Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.498
Filter
1.
Front Immunol ; 15: 1402834, 2024.
Article in English | MEDLINE | ID: mdl-39253083

ABSTRACT

Introduction: Group 3 innate lymphoid cells (ILC3s) are enriched in the intestinal mucosa and play important roles in host defense against infection and inflammatory diseases. Sirtuin 6 (SIRT6) is a nicotinamide adenine dinucleotide (NAD+)- dependent deacetylase and has been shown to control intestinal epithelial cell differentiation and survival. However, the role of SIRT6 in ILC3s remains unknown. Methods: To investigate the role of SIRT6 in gut ILC3s, we generated SIRT6 conditional knockout mice by crossing Rorccre and Sirt6flox/flox mice. Cell number and cytokine production was examined using flow cytometry. Citrobacter rodentium infection and dextran sodium sulfate-induced colitis models were used to determine the role of SIRT6 in gut defense. RT-qPCR, flow cytometry and immunohistochemistry were used to assess the intestinal inflammatory responses. Results: Here we show that SIRT6 inhibits IL-22 expression in intestinal ILC3s in a cell-intrinsic manner. Deletion of SIRT6 in ILC3s does not affect the cell numbers of total ILC3s and subsets, but results in increased IL-22 production. Furthermore, ablation of SIRT6 in ILC3s protects mice against Citrobacter rodentium infection and dextran sodium sulfate-induced colitis. Our results suggest that SIRT6 may play a role in ILC3 function by regulating gut immune responses against bacterial infection and inflammation. Discussion: Our finding provided insight into the relation of epigenetic regulators with IL-22 production and supplied a new perspective for a potential strategy against inflammatory bowel disease.


Subject(s)
Citrobacter rodentium , Colitis , Enterobacteriaceae Infections , Immunity, Innate , Interleukin-22 , Interleukins , Lymphocytes , Mice, Knockout , Sirtuins , Animals , Mice , Lymphocytes/immunology , Lymphocytes/metabolism , Interleukins/metabolism , Interleukins/immunology , Interleukins/genetics , Sirtuins/genetics , Sirtuins/metabolism , Colitis/immunology , Colitis/chemically induced , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Dextran Sulfate , Disease Models, Animal
2.
Allergol Immunopathol (Madr) ; 52(5): 94-102, 2024.
Article in English | MEDLINE | ID: mdl-39278858

ABSTRACT

BACKGROUND: Common variable immunodeficiency (CVID) is a heterogeneous disorder characterized by defective antibody production and impaired differentiation of B cells. B cell proliferation is an essential step for antibody synthesis. Depending on the nature of the stimulus, their response may be either T-cell-dependent or T-cell-independent. METHODS: We studied 23 CVID patients and 14 healthy donors (HD). The patients were categorized based on their percentage of memory B cells. In addition to standard immunophenotyping of circulating human B and T cell subsets, an in vitro CFSE dilution assay was used to assess the proliferative capacity of B cells and to compare the activation of the T cell-dependent and T cell-independent response among the patients. RESULTS: Patients with a reduction in memory B cells exhibited an increase in follicular T cells (Tfh) and showed low proliferation in response to PKW, CpG, and SAC stimuli (Condition II) (p= 0.0073). In contrast, patients with a normal percentage of memory B cells showed a high expression of IL-21R and low proliferation in response to CPG (Condition III); IL-21, CD40L, and anti-IgM (Condition IV) stimuli (p= 0.0163 and p = 0.0475, respectively). CONCLUSION: Defective proliferation in patients depends on the type of stimulus used and the phenotypic characteristics of the patients. Further studies are necessary to understand the disease mechanisms, which may guide us toward identifying genetic defects associated with CVID.


Subject(s)
Cell Proliferation , Common Variable Immunodeficiency , Lymphocyte Activation , Humans , Common Variable Immunodeficiency/immunology , Male , Female , Adult , Lymphocyte Activation/immunology , Middle Aged , Immunophenotyping , B-Lymphocytes/immunology , Young Adult , Cells, Cultured , Memory B Cells/immunology , Interleukins/metabolism , Interleukins/immunology , Adolescent , Immunologic Memory/immunology
3.
Front Immunol ; 15: 1441908, 2024.
Article in English | MEDLINE | ID: mdl-39224597

ABSTRACT

Introduction: The antiviral activity of recombinant bovine interferon lambda 3 (bovIFN-λ3) against bovine viral diarrhea virus (BVDV) has been demonstrated in vitro in Madin-Darby bovine kidney cells (MDBK) and in vivo in cattle. However, anti-BVDV activity of bovIFN-λ3 has not been studied in bovine respiratory tract epithelial cells, supposedly a primary target of BVDV infection when entering the host by the oronasal route. Methods: Here we investigated the anti-BVDV activity of bovIFN-λ3 in bovine turbinate-derived primary epithelial cells (BTu) using BVDV infection and immunoperoxidase staining, TCID50, RT-qPCR, DNA and transcriptome sequencing, and transfection with plasmids containing the two subunits, IL-28Rα and IL-10Rß that constitute the bovIFN-λ3 receptor. Results: Our immunoperoxidase staining, RT-qPCR, and TCID50 results show that while BVDV was successfully cleared in MDBK cells treated with bovIFN-λ3 and bovIFN-α, only the latter, bovIFN-α, cleared BVDV in BTu cells. Preincubation of MDBK cells with bovIFN-λ3 before BVDV infection was needed to induce optimal antiviral state. Both cell types displayed intact type I and III IFN signaling pathways and expressed similar levels of IL-10Rß subunit of the type III IFN receptor. Sequencing of PCR amplicon of the IL-28Rα subunit revealed intact transmembrane domain and lack of single nucleotide polymorphisms (SNPs) in BTu cells. However, RT-qPCR and transcriptomic analyses showed a lower expression of IL-28Rα transcripts in BTu cells as compared to MDBK cells. Interestingly, transfection of BTu cells with a plasmid encoding IL-28Rα subunit, but not IL-10Rß subunit, established the bovIFN-λ3 sensitivity showing similar anti-BVDV activity to the response in MDBK cells. Conclusion: Our results demonstrate that the sensitivity of cells to bovIFN-λ3 depends not only on the quality but also of the quantity of the IL-28Rα subunit of the heterodimeric receptor. A reduction in IL-28Rα transcript expression was detected in BTu as compared to MDBK cells, despite the absence of spliced variants or SNPs. The establishment of bovIFN-λ3 induced anti-BVDV activity in BTu cells transfected with an IL-28Rα plasmid suggests that the level of expression of this receptor subunit is crucial for the specific antiviral activity of type III IFN in these cells.


Subject(s)
Interferon Lambda , Interferons , Turbinates , Animals , Cattle , Interferons/metabolism , Interferons/immunology , Turbinates/virology , Turbinates/immunology , Turbinates/metabolism , Antiviral Agents/pharmacology , Diarrhea Viruses, Bovine Viral/immunology , Diarrhea Viruses, Bovine Viral/physiology , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Epithelial Cells/virology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Interleukins/genetics , Interleukins/pharmacology , Interleukins/immunology , Interleukins/metabolism , Cell Line , Bovine Virus Diarrhea-Mucosal Disease/immunology , Bovine Virus Diarrhea-Mucosal Disease/virology , Recombinant Proteins/pharmacology , Interleukin-10 Receptor beta Subunit/genetics , Interleukin-10 Receptor beta Subunit/metabolism , Receptors, Cytokine
4.
Cancer Cell ; 42(8): 1450-1466.e11, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39137729

ABSTRACT

Glioblastoma (GBM) is an aggressive brain cancer with limited therapeutic options. Natural killer (NK) cells are innate immune cells with strong anti-tumor activity and may offer a promising treatment strategy for GBM. We compared the anti-GBM activity of NK cells engineered to express interleukin (IL)-15 or IL-21. Using multiple in vivo models, IL-21 NK cells were superior to IL-15 NK cells both in terms of safety and long-term anti-tumor activity, with locoregionally administered IL-15 NK cells proving toxic and ineffective at tumor control. IL-21 NK cells displayed a unique chromatin accessibility signature, with CCAAT/enhancer-binding proteins (C/EBP), especially CEBPD, serving as key transcription factors regulating their enhanced function. Deletion of CEBPD resulted in loss of IL-21 NK cell potency while its overexpression increased NK cell long-term cytotoxicity and metabolic fitness. These results suggest that IL-21, through C/EBP transcription factors, drives epigenetic reprogramming of NK cells, enhancing their anti-tumor efficacy against GBM.


Subject(s)
Brain Neoplasms , CCAAT-Enhancer-Binding Protein-delta , Glioblastoma , Interleukins , Killer Cells, Natural , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Glioblastoma/immunology , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/therapy , Interleukins/genetics , Interleukins/metabolism , Interleukins/immunology , Humans , Animals , Mice , CCAAT-Enhancer-Binding Protein-delta/metabolism , CCAAT-Enhancer-Binding Protein-delta/genetics , Brain Neoplasms/immunology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Line, Tumor , Interleukin-15/genetics , Interleukin-15/metabolism , Interleukin-15/immunology , Xenograft Model Antitumor Assays
5.
Cancer Cell ; 42(8): 1333-1335, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39137725

ABSTRACT

Glioblastoma is the most common brain cancer, with a 5-year survival rate of less than 10%. This grim prognosis highlights the urgent need for novel therapeutic approaches. In this issue of Cancer Cell, Shanley et al.1 report an innovative engineering strategy to supercharge NK cell immunity against glioblastoma.


Subject(s)
Brain Neoplasms , Glioblastoma , Interleukins , Killer Cells, Natural , Glioblastoma/immunology , Glioblastoma/therapy , Humans , Killer Cells, Natural/immunology , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Interleukins/metabolism , Interleukins/immunology
6.
Dev Comp Immunol ; 161: 105249, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39154973

ABSTRACT

IL-26 is a cytokine that is crucial for the maintenance and function of the gut mucosal barrier. IL-26 signaling pathway relies on a heterodimeric receptor complex, which is composed of two distinct subunits, IL-10R2 and IL-20R1. However, there are no reports on the antibacterial immunity of IL-26 and its receptors in fish. For this purpose, in this study we identified IL-26 and its receptors IL-10R2 and IL-20R1 in Carassius cuvieri × Carassius auratus red var. (named WR-IL-26, WR-IL10R2 and WR-IL20R1, respectively). Phylogenetic analysis confirmed the conservation of these genes, with shared structural motifs similar to those found in higher vertebrates. Upon exposure to Aeromonas hydrophila, a common fish pathogen, there was a significant upregulation of WR-IL-26, WR-IL10R2 and WR-IL20R1 in the gut, indicating a potential role in the immune response to infection. A co-immunoprecipitation assay revealed that WR-IL-26 formed complexes with WR-IL10R2 and WR-IL20R1. In vivo experiments demonstrated that administration of WR-IL-26 activated the JAK1-STAT3 signaling pathway and protected the gut mucosa barrier from A. hydrophila infection. Conversely, silencing WR-IL10R2 and WR-IL20R1 via RNA interference significantly attenuated the activation of WR-IL-26-mediated JAK1-STAT3 pathway. These results provided new insights into the role of IL-26 and its receptors in the gut mucosa barrier and could offer novel therapeutic strategies for managing bacterial infections in aquaculture.


Subject(s)
Aeromonas hydrophila , Fish Diseases , Fish Proteins , Immunity, Innate , Interleukins , Intestinal Mucosa , Receptors, Interleukin , Signal Transduction , Animals , Fish Proteins/genetics , Fish Proteins/metabolism , Fish Proteins/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Aeromonas hydrophila/immunology , Aeromonas hydrophila/physiology , Interleukins/metabolism , Interleukins/immunology , Interleukins/genetics , Receptors, Interleukin/metabolism , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Fish Diseases/immunology , Signal Transduction/immunology , Phylogeny , Gram-Negative Bacterial Infections/immunology , Goldfish/immunology , Immunity, Mucosal , Interleukin-10 Receptor beta Subunit/metabolism , Interleukin-10 Receptor beta Subunit/genetics , Interleukin-10 Receptor beta Subunit/immunology
7.
Fish Shellfish Immunol ; 153: 109838, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39151839

ABSTRACT

IL-26 is a crucial inflammatory cytokine that participates in defending host cells against infections. We initially cloned and identified the cDNA sequences of interleukin (IL)-26 in channel catfish (Ictalurus punctatus). The open reading frame (ORF) of IpIL-26 was 537 bp in length, encoding 178 amino acids (aa). Constitutive expression of IpIL-26 was observed in tested tissues, with the highest level found in the gill and spleen. To explore the function of IpIL-26 in channel catfish, different stimuli were used to act on both channel catfish and channel catfish kidney cells (CCK). The expression of IpIL-26 could be up-regulated by bacteria and viruses in multiple tissues. In vitro, recombinant IpIL-26 (rIpIL-26) could induce the expression levels of inflammatory cytokines such as TNF-α, IL-1ß, IL-6, IL-20, and IL-22 playing vital roles in defending the host against infections. Our results demonstrated that IpIL-26 might be an essential cytokine, significantly affecting the immune defense of channel catfish against pathogen infections.


Subject(s)
Amino Acid Sequence , Fish Diseases , Fish Proteins , Gene Expression Regulation , Ictaluridae , Immunity, Innate , Interleukins , Phylogeny , Sequence Alignment , Animals , Ictaluridae/immunology , Ictaluridae/genetics , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/chemistry , Interleukins/genetics , Interleukins/immunology , Immunity, Innate/genetics , Fish Diseases/immunology , Sequence Alignment/veterinary , Gene Expression Regulation/immunology , Gene Expression Profiling/veterinary , Base Sequence , DNA, Complementary/genetics
8.
Signal Transduct Target Ther ; 9(1): 199, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39117617

ABSTRACT

High frequencies of stem-like memory T cells in infusion products correlate with superior patient outcomes across multiple T cell therapy trials. Herein, we analyzed a published CRISPR activation screening to identify transcriptional regulators that could be harnessed to augment stem-like behavior in CD8+ T cells. Using IFN-γ production as a proxy for CD8+ T cell terminal differentiation, LMO4 emerged among the top hits inhibiting the development of effectors cells. Consistently, we found that Lmo4 was downregulated upon CD8+ T cell activation but maintained under culture conditions facilitating the formation of stem-like T cells. By employing a synthetic biology approach to ectopically express LMO4 in antitumor CD8+ T cells, we enabled selective expansion and enhanced persistence of transduced cells, while limiting their terminal differentiation and senescence. LMO4 overexpression promoted transcriptional programs regulating stemness, increasing the numbers of stem-like CD8+ memory T cells and enhancing their polyfunctionality and recall capacity. When tested in syngeneic and xenograft tumor models, LMO4 overexpression boosted CD8+ T cell antitumor immunity, resulting in enhanced tumor regression. Rather than directly modulating gene transcription, LMO4 bound to JAK1 and potentiated STAT3 signaling in response to IL-21, inducing the expression of target genes (Tcf7, Socs3, Junb, and Zfp36) crucial for memory responses. CRISPR/Cas9-deletion of Stat3 nullified the enhanced memory signature conferred by LMO4, thereby abrogating the therapeutic benefit of LMO4 overexpression. These results establish LMO4 overexpression as an effective strategy to boost CD8+ T cell stemness, providing a new synthetic biology tool to bolster the efficacy of T cell-based immunotherapies.


Subject(s)
Adaptor Proteins, Signal Transducing , CD8-Positive T-Lymphocytes , LIM Domain Proteins , STAT3 Transcription Factor , Signal Transduction , LIM Domain Proteins/genetics , LIM Domain Proteins/immunology , CD8-Positive T-Lymphocytes/immunology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Mice , Animals , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Humans , Signal Transduction/immunology , Signal Transduction/genetics , Interleukins/genetics , Interleukins/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology
9.
JCI Insight ; 9(16)2024 08 22.
Article in English | MEDLINE | ID: mdl-39171524

ABSTRACT

Decidual regulatory T cells (Tregs) are essential for successful pregnancy outcome. A subset of Tregs, T cell immunoglobulin and mucin domain-containing protein 3-positive regulatory T cells (TregsTim-3+), plays a central role in the acceptance of the fetus during early stages of normal pregnancy. The molecular mechanism regulating the differentiation and function of TregsTim-3+ is unknown. Here, we investigated the role of the transcription factor B lymphocyte-induced maturation protein 1 (Blimp-1) on decidual TregTim-3+ differentiation. We demonstrated that Blimp-1 enhanced the coexpression of negative costimulatory molecules (Tim-3, T cell immunoreceptor with Ig and ITIM domains, and programmed cell death protein 1) on Tregs and improved their immunosuppressive functions, including increased IL-10 secretion, suppression of effector T cell proliferation, and promotion of macrophage polarization toward the M2 phenotype. Furthermore, we showed that IL-27 regulated the expression of Tim-3 and Blimp-1 through the STAT1 signaling pathway and that transfer of TregsBlimp-1+ into an abortion-prone mouse model effectively reduced embryo absorption rate. We postulated that abnormalities in the IL-27/Blimp-1 axis might be associated with recurrent pregnancy loss (RPL). These findings provided insights for developing more efficient immunotherapies for women with RPL.


Subject(s)
Cell Differentiation , Hepatitis A Virus Cellular Receptor 2 , Positive Regulatory Domain I-Binding Factor 1 , T-Lymphocytes, Regulatory , Female , Animals , Pregnancy , Positive Regulatory Domain I-Binding Factor 1/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Mice , Cell Differentiation/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , Humans , Decidua/immunology , Decidua/metabolism , Decidua/cytology , Interleukin-10/metabolism , Interleukins/metabolism , Interleukins/immunology , Interleukin-27/metabolism
10.
J Clin Invest ; 134(15)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-39087473

ABSTRACT

Inflammatory neuropathies, which include chronic inflammatory demyelinating polyneuropathy (CIDP) and Guillain Barré syndrome (GBS), result from autoimmune destruction of the PNS and are characterized by progressive weakness and sensory loss. CD4+ T cells play a key role in the autoimmune destruction of the PNS. Yet, key properties of pathogenic CD4+ T cells remain incompletely understood. Here, we used paired single-cell RNA-Seq (scRNA-Seq) and single-cell T cell receptor-sequencing (scTCR-Seq) of peripheral nerves from an inflammatory neuropathy mouse model to identify IL-21-expressing CD4+ T cells that were clonally expanded and multifunctional. These IL-21-expressing CD4+ T cells consisted of 2 transcriptionally distinct expanded cell populations, which expressed genes associated with T follicular helper (Tfh) and T peripheral helper (Tph) cell subsets. Remarkably, TCR clonotypes were shared between these 2 IL-21-expressing cell populations, suggesting a common lineage differentiation pathway. Finally, we demonstrated that IL-21 receptor-KO (IL-21R-KO) mice were protected from neuropathy development and had decreased immune infiltration into peripheral nerves. IL-21 signaling upregulated CXCR6, a chemokine receptor that promotes CD4+ T cell localization in peripheral nerves. Together, these findings point to IL-21 signaling, Tfh/Tph differentiation, and CXCR6-mediated cellular localization as potential therapeutic targets in inflammatory neuropathies.


Subject(s)
CD4-Positive T-Lymphocytes , Interleukins , Animals , Mice , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Lineage , Interleukins/genetics , Interleukins/immunology , Interleukins/metabolism , Mice, Knockout , Male , Female
11.
Zhongguo Fei Ai Za Zhi ; 27(7): 550-558, 2024 Jul 20.
Article in Chinese | MEDLINE | ID: mdl-39147710

ABSTRACT

Non-small cell lung cancer (NSCLC) is a prevalent and aggressive global malignancy. Conventional surgical treatments, radiotherapy, chemotherapy, and targeted therapies often fall short in halting disease progression due to inherent limitations, resulting in suboptimal prognosis. Despite the advent of immunotherapy drugs offering new hope for NSCLC treatment, current efficacy remains insufficient to meet all patient needs. Therefore, actively exploring novel immunotherapeutic approaches to further reduce mortality rates in NSCLC patients has become a crucial focus of NSCLC research. This article aims to systematically review the anti-tumor effects of interleukin-21 and follicular helper T cells in NSCLC immunotherapy by summarizing and analyzing relevant literatures from both domestic and international sources, as well as exploring the potential for enhancing NSCLC treatment prospects through immune checkpoint regulation via immunotherapeutic means.
.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Immunotherapy , Interleukins , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Lung Neoplasms/drug therapy , Interleukins/immunology , Interleukins/therapeutic use , Immunotherapy/methods , Animals
12.
Korean J Gastroenterol ; 84(2): 65-81, 2024 Aug 25.
Article in Korean | MEDLINE | ID: mdl-39176462

ABSTRACT

Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a chronic inflammatory disease of the gastrointestinal tract. The introduction of biologics, particularly anti-interleukin (IL) agents, has revolutionized IBD treatment. This review summarizes the role of ILs in IBD pathophysiology and describes the efficacy and positioning of anti-IL therapies. We discuss the functions of key ILs in IBD and their potential as therapeutic targets. The review then discusses anti-IL therapies, focusing primarily on ustekinumab (anti-IL-12/23), risankizumab (anti-IL-23), and mirikizumab (anti-IL-23). Clinical trial data demonstrate their efficacy in inducing and maintaining remission in Crohn's disease and ulcerative colitis. The safety profiles of these agents are generally favorable. However, long-term safety data for newer agents are still limited. The review also briefly discusses emerging therapies such as guselkumab and brazikumab. Network meta-analyses suggest that anti-IL therapies perform well compared to other biological agents. These agents may be considered first- or second-line therapies for many patients, especially those with comorbidities or safety concerns. Anti-IL therapies represent a significant advancement in IBD treatment, offering effective and relatively safe options for patients with moderate to severe disease.


Subject(s)
Antibodies, Monoclonal, Humanized , Biological Products , Inflammatory Bowel Diseases , Interleukins , Humans , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Biological Products/pharmacology , Biological Products/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Interleukins/antagonists & inhibitors , Interleukins/immunology , Interleukins/metabolism , Ustekinumab/therapeutic use
13.
Nat Immunol ; 25(9): 1565-1579, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39103576

ABSTRACT

Nucleophosmin 1 (NPM1) is commonly mutated in myelodysplastic syndrome (MDS) and acute myeloid leukemia. Concurrent inflammatory bowel diseases (IBD) and MDS are common, indicating a close relationship between IBD and MDS. Here we examined the function of NPM1 in IBD and colitis-associated colorectal cancer (CAC). NPM1 expression was reduced in patients with IBD. Npm1+/- mice were more susceptible to acute colitis and experimentally induced CAC than littermate controls. Npm1 deficiency impaired the function of interleukin-22 (IL-22)-producing group three innate lymphoid cells (ILC3s). Mice lacking Npm1 in ILC3s exhibited decreased IL-22 production and accelerated development of colitis. NPM1 was important for mitochondrial biogenesis and metabolism by oxidative phosphorylation in ILC3s. Further experiments revealed that NPM1 cooperates with p65 to promote mitochondrial transcription factor A (TFAM) transcription in ILC3s. Overexpression of Npm1 in mice enhanced ILC3 function and reduced the severity of dextran sulfate sodium-induced colitis. Thus, our findings indicate that NPM1 in ILC3s protects against IBD by regulating mitochondrial metabolism through a p65-TFAM axis.


Subject(s)
Colitis , Immunity, Mucosal , Mice, Knockout , Mitochondria , Nuclear Proteins , Nucleophosmin , Oxidative Phosphorylation , Animals , Mitochondria/metabolism , Mice , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Humans , Colitis/immunology , Colitis/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice, Inbred C57BL , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Interleukin-22 , Immunity, Innate , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Dextran Sulfate , Male , Interleukins/metabolism , Interleukins/genetics , Interleukins/immunology , Female
14.
Front Immunol ; 15: 1437046, 2024.
Article in English | MEDLINE | ID: mdl-39156888

ABSTRACT

Metabolic dysfunction-associated steatotic liver disease (MASLD) comprises a spectrum of liver diseases that span simple steatosis, metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis and may progress to cirrhosis and cancer. The pathogenesis of MASLD is multifactorial and is driven by environmental, genetic, metabolic and immune factors. This review will focus on the role of the type 3 cytokines IL-17 and IL-22 in MASLD pathogenesis and progression. IL-17 and IL-22 are produced by similar adaptive and innate immune cells such as Th17 and innate lymphoid cells, respectively. IL-17-related signaling is upregulated during MASLD resulting in increased chemokines and proinflammatory cytokines in the liver microenvironment, enhanced recruitment of myeloid cells and T cells leading to exacerbation of inflammation and liver disease progression. IL-17 may also act directly by activating hepatic stellate cells resulting in increased fibrosis. In contrast, IL-22 is a pleiotropic cytokine with a dominantly protective signature in MASLD and is currently being tested as a therapeutic strategy. IL-22 also exhibits beneficial metabolic effects and abrogates MASH-related inflammation and fibrosis development via inducing the production of anti-oxidants and anti-apoptotic factors. A sex-dependent effect has been attributed to both cytokines, most importantly to IL-22 in MASLD or related conditions. Altogether, IL-17 and IL-22 are key effectors in MASLD pathogenesis and progression. We will review the role of these two cytokines and cells that produce them in the development of MASLD, their interaction with host factors driving MASLD including sexual dimorphism, and their potential therapeutic benefits.


Subject(s)
Interleukin-17 , Interleukin-22 , Interleukins , Humans , Interleukin-17/metabolism , Interleukin-17/immunology , Interleukins/metabolism , Interleukins/immunology , Animals , Fatty Liver/immunology , Fatty Liver/metabolism , Fatty Liver/pathology , Metabolic Diseases/metabolism , Metabolic Diseases/immunology , Liver/pathology , Liver/metabolism , Liver/immunology
15.
Front Immunol ; 15: 1447431, 2024.
Article in English | MEDLINE | ID: mdl-39211040

ABSTRACT

Mammalian interleukin-22 (IL-22) attenuates organismal injury by inhibiting reactive oxygen species (ROS) and impeding the NLRP3 inflammasome activation. However, the role of fish IL-22 in this process remains unclear. We characterized MaIL-22, an IL-22 homolog in blunt snout bream (Megalobrama amblycephala). Despite its low sequence identity, it shares conserved structures and close evolutionary relationships with other teleost IL-22s. Furthermore, Aeromonas hydrophila (A. hydrophila) infection leads to tissue injury in M. amblycephala immune organs and concomitantly altered Mail-22 mRNA expression, suggesting that MaIL-22 was involved in the antimicrobial immune response. To explore MaIL-22's biological functions, we produced recombinant MaIL-22 (rMaIL-22) protein and demonstrated it significantly enhanced the survival of M. amblycephala post-A. hydrophila infection. To unravel its protective mechanisms, we explored the ROS/NLRP3 inflammasome axis and its downstream signaling responses. The results showed that rMaIL-22 treatment significantly elevated antioxidant enzyme (T-SOD, CAT and GSH-PX) activities to inhibit MDA activity and scavenge ROS in visceral tissues. Meanwhile, rMaIL-22 impeded the activation of NLRP3 inflammasome by suppressing NLRP3 protein and mRNA expression. This indicated that rMaIL-22 contributed to inhibit A. hydrophila-induced activation of the ROS/NLRP3 inflammasome axis. Consistent with these findings, rMaIL-22 treatment attenuated the expression of proinflammatory cytokines (il-1ß, tnf-α and il-6) and proapoptotic genes (caspase-3 and caspase-8) while promoting antiapoptotic genes (bcl-2b and mcl-1a) expression, ultimately mitigating tissue injury in visceral tissues. In conclusion, our research underscores MaIL-22's key role in microbial immune regulation, offering insights for developing IL-22-targeted therapies and breeding programs.


Subject(s)
Aeromonas hydrophila , Apoptosis , Fish Diseases , Gram-Negative Bacterial Infections , Inflammasomes , Inflammation , Interleukin-22 , Interleukins , NLR Family, Pyrin Domain-Containing 3 Protein , Reactive Oxygen Species , Animals , Aeromonas hydrophila/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Inflammasomes/immunology , Apoptosis/drug effects , Gram-Negative Bacterial Infections/immunology , Reactive Oxygen Species/metabolism , Interleukins/metabolism , Interleukins/immunology , Fish Diseases/immunology , Fish Diseases/microbiology , Inflammation/immunology , Fish Proteins/metabolism , Fish Proteins/immunology , Fish Proteins/genetics , Cyprinidae/immunology , Cyprinidae/metabolism , Signal Transduction
16.
J Immunol ; 213(5): 559-566, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38975727

ABSTRACT

Inactivating mutations of Foxp3, the master regulator of regulatory T cell development and function, lead to immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome in mice and humans. IPEX is a fatal autoimmune disease, with allogeneic stem cell transplant being the only available therapy. In this study, we report that a single dose of adeno-associated virus (AAV)-IL-27 to young mice with naturally occurring Foxp3 mutation (Scurfy mice) substantially ameliorates clinical symptoms, including growth retardation and early fatality. Correspondingly, AAV-IL-27 gene therapy significantly prevented naive T cell activation, as manifested by downregulation of CD62L and upregulation of CD44, and immunopathology typical of IPEX. Because IL-27 is known to induce IL-10, a key effector molecule of regulatory T cells, we evaluated the contribution of IL-10 induction by crossing IL-10-null allele to Scurfy mice. Although IL-10 deficiency does not affect the survival of Scurfy mice, it largely abrogated the therapeutic effect of AAV-IL-27. Our study revealed a major role for IL-10 in AAV-IL-27 gene therapy and demonstrated that IPEX is amenable to gene therapy.


Subject(s)
Forkhead Transcription Factors , Genetic Diseases, X-Linked , Genetic Therapy , Germ-Line Mutation , Interleukin-10 , T-Lymphocytes, Regulatory , Animals , Forkhead Transcription Factors/genetics , Mice , Interleukin-10/genetics , Interleukin-10/immunology , Genetic Therapy/methods , T-Lymphocytes, Regulatory/immunology , Genetic Diseases, X-Linked/therapy , Genetic Diseases, X-Linked/immunology , Genetic Diseases, X-Linked/genetics , Interleukins/immunology , Interleukins/genetics , Diarrhea/genetics , Diarrhea/therapy , Diarrhea/immunology , Intestinal Diseases/immunology , Intestinal Diseases/genetics , Intestinal Diseases/therapy , Dependovirus/genetics , Mice, Inbred C57BL , Immune System Diseases/immunology , Immune System Diseases/therapy , Immune System Diseases/genetics , Immune System Diseases/congenital , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/congenital , Mice, Knockout , Lymphocyte Activation/immunology , Humans , Interleukin-27/genetics
17.
Mol Immunol ; 173: 20-29, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39018744

ABSTRACT

SjÓ§gren's syndrome is a systemic autoimmune disease primarily targeting the salivary and lacrimal glands. Our previous investigations have shown that administration of interleukin-22 (IL-22), an IL-10 family cytokine known for its complex and context-dependent effects on tissues, either protective- or detrimental, to salivary glands leads to hypofunction and pathological changes of salivary glands in C57BL/6 mice and in non-obese diabetic (NOD) mice, the latter being a commonly used model of SjÓ§gren's syndrome. This study aims to delineate the pathophysiological roles of endogenously produced IL-22 in the development of salivary gland pathologies and dysfunction associated with SjÓ§gren's disease in the NOD mouse model. Our results reveal that neutralizing IL-22 offered a protective effect on salivary gland function without significantly affecting the immune cell infiltration of salivary glands or the autoantibody production. Blockade of IL-22 reduced the levels of phosphorylated STAT3 in salivary gland tissues of NOD mice, while its administration to salivary glands had the opposite effect. Correspondingly, the detrimental impact of exogenously applied IL-22 on salivary glands was almost completely abrogated by a specific STAT3 inhibitor. Moreover, IL-22 blockade led to a downregulation of protein amounts of Ten-Eleven-Translocation 2, a methylcytosine dioxygenase critical for mediating interferon-induced responses, in salivary gland epithelial cells. IL-22 neutralization also exerted a protective effect on the salivary gland epithelial cells that express high levels of surface EpCAM and bear the stem cell potential, and IL-22 treatment in vitro hampered the survival/expansion of these salivary gland stem cells, indicating a direct negative impact of IL-22 on these cells. In summary, this study has uncovered a critical pathogenic role of the endogenous IL-22 in the pathogenesis of Sjögren's disease-characteristic salivary gland dysfunction and provided initial evidence that this effect is dependent on STAT3 activation and potentially achieved through fostering Tet2-mediated interferon responses in salivary gland epithelial cells and negatively affecting the EpCAMhigh salivary gland stem cells.


Subject(s)
Interleukin-22 , Salivary Glands , Sjogren's Syndrome , Animals , Female , Humans , Mice , Disease Models, Animal , Interleukins/immunology , Interleukins/metabolism , Mice, Inbred C57BL , Mice, Inbred NOD , Salivary Glands/pathology , Salivary Glands/immunology , Salivary Glands/metabolism , Sjogren's Syndrome/immunology , STAT3 Transcription Factor/metabolism
18.
Mol Immunol ; 173: 71-79, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39067087

ABSTRACT

BACKGROUND: The treatment of food allergy (FA) needs improvement. The treatment of immune disorders can be improved by regulating epigenetic marks, which is a promising method. The objective of this research is to alleviate experimental FA by employing an inhibitor of DNA methyltransferase-1 (DNMT1). METHODS: Ovalbumin was used as the specific antigen to establish a mouse model of FA. Intestinal IL-35+ regulatory B cells (Breg cells) were isolated from FA mice, and characterized using immunological approaches. RESULTS: FA mice had a lower frequency of IL-35+ Breg cells, which was inversely correlated with their FA response. The quantity of IL-35 was lower in intestinal Breg cells from FA mice. Hypermethylation status was detected in the Il35 promoter, which was accompanied with high levels of H3K9me3. Enforced expression of DNMT1 hindered the promoter activity of the IL35 gene. Administration of an inhibitor of DNMT1 (RG108) restored the immune regulatory capacity of FA intestinal Bregs, and effectively suppressed the expression of DNMT1, and attenuated experimental FA. CONCLUSIONS: The elevated quantity of DNMT1 in intestinal Breg cells compromises the expression of IL-35 and affects the immune regulatory functions, which facilitates the development of FA. The immune regulatory functions of intestinal Breg cells are restored and experimental FA is attenuated by inhibiting DNMT1.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , Food Hypersensitivity , Interleukins , Promoter Regions, Genetic , Animals , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Food Hypersensitivity/immunology , Mice , Interleukins/immunology , Interleukins/metabolism , Interleukins/genetics , Promoter Regions, Genetic/genetics , B-Lymphocytes, Regulatory/immunology , Disease Models, Animal , Mice, Inbred BALB C , Female , Ovalbumin/immunology , Phthalimides/pharmacology , Intestines/immunology , Tryptophan/analogs & derivatives
19.
Front Immunol ; 15: 1397098, 2024.
Article in English | MEDLINE | ID: mdl-39044830

ABSTRACT

Background: Follicular helper T cells (Tfh) are pivotal in B cell responses. Activation of the purinergic receptor P2X7 on Tfh cells regulates their activity. We investigated the ATP-P2X7R axis in circulating Tfh (cTfh) cells during Respiratory Syncytial Virus (RSV) infection. Methods: We analyzed two cohorts: children with RSV infection (moderate, n=30; severe, n=21) and healthy children (n=23). We utilized ELISA to quantify the levels of PreF RSV protein-specific IgG antibodies, IL-21 cytokine, and soluble P2X7R (sP2X7R) in both plasma and nasopharyngeal aspirates (NPA). Additionally, luminometry was employed to determine ATP levels in plasma, NPA and supernatant culture. The frequency of cTfh cells, P2X7R expression, and plasmablasts were assessed by flow cytometry. To evaluate apoptosis, proliferation, and IL-21 production by cTfh cells, we cultured PBMCs in the presence of Bz-ATP and/or P2X7R antagonist (KN-62) and a flow cytometry analysis was performed. Results: In children with severe RSV disease, we observed diminished titers of neutralizing anti-PreF IgG antibodies. Additionally, severe infections, compared to moderate cases, were associated with fewer cTfh cells and reduced plasma levels of IL-21. Our investigation revealed dysregulation in the ATP-P2X7R pathway during RSV infection. This was characterized by elevated ATP levels in both plasma and NPA samples, increased expression of P2X7R on cTfh cells, lower levels of sP2X7R, and heightened ATP release from PBMCs upon stimulation, particularly evident in severe cases. Importantly, ATP exposure decreased cTfh proliferative response and IL-21 production, while promoting their apoptosis. The P2X7R antagonist KN-62 mitigated these effects. Furthermore, disease severity positively correlated with ATP levels in plasma and NPA samples and inversely correlated with cTfh frequency. Conclusion: Our findings indicate that activation of the ATP-P2X7R pathway during RSV infection may contribute to limiting the cTfh cell compartment by promoting cell death and dysfunction, ultimately leading to increased disease severity.


Subject(s)
Adenosine Triphosphate , Receptors, Purinergic P2X7 , Respiratory Syncytial Virus Infections , T Follicular Helper Cells , Humans , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/metabolism , Receptors, Purinergic P2X7/metabolism , Adenosine Triphosphate/metabolism , Male , Infant , Female , T Follicular Helper Cells/immunology , T Follicular Helper Cells/metabolism , Child, Preschool , Signal Transduction , Interleukins/metabolism , Interleukins/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Child , Respiratory Syncytial Virus, Human/immunology
20.
Cytokine ; 181: 156692, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38986251

ABSTRACT

IL-35 is a recently discovered protein made up of IL-12α and IL-27ß chains. It is encoded by IL12A and EBI3 genes. Interest in researching IL-35 has significantly increased in recent years, as evidenced by numerous scientific publications. Diabetes is on the rise globally, causing more illness and death in developing countries. The International Diabetes Federation (IDF) reports that diabetes is increasingly affecting children and teenagers, with varying rates across different regions. Therefore, scientists seek new diabetes treatments despite the growth of drug research. Recent research aims to emphasize IL-35 as a critical regulator of diabetes, especially type 1 and autoimmune diabetes. This review provides an overview of recent research on IL-35 and its link to diabetes and its associated complications. Studies suggest that IL-35 can offer protection against type-1 diabetes and autoimmune diabetes by regulating macrophage polarization, T-cell-related cytokines, and regulatory B cells (Bregs). This review will hopefully assist biomedical scientists in exploring the potential role of IL-35-mediated immunotherapy in treating diabetes. However, further research is necessary to determine the exact mechanism and plan clinical trials.


Subject(s)
Diabetes Mellitus, Type 1 , Immunotherapy , Interleukins , Humans , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Immunotherapy/methods , Interleukins/immunology , Interleukins/genetics , Interleukins/metabolism , Animals , B-Lymphocytes, Regulatory/immunology , Macrophages/immunology , Macrophages/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL