Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 15.992
1.
Hum Vaccin Immunother ; 20(1): 2346963, 2024 Dec 31.
Article En | MEDLINE | ID: mdl-38745461

COVID-19, caused by SARS-CoV-2, and meningococcal disease, caused by Neisseria meningitidis, are relevant infectious diseases, preventable through vaccination. Outer membrane vesicles (OMVs), released from Gram-negative bacteria, such as N. meningitidis, present adjuvant characteristics and may confer protection against meningococcal disease. Here, we evaluated in mice the humoral and cellular immune response to different doses of receptor binding domain (RBD) of SARS-CoV-2 adjuvanted by N. meningitidis C:2a:P1.5 OMVs and aluminum hydroxide, as a combined preparation for these pathogens. The immunization induced IgG antibodies of high avidity for RBD and OMVs, besides IgG that recognized the Omicron BA.2 variant of SARS-CoV-2 with intermediary avidity. Cellular immunity showed IFN-γ and IL-4 secretion in response to RBD and OMV stimuli, demonstrating immunologic memory and a mixed Th1/Th2 response. Offspring presented transferred IgG of similar levels and avidity as their mothers. Humoral immunity did not point to the superiority of any RBD dose, but the group immunized with a lower antigenic dose (0.5 µg) had the better cellular response. Overall, OMVs enhanced RBD immunogenicity and conferred an immune response directed to N. meningitidis too.


Antibodies, Viral , COVID-19 , Immunoglobulin G , Neisseria meningitidis , SARS-CoV-2 , Animals , Mice , Immunoglobulin G/blood , Neisseria meningitidis/immunology , Female , Antibodies, Viral/blood , Antibodies, Viral/immunology , COVID-19/prevention & control , COVID-19/immunology , SARS-CoV-2/immunology , Adjuvants, Immunologic/administration & dosage , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Immunity, Cellular , Immunity, Humoral , Mice, Inbred BALB C , Meningococcal Infections/prevention & control , Meningococcal Infections/immunology , Spike Glycoprotein, Coronavirus/immunology , Adjuvants, Vaccine/administration & dosage , Aluminum Hydroxide/administration & dosage , Aluminum Hydroxide/immunology , Immunization/methods , Antibody Affinity , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Meningococcal Vaccines/immunology , Meningococcal Vaccines/administration & dosage , Immunologic Memory , Th1 Cells/immunology
2.
Parasite Immunol ; 46(5): e13037, 2024 May.
Article En | MEDLINE | ID: mdl-38720446

The treatment for visceral leishmaniasis (VL) causes toxicity in patients, entails high cost and/or leads to the emergence of resistant strains. No human vaccine exists, and diagnosis presents problems related to the sensitivity or specificity of the tests. Here, we tested two phage clones, B1 and D11, which were shown to be protective against Leishmania infantum infection in a murine model as immunotherapeutics to treat mice infected with this parasite species. The phages were used alone or with amphotericin B (AmpB), while other mice received saline, AmpB, a wild-type phage (WTP) or WTP/AmpB. Results showed that the B1/AmpB and D11/AmpB combinations induced polarised Th1-type cellular and humoral responses, which were primed by high levels of parasite-specific IFN-γ, IL-12, TNF-α, nitrite and IgG2a antibodies, which reflected in significant reductions in the parasite load in distinct organs of the animals when analyses were performed 1 and 30 days after the treatments. Reduced organic toxicity was also found in these animals, as compared with the controls. In conclusion, preliminary data suggest the potential of the B1/AmpB and D11/AmpB combinations as immunotherapeutics against L. infantum infection.


Amphotericin B , Antibodies, Protozoan , Immunotherapy , Leishmania infantum , Leishmaniasis, Visceral , Mice, Inbred BALB C , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/drug therapy , Animals , Amphotericin B/therapeutic use , Amphotericin B/administration & dosage , Antibodies, Protozoan/blood , Leishmania infantum/immunology , Leishmania infantum/drug effects , Mice , Immunotherapy/methods , Female , Antiprotozoal Agents/therapeutic use , Antiprotozoal Agents/administration & dosage , Immunoglobulin G/blood , Parasite Load , Disease Models, Animal , Cell Surface Display Techniques , Cytokines/metabolism , Th1 Cells/immunology
3.
PLoS One ; 19(5): e0301687, 2024.
Article En | MEDLINE | ID: mdl-38718078

In the monitoring of human Toxoplasma gondii infection, it is crucial to confirm the development of a specific Th1/Th17 immune response memory. The use of a simple, specific, and sensitive assay to follow the T-cell activation is thus required. Current protocols are not always specific as stimulation with peptides is Human Leukocyte Antigen (HLA)-dependent, while stimulation with total-lysis antigens tends to stimulate seronegative donors resulting to false positives. Here, an improved ELISPOT protocol is reported, using peripheral blood mononuclear cells (PBMC) of T.gondii-infected donors, incubated with the inactivated parasite. The results showed that, contrary to standard protocols, a pre-incubation step at high cell density in presence of the inactivated parasite allowed a specific Th1/Th17 response with the secretion of IFN-γ, IL-2, IL-12 and IL-17 cytokines. This protocol allows to evaluate precisely the immune response after a T.gondii infection.


Enzyme-Linked Immunospot Assay , Th1 Cells , Th17 Cells , Toxoplasma , Toxoplasmosis , Humans , Th1 Cells/immunology , Th17 Cells/immunology , Enzyme-Linked Immunospot Assay/methods , Toxoplasmosis/immunology , Toxoplasma/immunology , Cytokines/immunology , Cytokines/metabolism , Leukocytes, Mononuclear/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism
4.
JCI Insight ; 9(9)2024 Apr 11.
Article En | MEDLINE | ID: mdl-38716729

Atopic dermatitis (AD) is an inflammatory skin condition with a childhood prevalence of up to 25%. Microbial dysbiosis is characteristic of AD, with Staphylococcus aureus the most frequent pathogen associated with disease flares and increasingly implicated in disease pathogenesis. Therapeutics to mitigate the effects of S. aureus have had limited efficacy and S. aureus-associated temporal disease flares are synonymous with AD. An alternative approach is an anti-S. aureus vaccine, tailored to AD. Experimental vaccines have highlighted the importance of T cells in conferring protective anti-S. aureus responses; however, correlates of T cell immunity against S. aureus in AD have not been identified. We identify a systemic and cutaneous immunological signature associated with S. aureus skin infection (ADS.aureus) in a pediatric AD cohort, using a combined Bayesian multinomial analysis. ADS.aureus was most highly associated with elevated cutaneous chemokines IP10 and TARC, which preferentially direct Th1 and Th2 cells to skin. Systemic CD4+ and CD8+ T cells, except for Th2 cells, were suppressed in ADS.aureus, particularly circulating Th1, memory IL-10+ T cells, and skin-homing memory Th17 cells. Systemic γδ T cell expansion in ADS.aureus was also observed. This study suggests that augmentation of protective T cell subsets is a potential therapeutic strategy in the management of S. aureus in AD.


Dermatitis, Atopic , Staphylococcal Skin Infections , Staphylococcus aureus , Dermatitis, Atopic/immunology , Dermatitis, Atopic/microbiology , Humans , Staphylococcus aureus/immunology , Child , Female , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Male , Child, Preschool , Skin/microbiology , Skin/immunology , Skin/pathology , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , Th17 Cells/immunology , Bayes Theorem , CD8-Positive T-Lymphocytes/immunology , Interleukin-10/metabolism , Interleukin-10/immunology , Intraepithelial Lymphocytes/immunology , Antigens, Differentiation, T-Lymphocyte , Membrane Glycoproteins
6.
Int Immunopharmacol ; 133: 112083, 2024 May 30.
Article En | MEDLINE | ID: mdl-38648714

Japanese encephalitis virus (JEV) infection is considered a global public health emergency. Severe peripheral neuropathy caused by JEV infection has increased disability and mortality rates in recent years. Because there are very few therapeutic options for JEV infection, prompt investigations of the ability of clinically safe, efficacious and globally available drugs to inhibit JEV infection and ameliorate peripheral neuropathy are urgently needed. In this study, we found that high doses of intravenous immunoglobulin, a function inhibitor of acid sphingomyelinase (FIASMA), inhibited acid sphingomyelinase (ASM) and ceramide activity in the serum and sciatic nerve of JEV-infected rats, reduced disease severity, reversed electrophysiological and histological abnormalities, significantly reduced circulating proinflammatory cytokine levels, inhibited Th1 and Th17 cell proliferation, and suppressed the infiltration of inflammatory CD4 + cells into the sciatic nerve. It also maintained the peripheral nerve-blood barrier without causing severe clinical side effects. In terms of the potential mechanisms, ASM was found to participate in immune cell differentiation and to activate immune cells, thereby exerting proinflammatory effects. Therefore, immunoglobulin is a FIASMA that reduces abnormal immune responses and thus targets the ASM/ceramide system to treat peripheral neuropathy caused by JEV infection.


Ceramides , Encephalitis Virus, Japanese , Encephalitis, Japanese , Immunoglobulins, Intravenous , Peripheral Nervous System Diseases , Sphingomyelin Phosphodiesterase , Animals , Ceramides/metabolism , Immunoglobulins, Intravenous/therapeutic use , Immunoglobulins, Intravenous/pharmacology , Encephalitis Virus, Japanese/immunology , Encephalitis Virus, Japanese/physiology , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/metabolism , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/immunology , Peripheral Nervous System Diseases/virology , Rats , Encephalitis, Japanese/drug therapy , Encephalitis, Japanese/immunology , Male , Sciatic Nerve/pathology , Cytokines/metabolism , Signal Transduction/drug effects , Humans , Th1 Cells/immunology , Rats, Sprague-Dawley , Th17 Cells/immunology
7.
Int Immunopharmacol ; 133: 112087, 2024 May 30.
Article En | MEDLINE | ID: mdl-38669951

EFHD2 (EF-hand domain family, member D2) has been identified as a calcium-binding protein with immunomodulatory effects. In this study, we characterized the phenotype of Efhd2-deficient mice in sepsis and examined the biological functions of EFHD2 in peripheral T cell activation and T helper (Th) cell differentiation. Increased levels of EFHD2 expression accompanied peripheral CD4+ T cell activation in the early stages of sepsis. Transcriptomic analysis indicated that immune response activation was impaired in Efhd2-deficient CD4+ T cells. Further, Efhd2-deficient CD4+ T cells isolated from the spleen of septic mice showed impaired T cell receptor (TCR)-induced Th differentiation, especially Th1 and Th17 differentiation. In vitro data also showed that Efhd2-deficient CD4+ T cells exhibit impaired Th1 and Th17 differentiation. In the CD4+ T cells and macrophages co-culture model for antigen presentation, the deficiency of Efhd2 in CD4+ T cells resulted in impaired formation of immunological synapses. In addition, Efhd2-deficient CD4+ T cells exhibited reduced levels of phospho-LCK and phospho-ZAP70, and downstream transcription factors including Nfat, Nfκb and Nur77 following TCR engagement. In summary, EFHD2 may promote TCR-mediated T cell activation subsequent Th1 and Th17 differentiation in the early stages of sepsis by regulating the intensity of TCR complex formation.


Calcium-Binding Proteins , Cell Differentiation , Lymphocyte Activation , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell , Sepsis , Signal Transduction , Animals , Sepsis/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/genetics , Lymphocyte Activation/immunology , Mice , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Th17 Cells/immunology , Cells, Cultured , T-Lymphocytes, Helper-Inducer/immunology , Macrophages/immunology , Th1 Cells/immunology , Male , Immunological Synapses/metabolism , Immunological Synapses/immunology
8.
Front Immunol ; 15: 1387534, 2024.
Article En | MEDLINE | ID: mdl-38650936

For several years, we have been committed to exploring the potential of Bordetella pertussis-derived outer membrane vesicles (OMVBp) as a promising third-generation vaccine against the reemerging pertussis disease. The results of our preclinical trials not only confirm its protective capacity against B. pertussis infection but also set the stage for forthcoming human clinical trials. This study delves into the examination of OMVBp as an adjuvant. To accomplish this objective, we implemented a two-dose murine schedule to evaluate the specific immune response induced by formulations containing OMVBp combined with 3 heterologous immunogens: Tetanus toxoid (T), Diphtheria toxoid (D), and the SARS-CoV-2 Spike protein (S). The specific levels of IgG, IgG1, and IgG2a triggered by the different tested formulations were evaluated using ELISA in dose-response assays for OMVBp and the immunogens at varying levels. These assays demonstrated that OMVBp exhibits adjuvant properties even at the low concentration employed (1.5 µg of protein per dose). As this effect was notably enhanced at medium (3 µg) and high concentrations (6 µg), we chose the medium concentration to determine the minimum immunogen dose at which the OMV adjuvant properties are significantly evident. These assays demonstrated that OMVBp exhibits adjuvant properties even at the lowest concentration tested for each immunogen. In the presence of OMVBp, specific IgG levels detected for the lowest amount of antigen tested increased by 2.5 to 10 fold compared to those found in animals immunized with formulations containing adjuvant-free antigens (p<0.0001). When assessing the adjuvant properties of OMVBp compared to the widely recognized adjuvant alum, we detected similar levels of specific IgG against D, T and S for both adjuvants. Experiments with OMVs derived from E. coli (OMVE.coli) reaffirmed that the adjuvant properties of OMVs extend across different bacterial species. Nonetheless, it's crucial to highlight that OMVBp notably skewed the immune response towards a Th1 profile (p<0.05). These collective findings emphasize the dual role of OMVBp as both an adjuvant and modulator of the immune response, positioning it favorably for incorporation into combined vaccine formulations.


Adjuvants, Immunologic , Bordetella pertussis , Immunoglobulin G , Th1 Cells , Whooping Cough , Bordetella pertussis/immunology , Animals , Adjuvants, Immunologic/administration & dosage , Mice , Th1 Cells/immunology , Whooping Cough/immunology , Whooping Cough/prevention & control , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Pertussis Vaccine/immunology , Pertussis Vaccine/administration & dosage , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Spike Glycoprotein, Coronavirus/immunology , Mice, Inbred BALB C , SARS-CoV-2/immunology , Bacterial Outer Membrane Proteins/immunology , Humans , COVID-19/immunology , COVID-19/prevention & control , Tetanus Toxoid/immunology
9.
Vet Res ; 55(1): 53, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38658996

Gene expression for Th1/Th2 cytokines (IL-4 and IFN-É£), regulatory cytokines (TGF-ß and IL-10) and the transcriptional factor FoxP3 was analyzed in the liver and hepatic lymph nodes (HLN) from sheep immunized with partially protective and non-protective vaccine candidates and challenged with Fasciola hepatica. FoxP3 T cells were also evaluated by immunohistochemistry (IHQ). The most remarkable difference between the partially protected vaccinated (V1) group and the non-protected vaccinated (V2) group was a more severe expansion of FoxP3 T cells recorded by IHQ in both the liver and HLN of the V2 group as compared to the V1 group, whereas no differences were found between the V2 group and the infected control (IC) group. Similar results were recorded for FoxP3 gene expression although significant differences among V1 and V2 groups were only significant in the HLN, while FoxP3 gene expression was very similar in the V2 and IC groups both in the liver and HLN. No significant differences for the remaining cytokines were recorded between the V1 and V2 groups, but in the liver the V2 group shows significant increases of IFN-É£ and IL-10 as compared to the uninfected control (UC) group whereas the V1 group did not. The lower expansion of FoxP3 T cells and lower increase of IFN-É£ and IL-10 in the partially protected vaccinated group may be related with lower hepatic lesions and fluke burdens recorded in this group as compared to the other two infected groups. The most relevant change in regulatory cytokine gene expression was the significant increase of TGF-ß in the liver of IC, V1 and V2 groups as compared to the UC group, which could be related to hepatic lesions.


Cytokines , Fasciola hepatica , Fascioliasis , Forkhead Transcription Factors , Sheep Diseases , Animals , Fascioliasis/veterinary , Fascioliasis/prevention & control , Fascioliasis/immunology , Fasciola hepatica/immunology , Sheep , Forkhead Transcription Factors/metabolism , Sheep Diseases/prevention & control , Sheep Diseases/immunology , Sheep Diseases/parasitology , Cytokines/metabolism , Liver/parasitology , Liver/immunology , Vaccines/immunology , Vaccines/administration & dosage , Th1 Cells/immunology , Lymph Nodes/immunology , Female , Th2 Cells/immunology
10.
Int J Mol Sci ; 25(8)2024 Apr 12.
Article En | MEDLINE | ID: mdl-38673887

Zinc is an essential trace element that plays a crucial role in T cell immunity. During T cell activation, zinc is not only structurally important, but zinc signals can also act as a second messenger. This research investigates zinc signals in T cell activation and their function in T helper cell 1 differentiation. For this purpose, peripheral blood mononuclear cells were activated via the T cell receptor-CD3 complex, and via CD28 as a costimulatory signal. Fast and long-term changes in intracellular zinc and calcium were monitored by flow cytometry. Further, interferon (IFN)-γ was analyzed to investigate the differentiation into T helper 1 cells. We show that fast zinc fluxes are induced via CD3. Also, the intracellular zinc concentration dramatically increases 72 h after anti-CD3 and anti-CD28 stimulation, which goes along with the high release of IFN-γ. Interestingly, we found that zinc signals can function as a costimulatory signal for T helper cell 1 differentiation when T cells are activated only via CD3. These results demonstrate the importance of zinc signaling alongside calcium signaling in T cell differentiation.


CD28 Antigens , CD3 Complex , Cell Differentiation , Interferon-gamma , Lymphocyte Activation , Zinc , Humans , Zinc/metabolism , Zinc/pharmacology , CD28 Antigens/metabolism , CD3 Complex/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Cell Differentiation/drug effects , Interferon-gamma/metabolism , Ionophores/pharmacology , T-Lymphocytes/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Signal Transduction/drug effects , Calcium/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/drug effects
11.
Int J Mol Sci ; 25(8)2024 Apr 19.
Article En | MEDLINE | ID: mdl-38674086

Biomaterials are extensively used as replacements for damaged tissue with bioactive glasses standing out as bone substitutes for their intrinsic osteogenic properties. However, biomaterial implantation has the following risks: the development of implant-associated infections and adverse immune responses. Thus, incorporating metallic ions with known antimicrobial properties can prevent infection, but should also modulate the immune response. Therefore, we selected silver, copper and tellurium as doping for bioactive glasses and evaluated the immunophenotype and cytokine profile of human T-cells cultured on top of these discs. Results showed that silver significantly decreased cell viability, copper increased the T helper (Th)-1 cell percentage while decreasing that of Th17, while tellurium did not affect either cell viability or immune response, as evaluated via multiparametric flow cytometry. Multiplex cytokines assay showed that IL-5 levels were decreased in the copper-doped discs, compared with its undoped control, while IL-10 tended to be lower in the doped glass, compared with the control (plastic) while undoped condition showed lower expression of IL-13 and increased MCP-1 and MIP-1ß secretion. Overall, we hypothesized that the Th1/Th17 shift, and specific cytokine expression indicated that T-cells might cross-activate other cell types, potentially macrophages and eosinophils, in response to the scaffolds.


Cytokines , Glass , Humans , Glass/chemistry , Cytokines/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Cell Survival/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Metals/chemistry , Copper/chemistry , Ions , Cells, Cultured , Th17 Cells/immunology , Th1 Cells/immunology , Th1 Cells/drug effects
12.
Int J Mol Sci ; 25(8)2024 Apr 20.
Article En | MEDLINE | ID: mdl-38674095

During periodontitis, the extracellular capsule of Porphyromonas gingivalis favors alveolar bone loss by inducing Th1 and Th17 patterns of lymphocyte response in the infected periodontium. Dendritic cells recognize bacterial antigens and present them to T lymphocytes, defining their activation and polarization. Thus, dendritic cells could be involved in the Th1 and Th17 response induced against the P. gingivalis capsule. Herein, monocyte-derived dendritic cells were obtained from healthy individuals and then stimulated with different encapsulated strains of P. gingivalis or two non-encapsulated isogenic mutants. Dendritic cell differentiation and maturation were analyzed by flow cytometry. The mRNA expression levels for distinct Th1-, Th17-, or T-regulatory-related cytokines and transcription factors, as well as TLR2 and TLR4, were assessed by qPCR. In addition, the production of IL-1ß, IL-6, IL-23, and TNF-α was analyzed by ELISA. The encapsulated strains and non-encapsulated mutants of P. gingivalis induced dendritic cell maturation to a similar extent; however, the pattern of dendritic cell response was different. In particular, the encapsulated strains of P. gingivalis induced higher expression of IRF4 and NOTCH2 and production of IL-1ß, IL-6, IL-23, and TNF-α compared with the non-encapsulated mutants, and thus, they showed an increased capacity to trigger Th1 and Th17-type responses in human dendritic cells.


Cytokines , Dendritic Cells , Porphyromonas gingivalis , Th17 Cells , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Porphyromonas gingivalis/immunology , Humans , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Th17 Cells/immunology , Th17 Cells/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Cytokines/metabolism , Cell Differentiation , Th1 Cells/immunology , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Cells, Cultured , Bacterial Capsules/immunology , Bacterial Capsules/metabolism , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/microbiology , Tumor Necrosis Factor-alpha/metabolism
13.
J Med Virol ; 96(5): e29627, 2024 May.
Article En | MEDLINE | ID: mdl-38659381

The immune mechanism underlying hepatitis B surface antigen (HBsAg) loss, particularly type I inflammatory response, during pegylated interferon-α (PEG-IFN) therapy remains unclear. In this study, we aimed to elucidate such immune mechanisms. Overall, 82 patients with chronic hepatitis B (CHB), including 41 with HBsAg loss (cured group) and 41 uncured patients, received nucleos(t)ide analogue and PEG-IFN treatments. Blood samples from all patients, liver tissues from 14 patients with CHB, and hepatic perfusate from 8 liver donors were collected for immune analysis. Jurkat, THP-1 and HepG2.2.15 cell lines were used in cell experiments. The proportion of IFN-γ+ Th1 cells was higher in the cured group than in the uncured group, which was linearly correlated with HBsAg decline and alanine aminotransferase (ALT) levels during treatment. However, CD8+ T cells were weakly associated with HBsAg loss. Serum and intrahepatic levels of Th1 cell-associated chemokines (C-X-C motif chemokine ligand [CXCL] 9, CXCL10, CXCL11, IFN-γ) were significantly lower in the cured patients than in patients with a higher HBsAg quantification during therapy. Serum from cured patients induced more M1 (CD68+CD86+ macrophage) cells than that from uncured patients. Patients with chronic HBV infection had significantly lower proportions of CD86+ M1 and CD206+ M2 macrophages in their livers than healthy controls. M1 polarization of intrahepatic Kupffer cells promoted HBsAg loss by upregulating the effector function of tissue-resident memory T cells with increased ALT levels. IFN-γ+ Th1 activates intrahepatic resident memory T cells to promote HBsAg loss by inducing M1 macrophage polarization.


Hepatitis B Surface Antigens , Hepatitis B, Chronic , Liver , Macrophages , Memory T Cells , Th1 Cells , Adult , Female , Humans , Male , Middle Aged , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Hepatitis B Surface Antigens/immunology , Hepatitis B virus/immunology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/drug therapy , Interferon-alpha , Interferon-gamma , Liver/immunology , Macrophages/immunology , Memory T Cells/immunology , Th1 Cells/immunology
14.
J Agric Food Chem ; 72(17): 10031-10045, 2024 May 01.
Article En | MEDLINE | ID: mdl-38629959

Casein (CN) is the primary allergenic protein in cow's milk, contributing to the worldwide escalating prevalence of food allergies. However, there remains limited knowledge regarding the effect of structural modifications on CN allergenicity. Herein, we prepared three modified CNs (mCN), including sodium dodecyl sulfate and dithiothreitol-induced linear CN (LCN), transglutaminase-cross-linked CN (TCN), and glucose-glycated CN (GCN). The electrophoresis results indicated widespread protein aggregation among mCN, causing variations in their molecular weights. The unique internal and external structural characteristics of mCN were substantiated by disparities in surface microstructure, alterations in the secondary structure, variations in free amino acid contents, and modifications in functional molecular groups. Despite the lower digestibility of TCN and GCN compared to LCN, they significantly suppressed IL-8 production in Caco-2 cells without significantly promoting their proliferation. Moreover, GCN showed the weakest capacity to induce LAD2 cell degranulation. Despite the therapeutic effect of TCN, GCN-treated mice displayed the most prominent attenuation of allergic reactions and a remarkably restored Th1/Th2 imbalance, while LCN administration resulted in severe allergic phenotypes and endotypes in both cellular and murine models. This study highlighted the detrimental effect of linear modifications and underscored the significance of glycation in relation to CN allergenicity.


Allergens , Caseins , Mice, Inbred BALB C , Th1 Cells , Th2 Cells , Animals , Humans , Mice , Th2 Cells/immunology , Caseins/immunology , Caseins/chemistry , Th1 Cells/immunology , Allergens/immunology , Allergens/chemistry , Caco-2 Cells , Female , Glycosylation , Cattle , Homeostasis , Food Hypersensitivity/immunology
15.
Int Immunopharmacol ; 132: 111995, 2024 May 10.
Article En | MEDLINE | ID: mdl-38581993

Elevation of arginase enzyme activity in the lung contributes to the pathogenesis of various chronic inflammatory diseases and infections. Inhibition of arginase expression and activity is able to alleviate those effects. Here, we investigated the immunomodulatory effect of arginase inhibitor in C. neoformans infection. In the pulmonary cryptococcosis model that was shown to recapitulate human infection, we found arginase expression was excessively induced in the lung during the late stage of infection. To inhibit the activity of arginase, we administered a specific arginase inhibitor, nor-NOHA, during C. neoformans infection. Inhibition of arginase reduced eosinophil infiltration and level of IL-13 secretion in the lungs. Whole lung transcriptome RNA-sequencing analysis revealed that treatment with nor-NOHA resulted in shifting the Th2-type gene expression patterns induced by C. neoformans infection to the Th1-type immune profile, with higher expression of cytokines Ifng, Il6, Tnfa, Csf3, chemokines Cxcl9 and Cxcl10 and transcription factor Stat1. More importantly, mice treated with arginase inhibitor had more infiltrating brain leukocytes and enhanced gene expression of Th1-associated cytokines and chemokines that are known to be essential for protection against C. neoformans infection. Inhibition of arginase dramatically attenuated spleen and brain infection, with improved survival. Taken together, these studies demonstrated that inhibiting arginase activity induced by C. neoformans infection can modulate host immune response by enhancing protective type-1 immune response during C. neoformans infection. The inhibition of arginase activity could be an immunomodulatory target to enhance protective anti-cryptococcal immune responses.


Arginase , Arginine/analogs & derivatives , Cryptococcosis , Cryptococcus neoformans , Mice, Inbred C57BL , Animals , Arginase/metabolism , Arginase/antagonists & inhibitors , Arginase/genetics , Cryptococcosis/immunology , Cryptococcosis/drug therapy , Cryptococcus neoformans/immunology , Cryptococcus neoformans/drug effects , Mice , Lung/immunology , Lung/pathology , Lung/drug effects , Cytokines/metabolism , Cytokines/immunology , Female , Disease Models, Animal , Lung Diseases, Fungal/immunology , Lung Diseases, Fungal/drug therapy , Humans , Th2 Cells/immunology , Th2 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/drug effects , Brain/immunology , Brain/drug effects , Brain/pathology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use
16.
Vet Microbiol ; 293: 110088, 2024 Jun.
Article En | MEDLINE | ID: mdl-38640639

Orf virus (ORFV), a member of the genus Parapoxvirus, possesses an excellent immune activation capability, which makes it a promising immunomodulation agent. In this study, we evaluated ORFV as a novel adjuvant to enhance the immune response of mice to a subunit vaccine using porcine circovirus type 2 (PCV2) capsid (Cap) protein as a model. Our results showed that both inactivated and live attenuated ORFV activated mouse bone marrow-derived dendritic cells and increased expression of immune-related cytokines interleukin (IL)-1ß, IL-6, and TNF-α. Enhanced humoral and cellular immune responses were induced in mice immunized with PCV2 Cap protein combined with inactivated or live attenuated ORFV adjuvant compared with the aluminum adjuvant. Increased secretion of Th1 and Th2 cytokines by splenic lymphocytes in immunized mice further indicated that the ORFV adjuvant promoted a mixed Th1/Th2 immune response. Moreover, addition of the ORFV adjuvant to the PCV2 subunit vaccine significantly reduced the viral load in the spleen and lungs of PCV2-challenged mice and prevented pathological changes in lungs. This study demonstrates that ORFV enhances the immunogenicity of a PCV2 subunit vaccine by improving the adaptive immune response, suggesting the potential application of ORFV as a novel adjuvant.


Adjuvants, Immunologic , Circoviridae Infections , Circovirus , Cytokines , Orf virus , Vaccines, Subunit , Viral Vaccines , Animals , Circovirus/immunology , Mice , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Circoviridae Infections/prevention & control , Circoviridae Infections/veterinary , Circoviridae Infections/immunology , Circoviridae Infections/virology , Adjuvants, Immunologic/administration & dosage , Cytokines/immunology , Orf virus/immunology , Capsid Proteins/immunology , Female , Immunity, Cellular , Dendritic Cells/immunology , Viral Load , Antibodies, Viral/blood , Immunity, Humoral , Swine , Adjuvants, Vaccine , Mice, Inbred BALB C , Th1 Cells/immunology
17.
Microbiol Spectr ; 12(5): e0364623, 2024 May 02.
Article En | MEDLINE | ID: mdl-38497717

Anti-interferon-γ autoantibody (AIGA) syndrome may be the basis of disseminated Talaromyces marneffei infection in human immunodeficiency virus (HIV)-negative adults. However, the pathogenesis of Th1 cell immunity in T. marneffei infection with AIGA syndrome is unknown. A multicenter study of HIV-negative individuals with T. marneffei infection was conducted between September 2018 and September 2020 in Guangdong and Guangxi, China. Patients were divided into AIGA-positive (AP) and AIGA-negative (AN) groups according to the AIGA titer and neutralizing activity. The relationship between AIGA syndrome and Th1 immune deficiency was investigated by using AP patient serum and purification of AIGA. Fifty-five HIV-negative adults with disseminated T. marneffei infection who were otherwise healthy were included. The prevalence of AIGA positivity was 83.6%. Based on their AIGA status, 46 and 9 patients were assigned to the AP and AN groups, respectively. The levels of Th1 cells, IFN-γ, and T-bet were higher in T. marneffei-infected patients than in healthy controls. However, the levels of CD4+ T-cell STAT-1 phosphorylation (pSTAT1) and Th1 cells were lower in the AP group than in the AN group. Both the serum of patients with AIGA syndrome and the AIGA purified from the serum of patients with AIGA syndrome could reduce CD4+ T-cell pSTAT1, Th1 cell differentiation and T-bet mRNA, and protein expression. The Th1 cell immune response plays a pivotal role in defense against T. marneffei infection in HIV-negative patients. Inhibition of the Th1 cell immune response may be an important pathological effect of AIGA syndrome.IMPORTANCEThe pathogenesis of Th1 cell immunity in Talaromyces marneffei infection with anti-interferon-γ autoantibody (AIGA) syndrome is unknown. This is an interesting study addressing an important knowledge gap regarding the pathogenesis of T. marneffei in non-HIV positive patients; in particular patients with AIGA. The finding of the Th1 cell immune response plays a pivotal role in defense against T. marneffei infection in HIV-negative patients, and inhibition of the Th1 cell immune response may be an important pathological effect of AIGA syndrome, which presented in this research could help bridge the current knowledge gap.


Autoantibodies , Interferon-gamma , Mycoses , Talaromyces , Th1 Cells , Humans , Talaromyces/immunology , Th1 Cells/immunology , Interferon-gamma/immunology , Autoantibodies/immunology , Autoantibodies/blood , Male , Adult , Female , China , Mycoses/immunology , Mycoses/microbiology , Middle Aged , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/genetics
18.
Int Immunol ; 36(6): 303-316, 2024 Apr 27.
Article En | MEDLINE | ID: mdl-38387051

Lymphocyte homing to peripheral lymph nodes (PLN) is critical for immune surveillance. However, autoimmune diseases such as multiple sclerosis (MS) can occur due to excessive immune responses in the PLN. Here we show that 6-sulfo sialyl Lewis X (6-sulfo sLex) glycans on high endothelial venules that function as ligands for l-selectin on lymphocytes play a critical role in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model of MS. In N-acetylglucosamine-6-O-sulfotransferase (GlcNAc6ST)-1 and GlcNAc6ST-2 double-knockout mice lacking the expression of 6-sulfo sLeX glycans, the EAE symptoms and the numbers of effector Th1 and Th17 cells in the draining lymph nodes (dLN) and spinal cords (SC) were significantly reduced. To determine whether 6-sulfo sLeX could serve as a target for MS, we also examined the effects of anti-glycan monoclonal antibody (mAb) SF1 against 6-sulfo sLeX in EAE. Administration of mAb SF1 significantly reduced EAE symptoms and the numbers of antigen-specific effector T cells in the dLN and SC in association with suppression of critical genes including Il17a and Il17f that are involved in the pathogenesis of EAE. Taken together, these results suggest that 6-sulfo sLeX glycan would serve as a novel target for MS.


Encephalomyelitis, Autoimmune, Experimental , Mice, Inbred C57BL , Mice, Knockout , Sialyl Lewis X Antigen , Sialyl Lewis X Antigen/analogs & derivatives , Th17 Cells , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Mice , Th17 Cells/immunology , Sialyl Lewis X Antigen/metabolism , Polysaccharides/metabolism , Interleukin-17/metabolism , Interleukin-17/immunology , Oligosaccharides , Carbohydrate Sulfotransferases , Th1 Cells/immunology , Sulfotransferases/metabolism , Sulfotransferases/genetics , Sulfotransferases/immunology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Female , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Spinal Cord/immunology , Spinal Cord/metabolism , Cell Movement/immunology
19.
J Mol Biol ; 436(4): 168446, 2024 02 15.
Article En | MEDLINE | ID: mdl-38242283

Adjuvants are vaccine components that can boost the type, magnitude, breadth, and durability of an immune response. We have previously demonstrated that certain adjuvant combinations can act synergistically to enhance and shape immunogenicity including promotion of Th1 and cytotoxic T-cell development. These combinations also promoted protective immunity in vulnerable populations such as newborns. In this study, we employed combined antigen-specific human in vitro models to identify adjuvant combinations that could synergistically promote the expansion of vaccine-specific CD4+ cells, induce cross-presentation on MHC class I, resulting in antigen-specific activation of CD8+ cells, and direct the balance of immune response to favor the production of Th1-promoting cytokines. A screen of 78 adjuvant combinations identified several T cell-potentiating adjuvant combinations. Remarkably, a combination of TLR9 and STING agonists (CpG + 2,3-cGAMP) promoted influenza-specific CD4+ and CD8+ T cell activation and selectively favored production of Th1-polarizing cytokines TNF and IL-12p70 over co-regulated cytokines IL-6 and IL-12p40, respectively. Phenotypic reprogramming towards cDC1-type dendritic cells by CpG + 2,3-cGAMP was also observed. Finally, we characterized the molecular mechanism of this adjuvant combination including the ability of 2,3-cGAMP to enhance DC expression of TLR9 and the dependency of antigen-presenting cell activation on the Sec22b protein important to endoplasmic reticulum-Golgi vesicle trafficking. The identification of the adjuvant combination CpG + 2,3-cGAMP may therefore prove key to the future development of vaccines against respiratory viral infections tailored for the functionally distinct immune systems of vulnerable populations such as older adults and newborns.


Adjuvants, Immunologic , Cross-Priming , Th1 Cells , Vaccine Development , Viral Vaccines , Humans , Infant, Newborn , Adjuvants, Immunologic/pharmacology , Cross-Priming/drug effects , Cytokines/metabolism , Dendritic Cells/immunology , Toll-Like Receptor 9 , Th1 Cells/immunology , Adolescent , Young Adult , Viral Vaccines/immunology
20.
Nature ; 626(7997): 160-168, 2024 Feb.
Article En | MEDLINE | ID: mdl-38233524

Guillain-Barré syndrome (GBS) is a rare heterogenous disorder of the peripheral nervous system, which is usually triggered by a preceding infection, and causes a potentially life-threatening progressive muscle weakness1. Although GBS is considered an autoimmune disease, the mechanisms that underlie its distinct clinical subtypes remain largely unknown. Here, by combining in vitro T cell screening, single-cell RNA sequencing and T cell receptor (TCR) sequencing, we identify autoreactive memory CD4+ cells, that show a cytotoxic T helper 1 (TH1)-like phenotype, and rare CD8+ T cells that target myelin antigens of the peripheral nerves in patients with the demyelinating disease variant. We characterized more than 1,000 autoreactive single T cell clones, which revealed a polyclonal TCR repertoire, short CDR3ß lengths, preferential HLA-DR restrictions and recognition of immunodominant epitopes. We found that autoreactive TCRß clonotypes were expanded in the blood of the same patient at distinct disease stages and, notably, that they were shared in the blood and the cerebrospinal fluid across different patients with GBS, but not in control individuals. Finally, we identified myelin-reactive T cells in the nerve biopsy from one patient, which indicates that these cells contribute directly to disease pathophysiology. Collectively, our data provide clear evidence of autoreactive T cell immunity in a subset of patients with GBS, and open new perspectives in the field of inflammatory peripheral neuropathies, with potential impact for biomedical applications.


Autoimmunity , CD8-Positive T-Lymphocytes , Guillain-Barre Syndrome , Peripheral Nerves , Peripheral Nervous System Diseases , Th1 Cells , Humans , Biopsy , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Guillain-Barre Syndrome/blood , Guillain-Barre Syndrome/cerebrospinal fluid , Guillain-Barre Syndrome/etiology , Guillain-Barre Syndrome/immunology , HLA-DR Antigens/immunology , Immunodominant Epitopes/immunology , Myelin Sheath/immunology , Peripheral Nerves/immunology , Peripheral Nerves/pathology , Peripheral Nervous System Diseases/complications , Peripheral Nervous System Diseases/immunology , Peripheral Nervous System Diseases/pathology , Receptors, Antigen, T-Cell/immunology , Th1 Cells/immunology , Th1 Cells/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , Immunologic Memory
...