Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20.172
Filter
1.
Carbohydr Polym ; 346: 122639, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39245530

ABSTRACT

Molecular weight (Mw) of ligand-mediated nanocarriers plays a pivotal role in their architecture and properties. In this study, self-assembled ovalbumin (OVA)-loaded nanoparticles were meticulously engineered by starch polyelectrolytes with different Mw. Results unveiled that, tailoring Mw of GRGDS pentapeptides-grafted carboxymethyl starch (G-CMS) displayed strong binding-affinity and transport efficiency through microfold cells (M cells) pathway in the simulated intestinal epithelial cell monolayer in which M cells were randomly located in the Caco-2 cells monolayer. Notably, nanoparticles assembled from G-CMS with relatively higher Mw exhibited more compact structures due to the stronger interactions between layers compared to that with relatively lower Mw, which rendered remarkably stable and only 19.01 % in vitro OVA leakage under conditions of the upper gastrointestinal tract. Subsequently, more intact nanoparticles reached M cells after in vitro digestion and exhibited higher transport efficiency through the M cells pathways (apparent permeability: 9.38 × 10-5 cm/s) than Caco-2 cells, attributing to specific- and non-specific binding affinity towards M cells. Therefore, optimal Mw tailoring of starch polyelectrolytes can mediate the molecular interactions among their assembled layers and the interactions with M cells to balance the structural compactness, release and transport efficacy of nanoparticles, holding promise for advancing M cells-targeting oral delivery technologies.


Subject(s)
Drug Carriers , Molecular Weight , Nanoparticles , Starch , Humans , Starch/chemistry , Starch/analogs & derivatives , Starch/metabolism , Caco-2 Cells , Nanoparticles/chemistry , Drug Carriers/chemistry , Ovalbumin/chemistry , Ovalbumin/metabolism , Drug Liberation , Biological Transport , M Cells
2.
Front Immunol ; 15: 1448320, 2024.
Article in English | MEDLINE | ID: mdl-39170611

ABSTRACT

Natural antibodies are used to compare immune systems across taxa, to study wildlife disease ecology, and as selection markers in livestock breeding. These immunoglobulins are present prior to immune stimulation. They are described as having low antigen specificity or polyreactive binding and are measured by binding to self-antigens or novel exogenous proteins. Most studies use only one or two antigens to measure natural antibodies and ignore potential effects of antigen specificity in analyses. It remains unclear how different antigen-specific natural antibodies are related or how diversity among natural antibodies may affect analyses of these immunoglobulins. Using genetically distinct lines of chickens as a model system, we tested the hypotheses that (1) antigen-specific natural antibodies are independent of each other and (2) antigen specificity affects the comparison of natural antibodies among animals. We used blood cell agglutination and enzyme-linked immunosorbent assays to measure levels of natural antibodies binding to four antigens: (i) rabbit erythrocytes, (ii) keyhole limpet hemocyanin, (iii) phytohemagglutinin, or (iv) ovalbumin. We observed that levels of antigen specific natural antibodies were not correlated. There were significant differences in levels of natural antibodies among lines of chickens, indicating genetic variation for natural antibody production. However, line distinctions were not consistent among antigen specific natural antibodies. These data show that natural antibodies are a pool of relatively distinct immunoglobulins, and that antigen specificity may affect interpretation of natural antibody function and comparative immunology.


Subject(s)
Chickens , Animals , Chickens/immunology , Rabbits , Antigens/immunology , Erythrocytes/immunology , Antibody Specificity/immunology , Ovalbumin/immunology , Antibodies/immunology , Hemocyanins/immunology , Phytohemagglutinins/immunology , Enzyme-Linked Immunosorbent Assay
3.
Mol Immunol ; 173: 110-116, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39106608

ABSTRACT

BACKGROUND: The prevalence of food allergies is on the rise, posing a significant challenge to public health. Rodents serve as the predominant animal model in food allergy research; yet, the application of rodent models proves to be a laborious and time-consuming endeavor. It is imperative to develop novel in vivo models. METHODS: Ovalbumin (OVA) was administered as the allergen, following the recommended dosage used in other species. During the sensitization phase, a dosage of 0.25 mg per 10 tails per 1 L was administered twice daily, and during the challenge phase, the dosage was increased to 3 times the initial level. The study explored two dimensions of sensitization: the mode of exposure, which can be either continuous or intermittent, and the duration of exposure, which includes 3 days, 5 days, and 7 days. We examined midgut pathological changes, immunoglobulins contents, and mRNA expressions associated to T helper cells (Th) 2 cytokines following exposure. RESULTS: A significant 109.3 % increase in the number of eosinophils was observed in the midgut histopathology following intermittent 5-day OVA exposure, which emerged as the most effective model. OVA exposure increased concentrations of immunoglobulin M (IgM) (105.2 %), IgZ (312.1 %), and IgD (304.3 %) in this model. The mRNA expressions of Th2-related interleukin (IL)-4 and IL-13 were also elevated by 132.8 % and 421.0 %, respectively. CONCLUSION: The intermittent 5-day OVA exposure was suggested to be the best constructed zebrafish food allergy model, which may be a potential tool for research into food allergies.


Subject(s)
Disease Models, Animal , Food Hypersensitivity , Ovalbumin , Zebrafish , Animals , Zebrafish/immunology , Food Hypersensitivity/immunology , Ovalbumin/immunology , Th2 Cells/immunology , Allergens/immunology , Cytokines/immunology , Cytokines/metabolism , Immunoglobulin E/immunology , Immunoglobulin E/blood
4.
Int J Med Sci ; 21(10): 1929-1944, 2024.
Article in English | MEDLINE | ID: mdl-39113893

ABSTRACT

Fine particulate matter (PM2.5) can damage airway epithelial barriers. The anion transport system plays a crucial role in airway epithelial barriers. However, the detrimental effect and mechanism of PM2.5 on the anion transport system are still unclear. In this study, airway epithelial cells and ovalbumin (OVA)-induced asthmatic mice were used. In transwell model, the adenosine triphosphate (ATP)-induced transepithelial anion short-circuit current (Isc) and airway surface liquid (ASL) significantly decreased after PM2.5 exposure. In addition, PM2.5 exposure decreased the expression levels of P2Y2R, CFTR and cytoplasmic free-calcium, but ATP can increase the expressions of these proteins. PM2.5 exposure increased the levels of Th2-related cytokines of bronchoalveolar lavage fluid, lung inflammation, collagen deposition and hyperplasisa of goblet cells. Interestingly, the administration of ATP showed an inhibitory effect on lung inflammation induced by PM2.5. Together, our study reveals that PM2.5 impairs the ATP-induced transepithelial anion Isc through downregulating P2Y2R/CFTR pathway, and this process may participate in aggravating airway hyperresponsiveness and airway inflammation. These findings may provide important insights on PM2.5-mediated airway epithelial injury.


Subject(s)
Asthma , Cystic Fibrosis Transmembrane Conductance Regulator , Particulate Matter , Receptors, Purinergic P2Y2 , Animals , Mice , Receptors, Purinergic P2Y2/metabolism , Receptors, Purinergic P2Y2/genetics , Asthma/metabolism , Asthma/pathology , Asthma/drug therapy , Asthma/chemically induced , Asthma/immunology , Particulate Matter/adverse effects , Particulate Matter/toxicity , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Humans , Adenosine Triphosphate/metabolism , Ovalbumin/immunology , Signal Transduction/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Down-Regulation/drug effects , Respiratory Mucosa/metabolism , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology
5.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 58-65, 2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39097895

ABSTRACT

This investigation aimed to explore the antioxidant, anti-inflammatory effects of Cade oil and its efficacy within a Wistar allergic asthma model. The antioxidant activity was assessed through various in vitro tests using chain-breaking antioxidant effects (radical scavenging and reducing abilities assays).  In vivo experiments involved Wistar rats categorized into four groups: negative control group, Ovalbumin-sensitised/challenged group, Cade oil-treated group, and Ovalbumin-sensitised/challenged Cade oil-treated group. These experiments aimed to evaluate oxidative stress parameters in the lungs and erythrocytes. The results indicated that the Cade oil exhibited significant antioxidant capabilities, evidenced by its radical scavenging activity against DPPH, ABTS, and Galvinoxyl radicals, with IC50 values ranging from 21.92 to 24.44 µg/mL. Besides, the reducing abilities methods showed A0,5 value ranging from 11.51 to 30.40  µg/mL for reducing power, Cupric ion reducing antioxidant capacity, and O-phenanthroline assays. Additionally, the IC50 value for ß-carotene scavenging was found to be (8.2 ± 0.25 µg/ml). Analysis revealed high levels of polyphenols and flavonoids in Cade oil, indicating rich polyphenol (275.21 ± 3.14 mg GAE/g DW) and flavonoid (28.23 ± 1.91 µg QE/mg) content. In vivo findings highlighted Cade oil's efficacy in reducing inflammatory cell recruitment, enhancing antioxidant status, reducing lipid peroxidation, and improving histopathological alterations within the allergic asthma model. These results demonstrated that Cade oil has a potent antioxidant, anti-inflammatory, and anti-asthmatic properties, suggesting its potential therapeutic application in asthma treatment.


Subject(s)
Anti-Asthmatic Agents , Anti-Inflammatory Agents , Antioxidants , Asthma , Disease Models, Animal , Juniperus , Rats, Wistar , Animals , Asthma/drug therapy , Asthma/metabolism , Antioxidants/pharmacology , Antioxidants/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Anti-Asthmatic Agents/pharmacology , Anti-Asthmatic Agents/therapeutic use , Anti-Asthmatic Agents/chemistry , Juniperus/chemistry , Rats , Plant Oils/pharmacology , Plant Oils/chemistry , Plant Oils/therapeutic use , Oxidative Stress/drug effects , Male , Ovalbumin , Lung/drug effects , Lung/pathology , Lung/metabolism
6.
Int J Mol Sci ; 25(16)2024 Aug 11.
Article in English | MEDLINE | ID: mdl-39201440

ABSTRACT

Th1 and Th2 cytokines determine the outcome of Leishmania major infection and immune protection depends mainly on memory T cells induced during vaccination. This largely hinges on the nature and type of memory T cells produced. In this study, transgenic Leishmania major strains expressing membrane-associated ovalbumin (mOVA) and soluble ovalbumin (sOVA) were used as a model to study whether fully differentiated Th1/Th2 and Th17 cells can recall immune memory and tolerate pathogen manipulation. Naïve OT-II T cells were polarised in vitro into Th1/Th2 cells, and these cells were transferred adoptively into recipient mice. Following the transferral of the memory cells, the recipient mice were challenged with OVA transgenic Leishmania major and a wild-type parasite was used a control. The in vitro-polarised T helper cells continued to produce the same cytokine signatures after being challenged by both forms of OVA-expressing Leishmania major parasites in vivo. This suggests that antigen-experienced cells remain the same or unaltered in the face of OVA-transgenic Leishmania major. Such ability of these antigen-experienced cells to remain resilient to manipulation by the parasite signifies that vaccines might be able to produce immune memory responses and defend against parasitic immune manipulation in order to protect the host from infection.


Subject(s)
Immunologic Memory , Leishmania major , Ovalbumin , Th1 Cells , Th17 Cells , Th2 Cells , Animals , Leishmania major/immunology , Ovalbumin/immunology , Mice , Th1 Cells/immunology , Th2 Cells/immunology , Th17 Cells/immunology , Cytokines/metabolism , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/parasitology , Disease Models, Animal , Mice, Inbred C57BL , Female , Mice, Transgenic
7.
PLoS Pathog ; 20(8): e1012457, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39186814

ABSTRACT

Chronic infection with Schistosoma mansoni parasites is associated with reduced allergic sensitization in humans, while schistosome eggs protects against allergic airway inflammation (AAI) in mice. One of the main secretory/excretory molecules from schistosome eggs is the glycosylated T2-RNAse Omega-1 (ω1). We hypothesized that ω1 induces protection against AAI during infection. Peritoneal administration of ω1 prior to sensitization with Ovalbumin (OVA) reduced airway eosinophilia and pathology, and OVA-specific Th2 responses upon challenge, independent from changes in regulatory T cells. ω1 was taken up by monocyte-derived dendritic cells, mannose receptor (CD206)-positive conventional type 2 dendritic cells (CD206+ cDC2), and by recruited peritoneal macrophages. Additionally, ω1 impaired CCR7, F-actin, and costimulatory molecule expression on myeloid cells and cDC2 migration in and ex vivo, as evidenced by reduced OVA+ CD206+ cDC2 in the draining mediastinal lymph nodes (medLn) and retainment in the peritoneal cavity, while antigen processing and presentation in cDC2 were not affected by ω1 treatment. Importantly, RNAse mutant ω1 was unable to reduce AAI or affect DC migration, indicating that ω1 effects are dependent on its RNAse activity. Altogether, ω1 hampers migration of OVA+ cDC2 to the draining medLn in mice, elucidating how ω1 prevents allergic airway inflammation in the OVA/alum mouse model.


Subject(s)
Cell Movement , Dendritic Cells , Ovalbumin , Schistosoma mansoni , Animals , Mice , Ovalbumin/immunology , Dendritic Cells/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Female , Mice, Inbred C57BL , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/prevention & control , Respiratory Hypersensitivity/parasitology , Th2 Cells/immunology , Inflammation/immunology
8.
Biomed Environ Sci ; 37(8): 811-822, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39198247

ABSTRACT

Objective: Air pollution is a leading public health issue. This study investigated the effect of air quality and pollutants on pulmonary function and inflammation in patients with asthma in Shanghai. Methods: The study monitored 27 asthma outpatients for a year, collecting data on weather, patient self-management [daily asthma diary, peak expiratory flow (PEF) monitoring, medication usage], spirometry and serum markers. To explore the potential mechanisms of any effects, asthmatic mice induced by ovalbumin (OVA) were exposed to PM 2.5. Results: Statistical and correlational analyses revealed that air pollutants have both acute and chronic effects on asthma. Acute exposure showed a correlation between PEF and levels of ozone (O 3) and nitrogen dioxide (NO 2). Chronic exposure indicated that interleukin-5 (IL-5) and interleukin-13 (IL-13) levels correlated with PM 2.5 and PM 10 concentrations. In asthmatic mouse models, exposure to PM 2.5 increased cytokine levels and worsened lung function. Additionally, PM 2.5 exposure inhibited cell proliferation by blocking the NF-κB and ERK phosphorylation pathways. Conclusion: Ambient air pollutants exacerbate asthma by worsening lung function and enhancing Th2-mediated inflammation. Specifically, PM 2.5 significantly contributes to these adverse effects. Further research is needed to elucidate the mechanisms by which PM 2.5 impacts asthma.


Subject(s)
Air Pollutants , Asthma , Lung , Asthma/chemically induced , China , Air Pollutants/adverse effects , Air Pollutants/toxicity , Animals , Humans , Female , Male , Adult , Mice , Middle Aged , Lung/drug effects , Lung/physiopathology , Mice, Inbred BALB C , Inflammation/chemically induced , Particulate Matter/toxicity , Particulate Matter/adverse effects , Cytokines/blood , Cytokines/metabolism , Ovalbumin
9.
Ultrason Sonochem ; 109: 107024, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39146820

ABSTRACT

Anthocyanins (AC) are vulnerable to degradation when affected by external factors. The present study employed ultrasound-assisted glycosylation of ovalbumin (OVA) and dextran (Dex) to generate conjugate carrier for AC to improve its stability. The results showed that sonication significantly improved the progression of Maillard reaction to OVA. Compared to traditional glycosylation, ultrasound treatment showed a higher degree of grafting, a lower number of free-SH, and smaller particle size and uniform distribution. The SDS-PAGE results indicated covalent interaction. Intrinsic fluorescence (INF), Fourier transform infrared spectroscopy (FTIR), and Circular dichroism (CD) analysis results suggested that ultrasound-assisted glycosylation altered the OVA structure. The scanning electron microscope (SEM) and X-ray diffractometer (XRD) observed that the ultrasound-assisted complex had a more compact and smoother structure and protein unfolding were better. The protein solubility increased significantly after glycosylation. Thermal gravimetric analysis (TGA) and Differential scanning calorimetry (DSC) indicated that the glycosylated conjugates can significantly improve the thermal stability of AC In addition, the AC showed an improved processing and storage stability when conjugated with glycosylated carrier. The glycosylated protein-anthocyanins complex may help provide new ideas and scientific basis for the development of naturally sourced anthocyanins-relevant products in pharmaceutical and food industry applications.


Subject(s)
Anthocyanins , Dextrans , Ovalbumin , Glycosylation , Dextrans/chemistry , Ovalbumin/chemistry , Anthocyanins/chemistry , Ultrasonic Waves , Solubility , Drug Carriers/chemistry
10.
Food Funct ; 15(17): 8810-8822, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39115430

ABSTRACT

Approximately two-thirds of patients with asthma, a common inflammatory airway disease, are thought to present with allergies. Probiotics and tryptophan metabolites are becoming increasingly important in treating allergic asthma. This study aimed to identify potential probiotic strains and tryptophan metabolites that could alleviate asthma symptoms. Based on in vitro fermentation experiments, we evaluated variations in probiotic capacity to metabolize tryptophan. Of the eight tested strains, Bifidobacterium animalis subsp. lactis CCFM1274 produced relatively high levels of indole-3-carboxaldehyde (I3C). A mouse model of allergic asthma was established by oral administration of ovalbumin (OVA) and was subjected to oral administration of probiotics. The results demonstrated that treatment with CCFM1274 reduced the tendency for body weight loss and mortality in OVA-induced asthmatic mice. Ingestion of CCFM1274 improved the infiltration of perivascular and peribronchial inflammatory cells in the lung sections stained with hematoxylin and eosin (H&E). This outcome was accompanied by a reduction in the serum levels of OVA-specific immunoglobulin E (OVA-sIgE) and in the levels of IL-10 and IL-17 in the bronchoalveolar lavage fluid (BALF). The linear discriminant analysis effect size (LEfSe) of the gut microbiota showed that CCFM1274 increased the relative abundance of Bifidobacterium. In conclusion, CCFM1274 remodeled intestinal tryptophan metabolism in mice and contributed to the improvement of allergic asthma.


Subject(s)
Asthma , Bifidobacterium animalis , Gastrointestinal Microbiome , Mice, Inbred BALB C , Probiotics , Tryptophan , Animals , Tryptophan/metabolism , Asthma/drug therapy , Mice , Probiotics/pharmacology , Female , Gastrointestinal Microbiome/drug effects , Ovalbumin , Disease Models, Animal , Immunoglobulin E , Bronchoalveolar Lavage Fluid/chemistry , Intestines/microbiology , Humans , Indoles/pharmacology
11.
Int Immunopharmacol ; 140: 112785, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39088915

ABSTRACT

OBJECTIVE: To observe the effects and mechanisms of Maresin2 on the function of DCs(Dendritic cells). METHOD: The levels of IL-6, IL-12, TNF-α and IL-1ß secreted by BMDCs (Bone marrow-derived Dendritic cells) after Maresin2 treatment were detected by ELISA. At the same time, the expressions of costimulatory molecules CD40 and CD86 on the surface, the ability of phagocytosis of ovalbumin(OVA) antigen, and antigen presentation function in BMDCs were analyzed by flow cytometry. Finally, MAPK and NF-κB pathway signaling phosphorylation in Maresin2-treated BMDCs were detected by western blot. RESULTS: The secretion levels of IL-6, IL-12, TNF-α and IL-1ß were significantly decreased in the Maresin2 treatment group after LPS treatment (P < 0.05). The expression levels of CD86 and CD40 were significantly decreased after Maresin2 treatment (P < 0.05). Maresin2 enhanced the phagocytosis ability of ovalbumin(OVA) (P < 0.05), but the ability of antigen presentation of BMDCs with the treatment of Maresin2 changed slightly (P > 0.05). Phosphorylation of p38, JNK, p65, ikka/ß and ERK peaked at 15 min in the LPS group, while phosphorylation of p-p38 and p-ERK weakened 30 min and 60 min after treatment with Maresin2. CONCLUSIONS: Maresin2 inhibits inflammatory cytokine secretion but enhances phagocytosis via the MAPK/NF-κB pathway in BMDCs, which may contribute to negatively regulating inflammation.


Subject(s)
Cytokines , Dendritic Cells , NF-kappa B , Phagocytosis , Signal Transduction , Animals , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , NF-kappa B/metabolism , Mice , Cytokines/metabolism , Signal Transduction/drug effects , Phagocytosis/drug effects , Cells, Cultured , Ovalbumin/immunology , Lipopolysaccharides/pharmacology , Lipopolysaccharides/immunology , Mice, Inbred C57BL , Cell Differentiation/drug effects , CD40 Antigens/metabolism , Antigen Presentation/drug effects , Docosahexaenoic Acids
12.
Biomolecules ; 14(8)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39199350

ABSTRACT

Graphene oxide (GO), a carbon-based material with oxygen-containing functional groups, can be applied in biomedicine for drug delivery, cancer therapy, and tissue regeneration. We have previously shown that nanoscale-sized graphene oxide (NGO), an oxidized graphene derivative, exhibits effective anti-inflammatory activity in a murine model of sepsis mediated by T helper (Th)1-promoting cytokines such as IFNγ and TNFα. However, whether NGO influences Th2-induced skin inflammation remains unclear. To address this issue, we employed an ovalbumin (OVA) plus aluminum hydroxide (Alum)-induced Th2-mediated skin inflammation model in conjunction with OVA-specific DO11.10 T cell receptor transgenic Balb/c mice. In vivo NGO injection upon OVA/Alum sensitization down-regulated OVA-elicited antigen-specific Th2 cells and GATA3-expressing Th2-type regulatory T cells. Next, we examined the effect of NGO injection on OVA/Alum-induced atopic dermatitis (AD)-like skin inflammation. NGO-injected mice exhibited significantly decreased Th2 disease phenotypes (e.g., a lower clinical score, decreased epidermal thickness and Th2 cell differentiation, and fewer infiltrated mast cells and basophils in skin lesions) compared with vehicle-injected control mice. Overall, our results suggest that NGOs are promising therapeutic materials for treating allergic diseases such as AD.


Subject(s)
Graphite , Mice, Inbred BALB C , Ovalbumin , Th2 Cells , Animals , Graphite/chemistry , Th2 Cells/immunology , Th2 Cells/drug effects , Mice , Alum Compounds/chemistry , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/pathology , Dermatitis, Atopic/immunology , Female , Inflammation/drug therapy , Inflammation/pathology , Inflammation/chemically induced , Down-Regulation/drug effects , Skin/drug effects , Skin/pathology , Skin/immunology , Nanoparticles/chemistry
13.
ACS Appl Mater Interfaces ; 16(35): 45917-45928, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39178210

ABSTRACT

Effective activation of an antigen-specific immune response hinges upon the intracellular delivery of cancer antigens to antigen-presenting cells (APCs), marking the initial stride in cancer vaccine development. Leveraging biomimetic topological morphology, we employed virus-like mesoporous silica nanoparticles (VMSNs) coloaded with antigens and toll-like receptor 9 (TLR9) agonists to craft a potent cancer vaccine. Our VMSNs could be efficiently internalized by APCs to a greater extent than their nonviral structured counterparts, thereby promoting the activation of APCs by upregulating the TLR9 pathway and cross-presenting ovalbumin (OVA) epitopes. In in vivo animal study, VMSN-based nanovaccines triggered substantial CD4+ and CD8+ lymphocyte populations in both lymph nodes and spleen while inducing the effector memory of adaptive T cells. Consequently, VMSN-based nanovaccines suppressed tumor progression and increased the survival rate of B16-OVA-bearing mice in both prophylactic and therapeutic studies. The combination of immune checkpoint blockade (ICB) with the VMSN-based nanovaccine has synergistic effects in significantly preventing tumor progression under therapeutic conditions. These findings highlight the potential of viral structure-mimicking mesoporous silica nanoparticles as promising candidates for antigen-delivering nanocarriers in vaccine development.


Subject(s)
Mice, Inbred C57BL , Nanoparticles , Silicon Dioxide , Silicon Dioxide/chemistry , Animals , Nanoparticles/chemistry , Mice , Cancer Vaccines/immunology , Cancer Vaccines/chemistry , Ovalbumin/chemistry , Ovalbumin/immunology , Porosity , Adaptive Immunity/drug effects , Humans , Antigen-Presenting Cells/immunology , Neoplasms/immunology , Female , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/agonists , Antigens, Neoplasm/immunology , Antigens, Neoplasm/chemistry , Cell Line, Tumor
14.
Mol Pharm ; 21(9): 4708-4725, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39115967

ABSTRACT

This study investigates the interaction between daphnetin and ovalbumin (OVA) as well as its potential to inhibit OVA fibrillation using both spectroscopic and computational analysis. A moderate binding affinity of 1 × 104 M-1 was observed between OVA and daphnetin, with a static quenched mechanism identified during the fluorescence quenching processes. Metal ions' (Cu2+ and Zn2+) presence led to an increase in the binding affinities of daphnetin toward OVA, mirroring a similar trend observed with the pH variation. Synchronous and 3D fluorescence studies indicated an increase in the polarity of the microenvironment surrounding the Trp residues during binding. Interestingly, circular dichroism and Fourier transform infrared studies showed a significant change in the secondary structure of OVA upon binding with daphnetin. The efficacy of daphnetin in inhibiting protein fibrillation was confirmed through thioflavin T and Congo Red binding assays along with fluorescence microscopic imaging analysis. The thermodynamic assessment showed positive ΔH° [+(29.34 ± 1.526) kJ mol-1] and ΔS° [+(181.726 ± 5.465) J mol-1] values, indicating the presence of the hydrophobic forces, while negative ΔG° signifies spontaneous binding interactions. These experimental findings were further correlated with computational analysis, revealing daphnetin dynamics within the binding site of OVA.


Subject(s)
Coumarins , Ovalbumin , Umbelliferones , Ovalbumin/metabolism , Umbelliferones/chemistry , Umbelliferones/metabolism , Hydrogen-Ion Concentration , Coumarins/chemistry , Coumarins/metabolism , Circular Dichroism , Protein Binding , Thermodynamics , Spectroscopy, Fourier Transform Infrared/methods , Molecular Docking Simulation , Zinc/chemistry , Zinc/metabolism , Hydrophobic and Hydrophilic Interactions , Spectrometry, Fluorescence , Binding Sites , Copper/chemistry , Protein Structure, Secondary
15.
Clin Transl Med ; 14(7): e1765, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39031979

ABSTRACT

BACKGROUND: The meticulous selection of appropriate vaccine adjuvants is crucial for optimizing immune responses. Traditionally, pemphigus vulgaris (PV), an autoimmune disorder, has been modelled using complete Freund's adjuvant (CFA). In this study, we aimed to discern potential variations in immune responses elicited by Toll-like receptor (TLR) ligands as compared to CFA. METHODS: A comprehensive investigation was conducted, comparing the effects of these adjuvants in conjunction with ovalbumin or desmoglein-3. Flow cytometry was employed to analyse distinct cell subsets, while enzyme-linked immunosorbent assay quantified antigen-specific antibodies and cytokine levels. Histological examination of harvested skin tissues and transcriptome analysis of skin lesions were performed to identify differentially expressed genes. RESULTS: TLR ligands demonstrated efficacy in inducing PV-like symptoms in wild-type mice, in contrast to CFA. This underscored the substantial impact of the adjuvant on self-antigen tolerance. Furthermore, we proposed an enhanced method for establishing a PV model through adoptive transfer, substituting CFA with TLR ligands. Our results revealed that in contrast to the perception that CFA being the most potent immunopotentiator reported, CFA promoted regulatory T cells (Treg), follicular regulatory T cells and IL-10-producing neutrophils, whereas TLR ligands downregulated CCL17 and IL-10. This suggested potential implications for the recruitment and activation of Treg subsets. While B cell and CD8+ T cell responses exhibited similarity, CFA induced less activation in dendritic cell subsets. A novel mouse model of PV and systemic comparison of immunostimulatory effects of adjuvants were provided by this study. CONCLUSIONS: The systematic comparison of CFA and TLR ligands shed light on the distinctive properties of these adjuvants, presenting innovative mouse models for the investigation of pemphigus. This study significantly contributes to adjuvant research and advances our understanding of PV pathogenesis. KEY POINTS/HIGHLIGHTS: Immunization with desmoglein 3 and Toll-like receptor (TLR) ligands effectively induces pemphigus symptoms in wild-type mice, whereas complete Freund's adjuvant (CFA) fails. TLR ligands heightened the autoreactivity of donor cells in the adoptive transfer pemphigus model. CFA promoted regulatory T cells and IL-10-producing neutrophils, whereas TLR ligands downregulated CCL17 and IL-10, leading to more effective immune responses.


Subject(s)
Adjuvants, Immunologic , Disease Models, Animal , Pemphigus , Toll-Like Receptors , Animals , Pemphigus/immunology , Mice , Toll-Like Receptors/metabolism , Toll-Like Receptors/immunology , Toll-Like Receptors/agonists , Adjuvants, Immunologic/pharmacology , Freund's Adjuvant/immunology , Mice, Inbred C57BL , Ligands , Ovalbumin/immunology , Female
16.
Allergol Immunopathol (Madr) ; 52(4): 91-96, 2024.
Article in English | MEDLINE | ID: mdl-38970271

ABSTRACT

Asthma is a widely prevalent chronic disease that brings great suffering to patients and may result in death if it turns severe. Jolkinolide B (JB) is one diterpenoid component separated from the dried roots of Euphorbia fischeriana Steud (Euphorbiaceae), and has anti--inflammatory, antioxidative, and antitumor properties. However, the detailed regulatory role and associated regulatory mechanism in the progression of asthma remain elusive. In this work, it was demonstrated that the extensive infiltration of bronchial inflammatory cells and the thickening of airway wall were observed in ovalbumin (OVA)-induced mice, but these impacts were reversed by JB (10 mg/kg) treatment, indicating that JB relieved the provocative symptoms in OVA-induced asthma mice. In addition, JB can control OVA-triggered lung function and pulmonary resistance. Moreover, JB attenuated OVA-evoked inflammation by lowering the levels of interleukin (IL)-4, IL-5, and IL-13. Besides, the activated nuclear factor kappa B (NF-κB) and transforming growth factor-beta-mothers against decapentaplegic homolog 3 (TGFß/smad3) pathways in OVA-induced mice are rescued by JB treatment. In conclusion, it was disclosed that JB reduced allergic airway inflammation and airway remodeling in asthmatic mice by modulating the NF-κB and TGFß/smad3 pathways. This work could offer new opinions on JB for lessening progression of asthma.


Subject(s)
Airway Remodeling , Asthma , Disease Models, Animal , Diterpenes , Mice, Inbred BALB C , NF-kappa B , Ovalbumin , Animals , Asthma/drug therapy , Asthma/immunology , Airway Remodeling/drug effects , Mice , Diterpenes/pharmacology , Diterpenes/administration & dosage , Diterpenes/therapeutic use , Ovalbumin/immunology , NF-kappa B/metabolism , Female , Transforming Growth Factor beta/metabolism , Cytokines/metabolism , Smad3 Protein/metabolism , Signal Transduction/drug effects , Humans , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Asthmatic Agents/pharmacology , Anti-Asthmatic Agents/therapeutic use , Euphorbia/chemistry
17.
J Gene Med ; 26(7): e3718, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38979822

ABSTRACT

BACKGROUND: Icariin (ICA) inhibits inflammatory response in various diseases, but the mechanism underlying ICA treating airway inflammation in asthma needs further understood. We aimed to predict and validate the potential targets of ICA against asthma-associated airway inflammation using network pharmacology and experiments. METHODS: The ovalbumin-induced asthma-associated airway inflammation mice model was established. The effects of ICA were evaluated by behavioral, airway hyperresponsiveness, lung pathological changes, inflammatory cell and cytokines counts. Next, the corresponding targets of ICA were mined via the SEA, CTD, HERB, PharmMapper, Symmap database and the literature. Pubmed-Gene and GeneCards databases were used to screen asthma and airway inflammation-related targets. The overlapping targets were used to build an interaction network, analyze gene ontology and enrich pathways. Subsequently, flow cytometry, quantitative real-time PCR and western blotting were employed for validation. RESULTS: ICA alleviated the airway inflammation of asthma; 402 targets of ICA, 5136 targets of asthma and 4531 targets of airway inflammation were screened; 216 overlapping targets were matched and predicted ICA possesses the potential to modulate asthmatic airway inflammation by macrophage activation/polarization. Additionally, ICA decreased M1 but elevated M2. Potential targets that were disrupted by asthma inflammation were restored by ICA treatment. CONCLUSIONS: ICA alleviates airway inflammation in asthma by inhibiting the M1 polarization of alveolar macrophages, which is related to metabolic reprogramming. Jun, Jak2, Syk, Tnf, Aldh2, Aldh9a1, Nos1, Nos2 and Nos3 represent potential targets of therapeutic intervention. The present study enhances understanding of the anti-airway inflammation effects of ICA, especially in asthma.


Subject(s)
Asthma , Disease Models, Animal , Flavonoids , Macrophage Activation , Macrophages, Alveolar , Network Pharmacology , Animals , Asthma/drug therapy , Asthma/metabolism , Mice , Flavonoids/pharmacology , Flavonoids/therapeutic use , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/immunology , Macrophage Activation/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Cytokines/metabolism , Ovalbumin , Lung/pathology , Lung/drug effects , Lung/metabolism , Female
18.
Molecules ; 29(13)2024 Jul 07.
Article in English | MEDLINE | ID: mdl-38999173

ABSTRACT

Ovalbumin (OVA), a protein vital for chick embryo nutrition, hydration, and antimicrobial protection, together with other egg-white proteins, migrates to the amniotic fluid and is orally absorbed by the embryo during embryogenesis. Recently, it has been shown that for optimal eggshell quality, the hen diet can be supplemented with manganese. Although essential for embryonic development, manganese in excess causes neurotoxicity. This study investigates whether OVA may be involved in the regulation of manganese levels. The binding of Mn(II) to OVA was investigated using electron paramagnetic resonance (EPR) spectroscopy. The results show that OVA binds a maximum of two Mn(II) ions, one with slightly weaker affinity, even in a 10-fold excess, suggesting it may have a protective role from Mn(II) overload. It seems that the binding of Mn(II), or the presence of excess Mn(II), does not affect OVA's tertiary structure, as evidenced from fluorescence and UV/vis measurements. Comparative analysis with bovine and human serum albumins revealed that they exhibit higher affinities for Mn(II) than OVA, most likely due to their essentially different physiological roles. These findings suggest that OVA does not play a role in the transport and storage of manganese; however, it may be involved in embryo protection from manganese-induced toxicity.


Subject(s)
Embryonic Development , Homeostasis , Manganese , Ovalbumin , Manganese/metabolism , Animals , Chick Embryo , Electron Spin Resonance Spectroscopy/methods , Humans , Protein Binding , Cattle , Chickens
19.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000141

ABSTRACT

Asthma is a chronic immunological disease related to oxidative stress and chronic inflammation; both processes promote airway remodeling with collagen deposition and matrix thickening, causing pulmonary damage and lost function. This study investigates the immunomodulation of C-phycocyanin (CPC), a natural blue pigment purified from cyanobacteria, as a potential alternative treatment to prevent the remodeling process against asthma. We conducted experiments using ovalbumin (OVA) to induce asthma in Sprague Dawley rats. Animals were divided into five groups: (1) sham + vehicle, (2) sham + CPC, (3) asthma + vehicle, (4) asthma + CPC, and (5) asthma + methylprednisolone (MP). Our findings reveal that asthma promotes hypoxemia, leukocytosis, and pulmonary myeloperoxidase (MPO) activity by increasing lipid peroxidation, reactive oxygen and nitrogen species, inflammation associated with Th2 response, and airway remodeling in the lungs. CPC and MP treatment partially prevented these physiological processes with similar action on the biomarkers evaluated. In conclusion, CPC treatment enhanced the antioxidant defense system, thereby preventing oxidative stress and reducing airway inflammation by regulating pro-inflammatory and anti-inflammatory cytokines, consequently avoiding asthma-induced airway remodeling.


Subject(s)
Airway Remodeling , Asthma , Disease Models, Animal , Ovalbumin , Oxidative Stress , Phycocyanin , Rats, Sprague-Dawley , Animals , Phycocyanin/pharmacology , Phycocyanin/therapeutic use , Asthma/drug therapy , Asthma/metabolism , Asthma/chemically induced , Oxidative Stress/drug effects , Ovalbumin/adverse effects , Rats , Airway Remodeling/drug effects , Inflammation/metabolism , Inflammation/drug therapy , Male , Lung/drug effects , Lung/pathology , Lung/metabolism , Cytokines/metabolism
20.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39000495

ABSTRACT

Patients with asthma experience elevated rates of mental illness. However, the molecular links underlying such lung-brain crosstalk remain ambiguous. Hypothalamic dysfunction is observed in many psychiatric disorders, particularly those with an inflammatory component due to many hypothalamic regions being unprotected by the blood-brain barrier. To gain a better insight into such neuropsychiatric sequelae, this study investigated gene expression differences in the hypothalamus following lung inflammation (asthma) induction in mice, using RNA transcriptome profiling. BALB/c mice were challenged with either bacterial lipopolysaccharide (LPS, E. coli) or ovalbumin (OVA) allergens or saline control (n = 7 per group), and lung inflammation was confirmed via histological examination of postmortem lung tissue. The majority of the hypothalamus was micro-dissected, and total RNA was extracted for sequencing. Differential expression analysis identified 31 statistically significant single genes (false discovery rate FDR5%) altered in expression following LPS exposure compared to controls; however, none were significantly changed following OVA treatment, suggesting a milder hypothalamic response. When gene sets were examined, 48 were upregulated and 8 were downregulated in both asthma groups relative to controls. REACTOME enrichment analysis suggests these gene sets are involved in signal transduction metabolism, immune response and neuroplasticity. Interestingly, we identified five altered gene sets directly associated with neurotransmitter signaling. Intriguingly, many of these altered gene sets can influence mental health and or/neuroinflammation in humans. These findings help characterize the links between asthma-induced lung inflammation and the brain and may assist in identifying relevant pathways and therapeutic targets for future intervention.


Subject(s)
Asthma , Disease Models, Animal , Hypothalamus , Lipopolysaccharides , Lung , Mice, Inbred BALB C , Transcriptome , Animals , Asthma/genetics , Asthma/metabolism , Asthma/pathology , Hypothalamus/metabolism , Mice , Lung/metabolism , Lung/pathology , Ovalbumin , Gene Expression Profiling , Female , Gene Expression Regulation
SELECTION OF CITATIONS
SEARCH DETAIL