Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 12.424
Filtrer
1.
Sci Rep ; 14(1): 15262, 2024 07 03.
Article de Anglais | MEDLINE | ID: mdl-38961116

RÉSUMÉ

Infectious coryza (IC) is an acute upper respiratory disease of chicken caused by Avibacterium (A.) paragallinarum. This disease results in an increased culling rate in meat chicken and a marked decrease in egg production (10% to more than 40%) in laying and breeding hens. Vaccines were first used against IC and effectively controlled the disease. Nanotechnology provides an excellent way to develop a new generation of vaccines. NPs have been widely used in vaccine design as adjuvants and antigen delivery vehicles and as antibacterial agents; thus, they can be used as inactivators for bacterial culture. In this research, the antibacterial effects of several nanoparticles (NPs), such as silicon dioxide with chitosan (SiO2-CS), oleoyl-chitosan (O.CS), silicon dioxide (SiO2), and iron oxide (Fe3O4), on A. paragallinarum were studied. Additionally, different A. paragallinarum vaccines were made using the same nanomaterials at a concentration of 400 µg/ml to help control infectious coryza disease in chicken. A concentration of 400 µg/ml of all the NPs tested was the best concentration for the inactivation of A. paragallinarum. Additionally, this study showed that the infectious coryza vaccine adjuvanted with SiO2 NPs had the highest immune response, followed by the infectious coryza vaccine adjuvanted with Fe3O4 NPs, the infectious coryza vaccine adjuvanted with SiO2-CS NPs, and the infectious coryza vaccine adjuvanted with O.CS NPs in comparison with the infectious coryza vaccine adjuvanted with liquid paraffin (a commercial vaccine).


Sujet(s)
Adjuvants immunologiques , Poulets , Chitosane , Nanoparticules , Maladies de la volaille , Animaux , Poulets/immunologie , Maladies de la volaille/prévention et contrôle , Maladies de la volaille/immunologie , Nanoparticules/composition chimique , Chitosane/composition chimique , Adjuvants immunologiques/pharmacologie , Vaccins antibactériens/immunologie , Vaccins antibactériens/administration et posologie , Silice/composition chimique , Adjuvants vaccinaux , Polymères/composition chimique , Vecteurs de médicaments/composition chimique , Pasteurellaceae/immunologie
2.
Oncoimmunology ; 13(1): 2373526, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948931

RÉSUMÉ

Prostate cancer (PCa) is characterized as a "cold tumor" with limited immune responses, rendering the tumor resistant to immune checkpoint inhibitors (ICI). Therapeutic messenger RNA (mRNA) vaccines have emerged as a promising strategy to overcome this challenge by enhancing immune reactivity and significantly boosting anti-tumor efficacy. In our study, we synthesized Tetra, an mRNA vaccine mixed with multiple tumor-associated antigens, and ImmunER, an immune-enhancing adjuvant, aiming to induce potent anti-tumor immunity. ImmunER exhibited the capacity to promote dendritic cells (DCs) maturation, enhance DCs migration, and improve antigen presentation at both cellular and animal levels. Moreover, Tetra, in combination with ImmunER, induced a transformation of bone marrow-derived dendritic cells (BMDCs) to cDC1-CCL22 and up-regulated the JAK-STAT1 pathway, promoting the release of IL-12, TNF-α, and other cytokines. This cascade led to enhanced proliferation and activation of T cells, resulting in effective killing of tumor cells. In vivo experiments further revealed that Tetra + ImmunER increased CD8+T cell infiltration and activation in RM-1-PSMA tumor tissues. In summary, our findings underscore the promising potential of the integrated Tetra and ImmunER mRNA-LNP therapy for robust anti-tumor immunity in PCa.


Sujet(s)
Adjuvants immunologiques , Antigènes néoplasiques , Vaccins anticancéreux , Cellules dendritiques , Tumeurs de la prostate , ARN messager , Animaux , Mâle , Tumeurs de la prostate/immunologie , Tumeurs de la prostate/thérapie , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/traitement médicamenteux , Antigènes néoplasiques/immunologie , Souris , Cellules dendritiques/immunologie , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/administration et posologie , ARN messager/génétique , ARN messager/métabolisme , ARN messager/administration et posologie , Vaccins anticancéreux/administration et posologie , Vaccins anticancéreux/immunologie , Humains , Souris de lignée C57BL , Lignée cellulaire tumorale , Vaccins à ARNm , Lymphocytes T CD8+/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Immunothérapie/méthodes , Activation des lymphocytes/effets des médicaments et des substances chimiques
3.
PLoS One ; 19(7): e0306664, 2024.
Article de Anglais | MEDLINE | ID: mdl-38968270

RÉSUMÉ

BACKGROUNDS: Malaria, a preventive and treatable disease, is still responsible for annual deaths reported in most tropical regions, principally in sub-Saharan Africa. Subunit recombinant transmission-blocking vaccines (TBVs) have been proposed as promising vaccines to succeed in malaria elimination and eradication. Here, a provisional study was designed to assess the immunogenicity and functional activity of alanyl aminopeptidase N (APN1) of Anopheles stephensi, as a TBV candidate, administered with MPL, CpG, and QS21 adjuvants in the murine model. METHODOLOGY/PRINCIPAL FINDINGS: The mouse groups were immunized with recombinant APN1 (rAPN1) alone or formulated with CpG, MPL, QS-21, or a combination of adjuvants (CMQ), and the elicited immune responses were evaluated after the third immunization. The standard membrane feeding assay (SMFA) measured the functional activity of antibodies against bacterial-expressed APN1 protein in adjuvanted vaccine groups on transmission of P. falciparum (NF54) to An. stephensi mosquitoes. Evaluation of mice vaccinated with rAPN1 formulated with distinct adjuvants manifested a significant increase in the high-avidity level of anti-APN1 IgG and IgG subclasses; however, rAPN1 induced the highest level of high-avidity anti-APN1 IgG1, IgG2a, and IgG2b antibodies in the immunized vaccine group 5 (APN1/CMQ). In addition, vaccine group 5 (receiving APN1/CMQ), had still the highest level of anti-APN1 IgG antibodies relative to other immunized groups after six months, on day 180. The SMFA data indicates a trend towards higher transmission-reducing activity in groups 2 and 5, which received the antigen formulated with CpG or a combination of three adjuvants. CONCLUSIONS/SIGNIFICANCE: The results have shown the capability of admixture to stimulate high-affinity and long-lasting antibodies against the target antigen to hinder Plasmodium parasite development in the mid-gut of An. stephensi. The attained results authenticated APN1/CMQ and APN1/CpG as a potent APN1-based TBV formulation which will be helpful in designing a vaccine in the future.


Sujet(s)
Adjuvants immunologiques , Anopheles , Antigènes CD13 , Vaccins contre le paludisme , Saponines , Animaux , Anopheles/parasitologie , Anopheles/immunologie , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/administration et posologie , Souris , Vaccins contre le paludisme/immunologie , Vaccins contre le paludisme/administration et posologie , Saponines/pharmacologie , Saponines/administration et posologie , Antigènes CD13/immunologie , Antigènes CD13/métabolisme , Femelle , Plasmodium falciparum/immunologie , Paludisme/prévention et contrôle , Paludisme/transmission , Paludisme/immunologie , Paludisme/parasitologie , Oligodésoxyribonucléotides/pharmacologie , Oligodésoxyribonucléotides/administration et posologie , Oligodésoxyribonucléotides/immunologie , Souris de lignée BALB C , Paludisme à Plasmodium falciparum/prévention et contrôle , Paludisme à Plasmodium falciparum/transmission , Paludisme à Plasmodium falciparum/immunologie , Paludisme à Plasmodium falciparum/parasitologie
4.
Theranostics ; 14(10): 3810-3826, 2024.
Article de Anglais | MEDLINE | ID: mdl-38994034

RÉSUMÉ

Rationale: Surgical resection is a primary treatment for solid tumors, but high rates of tumor recurrence and metastasis post-surgery present significant challenges. Manganese (Mn2+), known to enhance dendritic cell-mediated cancer immunotherapy by activating the cGAS-STING pathway, has potential in post-operative cancer management. However, achieving prolonged and localized delivery of Mn2+ to stimulate immune responses without systemic toxicity remains a challenge. Methods: We developed a post-operative microenvironment-responsive dendrobium polysaccharide hydrogel embedded with Mn2+-pectin microspheres (MnP@DOP-Gel). This hydrogel system releases Mn2+-pectin microspheres (MnP) in response to ROS, and MnP shows a dual effect in vitro: promoting immunogenic cell death and activating immune cells (dendritic cells and macrophages). The efficacy of MnP@DOP-Gel as a post-surgical treatment and its potential for immune activation were assessed in both subcutaneous and metastatic melanoma models in mice, exploring its synergistic effect with anti-PD1 antibody. Result: MnP@DOP-Gel exhibited ROS-responsive release of MnP, which could exert dual effects by inducing immunogenic cell death of tumor cells and activating dendritic cells and macrophages to initiate a cascade of anti-tumor immune responses. In vivo experiments showed that the implanted MnP@DOP-Gel significantly inhibited residual tumor growth and metastasis. Moreover, the combination of MnP@DOP-Gel and anti-PD1 antibody displayed superior therapeutic potency in preventing either metastasis or abscopal brain tumor growth. Conclusions: MnP@DOP-Gel represents a promising drug-free strategy for cancer post-operative management. Utilizing this Mn2+-embedding and ROS-responsive delivery system, it regulates surgery-induced immune responses and promotes sustained anti-tumor responses, potentially increasing the effectiveness of surgical cancer treatments.


Sujet(s)
Dendrobium , Hydrogels , Manganèse , Souris de lignée C57BL , Microsphères , Polyosides , Animaux , Souris , Hydrogels/composition chimique , Manganèse/composition chimique , Polyosides/composition chimique , Polyosides/pharmacologie , Dendrobium/composition chimique , Macrophages/immunologie , Macrophages/effets des médicaments et des substances chimiques , Mélanome/immunologie , Mélanome/traitement médicamenteux , Mélanome/thérapie , Immunothérapie/méthodes , Cellules dendritiques/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Femelle , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Espèces réactives de l'oxygène/métabolisme , Adjuvants immunologiques/administration et posologie , Adjuvants immunologiques/pharmacologie , Mélanome expérimental/immunologie , Mélanome expérimental/thérapie , Mélanome expérimental/traitement médicamenteux
5.
Int J Nanomedicine ; 19: 6589-6602, 2024.
Article de Anglais | MEDLINE | ID: mdl-38979535

RÉSUMÉ

Orthopedic infection is one of the most intractable orthopedic problems. Bacteria resistant to antibiotics also develop gradually. Chitosan is widely used in the Biomedical field because of its high biocompatibility, biodegradability, and antibacterial activity. Chitosan-based drug delivery systems are frequently utilized to produce controlled medication release. When combined with antibiotics, synergistic antibacterial effects can be achieved. Chitosan-based nanoparticles are one of the most widely used applications in drug delivery systems. The focus of this review is to provide information on new methods being developed for chitosan-based nanoparticles in the field of bone infection treatment, including chitosan nanoparticles for antibacterial purposes, Ch-loaded with antibiotics, Ch-loaded with metal, and used as immune adjuvants. It may Provide ideas for the fundamental research and the prospects of future clinical applications of orthopedic infections.


Sujet(s)
Antibactériens , Chitosane , Nanoparticules , Chitosane/composition chimique , Chitosane/pharmacologie , Humains , Nanoparticules/composition chimique , Antibactériens/composition chimique , Antibactériens/pharmacologie , Antibactériens/administration et posologie , Antibactériens/pharmacocinétique , Animaux , Systèmes de délivrance de médicaments/méthodes , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/composition chimique , Adjuvants immunologiques/administration et posologie , Vecteurs de médicaments/composition chimique
6.
Molecules ; 29(13)2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-38999177

RÉSUMÉ

A short 19 bp dsRNA with 3'-trinucleotide overhangs acting as immunostimulating RNA (isRNA) demonstrated strong antiproliferative action against cancer cells, immunostimulatory activity through activation of cytokines and Type-I IFN secretion, as well as anti-tumor and anti-metastatic effects in vivo. The aim of this study was to determine the tolerance of chemical modifications (2'-F, 2'-OMe, PS, cholesterol, and amino acids) located at different positions within this isRNA to its ability to activate the innate immune system. The obtained duplexes were tested in vivo for their ability to activate the synthesis of interferon-α in mice, and in tumor cell cultures for their ability to inhibit their proliferation. The obtained data show that chemical modifications in the composition of isRNA have different effects on its individual functions, including interferon-inducing and antiproliferative effects. The effect of modifications depends not only on the type of modification but also on its location and the surrounding context of the modifications. This study made it possible to identify leader patterns of modifications that enhance the properties of isRNA: F2/F2 and F2_S/F2 for interferon-inducing activity, as well as F2_S5/F2_S5, F2-NH2/F2-NH2, and Ch-F2/Ch-F2 for antiproliferative action. These modifications can improve the pharmacokinetic and pharmacodynamic properties, as well as increase the specificity of isRNA action to obtain the desired effect.


Sujet(s)
Prolifération cellulaire , ARN double brin , ARN double brin/pharmacologie , ARN double brin/composition chimique , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris , Humains , Lignée cellulaire tumorale , Interféron alpha/métabolisme , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/composition chimique , Interférons/métabolisme
7.
Sci Adv ; 10(27): eadg3747, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38959314

RÉSUMÉ

Vaccination can help prevent infection and can also be used to treat cancer, allergy, and potentially even drug overdose. Adjuvants enhance vaccine responses, but currently, the path to their advancement and development is incremental. We used a phenotypic small-molecule screen using THP-1 cells to identify nuclear factor-κB (NF-κB)-activating molecules followed by counterscreening lead target libraries with a quantitative tumor necrosis factor immunoassay using primary human peripheral blood mononuclear cells. Screening on primary cells identified an imidazopyrimidine, dubbed PVP-037. Moreover, while PVP-037 did not overtly activate THP-1 cells, it demonstrated broad innate immune activation, including NF-κB and cytokine induction from primary human leukocytes in vitro as well as enhancement of influenza and SARS-CoV-2 antigen-specific humoral responses in mice. Several de novo synthesis structural enhancements iteratively improved PVP-037's in vitro efficacy, potency, species-specific activity, and in vivo adjuvanticity. Overall, we identified imidazopyrimidine Toll-like receptor-7/8 adjuvants that act in synergy with oil-in-water emulsion to enhance immune responses.


Sujet(s)
Adjuvants immunologiques , Pyrimidines , Récepteur de type Toll-7 , Récepteur de type Toll-8 , Humains , Récepteur de type Toll-8/agonistes , Récepteur de type Toll-8/métabolisme , Animaux , Souris , Adjuvants immunologiques/pharmacologie , Récepteur de type Toll-7/agonistes , Pyrimidines/pharmacologie , Pyrimidines/composition chimique , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/immunologie , Imidazoles/pharmacologie , Imidazoles/composition chimique , Cellules THP-1 , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/métabolisme , Agranulocytes/immunologie , COVID-19/virologie , COVID-19/immunologie , Facteur de transcription NF-kappa B/métabolisme , Femelle , Découverte de médicament/méthodes , Immunité innée/effets des médicaments et des substances chimiques
8.
Egypt J Immunol ; 31(3): 28-40, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38985532

RÉSUMÉ

The study aimed to assess the immunomodulatory effects of Phoenix dactylifera (dates) fruit, a traditional remedy used by Moroccans to enhance immunity against pathogens. This research sought to evaluate the impacts of this fruit on immune cells and their functions. To achieve this, we conducted tests using date extracts on splenocytes, thymocytes, and macrophages, focusing on their functions: antibody production, phagocytosis, and T-lymphocyte toxicity. The results obtained demonstrated that the aqueous extract of P. dactylifera fruit exhibited significant immunostimulatory effects on humoral immunity. It achieved this by enhancing complement activity and increasing splenocyte (including B-lymphocytes) proliferation by 142.5% compared to control cells. Similarly, in the same conditions, there was notable stimulation of cellular immunity through thymocyte activity, resulting in a remarkable increase in cell proliferation (225%) and a boost in thymocyte function (245.9%), which plays a role in safeguarding against cancer. Moreover, the date extract demonstrated anti-inflammatory properties. This was evident in the increased phagocytosis activity mediated by macrophages under the ethyl acetate extract, effectively eliminating pathogens. Assessing the cosmetic potential of date extracts showed that the ethyl acetate extract possesses both anti-inflammatory and strong antioxidant effects, exhibited high photo absorption of ultraviolet-B rays. Based on these findings, we propose to study the utilization of this extract for sun protection as a sunscreen. Furthermore, the Fourier-transform infrared spectroscopy analysis indicated that the most active compounds present were flavonoids. These outcomes substantiate the traditional usage of this fruit for reinforcing immunity.


Sujet(s)
Immunité cellulaire , Immunité humorale , Phoeniceae , Extraits de plantes , Animaux , Immunité cellulaire/effets des médicaments et des substances chimiques , Immunité humorale/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Extraits de plantes/immunologie , Souris , Phoeniceae/composition chimique , Adjuvants immunologiques/pharmacologie , Phagocytose/effets des médicaments et des substances chimiques , Phagocytose/immunologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Rate/immunologie , Rate/effets des médicaments et des substances chimiques , Rate/cytologie , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Fruit/composition chimique , Fruit/immunologie , Mâle , Prolifération cellulaire/effets des médicaments et des substances chimiques
9.
Arch Virol ; 169(8): 163, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38990396

RÉSUMÉ

Antigenically divergent H7N9 viruses pose a potential threat to public health, with the poor immunogenicity of candidate H7N9 vaccines demonstrated in clinical trials underscoring the urgent need for more-effective H7N9 vaccines. In the present study, mice were immunized with various doses of a suspended-MDCK-cell-derived inactivated H7N9 vaccine, which was based on a low-pathogenic H7N9 virus, to assess cross-reactive immunity and cross-protection against antigenically divergent H7N9 viruses. We found that the CRX-527 adjuvant, a synthetic TLR4 agonist, significantly enhanced the humoral immune responses of the suspended-MDCK-cell-derived H7N9 vaccine, with significant antigen-sparing and immune-enhancing effects, including robust virus-specific IgG, hemagglutination-inhibiting (HI), neuraminidase-inhibiting (NI), and virus-neutralizing (VN) antibody responses, which are crucial for protection against influenza virus infection. Moreover, the CRX-527-adjuvanted H7N9 vaccine also elicited cross-protective immunity and cross-protection against a highly pathogenic H7N9 virus with a single vaccination. Notably, NI and VN antibodies might play an important role in cross-protection against lethal influenza virus infections. This study showed that a synthetic TLR4 agonist adjuvant has a potent immunopotentiating effect, which might be considered worth further development as a means of increasing vaccine effectiveness.


Sujet(s)
Anticorps antiviraux , Immunité humorale , Sous-type H7N9 du virus de la grippe A , Vaccins antigrippaux , Souris de lignée BALB C , Infections à Orthomyxoviridae , Récepteur de type Toll-4 , Vaccins inactivés , Animaux , Sous-type H7N9 du virus de la grippe A/immunologie , Récepteur de type Toll-4/agonistes , Récepteur de type Toll-4/immunologie , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Souris , Anticorps antiviraux/immunologie , Chiens , Cellules rénales canines Madin-Darby , Vaccins inactivés/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Femelle , Anticorps neutralisants/immunologie , Protection croisée/immunologie , Adjuvants immunologiques/administration et posologie , Adjuvants immunologiques/pharmacologie , Adjuvants vaccinaux , Immunoglobuline G/immunologie , Immunoglobuline G/sang
10.
ACS Nano ; 18(26): 16589-16609, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38885198

RÉSUMÉ

Adjuvants are effective tools to enhance vaccine efficacy and control the type of immune responses such as antibody and T helper 1 (Th1)- or Th2-type responses. Several studies suggest that interferon (IFN)-γ-producing Th1 cells play a significant role against infections caused by intracellular bacteria and viruses; however, only a few adjuvants can induce a strong Th1-type immune response. Recently, several studies have shown that lipid nanoparticles (LNPs) can be used as vaccine adjuvants and that each LNP has a different adjuvant activity. In this study, we screened LNPs to develop an adjuvant that can induce Th1 cells and antibodies using a conventional influenza split vaccine (SV) as an antigen in mice. We observed that LNP with 1,2-di-O-octadecenyl-3-trimethylammonium-propane (DOTMA) as a component lipid (DOTMA-LNP) elicited robust SV-specific IgG1 and IgG2 responses compared with SV alone in mice and was as efficient as SV adjuvanted with other adjuvants in mice. Furthermore, DOTMA-LNPs induced robust IFN-γ-producing Th1 cells without inflammatory responses compared to those of other adjuvants, which conferred strong cross-protection in mice. We also demonstrated the high versatility of DOTMA-LNP as a Th1 cell-inducing vaccine adjuvant using vaccine antigens derived from severe acute respiratory syndrome coronavirus 2 and Streptococcus pneumoniae. Our findings suggest the potential of DOTMA-LNP as a safe and effective Th1 cell-inducing adjuvant and show that LNP formulations are potentially potent adjuvants to enhance the effectiveness of other subunit vaccines.


Sujet(s)
Nanoparticules , Composés d'ammonium quaternaire , Lymphocytes auxiliaires Th1 , Animaux , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique , Souris , Composés d'ammonium quaternaire/composition chimique , Composés d'ammonium quaternaire/pharmacologie , Femelle , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/composition chimique , Lipides/composition chimique , Souris de lignée BALB C , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/composition chimique , Adjuvants vaccinaux/composition chimique , Adjuvants vaccinaux/pharmacologie , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/composition chimique , COVID-19/prévention et contrôle , COVID-19/immunologie , Liposomes
11.
Int J Biol Macromol ; 273(Pt 1): 133067, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38866287

RÉSUMÉ

Adjuvants, as the essential component of vaccines, are crucial in enhancing the magnitude, breadth and durability of immune responses. Unfortunately, commonly used Alum adjuvants predominantly provoke humoral immune response, but fail to evoke cellular immune response, which is crucial for the prevention of various chronic infectious diseases and cancers. Thus, it is necessary to develop effective adjuvants to simultaneously induce humoral and cellular immune response. In this work, we obtained a water soluble polysaccharide isolated and purified from Poria cocos, named as PCP, and explored the possibility of PCP as a vaccine adjuvant. The PCP, with Mw of 20.112 kDa, primarily consisted of →6)-α-D-Galp-(1→, with a small amount of →3)-ß-D-Glcp-(1 â†’ and →4)-ß-D-Glcp-(1→. Our results demonstrated that the PCP promoted the activation of dendritic cells (DCs) and macrophages in vitro. As the adjuvant to ovalbumin, the PCP facilitated the activation of DCs in lymph nodes, and evoked strong antibody response with a combination of Th1 and Th2 immune responses. Moreover, compared to Alum adjuvant, the PCP markedly induced a potent cellular response, especially the cytotoxic T lymphocytes response. Therefore, we confirmed that the PCP has great potential to be an available adjuvant for simultaneously inducing humoral and cellular immune responses.


Sujet(s)
Adjuvants immunologiques , Cellules dendritiques , Polyosides , Solubilité , Eau , Animaux , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/composition chimique , Polyosides/pharmacologie , Polyosides/composition chimique , Polyosides/isolement et purification , Souris , Eau/composition chimique , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Femelle , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Wolfiporia/composition chimique , Ovalbumine/immunologie , Immunité cellulaire/effets des médicaments et des substances chimiques , Immunité humorale/effets des médicaments et des substances chimiques , Poria/composition chimique
12.
ACS Nano ; 18(26): 16878-16894, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38899978

RÉSUMÉ

Aluminum salts still remain as the most popular adjuvants in marketed human prophylactic vaccines due to their capability to trigger humoral immune responses with a good safety record. However, insufficient induction of cellular immune responses limits their further applications. In this study, we prepare a library of silicon (Si)- or calcium (Ca)-doped aluminum oxyhydroxide (AlOOH) nanoadjuvants. They exhibit well-controlled physicochemical properties, and the dopants are homogeneously distributed in nanoadjuvants. By using Hepatitis B surface antigen (HBsAg) as the model antigen, doped AlOOH nanoadjuvants mediate higher antigen uptake and promote lysosome escape of HBsAg through lysosomal rupture induced by the dissolution of the dopant in the lysosomes in bone marrow-derived dendritic cells (BMDCs). Additionally, doped nanoadjuvants trigger higher antigen accumulation and immune cell activation in draining lymph nodes. In HBsAg and varicella-zoster virus glycoprotein E (gE) vaccination models, doped nanoadjuvants induce high IgG titer, activations of CD4+ and CD8+ T cells, cytotoxic T lymphocytes, and generations of effector memory T cells. Doping of aluminum salt-based adjuvants with biological safety profiles and immunostimulating capability is a potential strategy to mediate robust humoral and cellular immunity. It potentiates the applications of engineered adjuvants in the development of vaccines with coordinated immune responses.


Sujet(s)
Adjuvants immunologiques , Hydroxyde d'aluminium , Calcium , Antigènes de surface du virus de l'hépatite B , Silicium , Adjuvants immunologiques/composition chimique , Adjuvants immunologiques/pharmacologie , Animaux , Silicium/composition chimique , Souris , Antigènes de surface du virus de l'hépatite B/immunologie , Antigènes de surface du virus de l'hépatite B/composition chimique , Calcium/composition chimique , Hydroxyde d'aluminium/composition chimique , Hydroxyde d'aluminium/pharmacologie , Souris de lignée C57BL , Femelle , Vaccins/immunologie , Vaccins/composition chimique , Cellules dendritiques/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique , Humains , Oxyde d'aluminium
13.
J Med Chem ; 67(12): 9976-9990, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38886162

RÉSUMÉ

This study describes the design and synthesis of five TF-based cancer vaccine candidates using a lipid A mimetic as the carrier and a built-in adjuvant. All synthesized conjugates elicited robust and consistent TF-specific immune responses in mice without external adjuvants. Immunological studies subsequently conducted in wild-type and TLR4 knockout C57BL/6 mice demonstrated that the activation of TLR4 was the main reason that the synthesized lipid A mimetics increased the TF-specific immune responses. All antisera induced by these conjugates can specifically recognize, bind to, and induce the lysis of TF-positive cancer cells. Moreover, representative conjugates 2 and 3 could effectively reduce the growth of tumors and prolong the survival time of mice in vivo, and the efficacies were better than glycoprotein TF-CRM197 with alum adjuvant. Lipid A mimetics could therefore be a promising platform for the development of new carbohydrate-based vaccine carriers with self-adjuvanting properties for the treatment of cancer.


Sujet(s)
Adjuvants immunologiques , Vaccins anticancéreux , Conception de médicament , Lipide A , Souris de lignée C57BL , Animaux , Lipide A/analogues et dérivés , Lipide A/composition chimique , Lipide A/pharmacologie , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/pharmacologie , Vaccins anticancéreux/synthèse chimique , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/synthèse chimique , Adjuvants immunologiques/composition chimique , Souris , Souris knockout , Humains , Femelle , Récepteur de type Toll-4/métabolisme , Lignée cellulaire tumorale
14.
Cell Commun Signal ; 22(1): 305, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38831299

RÉSUMÉ

As a major component of innate immunity and a positive regulator of interferons, the Stimulator of interferon gene (STING) has an immunotherapy potential to govern a variety of infectious diseases. Despite the recent advances regarding vaccines against COVID-19, nontoxic novel adjuvants with the potential to enhance vaccine efficacy are urgently desired. In this connection, it has been well-documented that STING agonists are applied to combat COVID-19. This approach is of major significance for boosting immune responses most likely through an autophagy-dependent manner in susceptible individuals against infection induced by severe acute respiratory syndrome Coronavirus (SARS­CoV­2). Given that STING agonists exert substantial immunomodulatory impacts under a wide array of pathologic conditions, these agents could be considered novel adjuvants for enhancing immunogenicity against the SARS-related coronavirus. Here, we intend to discuss the recent advances in STING agonists' recruitment to boost innate immune responses upon vaccination against SARS-related coronavirus infections. In light of the primordial role of autophagy modulation, the potential of being an antiviral vaccine adjuvant was also explored.


Sujet(s)
Autophagie , COVID-19 , Protéines membranaires , SARS-CoV-2 , Autophagie/immunologie , Autophagie/effets des médicaments et des substances chimiques , Humains , Protéines membranaires/immunologie , SARS-CoV-2/immunologie , COVID-19/immunologie , COVID-19/prévention et contrôle , Animaux , Vaccins contre la COVID-19/immunologie , Immunité innée/effets des médicaments et des substances chimiques , Adjuvants vaccinaux/usage thérapeutique , Adjuvants vaccinaux/pharmacologie , Adjuvants immunologiques/pharmacologie
15.
Int J Pharm ; 660: 124318, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-38852750

RÉSUMÉ

Avian influenza virus subtype H9N2 has the ability to infect birds and humans, further causing significant losses to the poultry industry and even posing a great threat to human health. Oral vaccine received particular interest for preventing majority infection due to its ability to elicit both mucosal and systemic immune responses, but their development is limited by the bad gastrointestinal (GI) environment, compact epithelium and mucus barrier, and the lack of effective mucosal adjuvants. Herein, we developed the dendritic fibrous nano-silica (DFNS) grafted with Cistanche deserticola polysaccharide (CDP) nanoparticles (CDP-DFNS) as an adjuvant for H9N2 vaccine. Encouragingly, CDP-DFNS facilitated the proliferation of T and B cells, and further induced the activation of T lymphocytes in vitro. Moreover, CDP-DFNS/H9N2 significantly promoted the antigen-specific antibodies levels in serum and intestinal mucosal of chickens, indicating the good ability to elicit both systemic and mucosal immunity. Additional, CDP-DFNS facilitate the activation of CD4 + and CD8 + T cells both in spleen and intestinal mucosal, and the indexes of immune organs. This study suggested that CDP-DFNS may be a new avenue for development of oral vaccine against pathogens that are transmitted via mucosal route.


Sujet(s)
Adjuvants immunologiques , Poulets , Immunité muqueuse , Sous-type H9N2 du virus de la grippe A , Vaccins antigrippaux , Grippe chez les oiseaux , Nanoparticules , Polyosides , Silice , Animaux , Sous-type H9N2 du virus de la grippe A/immunologie , Sous-type H9N2 du virus de la grippe A/effets des médicaments et des substances chimiques , Polyosides/administration et posologie , Polyosides/pharmacologie , Polyosides/composition chimique , Polyosides/immunologie , Silice/administration et posologie , Silice/composition chimique , Nanoparticules/administration et posologie , Vaccins antigrippaux/administration et posologie , Vaccins antigrippaux/immunologie , Immunité muqueuse/effets des médicaments et des substances chimiques , Grippe chez les oiseaux/prévention et contrôle , Grippe chez les oiseaux/immunologie , Adjuvants immunologiques/administration et posologie , Adjuvants immunologiques/pharmacologie , Administration par voie orale , Muqueuse intestinale/immunologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie
16.
Acta Biomater ; 183: 330-340, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38838909

RÉSUMÉ

Although vaccination with inactivated vaccines is a popular preventive method against pseudorabies virus (PRV) infection, inactivated vaccines have poor protection efficiency because of their weak immunogenicity. The development of an effective adjuvant is urgently needed to improve the efficacy of inactivated PRV vaccines. In this study, a promising nanocomposite adjuvant named as MIL@A-SW01-C was developed by combining polyacrylic acid-coated metal-organic framework MIL-53(Al) (MIL@A) and squalene (oil)-in-water emulsion (SW01) and then mixing it with a carbomer solution. One part of the MIL@A was loaded onto the oil/water interface of SW01 emulsion via hydrophobic interaction and coordination, while another part was dispersed in the continuous water phase using carbomer. MIL@A-SW01-C showed good biocompatibility, high PRV (antigen)-loading capability, and sustained antigen release. Furthermore, the MIL@A-SW01-C adjuvanted PRV vaccine induced high specific serum antibody titers, increased splenocyte proliferation and cytokine secretion, and a more balanced Th1/Th2 immune response compared with commercial adjuvants, such as alum and biphasic 201. In the mouse challenge experiment, two- and one-shot vaccinations resulted in survival rates of 73.3 % and 86.7 %, respectively. After one-shot vaccination, the host animal pigs were also challenged with wild PRV. A protection rate of 100 % was achieved, which was much higher than that observed with commercial adjuvants. This study not only establishes the superiority of MIL@A-SW01-C composite nanoadjuvant for inactivated PRV vaccine in mice and pigs but also presents an effective method for developing promising nanoadjuvants. STATEMENT OF SIGNIFICANCE: We have developed a nanocomposite of MIL-53(Al) and oil-in-water emulsion (MIL@A-SW01-C) as a promising adjuvant for the inactivated PRV vaccines. MIL@A-SW01-C has good biocompatibility, high PRV (antigen) loading capability, and prolonged antigen release. The developed nanoadjuvant induced much higher specific IgG antibody titers, increased splenocyte proliferation and cytokine secretion, and a more balanced Th1/Th2 immune response than commercial adjuvants alum and biphasic 201. In mouse challenge experiments, survival rates of 73.3 % and 86.7 % were achieved from two-shot and one-shot vaccinations, respectively. At the same time, a protection rate of 100 % was achieved with the host animal pigs challenged with wild PRV.


Sujet(s)
Adjuvants immunologiques , Émulsions , Animaux , Adjuvants immunologiques/pharmacologie , Émulsions/composition chimique , Souris , Suidae , Herpèsvirus porcin de type 1/immunologie , Vaccins contre la maladie d'Aujeszky/immunologie , Souris de lignée BALB C , Huiles/composition chimique , Femelle , Eau/composition chimique , Vaccins inactivés/immunologie , Maladie d'Aujeszky/prévention et contrôle , Maladie d'Aujeszky/immunologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Cytokines/métabolisme
17.
Eur J Pharm Biopharm ; 201: 114365, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38876362

RÉSUMÉ

Vaccines against influenza and many other infectious diseases require multiple boosters in addition to the primary dose to improve efficacy, but this approach is not ideal for compliance. The multiple doses could potentially be replaced by sustained or pulsatile release of antigens encapsulated in degradable microparticles (MPs). The efficacy of a vaccine is improved by adding an adjuvant, which can be co-delivered from the particles to enhance immunogenicity. Here, we developed degradable poly-lactic-co-glycolic acid (PLGA) (7-17 kDa) MPs capable of sustained release of ultraviolet killed influenza virus (A/PR/8/34) (kPR8) vaccine and the natural killer T (NKT) cell agonist alpha-galactosylceramide (α-GalCer) and tested their effectiveness at providing long-term protection against influenza virus infection in mice. Multiple formulations were developed for encapsulating the virus and adjuvant separately, and in combination. The MPs exhibited sustained release of both the virus and the adjuvant lasting more than a month. Co-encapsulation significantly increased the encapsulation efficiency (EE) of the vaccine but reduced the release duration. On the other hand, co-encapsulation led to a reduction in EE for the α-GalCer and a change in release profile to a higher initial burst followed by a linear release compared to a low initial burst and slower linear release. The α-GalCer also had considerably longer release duration compared to the vaccine. Mice injected with particle formulations co-encapsulating kPR8 and α-GalCer were protected from a lethal influenza virus infection 30 weeks after vaccination. This study demonstrates that PLGA MP based vaccines are promising for providing effective vaccination and possibly for replacing multiple doses with a single injection.


Sujet(s)
Préparations à action retardée , Galactosylcéramides , Vaccins antigrippaux , Cellules T tueuses naturelles , Infections à Orthomyxoviridae , Copolymère d'acide poly(lactique-co-glycolique) , Animaux , Galactosylcéramides/administration et posologie , Galactosylcéramides/immunologie , Galactosylcéramides/composition chimique , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique , Souris , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Vaccins antigrippaux/composition chimique , Cellules T tueuses naturelles/immunologie , Cellules T tueuses naturelles/effets des médicaments et des substances chimiques , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Femelle , Souris de lignée BALB C , Adjuvants immunologiques/administration et posologie , Adjuvants immunologiques/pharmacologie , Souris de lignée C57BL , Vaccins inactivés/immunologie , Vaccins inactivés/administration et posologie
18.
Commun Biol ; 7(1): 709, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38851856

RÉSUMÉ

Vaccination reduces morbidity and mortality due to infections, but efficacy may be limited due to distinct immunogenicity at the extremes of age. This raises the possibility of employing adjuvants to enhance immunogenicity and protection. Early IFNγ production is a hallmark of effective vaccine immunogenicity in adults serving as a biomarker that may predict effective adjuvanticity. We utilized mass cytometry (CyTOF) to dissect the source of adjuvant-induced cytokine production in human blood mononuclear cells (BMCs) from newborns (~39-week-gestation), adults (~18-63 years old) and elders (>65 years of age) after stimulation with pattern recognition receptors agonist (PRRa) adjuvants. Dimensionality reduction analysis of CyTOF data mapped the BMC compartment, elucidated age-specific immune responses and profiled PRR-mediated activation of monocytes and DCs upon adjuvant stimulation. Furthermore, we demonstrated PRRa adjuvants mediated innate IFNγ induction and mapped NK cells as the key source of TLR7/8 agonist (TLR7/8a) specific innate IFNγ responses. Hierarchical clustering analysis revealed age and TLR7/8a-specific accumulation of innate IFNγ producing γδ T cells. Our study demonstrates the application of mass cytometry and cutting-edge computational approaches to characterize immune responses across immunologically distinct age groups and may inform identification of the bespoke adjuvantation systems tailored to enhance immunity in distinct vulnerable populations.


Sujet(s)
Adjuvants immunologiques , Agranulocytes , Humains , Agranulocytes/immunologie , Agranulocytes/métabolisme , Adulte , Adulte d'âge moyen , Adjuvants immunologiques/pharmacologie , Sujet âgé , Jeune adulte , Adolescent , Interféron gamma/métabolisme , Nouveau-né , Femelle , Mâle , Facteurs âges , Immunité innée
19.
ACS Appl Bio Mater ; 7(6): 3877-3889, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38832760

RÉSUMÉ

Adjuvants and immunomodulators that effectively drive a Th17-skewed immune response are not part of the standard vaccine toolkit. Vaccine adjuvants and delivery technologies that can induce Th17 or Th1/17 immunity and protection against bacterial pathogens, such as tuberculosis (TB), are urgently needed. Th17-polarized immune response can be induced using agonists that bind and activate C-type lectin receptors (CLRs) such as macrophage inducible C-type lectin (Mincle). A simple but effective strategy was developed for codelivering Mincle agonists with the recombinant Mycobacterium tuberculosis fusion antigen, M72, using tunable silica nanoparticles (SNP). Anionic bare SNP, hydrophobic phenyl-functionalized SNP (P-SNP), and cationic amine-functionalized SNP (A-SNP) of different sizes were coated with three synthetic Mincle agonists, UM-1024, UM-1052, and UM-1098, and evaluated for adjuvant activity in vitro and in vivo. The antigen and adjuvant were coadsorbed onto SNP via electrostatic and hydrophobic interactions, facilitating multivalent display and delivery to antigen presenting cells. The cationic A-SNP showed the highest coloading efficiency for the antigen and adjuvant. In addition, the UM-1098-adsorbed A-SNP formulation demonstrated slow-release kinetics in vitro, excellent stability over 12 months of storage, and strong IL-6 induction from human peripheral blood mononuclear cells. Co-adsorption of UM-1098 and M72 on A-SNP significantly improved antigen-specific humoral and Th17-polarized immune responses in vivo in BALB/c mice relative to the controls. Taken together, A-SNP is a promising platform for codelivery and proper presentation of adjuvants and antigens and provides the basis for their further development as a vaccine delivery platform for immunization against TB or other diseases for which Th17 immunity contributes to protection.


Sujet(s)
Antigènes bactériens , Lectines de type C , Nanoparticules , Silice , Cellules Th17 , Lectines de type C/métabolisme , Lectines de type C/immunologie , Lectines de type C/agonistes , Nanoparticules/composition chimique , Cellules Th17/immunologie , Animaux , Silice/composition chimique , Souris , Antigènes bactériens/immunologie , Antigènes bactériens/administration et posologie , Antigènes bactériens/composition chimique , Mycobacterium tuberculosis/immunologie , Adjuvants immunologiques/composition chimique , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/administration et posologie , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Taille de particule , Test de matériaux , Humains , Femelle , Protéines membranaires/immunologie , Protéines membranaires/agonistes
20.
Pharm Res ; 41(6): 1163-1181, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38839718

RÉSUMÉ

OBJECTIVE: This study aims to utilize PEGylated poly (lactic-co-glycolic acid) (PLGA) nanoparticles as a delivery system for simultaneous administration of the BRAFV600E peptide, a tumor-specific antigen, and imiquimod (IMQ). The objective is to stimulate dendritic cell (DC) maturation, activate macrophages, and facilitate antigen presentation in C57BL6 mice. METHODS: PEG-PLGA-IMQ-BRAFV600E nanoparticles were synthesized using a PLGA-PEG-PLGA tri-block copolymer, BRAFV600E, and IMQ. Characterization included size measurement and drug release profiling. Efficacy was assessed in inhibiting BPD6 melanoma cell growth and activating immature bone marrow DCs, T cells, macrophages, and splenocyte cells through MTT and ELISA assays. In vivo, therapeutic and immunogenic effects potential was evaluated, comparing it to IMQ + BRAFV600E and PLGA-IMQ-BRAFV600E nanoparticles in inhibiting subcutaneous BPD6 tumor growth. RESULTS: The results highlight the successful synthesis of PEG-PLGA-IMQ-BRAFV600E nanoparticles (203 ± 11.1 nm), releasing 73.4% and 63.2% of IMQ and BARFV600E, respectively, within the initial 48 h. In vitro, these nanoparticles demonstrated a 1.3-fold increase in potency against BPD6 cells, achieving ~ 2.8-fold enhanced cytotoxicity compared to PLGA-IMQ-BRAFV600E. Moreover, PEG-PLGA-IMQ-BRAFV600E exhibited a 1.3-fold increase in potency for enhancing IMQ cytotoxic effects and a 1.1- to ~ 2.4-fold increase in activating DCs, T cells, macrophages, and splenocyte cells compared to IMQ-BRAFV600E and PLGA-IMQ-BRAFV600E. In vivo, PEG-PLGA-IMQ-BRAFV600E displayed a 1.3- to 7.5-fold increase in potency for inhibiting subcutaneous BPD6 tumor growth compared to the other formulations. CONCLUSIONS: The findings suggest that PEG-PLGA nanoparticles effectively promote DC maturation, T cell activation, and potentially macrophage activation. The study highlights the promising role of this nanocomposite in vaccine development.


Sujet(s)
Cellules dendritiques , Imiquimod , Mélanome , Souris de lignée C57BL , Nanoparticules , Polyéthylène glycols , Protéines proto-oncogènes B-raf , Animaux , Cellules dendritiques/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Polyéthylène glycols/composition chimique , Protéines proto-oncogènes B-raf/génétique , Mélanome/immunologie , Mélanome/traitement médicamenteux , Nanoparticules/composition chimique , Lignée cellulaire tumorale , Souris , Imiquimod/pharmacologie , Adjuvants immunologiques/administration et posologie , Adjuvants immunologiques/pharmacologie , Femelle , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Libération de médicament , Humains , Tumeurs cutanées/immunologie , Tumeurs cutanées/prévention et contrôle , Tumeurs cutanées/traitement médicamenteux
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...