Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.818
Filter
1.
J Exp Med ; 221(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38949650

ABSTRACT

Germline activating mutations in STAT3 cause a multi-systemic autoimmune and autoinflammatory condition. By studying a mouse model, Toth et al. (https://doi.org/10.1084/jem.20232091) propose a role for dysregulated IL-22 production by Th17 cells in causing some aspects of immune-mediated skin inflammation in human STAT3 GOF syndrome.


Subject(s)
Interleukin-22 , STAT3 Transcription Factor , Skin , Th17 Cells , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Animals , Humans , Th17 Cells/immunology , Th17 Cells/metabolism , Skin/metabolism , Skin/pathology , Interleukins/genetics , Interleukins/metabolism , Gain of Function Mutation , Mice , Inflammation/metabolism
2.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(3): 744-748, 2024 May 20.
Article in Chinese | MEDLINE | ID: mdl-38948276

ABSTRACT

Objective: To investigate the roles of histone H3K27me3 methylation and its regulatory enzymes JMJD3 and EZH2 in the differentiation of Th17 cells in ankylosing spondylitis (AS), to unveil their potential involvement in the pathogenesis of AS, and to provide new strategies and targets for the clinical treatment of AS by analyzing the methylation state of H3K27me3 and its interactions with Th17-related factors. Methods: A total of 84 AS patients (42 active AS patiens and 42 patients in the stable phase of AS) were enrolled for the study, while 84 healthy volunteers were enrolled as the controls. Blood samples were collected. Peripheral blood mononuclear cells were isolated. ELISA assay was performed to examine Th17 cells and the relevant cytokines IL-21, IL-22, and IL-17. The mRNA expressions of RORc, JAK2, and STAT3 were analyzed by RT-PCR, the protein expressions of RORc, JAK2/STAT3 pathway protein, H3K27me3 and the relevant protease (EZH2 and JMJD3) were determined by Western blot. Correlation between H3K27me3, EZH2 and JMJD3 and the key signaling pathway molecules of Th cell differentiation was analyzed by Pearson correlation analysis. Results: The mRNA expressions of RORc, JAK2, and STAT3 were significantly higher in the active phase group than those in the stable phase group ( P<0.05). The relative grayscale values of H3K27me3 and EZH2 in the active phase group were lower than those of the stable phase group, which were lower than those of the control group, with the differences being statistically significant ( P<0.05). The relative grayscale values of JMJD3, RORc, JAK2, pJAK2, STAT3, and pSTAT3 proteins were significantly higher in the active phase group than those in the stable phase group, which were higher than those in the control group (all P<0.05). The proportion of Th17 and the expression level of inflammatory factors in the active period group were higher than those in the other two groups (P<0.05). H3K27me3 was negatively correlated with RORc, JAK2, STAT3, and IL-17, JMJD3 was positvely correlated with JAK2, STAT3, and IL-17, and EZH2 was negatively correlated with JAK2, STAT3, and IL-17 (all P<0.05). Conclusion: The low expression of H3K27me3 in AS is influenced by the gene loci JMJD3 and EZH2, which can regulate the differentiation of Th17 cells and thus play a role in the pathogenesis and progression of AS.


Subject(s)
Cell Differentiation , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Histones , Interleukin-17 , Jumonji Domain-Containing Histone Demethylases , Nuclear Receptor Subfamily 1, Group F, Member 3 , STAT3 Transcription Factor , Spondylitis, Ankylosing , Th17 Cells , Humans , Spondylitis, Ankylosing/genetics , Spondylitis, Ankylosing/metabolism , Th17 Cells/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Histones/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Interleukin-17/metabolism , Interleukin-17/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Janus Kinase 2/metabolism , Janus Kinase 2/genetics , Methylation , Interleukins/metabolism , Interleukins/genetics , Interleukin-22 , Male , Female , Adult
3.
PLoS Pathog ; 20(7): e1012302, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38949991

ABSTRACT

Oropharyngeal candidiasis (OPC) is the most common human fungal infection, arising typically from T cell immune impairments. IL-17 and IL-22 contribute individually to OPC responses, but here we demonstrate that the combined actions of both cytokines are essential for resistance to OPC. Mice lacking IL-17RA and IL-22RA1 exhibited high fungal loads in esophagus- and intestinal tract, severe weight loss, and symptoms of colitis. Ultimately, mice succumbed to infection. Dual loss of IL-17RA and IL-22RA impaired expression of small proline rich proteins (SPRRs), a class of antimicrobial effectors not previously linked to fungal immunity. Sprr2a1 exhibited direct candidacidal activity in vitro, and Sprr1-3a-/- mice were susceptible to OPC. Thus, cooperative actions of Type 17 cytokines mediate oral mucosal anti-Candida defenses and reveal a role for SPRRs.


Subject(s)
Candidiasis, Oral , Interleukin-17 , Interleukin-22 , Interleukins , Mice, Knockout , Animals , Mice , Candida albicans/immunology , Candidiasis, Oral/immunology , Candidiasis, Oral/microbiology , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukins/immunology , Interleukins/metabolism , Mice, Inbred C57BL , Receptors, Interleukin/immunology , Receptors, Interleukin/metabolism , Receptors, Interleukin-17/immunology , Receptors, Interleukin-17/metabolism
4.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(7): 653-662, 2024 Jul 09.
Article in Chinese | MEDLINE | ID: mdl-38949133

ABSTRACT

Objective: To investigate the regulatory effect and mechanism of interleukin-22 (IL-22) on the gingival epithelial barrier in the context of periodontal inflammation. Methods: IL-22 knockout (IL-22 KO) mice were constructed, and periodontitis mice models were established through oral gavage with polymicrobial inoculation. DNAs were extracted from the oral plaques of IL-22 KO periodontitis mice group (n=7) and their wild-type littermates periodontitis group (n=7) to establish a periodontitis-related oral microbiota database"PD-RiskMicroDB", determining the relationship between changes in oral microbiota and microbial function in two groups using 16S rRNA sequencing results. Gingival epithelial cells (GEC) were cultured by modified trypsinization method, and were stimulated with 100 µg/L IL-22, Porphyromonas gingivalis (Pg) (multiplicity of infection:100), separately or together for 3 and 12 hours. The experimental groups were as follows: control group (no stimulation), IL-22 group, Pg group and Pg+IL-22 group. The expression of barrier protein E-cadherin in each group at 3 h was detected by immunofluorescence, real-time fluorescence quantitative PCR (RT-qPCR) and Western blotting. Fluorescein isothiocyanate-dextran-mediated epithelial cell permeability experiment was conducted to clarify the changes in permeability of GEC in each group at 3 and 12 h. The mRNA expressions of E-cadherin in the gingival epithelium of wild-type littermates periodontitis group and IL-22 KO periodontitis group were detected by RT-qPCR. Fifteen C57BL/6 wild-type mice were randomly divided into control group (n=5), periodontitis group (n=5) and periodontitis+IL-22 treatment group (n=5). RT-qPCR and immunohistochemistry (IHC) staining were used to detect the expression level of E-cadherin in the gingival epithelium of each group. Results: 16S rRNA sequencing results showed that the composition of oral microbiota changed in IL-22 KO periodontitis group, of which the abundance of bacterial genera related to periodontal tissue invasion was significantly increased (linear discriminant analysis score: 2.22, P=0.009), compared with wild-type littermates periodontitis group. In vitro cell experiments showed that after Pg infection for 3 hours, the cell connections of GEC in Pg group were interrupted, and the fluorescence intensity of E-cadherin was reduced in Pg group compared with the control group. Meanwhile, the mRNA and protein expression levels of E-cadherin (mRNA: 0.69±0.12; protein: 0.60±0.12) were downregulated compared with the control group [mRNA: 1.00±0.00 (P=0.043); protein: 1.04±0.08 (P=0.003)], respectively. The fluorescence intensity of E-cadherin in the Pg+IL-22 group was enhanced compared with Pg group, and expression levels of E-cadherin mRNA (1.16±0.10) and protein (0.98±0.07) in Pg+IL-22 group showed a significant increase compared with Pg group [mRNA: 0.69±0.12 (P=0.005); protein: 0.60±0.12 (P=0.007)]. The result of epithelial permeability test showed that there was no statistical difference in epithelial permeability among control group, Pg group, IL-22 group and Pg+IL-22 group with treatment for 3 hours (F=0.20, P=0.893). While when the treatment time turned to be 12 hours, the epithelial barrier permeability showed a significant increase in Pg group (1.39±0.15) compared with control group (1.00±0.00, P=0.027), and a decrease in Pg+IL-22 group (1.02±0.18) compared with Pg group (1.39±0.15, P=0.034). In vivo, the mRNA expression of E-cadherin in the gingival epithelium of IL-22 KO periodontitis group decreased significantly (0.32±0.21) compared with wild-type littermates periodontitis group (1.01±0.01) (t=5.70, P=0.005). Moreover, RT-qPCR and IHC staining results showed that the mRNA expression level of E-cadherin (0.40±0.07) and absorbance value of E-cadherin positive expression (0.02±0.00) in gingival epithelial tissue of periodontitis group were both significantly down-regulated compared with control group [mRNA: 1.00±0.00 (P=0.005); absorbance value of E-cadherin positive expression: 0.04±0.01 (P=0.006)]. Meanwhile, the mRNA expression level of E-cadherin (1.06±0.24) and the absorbance value of E-cadherin positive expression (0.03±0.01) were both observed increase in periodontitis+IL-22 treatment group compared with periodontitis group (P=0.003, P=0.039). Conclusions: IL-22 may exert a protective effect on the gingival epithelial barrier in an inflammatory environment by regulating the invasiveness of oral microbiota and the expression of host barrier protein.


Subject(s)
Cadherins , Gingiva , Interleukin-22 , Interleukins , Mice, Knockout , Microbiota , Periodontitis , Porphyromonas gingivalis , Animals , Interleukins/metabolism , Cadherins/metabolism , Gingiva/cytology , Gingiva/metabolism , Gingiva/microbiology , Mice , Periodontitis/microbiology , Periodontitis/metabolism , Epithelial Cells/metabolism , RNA, Ribosomal, 16S
5.
Respir Res ; 25(1): 275, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003443

ABSTRACT

Pleurisy can be categorized as primary or secondary, arising from immunological, tumorous, or microbial conditions. It often results in lung structure damage and the development of various respiratory issues. Among the different types, tuberculous pleurisy has emerged as a prominent focus for both clinical and scientific investigations. The IL-10 family, known for its anti-inflammatory properties in the human immune system, is increasingly being studied for its involvement in the pathogenesis of pleurisy. This review aims to present a detailed overview of the intricate role of IL-10 family members (specifically IL-10, IL-22, and IL-26) in human and animal pleuritic diseases or relevant animal models. These insights could serve as valuable guidance and references for further studies on pleurisy and potential therapeutic strategies.


Subject(s)
Interleukin-10 , Interleukin-22 , Interleukins , Tuberculosis, Pleural , Humans , Tuberculosis, Pleural/diagnosis , Tuberculosis, Pleural/immunology , Tuberculosis, Pleural/metabolism , Tuberculosis, Pleural/drug therapy , Interleukins/metabolism , Interleukins/immunology , Animals , Interleukin-10/metabolism , Pleurisy/immunology , Pleurisy/diagnosis , Pleurisy/metabolism
6.
Front Immunol ; 15: 1395921, 2024.
Article in English | MEDLINE | ID: mdl-38966644

ABSTRACT

IL-27, a member of the IL-6/IL-12 cytokine superfamily, is primarily secreted by antigen presenting cells, specifically by dendric cells, macrophages and B cells. IL-27 has antiviral activities and modulates both innate and adaptive immune responses against viruses. The role of IL-27 in the setting of viral infections is not well defined and both pro-inflammatory and anti-inflammatory functions have been described. Here, we discuss the latest advancements in the role of IL-27 in several viral infection models of human disease. We highlight important aspects of IL-27 expression regulation, the critical cell sources at different stages of the infection and their impact in cell mediated immunity. Lastly, we discuss the need to better define the antiviral and modulatory (pro-inflammatory vs anti-inflammatory) properties of IL-27 in the context of human chronic viral infections.


Subject(s)
Adaptive Immunity , Virus Diseases , Humans , Virus Diseases/immunology , Animals , Gene Expression Regulation , Interleukin-27/metabolism , Viruses/immunology , Interleukins/immunology , Interleukins/metabolism
7.
J Immunol Res ; 2024: 9527268, 2024.
Article in English | MEDLINE | ID: mdl-38966668

ABSTRACT

Aberrant accumulation of circulating follicular helper T cells (cTfh) has been found in the peripheral blood mononuclear cells (PBMCs) of Graves' disease (GD) patients. However, the underlying mechanism that contributes to the imbalance of cTfh cells remains unknown. Previously, studies described a GD-related circular RNAs (circRNAs)-circZNF644 that might be associated with cTfh cells. This study aimed to investigate the role of circZNF644 on cTfh cells in GD patients. Here, we found that circZNF644 was highly stable expression in the PBMCs of GD patients, which was positively correlated with the serum levels of TSH receptor autoantibodies (TRAb). Knockdown of circZNF644 caused a reduction of the proportion of cTfh cells in vitro. Mechanistically, circZNF644 served as a ceRNA for miR-29a-3p to promote ICOS expression, resulting in increased cTfh cells. In the PBMCs of GD patients, circZNF644 expression was positively correlated with ICOS expression and the percentage of cTfh cells, but negatively related to miR-29a-3p expression. Additionally, a strong relationship between circZNF644 and IL-21 was revealed in GD patients, and silencing of circZNF644 inhibited IL-21 expression. Our study elucidated that elevated expression of circZNF644 is a key feature in the development of GD and may contribute to the pathogenic role of cTfh cells in GD.


Subject(s)
Graves Disease , MicroRNAs , RNA, Circular , T Follicular Helper Cells , Humans , Graves Disease/genetics , Graves Disease/immunology , RNA, Circular/genetics , Male , Female , T Follicular Helper Cells/immunology , Adult , MicroRNAs/genetics , Middle Aged , Autoantibodies/immunology , Autoantibodies/blood , Inducible T-Cell Co-Stimulator Protein/metabolism , Inducible T-Cell Co-Stimulator Protein/genetics , Interleukins/genetics , Interleukins/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Gene Expression Regulation
8.
Mediators Inflamm ; 2024: 3282679, 2024.
Article in English | MEDLINE | ID: mdl-38962170

ABSTRACT

Ulcerative colitis (UC) is a chronic intestinal inflammatory disease with complex etiology. Interleukin-35 (IL-35), as a cytokine with immunomodulatory function, has been shown to have therapeutic effects on UC, but its mechanism is not yet clear. Therefore, we constructed Pichia pastoris stably expressing IL-35 which enables the cytokines to reach the diseased mucosa, and explored whether upregulation of T-cell protein tyrosine phosphatase (TCPTP) in macrophages is involved in the mechanisms of IL-35-mediated attenuation of UC. After the successful construction of engineered bacteria expressing IL-35, a colitis model was successfully induced by giving BALB/c mice a solution containing 3% dextran sulfate sodium (DSS). Mice were treated with Pichia/IL-35, empty plasmid-transformed Pichia (Pichia/0), or PBS by gavage, respectively. The expression of TCPTP in macrophages (RAW264.7, BMDMs) and intestinal tissues after IL-35 treatment was detected. After administration of Pichia/IL-35, the mice showed significant improvement in weight loss, bloody stools, and shortened colon. Colon pathology also showed that the inflammatory condition of mice in the Pichia/IL-35 treatment group was alleviated. Notably, Pichia/IL-35 treatment not only increases local M2 macrophages but also decreases the expression of inflammatory cytokine IL-6 in the colon. With Pichia/IL-35 treatment, the proportion of M1 macrophages, Th17, and Th1 cells in mouse MLNs were markedly decreased, while Tregs were significantly increased. In vitro experiments, IL-35 significantly promoted the expression of TCPTP in macrophages stimulated with LPS. Similarly, the mice in the Pichia/IL-35 group also expressed more TCPTP than that of the untreated group and the Pichia/0 group.


Subject(s)
Interleukins , Macrophages , Mice, Inbred BALB C , Animals , Mice , Interleukins/metabolism , Macrophages/metabolism , RAW 264.7 Cells , Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate , Disease Models, Animal , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/chemically induced , Male , Up-Regulation , Saccharomycetales
9.
Mol Med Rep ; 30(3)2024 Sep.
Article in English | MEDLINE | ID: mdl-38994764

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive form of cancer with a low survival rate. A successful treatment strategy should not be limited to targeting cancer cells alone, but should adopt a more comprehensive approach, taking into account other influential factors. These include the extracellular matrix (ECM) and immune microenvironment, both of which are integral components of the tumor microenvironment. The present review describes the roles of pancreatic stellate cells, differentiated cancer­associated fibroblasts and the interleukin family, either independently or in combination, in the progression of precursor lesions in pancreatic intraepithelial neoplasia and PDAC. These elements contribute to ECM deposition and immunosuppression in PDAC. Therapeutic strategies that integrate interleukin and/or stromal blockade for PDAC immunomodulation and fibrogenesis have yielded inconsistent results. A deeper comprehension of the intricate interplay between fibrosis, and immune responses could pave the way for more effective treatment targets, by elucidating the mechanisms and causes of ECM fibrosis during PDAC progression.


Subject(s)
Carcinoma, Pancreatic Ductal , Fibrosis , Interleukins , Pancreatic Neoplasms , Pancreatic Stellate Cells , Tumor Microenvironment , Humans , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Tumor Microenvironment/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Interleukins/metabolism , Interleukins/immunology , Animals , Extracellular Matrix/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/pathology
10.
Yi Chuan ; 46(6): 478-489, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38886151

ABSTRACT

Metronidazole (MTZ), a commonly used anti-infective drug in clinical practice, has also been employed as a prodrug in cell-targeted ablation systems in scientific research, exhibiting significant application value. However, it has been demonstrated that MTZ can induce neurotoxic symptoms to some extent during its use, and there is currently a lack of effective means to circumvent its toxicity in both clinical and research settings, which limits its application. Therefore, exploring the specific mechanisms underlying MTZ-induced neurotoxic symptoms and elucidating countermeasures will enhance the practical value of MTZ. In this study, using a zebrafish spinal cord injury regeneration model, we confirmed that MTZ neurotoxicity leads to impaired axon regeneration in the central nervous system. By overexpressing il34 in the central nervous system of zebrafish, we eliminated the inhibitory effect of MTZ on axonal regeneration and demonstrated that the pro-regenerative effect against MTZ neurotoxicity is not caused by excessive macrophages/microglia chemoattracted by interleukin 34(Il34). Transcriptome sequencing analysis and GO enrichment analysis of differentially expressed genes between groups revealed that Il34 may counteract MTZ neurotoxicity and promote spinal cord injury repair through biological processes that enhance cellular adhesion and cell location. In summary, our work uncovers a possible cause of MTZ neurotoxicity and provides a new perspective for eliminating MTZ toxicity.


Subject(s)
Metronidazole , Spinal Cord Injuries , Spinal Cord Regeneration , Zebrafish , Animals , Metronidazole/pharmacology , Metronidazole/adverse effects , Spinal Cord Regeneration/drug effects , Spinal Cord Injuries/metabolism , Interleukins/genetics , Interleukins/metabolism , Central Nervous System/drug effects , Central Nervous System/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism
11.
Int Immunopharmacol ; 136: 112305, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38823178

ABSTRACT

The second-leading cause of death, cancer, poses a significant threat to human life. Innovations in cancer therapies are crucial due to limitations in traditional approaches. Newcastle disease virus (NDV), a nonpathogenic oncolytic virus, exhibits multifunctional anticancer properties by selectively infecting, replicating, and eliminating tumor cells. To enhance NDV's antitumor activity, four oncolytic NDV viruses were developed, incorporating IL24 and/or GM-CSF genes at different gene loci using reverse genetics. In vitro experiments revealed that oncolytic NDV virus augmented the antitumor efficacy of the parental virus rClone30, inhibiting tumor cell proliferation, inducing tumor cell fusion, and promoting apoptosis. Moreover, NDV carrying the IL24 gene inhibited microvessel formation in CAM experiments. Evaluation in a mouse model of liver cancer confirmed the therapeutic efficacy of oncolytic NDV viral therapy. Tumors in mice treated with oncolytic NDV virus significantly decreased in size, accompanied by tumor cell detachment and apoptosis evident in pathological sections. Furthermore, oncolytic NDV virus enhanced T cell and dendritic cell production and substantially improved the survival rate of mice with hepatocellular carcinoma, with rClone30-IL24(P/M) demonstrating significant therapeutic effects. This study establishes a basis for utilizing oncolytic NDV virus as an antitumor agent in clinical practice.


Subject(s)
Interleukins , Newcastle disease virus , Oncolytic Virotherapy , Oncolytic Viruses , Animals , Newcastle disease virus/genetics , Newcastle disease virus/physiology , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , Humans , Mice , Cell Line, Tumor , Interleukins/genetics , Interleukins/metabolism , Liver Neoplasms/therapy , Mice, Inbred BALB C , Carcinoma, Hepatocellular/therapy , Apoptosis , Neovascularization, Pathologic/therapy , Cell Proliferation , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Dendritic Cells/immunology , T-Lymphocytes/immunology
12.
Cell Rep ; 43(6): 114292, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38823020

ABSTRACT

Interleukin (IL)-22 promotes host-microbiota homeostasis. We sought to identify microbiota metabolite(s) that drive intestinal IL-22 production. We observed that exposing Peyer's patch cells (PPCs), ex vivo, to fecal supernatants (FSs) recapitulates fermentable fiber- and microbiota-dependent IL-22 production, and cellular sources thereof, thus supporting the use of this model. An interrogation of FSs generated from mice fed the fermentable fiber inulin (FS-Inu) revealed that its IL-22-inducing activity is mediated by heat-labile protein. Fractionation of FS-Inu by ion-exchange chromatography, and subsequent proteomic analysis of IL-22-inducing fractions, indicates that outer membrane protein A (OmpA) might be a microbial driver of IL-22 expression. Concomitantly, recombinant OmpA from Parabacteroides goldsteinii, which is enriched by an inulin diet, induces IL-22 production and expression of the IL-22-dependent genes REG3γ and -ß, in PPCs and mice. Thus, OmpA is one bacterial inducer of IL-22 expression, potentially linking diet, mucosal immune homeostasis, and gut health.


Subject(s)
Bacterial Outer Membrane Proteins , Interleukin-22 , Interleukins , Animals , Interleukins/metabolism , Bacterial Outer Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Pancreatitis-Associated Proteins/metabolism , Feces/microbiology , Inulin/metabolism , Gastrointestinal Microbiome
14.
Cancer Immunol Immunother ; 73(8): 138, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38833177

ABSTRACT

Despite the success of immune checkpoint inhibitors (ICIs) in treating solid tumors, lots of patients remain unresponsive to this therapy. Microwave ablation (MWA) stimulates systemic adaptive immunity against tumor cells by releasing tumor antigens. Additionally, IL-21 has demonstrated importance in stimulating T-cell effector function. The combination of these three therapies-MWA, IL-21, and anti-PD-1 monoclonal antibodies (mAbs)-has yet to be explored in the context of cancer treatment.In this study, we explored the impact of thermal ablation on IL-21R expression in tumor-infiltrating lymphocytes (TILs). Subsequently, we assessed alterations in the tumor microenvironment (TME) and peripheral lymphoid organs. Additionally, we conducted a thorough examination of tumor-infiltrating CD45+ immune cells across various treatment groups using single-cell RNA sequencing (scRNA-seq). Moreover, we determined the potential anti-tumor effects of the triple combination involving MWA, IL-21, and anti-PD-1 mAbs.Our findings revealed that MWA upregulated the expression of IL-21R on various immune cells in the untreated tumors. The combination of MWA with IL-21 exhibited a robust abscopal anti-tumor effect, enhancing the effector function of CD8+ T cells and facilitating dendritic cells' maturation and antigen presentation in the untreated tumor. Notably, the observed abscopal anti-tumor effect resulting from the combination is contingent upon T-cell recirculation, indicating the reliance of systemic adaptive immunity for this treatment regimen. Additionally, the combination of MWA, IL-21, and PD-1 mAbs demonstrated profound abscopal anti-tumor efficacy. Our findings provide support for further clinical investigation into a triple combination therapy involving MWA, IL-21, and ICIs for the treatment of metastatic cancer.


Subject(s)
Immune Checkpoint Inhibitors , Interleukins , Programmed Cell Death 1 Receptor , Tumor Microenvironment , Interleukins/metabolism , Animals , Mice , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Humans , Tumor Microenvironment/immunology , Combined Modality Therapy , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Female , Neoplasms/immunology , Neoplasms/therapy , Mice, Inbred C57BL , Cell Line, Tumor
15.
Cell Commun Signal ; 22(1): 307, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38831315

ABSTRACT

BACKGROUND: Interleukin 24 (IL-24) has been implicated in the nociceptive signaling. However, direct evidence and the precise molecular mechanism underlying IL-24's role in peripheral nociception remain unclear. METHODS: Using patch clamp recording, molecular biological analysis, immunofluorescence labeling, siRNA-mediated knockdown approach and behavior tests, we elucidated the effects of IL-24 on sensory neuronal excitability and peripheral pain sensitivity mediated by T-type Ca2+ channels (T-type channels). RESULTS: IL-24 enhances T-type channel currents (T-currents) in trigeminal ganglion (TG) neurons in a reversible and dose-dependent manner, primarily by activating the interleukin-22 receptor 1 (IL-22R1). Furthermore, we found that the IL-24-induced T-type channel response is mediated through tyrosine-protein kinase Lyn, but not its common downstream target JAK1. IL-24 application significantly activated protein kinase A; this effect was independent of cAMP and prevented by Lyn antagonism. Inhibition of PKA prevented the IL-24-induced T-current response, whereas inhibition of protein kinase C or MAPK kinases had no effect. Functionally, IL-24 increased TG neuronal excitability and enhanced pain sensitivity to mechanical stimuli in mice, both of which were suppressed by blocking T-type channels. In a trigeminal neuropathic pain model induced by chronic constriction injury of the infraorbital nerve, inhibiting IL-22R1 signaling alleviated mechanical allodynia, which was reversed by blocking T-type channels or knocking down Cav3.2. CONCLUSION: Our findings reveal that IL-24 enhances T-currents by stimulating IL-22R1 coupled to Lyn-dependent PKA signaling, leading to TG neuronal hyperexcitability and pain hypersensitivity. Understanding the mechanism of IL-24/IL-22R1 signaling in sensory neurons may pave the way for innovative therapeutic strategies in pain management.


Subject(s)
Calcium Channels, T-Type , Cyclic AMP-Dependent Protein Kinases , Receptors, Interleukin , Sensory Receptor Cells , Signal Transduction , Trigeminal Ganglion , src-Family Kinases , Animals , Calcium Channels, T-Type/metabolism , Calcium Channels, T-Type/genetics , src-Family Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Trigeminal Ganglion/metabolism , Male , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Receptors, Interleukin/metabolism , Mice , Mice, Inbred C57BL , Interleukins/metabolism
16.
Sci Rep ; 14(1): 13133, 2024 06 07.
Article in English | MEDLINE | ID: mdl-38849432

ABSTRACT

The short-lived nature and heterogeneity of Natural Killer (NK) cells limit the development of NK cell-based therapies, despite their proven safety and efficacy against cancer. Here, we describe the biological basis, detailed phenotype and function of long-lived anti-tumour human NK cells (CD56highCD16+), obtained without cell sorting or feeder cells, after priming of peripheral blood cells with Bacillus Calmette-Guérin (BCG). Further, we demonstrate that survival doses of a cytokine combination, excluding IL18, administered just weekly to BCG-primed NK cells avoids innate lymphocyte exhaustion and leads to specific long-term proliferation of innate cells that exert potent cytotoxic function against a broad range of solid tumours, mainly through NKG2D. Strikingly, a NKG2C+CD57-FcεRIγ+ NK cell population expands after BCG and cytokine stimulation, independently of HCMV serology. This strategy was exploited to rescue anti-tumour NK cells even from the suppressor environment of cancer patients' bone marrow, demonstrating that BCG confers durable anti-tumour features to NK cells.


Subject(s)
Cell Proliferation , Killer Cells, Natural , Killer Cells, Natural/immunology , Killer Cells, Natural/drug effects , Humans , Cell Proliferation/drug effects , Neoplasms/immunology , Neoplasms/drug therapy , BCG Vaccine/immunology , BCG Vaccine/administration & dosage , Mycobacterium bovis/immunology , Lymphocyte Activation/drug effects , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Interleukins/metabolism , CD56 Antigen/metabolism , NK Cell Lectin-Like Receptor Subfamily C/metabolism
17.
Int J Biol Sci ; 20(8): 3094-3112, 2024.
Article in English | MEDLINE | ID: mdl-38904012

ABSTRACT

Atopic dermatitis (AD) is a common inflammation skin disease that involves dysregulated interplay between immune cells and keratinocytes. Interleukin-38 (IL-38), a poorly characterized IL-1 family cytokine, its role and mechanism in the pathogenesis of AD is elusive. Here, we show that IL-38 is mainly secreted by epidermal keratinocytes and highly expressed in the skin and downregulated in AD lesions. We generated IL-38 keratinocyte-specific knockout mice (K14Cre/+-IL-38f/f ) and induced AD models by 2,4-dinitrofluorobenzene (DNFB). Unexpectedly, after treatment with DNFB, K14Cre/+-IL-38f/f mice were less susceptible to cutaneous inflammation of AD. Moreover, keratinocyte-specific deletion of IL-38 suppressed the migration of Langerhans cells (LCs) into lymph nodes which results in disturbed differentiation of CD4+T cells and decreased the infiltration of immune cells into AD lesions. LCs are a type of dendritic cell that reside specifically in the epidermis and regulate immune responses. We developed LC-like cells in vitro from mouse bone marrow (BM) and treated with recombined IL-38. The results show that IL-38 depended on IL-36R, activated the phosphorylated expression of IRAK4 and NF-κB P65 and upregulated the expression of CCR7 to promoting the migration of LCs, nevertheless, the upregulation disappeared with the addition of IL-36 receptor antagonist (IL-36RA), IRAK4 or NF-κB P65 inhibitor. Furthermore, after treatment with IRAK4 inhibitors, the experimental AD phenotypes were alleviated and so IRAK4 is considered a promising target for the treatment of inflammatory diseases. Overall, our findings indicated a potential pathway that IL-38 depends on IL-36R, leading to LCs migration to promote AD by upregulating CCR7 via IRAK4/NF-κB and implied the prevention and treatment of AD, supporting potential clinical utilization of IRAK4 inhibitors in AD treatment.


Subject(s)
Cell Movement , Dermatitis, Atopic , Langerhans Cells , Animals , Dermatitis, Atopic/metabolism , Langerhans Cells/metabolism , Mice , Mice, Knockout , Interleukin-1/metabolism , Keratinocytes/metabolism , Dinitrofluorobenzene , NF-kappa B/metabolism , Interleukins/metabolism
18.
Life Sci ; 350: 122766, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38834097

ABSTRACT

Psoriatic arthritis (PsA) is a chronic inflammatory arthropathy affecting the skin, entheses, and joints. Over the past decade, experimental evidence has revealed the activation of several immune cells and signaling cascades in modulating the pathophysiology of PsA. Recently, targeted therapies have been developed to combat the severity of disease. However, with diverse etiologies, flareups, and relapses, there has been an increased prevalence and mortality associated with PsA in recent years. Therefore, it is imperative to investigate new potential mediators and combination therapies to manage PsA pathogenesis. IL-21, an immunomodulatory cytokine, has pleiotropic effects on immune cells and the protein cascades involved in PsA pathogenesis. Recently, emerging evidence of increased IL-21 levels in patients with PsA has engendered much enthusiasm for its potential as a therapeutic target. Here, we unmasked IL-21 as a significant modulator of PsA pathogenesis and reviewed the comorbidities associated with the disease, further cataloging future therapeutic modalities to ameliorate PsA progression.


Subject(s)
Arthritis, Psoriatic , Interleukins , Arthritis, Psoriatic/metabolism , Arthritis, Psoriatic/drug therapy , Humans , Interleukins/metabolism , Animals , Signal Transduction
19.
Front Immunol ; 15: 1388496, 2024.
Article in English | MEDLINE | ID: mdl-38873613

ABSTRACT

The intricate immune mechanisms governing mucosal healing following intestinal damage induced by cytotoxic drugs remain poorly understood. The goal of this study was to investigate the role of lymphotoxin beta receptor (LTßR) signaling in chemotherapy-induced intestinal damage. LTßR deficient mice exhibited heightened body weight loss, exacerbated intestinal pathology, increased proinflammatory cytokine expression, reduced IL-22 expression, and proliferation of intestinal epithelial cells following methotrexate (MTX) treatment. Furthermore, LTßR-/-IL-22-/- mice succumbed to MTX treatment, suggesting that LTßR- and IL-22- dependent pathways jointly promote mucosal repair. Although both LTßR ligands LIGHT and LTß were upregulated in the intestine early after MTX treatment, LIGHT-/- mice, but not LTß-/- mice, displayed exacerbated disease. Further, we revealed the critical role of T cells in mucosal repair as T cell-deficient mice failed to upregulate intestinal LIGHT expression and exhibited increased body weight loss and intestinal pathology. Analysis of mice with conditional inactivation of LTßR revealed that LTßR signaling in intestinal epithelial cells, but not in Lgr5+ intestinal stem cells, macrophages or dendritic cells was critical for mucosal repair. Furthermore, inactivation of the non-canonical NF-kB pathway member RelB in intestinal epithelial cells promoted MTX-induced disease. Based on these results, we propose a model wherein LIGHT produced by T cells activates LTßR-RelB signaling in intestinal epithelial cells to facilitate mucosal repair following chemotherapy treatment.


Subject(s)
Intestinal Mucosa , Lymphotoxin beta Receptor , Methotrexate , Mice, Knockout , Signal Transduction , Transcription Factor RelB , Animals , Intestinal Mucosa/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/drug effects , Lymphotoxin beta Receptor/metabolism , Lymphotoxin beta Receptor/genetics , Mice , Transcription Factor RelB/metabolism , Transcription Factor RelB/genetics , Methotrexate/adverse effects , Epithelial Cells/metabolism , Mice, Inbred C57BL , Interleukin-22 , Interleukins/metabolism , Interleukins/genetics
20.
J Exp Med ; 221(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38861030

ABSTRACT

Germline gain-of-function (GOF) variants in STAT3 cause an inborn error of immunity associated with early-onset poly-autoimmunity and immune dysregulation. To study tissue-specific immune dysregulation, we used a mouse model carrying a missense variant (p.G421R) that causes human disease. We observed spontaneous and imiquimod (IMQ)-induced skin inflammation associated with cell-intrinsic local Th17 responses in STAT3 GOF mice. CD4+ T cells were sufficient to drive skin inflammation and showed increased Il22 expression in expanded clones. Certain aspects of disease, including increased epidermal thickness, also required the presence of STAT3 GOF in epithelial cells. Treatment with a JAK inhibitor improved skin disease without affecting local Th17 recruitment and cytokine production. These findings collectively support the involvement of Th17 responses in the development of organ-specific immune dysregulation in STAT3 GOF and suggest that the presence of STAT3 GOF in tissues is important for disease and can be targeted with JAK inhibition.


Subject(s)
Gain of Function Mutation , Imiquimod , STAT3 Transcription Factor , Th17 Cells , Animals , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Th17 Cells/immunology , Mice , Humans , Imiquimod/pharmacology , Skin/pathology , Skin/metabolism , Skin/immunology , Interleukin-22 , Dermatitis/immunology , Dermatitis/genetics , Dermatitis/pathology , Dermatitis/metabolism , Mice, Inbred C57BL , Interleukins/genetics , Interleukins/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/immunology , Inflammation/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...