Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.950
Filtrar
1.
Front Immunol ; 15: 1406643, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39263220

RESUMEN

Introduction: Immunogenicity, the unwanted immune response triggered by therapeutic antibodies, poses significant challenges in biotherapeutic development. This response can lead to the production of anti-drug antibodies, potentially compromising the efficacy and safety of treatments. The internalization of therapeutic antibodies into dendritic cells (DCs) is a critical factor influencing immunogenicity. Using monoclonal antibodies, with differences in non-specific cellular uptake, as tools to explore the impact on the overall risk of immunogenicity, this study explores how internalization influences peptide presentation and subsequently T cell activation. Materials and methods: To investigate the impact of antibody internalization on immunogenicity, untargeted toolantibodies with engineered positive or negative charge patches were utilized. Immature monocyte-derived DCs (moDCs), known for their physiologically relevant high endocytic activity, were employed for internalization assays, while mature moDCs were used for MHC-II associated peptide proteomics (MAPPs) assays. In addition to the lysosomal accumulation and peptide presentation, subsequent CD4+ T cell activation has been assessed. Consequently, a known CD4+ T cell epitope from ovalbumin was inserted into the tool antibodies to evaluate T cell activation on a single, shared epitope. Results: Antibodies with positive charge patches exhibited higher rates of lysosomal accumulation and epitope presentation compared to those with negative charge patches or neutral surface charge. Furthermore, a direct correlation between internalization rate and presentation on MHC-II molecules could be established. To explore the link between internalization, peptide presentation and CD4+ T cell activation, tool antibodies containing the same OVA epitope were used. Previous observations were not altered by the insertion of the OVA epitope and ultimately, an enhanced CD4+ T cell response correlated with increased internalization in DCs and peptide presentation. Discussion: These findings demonstrate that the biophysical properties of therapeutic antibodies, particularly surface charge, play a crucial role in their internalization into DCs. Antibodies internalized faster and processed by DCs, are also more prone to be presented on their surface leading to a higher risk of triggering an immune response. These insights underscore the importance of considering antibody surface charge and other properties that enhance cellular accumulation during the preclinical development of biotherapeutics to mitigate immunogenicity risks.


Asunto(s)
Presentación de Antígeno , Células Dendríticas , Activación de Linfocitos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Presentación de Antígeno/inmunología , Activación de Linfocitos/inmunología , Anticuerpos Monoclonales/inmunología , Linfocitos T CD4-Positivos/inmunología , Epítopos de Linfocito T/inmunología , Factores de Riesgo , Endocitosis/inmunología , Ovalbúmina/inmunología
2.
Eur J Med Chem ; 278: 116792, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39217861

RESUMEN

Enhancing the efficacy of subunit vaccines relies significantly on the utilization of potent adjuvants, particularly those capable of triggering multiple immune pathways. To achieve synergistic immune augmentation by Toll-like receptor 4 agonist (TLR4a) and nucleotide-binding oligomerization-domain-containing protein 2 agonist (NOD2a), in this work, we conjugated RC529 (TLR4a) and MDP (NOD2a) to give RC529-MDP, and evaluated its adjuvanticity for OVA antigen. Compared to the unconjugated RC529+MDP, RC529-MDP remarkably enhanced innate immune responses with 6.8-fold increase in IL-6 cytokine, and promoted the maturation of antigen-presenting cells (APCs), possibly because of the conjugation of multiple agonists ensuring their delivery to the same cell and activation of various signaling pathways within that cell. Furthermore, RC529-MDP improved OVA-specific antibody response, T cells response and the memory T cells ratio relative to the unconjugated mixture. Therefore, covalently conjugating TLR4 agonist and NOD2 agonist was an effective strategy to enhance immune responses, providing the potential to design and develop more effective vaccines.


Asunto(s)
Acetilmuramil-Alanil-Isoglutamina , Adyuvantes Inmunológicos , Proteína Adaptadora de Señalización NOD2 , Receptor Toll-Like 4 , Proteína Adaptadora de Señalización NOD2/agonistas , Proteína Adaptadora de Señalización NOD2/metabolismo , Receptor Toll-Like 4/agonistas , Animales , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/química , Acetilmuramil-Alanil-Isoglutamina/farmacología , Acetilmuramil-Alanil-Isoglutamina/química , Ratones , Ovalbúmina/inmunología , Humanos , Ratones Endogámicos C57BL , Estructura Molecular , Relación Dosis-Respuesta a Droga , Relación Estructura-Actividad
3.
Allergol Immunopathol (Madr) ; 52(5): 29-35, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39278848

RESUMEN

Allergic rhinitis (AR) is a chronic, non-infectious inflammatory condition of the nasal mucosa mediated by IgE. There is a need for the development of novel medications to treat this ailment. Isoorientin is a naturally occurring flavonoid that possesses antioxidant, anti--inflammatory, and various other advantageous characteristics. However, its potential effects on AR remain unclear. This study evaluates the therapeutic effects of isoorientin on ovalbumin (OVA)-induced allergic rhinitis (AR) in mice and explores the underlying mechanism. Our study revealed that isoorientin administration effectively decreased the frequency of nose rubbing and sneezing in AR mice. The groups treated with isoorientin showed a significant decrease in serum levels of IgE and histamine, with reductions of 40% and 30%, respectively. Isoorientin ameliorated inflammation of the nasal mucosa and restored the Th1/Th2 balance. In addition, isoorientin inhibited the activation of the NF-κB pathway in nasal tissues. In summary, Isoorientin alleviates OVA-stimulated AR in mice by restoring Th1/Th2 balance and blocking the NF-κB pathway. Thus, isoorientin exhibits promise as a natural therapeutic agent for allergic rhinitis.


Asunto(s)
Modelos Animales de Enfermedad , Inmunoglobulina E , Luteolina , Ratones Endogámicos BALB C , FN-kappa B , Mucosa Nasal , Ovalbúmina , Rinitis Alérgica , Balance Th1 - Th2 , Animales , Luteolina/farmacología , Ovalbúmina/inmunología , Ratones , Rinitis Alérgica/inmunología , Rinitis Alérgica/tratamiento farmacológico , Balance Th1 - Th2/efectos de los fármacos , Mucosa Nasal/inmunología , Mucosa Nasal/efectos de los fármacos , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , FN-kappa B/metabolismo , Células Th2/inmunología , Femenino , Humanos , Alérgenos/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Células TH1/inmunología , Células TH1/efectos de los fármacos , Histamina/metabolismo , Histamina/sangre
4.
Allergol Immunopathol (Madr) ; 52(5): 51-58, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39278851

RESUMEN

Allergic asthma is an important public health problem and is a complicated respiratory sickness that is characterized by bronchial inflammation, bronchoconstriction, and breathlessness. Asthma is orchestrated by type 2 immune response and remodeling is one of the important outputted problem in chronic asthma. Thymol is a naturally occurring monocyclic phenolic, it has a series of biological properties, and its immunomodulatory and anti-remodeling effects on allergic asthma were evaluated. The OVA-LPS-induced asthmatic mice were treated with thymol. Methacholine challenge test, eosinophil count, and levels of IL-4, IL-5, IL-13, and IL-33 in bronchoalveolar lavage fluid, total and OVA-specific IgE levels in serum, remodeling factors, gene expression of TGF-ß, Smad2, Smad3, and lung histopathology were done. Treatment with thymol could control AHR, eosinophil percentage levels of Th2 cytokines and Igs, remodeling factors, expression of TGF-ß, Smad2 and Smad3 genes, inflammation, goblet cell hyperplasia, and mucus production in asthmatic mice. Thymol can control asthma pathogens and related remodeling and fibrosis bio-factors and can be a potential treatment of asthma.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Asma , Modelos Animales de Enfermedad , Ratones Endogámicos BALB C , Transducción de Señal , Proteína smad3 , Timol , Factor de Crecimiento Transformador beta , Animales , Timol/farmacología , Asma/inmunología , Asma/tratamiento farmacológico , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Proteína smad3/metabolismo , Ratones , Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Citocinas/metabolismo , Femenino , Ovalbúmina/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Líquido del Lavado Bronquioalveolar/citología , Eosinófilos/inmunología , Eosinófilos/efectos de los fármacos , Humanos , Inmunoglobulina E/inmunología , Inmunoglobulina E/sangre , Proteína Smad2/metabolismo
5.
Int J Mol Sci ; 25(16)2024 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-39201440

RESUMEN

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.


Asunto(s)
Memoria Inmunológica , Leishmania major , Ovalbúmina , Células TH1 , Células Th17 , Células Th2 , Animales , Leishmania major/inmunología , Ovalbúmina/inmunología , Ratones , Células TH1/inmunología , Células Th2/inmunología , Células Th17/inmunología , Citocinas/metabolismo , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/parasitología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Femenino , Ratones Transgénicos
6.
Int Immunopharmacol ; 140: 112785, 2024 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-39088915

RESUMEN

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.


Asunto(s)
Citocinas , Células Dendríticas , FN-kappa B , Fagocitosis , Transducción de Señal , Animales , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , FN-kappa B/metabolismo , Ratones , Citocinas/metabolismo , Transducción de Señal/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Células Cultivadas , Ovalbúmina/inmunología , Lipopolisacáridos/farmacología , Lipopolisacáridos/inmunología , Ratones Endogámicos C57BL , Diferenciación Celular/efectos de los fármacos , Antígenos CD40/metabolismo , Presentación de Antígeno/efectos de los fármacos , Ácidos Docosahexaenoicos
7.
ACS Appl Mater Interfaces ; 16(35): 45917-45928, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39178210

RESUMEN

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.


Asunto(s)
Ratones Endogámicos C57BL , Nanopartículas , Dióxido de Silicio , Dióxido de Silicio/química , Animales , Nanopartículas/química , Ratones , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/química , Ovalbúmina/química , Ovalbúmina/inmunología , Porosidad , Inmunidad Adaptativa/efectos de los fármacos , Humanos , Células Presentadoras de Antígenos/inmunología , Neoplasias/inmunología , Femenino , Receptor Toll-Like 9/inmunología , Receptor Toll-Like 9/agonistas , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/química , Línea Celular Tumoral
8.
Mol Immunol ; 173: 110-116, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39106608

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Hipersensibilidad a los Alimentos , Ovalbúmina , Pez Cebra , Animales , Pez Cebra/inmunología , Hipersensibilidad a los Alimentos/inmunología , Ovalbúmina/inmunología , Células Th2/inmunología , Alérgenos/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Inmunoglobulina E/inmunología , Inmunoglobulina E/sangre
9.
ACS Nano ; 18(37): 25826-25840, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39196858

RESUMEN

Engineering nanovaccines capable of targeting dendritic cells (DCs) is desperately required to maximize antigen cross-presentation to effector immune cells, elicit strong immune responses, and avoid adverse reactions. Here, we showed that glucose transporter 1 (Glut-1) on DCs is a reliable target for delivering antigens to DCs, and thus, a versatile antigen delivery strategy using glucosylated nanovaccines was developed for DC-targeted antigen delivery and tumor immunotherapy. The developed glucosylated ovalbumin-loaded nanovaccines highly accumulated in lymph nodes and efficiently engaged with Glut-1 on DCs to accelerate intracellular antigen delivery and promote DC maturation and antigen presentation, which elicited potent antitumor immunity to prevent and inhibit ovalbumin-expressing melanoma. Moreover, immunotherapeutic experiments in DC- and macrophage-depleted animal models confirmed that the glucosylated nanovaccines functioned mainly through DCs. In addition, the neoantigen-delivering glucosylated nanovaccines were further engineered to elicit tumor-specific immune responses against MC38 tumors. This study offers a DC-targeted antigen delivery strategy for cancer immunotherapy.


Asunto(s)
Vacunas contra el Cáncer , Células Dendríticas , Inmunoterapia , Ratones Endogámicos C57BL , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Animales , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/administración & dosificación , Ratones , Ovalbúmina/inmunología , Ovalbúmina/química , Nanopartículas/química , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/química , Femenino , Presentación de Antígeno/inmunología , Línea Celular Tumoral , Humanos , Nanovacunas
10.
Int Immunopharmacol ; 141: 113018, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39216235

RESUMEN

Turmeric (Curcuma longa L.) extract (CLE) has been shown to elicit several pharmacological properties and is widely used in Asian traditional medicine. Herein, we assessed the impact of CLE on airway inflammation in BALB/c mice and A549 cells to clarify the underlying mechanism. An asthmatic mouse model was established by administering ovalbumin (OVA). CLE (100 or 300 mg/kg/day) was orally administered daily from days 18 to 23, with dexamethasone (3 mg/kg/day) used as the positive control. Human airway epithelial cells, A549, were stimulated using recombinant tumor necrosis factor-α. The CLE100 and CLE400 groups exhibited a significant downregulation in eosinophil counts, cytokine levels, and immunoglobulin-E levels. Moreover, CLE administration dose-dependently suppressed oxidative stress and airway inflammation in the lung tissue. CLE administration inhibited the phosphorylation of mitogen-activated protein kinases (MAPKs) and the expression and activity of matrix metalloproteinase (MMP)-9. In vitro, CLE treatment reduced mRNA levels of proinflammatory cytokines, MAPK phosphorylation, and the expression and activity of MMP-2 and MMP-9. Additionally, 50 µg/mL CLE and 2.5 µg/mL curcumin showed similar anti-inflammatory effects. Collectively, our findings revealed that CLE could suppress airway inflammation in asthmatic mice and A549 cells via oxidative stress-driven MAPK/MMPs signaling, suggesting that CLE could be developed as a potential treatment option for patients with asthma.


Asunto(s)
Antiinflamatorios , Asma , Curcuma , Metaloproteinasa 9 de la Matriz , Ratones Endogámicos BALB C , Estrés Oxidativo , Extractos Vegetales , Animales , Humanos , Estrés Oxidativo/efectos de los fármacos , Asma/tratamiento farmacológico , Asma/metabolismo , Asma/inmunología , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Curcuma/química , Células A549 , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Ratones , Citocinas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Ovalbúmina/inmunología , Modelos Animales de Enfermedad , Femenino , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo , Pulmón/inmunología , Inmunoglobulina E/sangre , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos
11.
Microb Pathog ; 195: 106881, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39197690

RESUMEN

BACKGROUND: The etiology of allergic rhinitis (AR) is not fully understood. Studies have shown that the maturation of children's immune systems is closely related to microecology. However, few studies have focused simultaneously on changes in respiratory and gut microbiota in AR and their correlation between microecological changes and Th1/Th2/Treg. OBJECTIVE: The aim is to investigate the pathogenesis of AR based on respiratory microecology, gut microecology, and Th1/Th2/Treg levels by applying microbiome techniques and correlation analysis. METHODS: Standardized OVA-induced AR mice were established. Serum OVA-sIgE, IL-4, IFN-γ, IL-10 were measured by ELISA, Tregs in lymph nodes were determined by flow cytometry, and the histological characteristics of nasal tissues were evaluated by Hematoxylin & Eosin (H&E). Nasal symptoms were observed to determine the reliability of the AR mouse model. Nasal lavage fluid (NALF) and fecal samples were collected after the last OVA challenge. The composition of respiratory microbiota in NALF and gut microbial in feces samples via 16S rRNA gene sequencing between the two groups, further explored the relationship between microbiota and Th1/Th2/Treg levels. RESULTS: In the AR group, the incidence of nose rubbing and sneezing in each mouse was significantly increased compared with the control group (all P < 0.001) and the inflammatory cell infiltration of NALF shows a significant increase in eosinophilic and neutrophilic infiltrates upon the AR group; H&E showed that the nasal mucosa of AR mice infiltration of massive eosinophils cells and neutrophils cells. OVA-sIgE and IL-4 in the AR group were increased (P < 0.01, P < 0.05) and IFN-γ, IL-10 were significantly decreased (P < 0.01, P < 0.05). Tregs showed a downward trend in the AR group, but there was no statistical difference. Compared with the control group, the respiratory microbiota of AR mice did not change significantly, while the gut microbiota changed significantly. In gut microbiota, compared to the control group, Shannon index in the AR group revealed a significant decrease at the genus level (P < 0.01), and Simpson index was significantly increased at all levels (all P < 0.05). PCoA also showed significant differences in beta diversity between the two groups (all P < 0.05). Compared to the control group, Deferribacteres at phylum level, Roseburia, Ruminiclostridium, Anaerotruncus at genus level were significantly decreased in the AR group (all P < 0.05). Spearman's rank correlation showed that OVA-sIgE was positively correlated with Bacteroidetes, Muribaculaceae and Erysipelotrichaceae (all P < 0.05); IL-4 was significantly negatively correlated with Epsilonbacteraeota and Deferribacteres (all P < 0.05). Treg was significantly positively correlated with Patescibacteria, Lachnospiraceae, and Saccharimonadaceae in gut microecology. CONCLUSION: Our results showed that the respiratory microbiota of AR mice was not significantly altered, but the gut microbiota varied significantly and there was a correlation between gut microbiota and Th1/Th2/Treg.


Asunto(s)
Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Ovalbúmina , ARN Ribosómico 16S , Sistema Respiratorio , Rinitis Alérgica , Linfocitos T Reguladores , Células TH1 , Células Th2 , Animales , Ratones , Microbioma Gastrointestinal/inmunología , Linfocitos T Reguladores/inmunología , Células Th2/inmunología , Rinitis Alérgica/inmunología , Rinitis Alérgica/microbiología , Células TH1/inmunología , Ovalbúmina/inmunología , ARN Ribosómico 16S/genética , Sistema Respiratorio/microbiología , Sistema Respiratorio/inmunología , Femenino , Ratones Endogámicos BALB C , Citocinas/metabolismo , Interleucina-10/genética , Inmunoglobulina E/sangre , Heces/microbiología , Líquido del Lavado Nasal/inmunología , Líquido del Lavado Nasal/microbiología , Interferón gamma/genética , Interleucina-4
12.
PLoS Pathog ; 20(8): e1012457, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39186814

RESUMEN

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.


Asunto(s)
Movimiento Celular , Células Dendríticas , Ovalbúmina , Schistosoma mansoni , Animales , Ratones , Ovalbúmina/inmunología , Células Dendríticas/inmunología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Femenino , Ratones Endogámicos C57BL , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/prevención & control , Hipersensibilidad Respiratoria/parasitología , Células Th2/inmunología , Inflamación/inmunología
13.
Immunobiology ; 229(5): 152837, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39089130

RESUMEN

It is known that conventional antigen presentation involves phagocytosis of antigens followed by its internalization in endocytic compartments and presentation of epitopes through MHC class II molecules for CD4 T cells. However, since 1976 a cross-presentation pathway has been studied, in which CD8 T cells are activated via MHC class I with antigens acquired through phagocytosis or endocytosis by dendritic cells (DCs). Among some important molecules involved in the cross-presentation, the C-type lectin receptor of the Dectin-1 cluster (CLECs), particularly the CLEC9A receptor, not only is expressed in dendritic cells but also presents a pivotal role in this context. In special, CLEC12A has been highlighted as a malaria pigment hemozoin (HZ) receptor. During Plasmodium infection, hemozoin crystals defend the parasite against heme toxicity within erythrocytes, as well as the released native HZ elicits pro-inflammatory responses and can induce cross-presentation. Particularly, this crystal can be synthesized from hematin anhydride and mimics the native form, and the gaps generated between the nanocrystal domains during its synthesis allow for substance coupling followed by its coating. Therefore, this study aimed to assess whether synthetic hemozoin (sHz) or hematin anhydride could be a nanocarrier and promote cross-presentation in dendritic cells. Firstly, it was verified that sHz can carry coated and coupled antigens, the compounds can associate to LAMP1-positive vesicles and decrease overall intracellular pH, which can potentially enhance the cross-presentation of ovalbumin and Leishmania infantum antigens. Thus, this study adds important data in the molecular intricacies of antigen presentation by showing not only the sHz immunomodulatory properties but also its potential applications as an antigen carrier.


Asunto(s)
Presentación de Antígeno , Reactividad Cruzada , Células Dendríticas , Hemoproteínas , Hemoproteínas/inmunología , Reactividad Cruzada/inmunología , Animales , Células Dendríticas/inmunología , Ratones , Nanopartículas/química , Humanos , Malaria/inmunología , Lectinas Tipo C/metabolismo , Lectinas Tipo C/inmunología , Ovalbúmina/inmunología
14.
Int J Med Sci ; 21(10): 1929-1944, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39113893

RESUMEN

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.


Asunto(s)
Asma , Regulador de Conductancia de Transmembrana de Fibrosis Quística , Material Particulado , Receptores Purinérgicos P2Y2 , Animales , Ratones , Receptores Purinérgicos P2Y2/metabolismo , Receptores Purinérgicos P2Y2/genética , Asma/metabolismo , Asma/patología , Asma/tratamiento farmacológico , Asma/inducido químicamente , Asma/inmunología , Material Particulado/efectos adversos , Material Particulado/toxicidad , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Humanos , Adenosina Trifosfato/metabolismo , Ovalbúmina/inmunología , Transducción de Señal/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/patología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología
15.
Front Immunol ; 15: 1448320, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39170611

RESUMEN

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.


Asunto(s)
Pollos , Animales , Pollos/inmunología , Conejos , Antígenos/inmunología , Eritrocitos/inmunología , Especificidad de Anticuerpos/inmunología , Ovalbúmina/inmunología , Anticuerpos/inmunología , Hemocianinas/inmunología , Fitohemaglutininas/inmunología , Ensayo de Inmunoadsorción Enzimática
16.
Sci Rep ; 14(1): 17241, 2024 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-39060348

RESUMEN

Studies have demonstrated that prior to puberty, girls have a lower incidence and severity of asthma symptoms compared to boys. This study aimed to explore the role of progesterone (P4), a sex hormone, in reducing inflammation and altering the immune microenvironment in a mouse model of allergic asthma induced by OVA. Female BALB/c mice with or without ovariectomy to remove the influence of sex hormones were used for the investigations. Serum, bronchoalveolar lavage fluid (BALF), and lung tissue samples were collected for analysis. The results indicated that P4 treatment was effective in decreasing inflammation and mucus secretion in the lungs of OVA-induced allergic asthma mice. P4 treatment also reduced the influx of inflammatory cells into the BALF and increased the levels of Th1 and Th17 cytokines while decreasing the levels of Th2 and Treg cytokines in both BALF and lung microenvironment CD45+ T cells. Furthermore, P4 inhibited the infiltration of inflammatory cells into the lungs, suppressed NETosis, and reduced the number of pulmonary CD4+ T cells while increasing the number of regulatory T cells. The neutrophil elastase inhibitor GW311616A also suppressed airway inflammation and mucus production and modified the secretion of immune Th1, Th2, Th17, and Treg cytokines in lung CD45+ immune cells. These changes led to an alteration of the immunological milieu with increased Th1 and Th17 cells, accompanied by decreased Th2, Treg, and CD44+ T cells, similar to the effects of P4 treatment. Treatment with P4 inhibited NETosis by suppressing the p38 pathway activation, leading to reduced reactive oxygen species production. Moreover, P4 treatment hindered the release of double-stranded DNA during NETosis, thereby influencing the immune microenvironment in the lungs. These findings suggest that P4 treatment may be beneficial in reducing inflammation associated with allergic asthma by modulating the immune microenvironment. In conclusion, this research indicates the potential of P4 as a therapeutic agent for ameliorating inflammation in OVA-induced allergic asthma mice.


Asunto(s)
Asma , Ovalbúmina , Progesterona , Animales , Femenino , Ratones , Asma/inmunología , Asma/tratamiento farmacológico , Asma/metabolismo , Líquido del Lavado Bronquioalveolar , Microambiente Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Trampas Extracelulares/efectos de los fármacos , Trampas Extracelulares/metabolismo , Trampas Extracelulares/inmunología , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/metabolismo , Pulmón/inmunología , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones Endogámicos BALB C , Ovalbúmina/inmunología , Progesterona/farmacología , Células Th17/inmunología , Células Th17/efectos de los fármacos , Células Th17/metabolismo
17.
Int Immunopharmacol ; 139: 112624, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39002519

RESUMEN

Airway epithelial-mesenchymal transition (EMT) is the important pathological feature of airway remodeling in asthma. While macrolides are not commonly used to treat asthma, they have been shown to have protective effects on the airways, in which mechanisms are not yet fully understood. This study aims to investigate the impact of clarithromycin on airway EMT in asthma and its potential mechanism. The results revealed an increase in Kv1.3 expression in the airways of ovalbumin (OVA)-induced asthmatic mice, with symptoms and pathological changes being alleviated after treatment with the Kv1.3 inhibitor 5-(4-phenoxybutoxy)psoralen (PAP-1). Clarithromycin was found to attenuate airway epithelial-mesenchymal transition through the inhibition of Kv1.3 and PI3K/Akt signaling. Further experiments in vitro confirmed that PAP-1 could mitigate EMT by modulating the PI3K/Akt signaling in airway epithelial cells undergoing transformation into mesenchymal cells. These findings confirmed that clarithromycin might have a certain protective effect on asthma-related airway remodeling and represent a promising treatment strategy.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Asma , Claritromicina , Transición Epitelial-Mesenquimal , Canal de Potasio Kv1.3 , Ratones Endogámicos BALB C , Ovalbúmina , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Animales , Transición Epitelial-Mesenquimal/efectos de los fármacos , Asma/tratamiento farmacológico , Asma/metabolismo , Asma/inducido químicamente , Asma/patología , Ovalbúmina/inmunología , Claritromicina/farmacología , Claritromicina/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Ratones , Canal de Potasio Kv1.3/metabolismo , Canal de Potasio Kv1.3/antagonistas & inhibidores , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Humanos , Modelos Animales de Enfermedad , Femenino , Ficusina/farmacología , Ficusina/uso terapéutico , Células Epiteliales/efectos de los fármacos
18.
Acta Biomater ; 185: 203-214, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39053817

RESUMEN

Vaccines play a critical role in combating infectious diseases and cancers, yet improving their efficacy remains challenging. Here, we introduce a separable nanocomposite hydrogel microneedle (NHMN) patch designed for intradermal and sustained delivery of ovalbumin (OVA), a model antigen, to enhance adaptive immune responses. The NHMN patch consists of an array of OVA-loaded microneedles made from photo-cross-linked methacrylated hyaluronic acid and laponite (LAP), supported by a hyaluronic acid backing. The incorporation of LAP not only enhances the mechanical strength of the pure hydrogel microneedles but also significantly prolongs OVA release. Furthermore, in vitro cell experiments demonstrate that NHMNs effectively activate dendritic cells without compromising cell viability. Upon skin penetration, NHMNs detach from the backing as the hyaluronic acid rapidly dissolves upon contact with the skin interstitial fluid, thereby acting as antigen reservoirs to release antigens to abundant skin dendritic cells. NHMNs containing 0.5% w/v LAP achieved a 15-day OVA release in vivo. Immunization studies demonstrate that the intradermal and sustained release of OVA via NHMNs elicited stronger and longer-lasting adaptive immune responses compared to conventional bolus injection. Given its easy to use, painless and minimally invasive features, the NHMN patch shows promise in improving vaccination accessibility and efficacy against a range of diseases. STATEMENT OF SIGNIFICANCE: The study introduces a separable nanocomposite hydrogel microneedle (NHMN) patch. This patch consists of an array of ovalbumin (OVA, a model antigen)-loaded microneedles made from photo-cross-linked methacrylated hyaluronic acid and laponite, with a hyaluronic acid backing, designed for intradermal and sustained delivery of antigens. This patch addresses several key challenges in traditional vaccination methods, including poor antigen uptake and presentation, and rapid systematic clearance. The incorporation of laponite enhances mechanical strength of microneedles, promotes dendritic cell activation, and significantly slows down antigen release. NHMN-based vaccination elicits stronger and longer-lasting adaptive immune responses compared to conventional bolus injection. This NHMN patch holds great potential for improving the efficacy, accessibility, and patient comfort of vaccinations against a range of diseases.


Asunto(s)
Inmunidad Adaptativa , Hidrogeles , Nanocompuestos , Agujas , Ovalbúmina , Animales , Ovalbúmina/inmunología , Ovalbúmina/administración & dosificación , Inmunidad Adaptativa/efectos de los fármacos , Hidrogeles/química , Nanocompuestos/química , Antígenos/administración & dosificación , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Femenino , Inyecciones Intradérmicas , Ácido Hialurónico/química , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacología , Silicatos
19.
J Control Release ; 373: 547-563, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39059501

RESUMEN

Melanoma, known for its aggressive metastatic nature, presents a formidable challenge in cancer treatment, where conventional therapies often fall short. This study introduces a pioneering approach utilizing metal-free nanosystem as tumor vaccines, spotlighting their potential in revolutionizing melanoma treatment. This work employed organic nitroxides, specifically 4-carboxy-TEMPO, in combination with chitosan (CS), to create a novel nanocomposite material - the CS-TEMPO-OVA nanovaccines. This composition not only improves biocompatibility and extends blood circulation time of TEMPO but also marks a significant departure from traditional gadolinium-based contrast agents in MRI technology, addressing safety concerns. CS-TEMPO-OVA nanovaccines demonstrate excellent biocompatibility at both the cellular and organoid level. They effectively stimulate bone marrow-derived dendritic cells (BMDCs), which in turn promote the maturation and activation of T cells. This ultimately leads to a strong production of essential cytokines. These nanovaccines serve a dual purpose as both therapeutic and preventive. By inducing an immune response, activating cytotoxic T cells, and promoting macrophage M1 polarization, they effectively inhibit melanoma growth and enhance survival in mouse models. When combined with αPD-1, the CS-TEMPO-OVA nanovaccines significantly bolster the infiltration of cytotoxic T lymphocytes (CTLs) within tumors, sparking a powerful systemic antitumor response that effectively curbs tumor metastasis. The ability of these nanovaccines to control both primary (subcutaneous) and metastatic B16-OVA tumors highlights their remarkable efficacy. Furthermore, the CS-TEMPO-OVA nanovaccine can be administered in vivo via both intravenous and intramuscular routes, both of which effectively enhance the T1 contrast of magnetic resonance imaging in tumor tissue. This study offers invaluable insights into the integrated application of these nanovaccines in both clinical diagnostics and treatment, marking a significant stride in cancer research and patient care.


Asunto(s)
Quitosano , Células Dendríticas , Inmunoterapia , Imagen por Resonancia Magnética , Ratones Endogámicos C57BL , Ovalbúmina , Nanomedicina Teranóstica , Animales , Células Dendríticas/inmunología , Inmunoterapia/métodos , Imagen por Resonancia Magnética/métodos , Nanomedicina Teranóstica/métodos , Ovalbúmina/inmunología , Ovalbúmina/administración & dosificación , Quitosano/química , Quitosano/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Femenino , Óxidos N-Cíclicos/química , Óxidos N-Cíclicos/administración & dosificación , Melanoma Experimental/terapia , Melanoma Experimental/inmunología , Ratones , Línea Celular Tumoral , Óxidos de Nitrógeno/administración & dosificación , Óxidos de Nitrógeno/química
20.
J Control Release ; 373: 568-582, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39067792

RESUMEN

Cancer vaccine is regarded as an effective immunotherapy approach mediated by dendritic cells (DCs) which are crucial for antigen presentation and the initiation of adaptive immune responses. However, lack of DC-targeting properties significantly hampers the efficacy of cancer vaccines. Here, by using the phage display technique, peptides targeting the endocytic receptor DEC-205 primarily found on cDC1s were initially screened. An optimized hydrolysis-resistant peptide, hr-8, was identified and conjugated to PLGA-loaded antigen (Ag) and CpG adjuvant nanoparticles, resulting in a DC-targeting nanovaccine. The nanovaccine hr-8-PLGA@Ag/CpG facilitates dendritic cell maturation and improves antigen cross-presentation. The nanovaccine can enhance the antitumor immune response mediated by CD8+ T cells by encapsulating the nanovaccine with either exogenous OVA protein antigen or endogenous gp100/E7 antigenic peptide. As a result, strong antitumor effects are observed in both anti-PD-1 responsive B16-OVA and anti-PD-1 non-responsive B16 and TC1 immunocompetent tumor models. In summary, this study presents the initial documentation of a nanovaccine that targets dendritic cells via the novel DEC-205 binding peptide. This approach offers a new method for developing cancer vaccines that can potentially improve the effectiveness of cancer immunotherapy.


Asunto(s)
Vacunas contra el Cáncer , Células Dendríticas , Inmunoterapia , Lectinas Tipo C , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor , Nanopartículas , Péptidos , Receptores de Superficie Celular , Células Dendríticas/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Animales , Lectinas Tipo C/inmunología , Receptores de Superficie Celular/inmunología , Inmunoterapia/métodos , Nanopartículas/química , Péptidos/química , Péptidos/administración & dosificación , Antígenos de Histocompatibilidad Menor/inmunología , Línea Celular Tumoral , Ovalbúmina/inmunología , Ovalbúmina/administración & dosificación , Femenino , Melanoma Experimental/terapia , Melanoma Experimental/inmunología , Ratones , Antígenos CD/inmunología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Linfocitos T CD8-positivos/inmunología , Nanovacunas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA