mice were orally inoculated with a recombinant Lactobacillus plantarum (L. plantarum) vaccine expressing the T. spiralis cathepsin F-like protease 1 gene (rTs-CPF1), which was given three times at 10-day intervals. Lycium barbarum polysaccharide (LBP) was administered orally for 37 days. At 37 days after the first immunization, mice were infected with 350 T. spiralis muscle larvae (ML). Specific IgG and sIgA antibody levels against the T. spiralis CPF1 protein were increased in mice immunized with rTs-CPF1+LBP compared to those immunized with rTs-CPF1 alone. Furthermore, LBP increased IFN-γ and IL-4 expression levels, and the number of intestinal and intramuscular worms was significantly reduced in the rTs-CPF1+LBP group compared to that in the rTs-CPF1 group. In the rTs-CPF1+LBP group, the reduction rates of adult worms and muscle larvae were 47.31 % and 68.88 %, respectively. To summarize, LBP promotes the immunoprotective effects of the T. spiralis vaccine and may be considered as a novel adjuvant in parasitic vaccines.


Subject(s)
Lactobacillus plantarum , Trichinella spiralis , Trichinellosis , Mice , Humans , Animals , Trichinella spiralis/genetics , Trichinellosis/prevention & control , Trichinellosis/parasitology , Cathepsin F , Lactobacillus plantarum/genetics , Antigens, Helminth/genetics , Vaccines, Synthetic , Adjuvants, Immunologic/pharmacology , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL