Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
1.
Sci Rep ; 14(1): 14248, 2024 06 20.
Article in English | MEDLINE | ID: mdl-38902343

ABSTRACT

Treatment of advanced triple-negative breast cancer (TNBC) is a great challenge in clinical practice. The immune checkpoints are a category of immunosuppressive molecules that cancer could hijack and impede anti-tumor immunity. Targeting immune checkpoints, such as anti-programmed cell death 1 (PD-1) therapy, is a promising therapeutic strategy in TNBC. The efficacy and safety of PD-1 monoclonal antibody (mAb) with chemotherapy have been validated in TNBC patients. However, the precise mechanisms underlying the synergistic effect of chemotherapy and anti-PD-1 therapy have not been elucidated, causing the TNBC patients that might benefit from this combination regimen not to be well selected. In the present work, we found that IL-23, an immunological cytokine, is significantly upregulated after chemotherapy in TNBC cells and plays a vital role in enhancing the anti-tumor immune response of cytotoxic T cells (CTLs), especially in combination with PD-1 mAb. In addition, the combination of IL-23 and PD-1 mAb could synergistically inhibit the expression of Phosphoinositide-3-Kinase Regulatory Subunit 1 (PIK3R1), which is a regulatory subunit of PI3K and inhibit p110 activity, and promote phosphorylation of AKT in TNBC-specific CTLs. Our findings might provide a molecular marker that could be used to predict the effects of combination chemotherapy therapy and PD-1 mAb in TNBC.


Subject(s)
Interleukin-23 Subunit p19 , Phosphatidylinositol 3-Kinases , Programmed Cell Death 1 Receptor , Proto-Oncogene Proteins c-akt , Signal Transduction , T-Lymphocytes, Cytotoxic , Triple Negative Breast Neoplasms , Humans , Proto-Oncogene Proteins c-akt/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/immunology , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Programmed Cell Death 1 Receptor/metabolism , Cell Line, Tumor , Female , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Interleukin-23 Subunit p19/metabolism , Animals , Mice , Antibodies, Monoclonal/pharmacology
2.
Sci Rep ; 14(1): 12293, 2024 05 29.
Article in English | MEDLINE | ID: mdl-38811719

ABSTRACT

HLA-B27 is a major risk factor for spondyloarthritis (SpA), yet the underlying mechanisms remain unclear. HLA-B27 misfolding-induced IL-23, which is mediated by endoplasmic reticulum (ER) stress has been hypothesized to drive SpA pathogenesis. Expression of HLA-B27 and human ß2m (hß2m) in rats (HLA-B27-Tg) recapitulates key SpA features including gut inflammation. Here we determined whether deleting the transcription factor CHOP (Ddit3-/-), which mediates ER-stress induced IL-23, affects gut inflammation in HLA-B27-Tg animals. ER stress-mediated Il23a overexpression was abolished in CHOP-deficient macrophages. Although CHOP-deficiency also reduced Il23a expression in immune cells isolated from the colon of B27+ rats, Il17a levels were not affected, and gut inflammation was not reduced. Rather, transcriptome analysis revealed increased expression of pro-inflammatory genes, including Il1a, Ifng and Tnf in HLA-B27-Tg colon tissue in the absence of CHOP, which was accompanied by higher histological Z-scores. RNAScope localized Il17a mRNA to the lamina propria of the HLA-B27-Tg rats and revealed similar co-localization with Cd3e (CD3) in the presence and absence of CHOP. This demonstrates that CHOP-deficiency does not improve, but rather exacerbates gut inflammation in HLA-B27-Tg rats, indicating that HLA-B27 is not promoting gut disease through ER stress-induced IL-23. Hence, CHOP may protect rats from more severe HLA-B27-induced gut inflammation.


Subject(s)
Colitis , Endoplasmic Reticulum Stress , HLA-B27 Antigen , Spondylarthritis , Transcription Factor CHOP , Animals , HLA-B27 Antigen/genetics , HLA-B27 Antigen/metabolism , Transcription Factor CHOP/metabolism , Transcription Factor CHOP/genetics , Colitis/metabolism , Colitis/genetics , Colitis/chemically induced , Colitis/pathology , Rats , Spondylarthritis/metabolism , Spondylarthritis/pathology , Spondylarthritis/genetics , Disease Models, Animal , Interleukin-23/metabolism , Interleukin-23/genetics , Humans , Interleukin-23 Subunit p19/genetics , Interleukin-23 Subunit p19/metabolism , Rats, Transgenic , Interleukin-17/metabolism , Interleukin-17/genetics , Colon/pathology , Colon/metabolism , Macrophages/metabolism , Macrophages/immunology
3.
Int J Mol Sci ; 25(10)2024 May 18.
Article in English | MEDLINE | ID: mdl-38791570

ABSTRACT

INTRODUCTION: Optimizing treatment with biological agents is an ideal goal for patients with ulcerative colitis (UC). Recent data suggest that mucosal inflammation patterns and serum cytokine profiles differ between patients who respond and those who do not. Ustekinumab, a monoclonal antibody targeting the p40 subunit of interleukin (IL)-12 and IL-23, has shown promise, but predicting treatment response remains a challenge. We aimed to identify prognostic markers of response to ustekinumab in patients with active UC, utilizing information from their mucosal transcriptome. METHODS: We performed a prospective observational study of 36 UC patients initiating treatment with ustekinumab. Colonic mucosal biopsies were obtained before treatment initiation for a gene expression analysis using a microarray panel of 84 inflammatory genes. A differential gene expression analysis (DGEA), correlation analysis, and network centrality analysis on co-expression networks were performed to identify potential biomarkers. Additionally, machine learning (ML) models were employed to predict treatment response based on gene expression data. RESULTS: Seven genes, including BCL6, CXCL5, and FASLG, were significantly upregulated, while IL23A and IL23R were downregulated in non-responders compared to responders. The co-expression analysis revealed distinct patterns between responders and non-responders, with key genes like BCL6 and CRP highlighted in responders and CCL11 and CCL22 in non-responders. The ML algorithms demonstrated a high predictive power, emphasizing the significance of the IL23R, IL23A, and BCL6 genes. CONCLUSIONS: Our study identifies potential biomarkers associated with ustekinumab response in UC patients, shedding light on its underlying mechanisms and variability in treatment outcomes. Integrating transcriptomic approaches, including gene expression analyses and ML, offers valuable insights for personalized treatment strategies and highlights avenues for further research to enhance therapeutic outcomes for patients with UC.


Subject(s)
Colitis, Ulcerative , Computational Biology , Ustekinumab , Humans , Ustekinumab/therapeutic use , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/genetics , Male , Female , Computational Biology/methods , Adult , Middle Aged , Treatment Outcome , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Prospective Studies , Transcriptome , Gene Expression Profiling/methods , Interleukin-23 Subunit p19/genetics , Interleukin-23 Subunit p19/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/drug effects , Machine Learning , Prognosis
4.
Cytokine ; 179: 156619, 2024 07.
Article in English | MEDLINE | ID: mdl-38669908

ABSTRACT

Interleukin (IL)-23 is implicated in the pathogenesis of several inflammatory diseases and is usually linked with helper T cell (Th17) biology. However, there is some data linking IL-23 with innate immune biology in such diseases. We therefore examined the effects of IL-23p19 genetic deletion and/or neutralization on in vitro macrophage activation and in an innate immune-driven peritonitis model. We report that endogenous IL-23 was required for maximal macrophage activation by zymosan as determined by pro-inflammatory cytokine production, including a dramatic upregulation of granulocyte-colony stimulating factor (G-CSF). Furthermore, both IL-23p19 genetic deletion and neutralization in zymosan-induced peritonitis (ZIP) led to a specific reduction in the neutrophil numbers, as well as a reduction in the G-CSF levels in exudate fluids. We conclude that endogenous IL-23 can contribute significantly to macrophage activation during an inflammatory response, mostly likely via an autocrine/paracrine mechanism; of note, endogenous IL-23 can directly up-regulate macrophage G-CSF expression, which in turn is likely to contribute to the regulation of IL-23-dependent neutrophil number and function during an inflammatory response, with potential significance for IL-23 targeting particularly in neutrophil-associated inflammatory diseases.


Subject(s)
Inflammation , Interleukin-23 , Myeloid Cells , Neutrophils , Zymosan , Animals , Inflammation/metabolism , Inflammation/immunology , Interleukin-23/metabolism , Mice , Neutrophils/metabolism , Neutrophils/immunology , Myeloid Cells/metabolism , Peritonitis/metabolism , Peritonitis/immunology , Mice, Inbred C57BL , Granulocyte Colony-Stimulating Factor/metabolism , Macrophage Activation , Macrophages/metabolism , Macrophages/immunology , Interleukin-23 Subunit p19/metabolism , Interleukin-23 Subunit p19/genetics , Mice, Knockout
5.
Biochem Pharmacol ; 222: 116072, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38387530

ABSTRACT

BACKGROUND: Interleukin-23p19 (IL-23p19) has been demonstrated to be involved in the occurrence and development of cardiovascular diseases such as myocardial infarction and atherosclerosis. This study aimed to examine whether IL-23p19 regulates cardiac remodeling processes and explore its possible mechanisms. METHODS AND RESULTS: Transverse aortic constriction was performed to construct a mouse cardiac remodeling model, and sham surgery was used as a control. The results showed that IL-23p19 expression was increased in the heart after surgery and may be mainly produced by cardiac macrophages. Knockout of IL-23p19 attenuated M1 macrophage polarization, reduced ferroptosis, improved the process of cardiac remodeling and alleviated cardiac dysfunction in TAC mice. Cell culture experiments found that macrophages were the main cause of ferroptosis when phenylephrine (PE) was added, and blocking ferroptosis with ferrostatin-1 (Fer-1), a ferroptosis inhibitor, significantly inhibited M1 macrophage polarization. Treatment with Fer-1 also improved cardiac remodeling and alleviated cardiac dysfunction in IL-23p19-/- mice subjected to TAC surgery. Finally, TAC IL-23p19-/- mice that were administered macrophages isolated from WT mice exhibited an increased proportion of M1 macrophages and aggravated cardiac remodeling, and these effects were reversed when Fer-1 was administered. CONCLUSION: Knockout of IL-23p19 may attenuate M1 macrophage polarization to improve the cardiac remodeling process by reducing macrophage ferroptosis, and IL-23p19 may be a potential target for the prevention and treatment of cardiac remodeling.


Subject(s)
Ferroptosis , Myocardial Infarction , Animals , Mice , Interleukin-23 Subunit p19/metabolism , Interleukin-23 Subunit p19/pharmacology , Interleukins/metabolism , Macrophages , Mice, Knockout , Myocardial Infarction/metabolism , Ventricular Remodeling
6.
Cutan Ocul Toxicol ; 43(2): 124-128, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38284163

ABSTRACT

PURPOSE: To study the effects of the anti-IL-23A antibody risankizumab on the IL-36γ/IL-23A/IL-17A signalling cascade we used a newly developed 3D skin model consisting of primary human keratinocytes, fibroblasts and γδ-T-cells. METHODS: In this in vitro study we developed new full-thickness 3D skin models containing normal human epidermal keratinocytes (NHEK), normal human dermal fibroblasts (NHDF) and IL-23A responsive and IL-17A producing γδ-T-cells. The effects of IL-36γ stimulation with and without risankizumab treatment on IL-23A and IL-17A expression were examined at the RNA and protein levels. RESULTS: In preliminary monolayer experiments stimulation of γδ-T-cells with IL-23A promoted the IL-17A expression that was inhibited after risankizumab treatment. Using 3D skin models containing γδ-T-cells, we found that stimulation with IL-36γ significantly increased not only IL-23A but also IL-17A expression. These effects were inhibited by concomitant treatment with risankizumab. CONCLUSIONS: Our results showed that blockade of IL-23A has inhibitory effects on the IL-36γ/IL-23A feedforward loop. Our newly developed 3D skin model containing IL-23A responsive and IL-17A producing γδ-T-cells enables molecular analysis of targeted therapies aimed at the IL-36γ/IL-23A/IL-17A signalling cascade in psoriasis.


Subject(s)
Antibodies, Monoclonal , Interleukin-17 , Interleukin-23 Subunit p19 , Keratinocytes , Skin , Humans , Interleukin-17/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Antibodies, Monoclonal/pharmacology , Interleukin-23 Subunit p19/metabolism , Skin/drug effects , Skin/metabolism , Skin/immunology , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Interleukin-1/metabolism , Intraepithelial Lymphocytes/drug effects , Intraepithelial Lymphocytes/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Psoriasis/drug therapy , Psoriasis/immunology
7.
J Crohns Colitis ; 18(6): 908-919, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38165390

ABSTRACT

BACKGROUND AND AIMS: Free D-amino acids, which have different functions from L-amino acids, have recently been discovered in various tissues. However, studies on the potential interactions between intestinal inflammation and D-amino acids are limited. We examined the inhibitory effects of D-alanine on the pathogenesis of intestinal inflammation. METHODS: We investigated serum D-amino acid levels in 40 patients with ulcerative colitis and 34 healthy volunteers. For 7 days [d], acute colitis was induced using dextran sulphate sodium in C57BL/6J mice. Plasma D-amino acid levels were quantified in mice with dextran sulphate sodium-induced colitis, and these animals were administered D-alanine via intraperitoneal injection. IFN-γ, IL-12p35, IL-17A, and IL-23p19 mRNA expression in the colonic mucosa was measured using real-time polymerase chain reaction [PCR]. In vitro proliferation assays were performed to assess naïve CD4+ T cell activation under Th-skewing conditions. Bone marrow cells were stimulated with mouse macrophage-colony stimulating factor to generate mouse bone marrow-derived macrophages. RESULTS: Serum D-alanine levels were significantly lower in patients with ulcerative colitis than in healthy volunteers. Dextran sulphate sodium-treated mice had significantly lower plasma D-alanine levels than control mice. D-alanine-treated mice had significantly lower disease activity index than control mice. IFN-γ, IL-12p35, IL-17A, and IL-23p19 mRNA expression levels were significantly lower in D-alanine-administered mice than in control mice. D-alanine suppressed naïve T cell differentiation into Th1 cells in vitro, and inhibited the production of IL-12p35 and IL-23p19 in bone marrow-derived macrophages. CONCLUSIONS: Our results suggest that D-alanine prevents dextran sulphate sodium-induced colitis in mice and suppresses IL-12p35 and IL-23p19 production in macrophages.


Subject(s)
Alanine , Colitis, Ulcerative , Dextran Sulfate , Interleukin-23 , Macrophages , Mice, Inbred C57BL , Animals , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Colitis, Ulcerative/drug therapy , Humans , Mice , Macrophages/metabolism , Macrophages/drug effects , Male , Adult , Female , Alanine/pharmacology , Interleukin-23/metabolism , Interleukin-12/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Middle Aged , Disease Models, Animal , Case-Control Studies , RNA, Messenger/metabolism , Interleukin-12 Subunit p35/metabolism , Interleukin-23 Subunit p19/metabolism , Young Adult
8.
Kaohsiung J Med Sci ; 39(8): 789-800, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37098777

ABSTRACT

The interleukin-23 (IL-23)/IL-17 immune axis has been linked to the pathology of psoriasis, but how this axis contributes to skin inflammation in this disease remains unclear. We measured inflammatory cytokines associated with the IL-23/IL-17 immune axis in the serum of patients with psoriasis using enzyme-linked immunosorbent assays. Psoriasis was induced in male C57BL/6J mice using imiquimod (IMQ) cream, and animals received intraperitoneal injections of recombinant mouse anti-IL-23A or anti-IL-17A antibodies for 7 days. The potential effects of the IL-23/IL-17 immune axis on skin inflammation were assessed based on pathology scoring, hematoxylin-eosin staining of skin samples, and quantitation of inflammatory cytokines. Western blotting was used to evaluate levels of the following factors in skin: ACT1, TRAF6, TAK1, NF-κB, and pNF-κB. The serum of psoriasis patients showed elevated levels of several cytokines involved in the IL-23/IL-17 immune axis: IL-2, IL-4, IL-8, IL-12, IL-17, IL-22, IL-23, and interferon-γ. Levels of IL-23p19 and IL-17 were increased in serum and skin of IMQ-treated mice, while ACT1, TRAF6, TAK1, NF-κB, and pNF-κB were upregulated in the skin. A large proportion of NF-κB p65 localized in nucleus of involucrin+ cells in the epidermis and in F4/80+ cells of the dermis of psoriatic lesional skin. Treating these animals with anti-IL-23 or anti-IL-17 antibodies improved pathological score and immune imbalance, mitigated skin inflammation and downregulated ACT1, TRAF6, TAK1, NF-κB, and pNF-κB in skin. Our results suggest that skin inflammation mediated by the IL-23/IL-17 immune axis in psoriasis involves activation of the ACT1/TRAF6/TAK1/NF-κB pathway in keratinocytes and macrophage.


Subject(s)
Imiquimod , Interleukin-17 , Interleukin-23 , NF-kappa B , Psoriasis , Animals , Male , Mice , Cytokines/metabolism , Disease Models, Animal , Imiquimod/adverse effects , Inflammation/pathology , Interleukin-23/genetics , Interleukin-23 Subunit p19/genetics , Interleukin-23 Subunit p19/metabolism , Keratinocytes/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Psoriasis/pathology , Skin/pathology , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Interleukin-17/metabolism
9.
Cells ; 11(19)2022 09 29.
Article in English | MEDLINE | ID: mdl-36231033

ABSTRACT

Chronic low-grade inflammation induced by obesity is a central risk factor for the development of metabolic syndrome. High low-density lipoprotein cholesterol (LDL-c) induces inflammation, which is a common denominator in metabolic syndrome. IL-23 plays a significant role in the pathogenesis of meta-inflammatory diseases; however, its relationship with LDL-c remains elusive. In this cross-sectional study, we determined whether the adipose tissue IL-23 expression was associated with other inflammatory mediators in people with increased plasma LDL-c concentrations. Subcutaneous adipose tissue biopsies were collected from 60 people, sub-divided into two groups based on their plasma LDL-c concentrations (<2.9 and ≥2.9 mmol/L). Adipose expression of IL-23 and inflammatory markers were determined using real-time qRT-PCR; plasma concentrations of total cholesterol (TC), triglyceride (TG), high-density lipoprotein cholesterol (HDL-c) and LDL-c were determined using the standard method; and adiponectin levels were measured by enzyme-linked immunosorbent assay (ELISA). Adipose IL-23 transcripts were found to be increased in people with high LDL-c, compared to low LDL-c group (H-LDL-c: 1.63 ± 0.10-Fold; L-LDL-c: 1.27 ± 0.09-Fold; p < 0.01); IL-23 correlated positively with LDL-c (r = 0.471, p < 0.0001). Immunochemistry analysis showed that AT IL-23 protein expression was also elevated in the people with H-LDL-c. IL-23 expression in the high LDL-c group was associated with multiple adipose inflammatory biomarkers (p ≤ 0.05), including macrophage markers (CD11c, CD68, CD86, CD127), TLRs (TLR8, TLR10), IRF3, pro-inflammatory cytokines (TNF-α, IL-12, IL-18), and chemokines (CXCL8, CCL3, CCL5, CCL15, CCL20). Notably, in this cohort, IL-23 expression correlated inversely with plasma adiponectin. In conclusion, adipose IL-23 may be an inflammatory biomarker for disease progression in people with high LDL-c.


Subject(s)
Hyperlipidemias , Interleukin-23 Subunit p19/metabolism , Metabolic Syndrome , Adiponectin/metabolism , Adipose Tissue/metabolism , Biomarkers/metabolism , Chemokines/metabolism , Cholesterol/metabolism , Cholesterol, HDL , Cholesterol, LDL/metabolism , Cross-Sectional Studies , Cytokines/metabolism , Humans , Hyperlipidemias/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Interleukin-12/metabolism , Interleukin-18/metabolism , Interleukin-23/metabolism , Metabolic Syndrome/metabolism , Toll-Like Receptor 8/metabolism , Triglycerides/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
Musculoskeletal Care ; 20 Suppl 1: S12-S21, 2022 11.
Article in English | MEDLINE | ID: mdl-36069174

ABSTRACT

BACKGROUND: Psoriatic arthritis (PsA) is a chronic inflammatory arthritis characterised by musculoskeletal and extra-articular manifestations, most notably psoriasis. While the underlying pathogenetic mechanisms are not yet fully understood, a central role has been identified for the IL-23/IL-17 pathway. OBJECTIVES: We briefly describe the role of IL-23 in the pathogenesis of PsA and go on to describe the available anti-IL-23 agents and their place in the management of PsA. METHODS: This is a narrative review of the current literature, focussing on the results of the phase 3 studies in PsA for the IL-12/23 p40 inhibitor ustekinumab and the more recent IL-23 p19 inhibitors guselkumab, risankizumab and tildrakizumab. RESULTS: IL-23 triggers expression of IL-17 and other effector cytokines in a variety of cells, leading to tissue inflammation and injury. Targeting IL-23, particularly with p19 inhibitors, appears to be an effective and safe strategy for multiple clinical domains in PsA, most notably the skin, with some differences in efficacy emerging between these agents. CONCLUSION: The development of IL-23 inhibitors represents a significant advance in the management of psoriatic disease. In the absence of head-to-head studies, future data emerging from real-world experiences of individual IL-23 p19 inhibitors will help inform the use of these agents in relation to other biologics in PsA.


Subject(s)
Arthritis, Psoriatic , Interleukin Inhibitors , Humans , Arthritis, Psoriatic/drug therapy , Interleukin Inhibitors/therapeutic use , Interleukin-17/metabolism , Interleukin-23 Subunit p19/metabolism , Ustekinumab/therapeutic use , Clinical Trials, Phase III as Topic , Interleukin-23/metabolism
11.
Int J Mol Sci ; 23(17)2022 Aug 26.
Article in English | MEDLINE | ID: mdl-36077117

ABSTRACT

Interleukin-23 (IL-23) plays a pivotal role in rheumatoid arthritis (RA). IL-23 and microRNA-223 (miR-223) are both up-regulated and mediate osteoclastogenesis in mice with collagen-induced arthritis (CIA). The aim of this study was to examine the association between IL-23 and miR-223 in contributing to osteoclastogenesis and arthritis. Levels of IL-23p19 in joints of mice with CIA were determined. Lentiviral vectors expressing short hairpin RNA (shRNA) targeting IL-23p19 and lisofylline (LSF) were injected intraperitoneally into arthritic mice. Bone marrow-derived macrophages (BMMs) were treated with signal transducers and activators of transcription 4 (STAT4) specific shRNA and miR-223 sponge carried by lentiviral vectors in response to IL-23 stimulation. Treatment responses were determined by evaluating arthritis scores and histopathology in vivo, and detecting osteoclast differentiation and miR-223 levels in vitro. The binding of STAT4 to the promoter region of primary miR-223 (pri-miR-223) was determined in the Raw264.7 cell line. IL-23p19 expression was increased in the synovium of mice with CIA. Silencing IL-23p19 and inhibiting STAT4 activity ameliorates arthritis by reducing miR-223 expression. BMMs from mice in which STAT4 and miR-223 were silenced showed decreased osteoclast differentiation in response to IL-23 stimulation. IL-23 treatment increased the expression of miR-223 and enhanced the binding of STAT4 to the promoter of pri-miR-223. This study is the first to demonstrate that IL-23 promotes osteoclastogenesis by transcriptional regulation of miR-223 in murine macrophages and mice with CIA. Furthermore, our data indicate that LSF, a selective inhibitor of STAT4, should be an ideal therapeutic agent for treating RA through down-regulating miR-223-associated osteoclastogenesis.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Interleukin-23 Subunit p19/metabolism , MicroRNAs , Animals , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/metabolism , Interleukin-23/genetics , Interleukin-23/metabolism , Mice , MicroRNAs/metabolism , Osteoclasts/metabolism , Osteogenesis , RNA, Small Interfering/metabolism
12.
J Clin Endocrinol Metab ; 107(10): e4037-e4047, 2022 09 28.
Article in English | MEDLINE | ID: mdl-35809263

ABSTRACT

CONTEXT: Teprotumumab, an IGF-I receptor (IGF-IR) inhibitor, is effective in thyroid-associated ophthalmopathy (TAO). The drug can modulate induction by TSH of IL-6 and IL-8 in CD34+ fibrocytes and their putative derivatives, CD34+ orbital fibroblasts (CD34+ OF). Fibrocytes express multiple thyroid autoantigens and cytokines implicated in TAO, which are downregulated by Slit2. Inflammation and disordered hyaluronan (HA) accumulation occur in TAO. Whether teprotumumab alters these processes directly in fibrocytes/CD34+ OF remains uncertain. OBJECTIVE: Determine teprotumumab effects on expression/synthesis of several TAO-relevant molecules in fibrocytes and GD-OF. DESIGN/SETTING/PARTICIPANTS: Patients with TAO and healthy donors were recruited from an academic endocrine and oculoplastic practice. MAIN OUTCOME MEASURES: Real-time PCR, specific immunoassays. RESULTS: Teprotumumab attenuates basal and TSH-inducible autoimmune regulator protein, thyroglobulin, sodium iodide symporter, thyroperoxidase, IL-10, and B-cell activating factor levels in fibrocytes. It downregulates IL-23p19 expression/induction while enhancing IL-12p35, intracellular and secreted IL-1 receptor antagonists, and Slit2. These effects are mirrored by linsitinib. HA production is marginally enhanced by teprotumumab, the consequence of enhanced HAS2 expression. CONCLUSION: Teprotumumab affects specific gene expression in fibrocytes and GD-OF in a target-specific, nonmonolithic manner, whereas IGF-IR control of these cells appears complex. The current results suggest that the drug may act on cytokine expression and HA production systemically and locally, within the TAO orbit. These findings extend our insights into the mechanisms through which IGF-IR inhibition might elicit clinical responses in TAO, including a potential role of Slit2 in attenuating inflammation and tissue remodeling.


Subject(s)
Graves Ophthalmopathy , Antibodies, Monoclonal, Humanized , Autoantigens/metabolism , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , B-Cell Activating Factor/pharmacology , Cells, Cultured , Fibroblasts/metabolism , Gene Expression , Graves Ophthalmopathy/drug therapy , Graves Ophthalmopathy/genetics , Humans , Hyaluronic Acid/metabolism , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Interleukin-10/metabolism , Interleukin-12 Subunit p35/genetics , Interleukin-12 Subunit p35/metabolism , Interleukin-12 Subunit p35/pharmacology , Interleukin-23 Subunit p19/genetics , Interleukin-23 Subunit p19/metabolism , Interleukin-23 Subunit p19/pharmacology , Interleukin-6/metabolism , Interleukin-8/metabolism , Orbit/metabolism , Receptor, IGF Type 1/genetics , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , Thyroglobulin/genetics , Thyrotropin/metabolism
13.
Ann Rheum Dis ; 81(11): 1564-1575, 2022 11.
Article in English | MEDLINE | ID: mdl-35817472

ABSTRACT

OBJECTIVES: The central nervous system disorder in systemic lupus erythematosus (SLE), called neuropsychiatric lupus (NPSLE), is one of the most severe phenotypes with various clinical symptoms, including mood disorder, psychosis and delirium as diffuse neuropsychological manifestations (dNPSLE). Although stress is one of the aggravating factors for neuropsychiatric symptoms, its role in the pathogenesis of dNPSLE remains to be elucidated. We aimed to investigate stress effects on the neuropsychiatric pathophysiology in SLE using lupus-prone mice and patients' data. METHODS: Sleep disturbance stress (SDS) for 2 weeks was placed on 6-8-week-old female MRL/lpr and control mice. Behavioural phenotyping, histopathological analyses and gene and protein expression analyses were performed to assess SDS-induced neuroimmunological alterations. We also evaluated cytokines of the cerebrospinal fluid and brain regional volumes in patients with dNPSLE and patients with non-dNPSLE. RESULTS: SDS-subjected MRL/lpr mice exhibited less anxiety-like behaviour, whereas stressed control mice showed increased anxiety. Furthermore, stress strongly activated the medial prefrontal cortex (mPFC) in SDS-subjected MRL/lpr. A transcriptome analysis of the PFC revealed the upregulation of microglial activation-related genes, including Il12b. We confirmed that stress-induced microglial activation and the upregulation of interleukin (IL) 12/23p40 proteins and increased dendritic spines in the mPFC of stressed MRL/lpr mice. IL-12/23p40 neutralisation and tyrosine kinase 2 inhibition mitigated the stress-induced neuropsychiatric phenotypes of MRL/lpr mice. We also found a higher level of cerebrospinal fluid IL-12/23p40 and more atrophy in the mPFC of patients with dNPSLE than those with non-dNPSLE. CONCLUSIONS: The microglial IL-12/23 axis in the mPFC might be associated with the pathogenesis and a promising therapeutic target for dNPSLE.


Subject(s)
Lupus Erythematosus, Systemic , Microglia , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Interleukin-12 , Interleukin-23 Subunit p19/metabolism , Lupus Erythematosus, Systemic/drug therapy , Mice , Mice, Inbred MRL lpr , Microglia/metabolism , Stress, Physiological , TYK2 Kinase
14.
J Immunol ; 208(3): 660-671, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35022276

ABSTRACT

Invasive candidiasis has high mortality rates in immunocompromised patients, causing serious health problems. In mouse models, innate immunity protects the host by rapidly mobilizing a variety of resistance and tolerance mechanisms to systemic Candida albicans infection. We have previously demonstrated that exogenous IL-33 regulates multiple steps of innate immunity involving resistance and tolerance processes. In this study, we systematically analyzed the in vivo functions of endogenous IL-33 using Il33 -/- mice and in vitro immune cell culture. Tubular epithelial cells mainly secreted IL-33 in response to systemic C. albicans infection. Il33 -/- mice showed increased mortality and morbidity, which were due to impaired fungal clearance. IL-33 initiated an innate defense mechanism by costimulating dendritic cells to produce IL-23 after systemic C. albicans infection, which in turn promoted the phagocytosis of neutrophils through secretion of GM-CSF by NK cells. The susceptibility of Il33 -/- mice was also associated with increased levels of IL-10, and neutralization of IL-10 resulted in enhanced fungal clearance in Il33 -/- mice. However, depletion of IL-10 overrode the effect of IL-33 on fungal clearance. In Il10 -/- mouse kidneys, MHC class II+F4/80+ macrophages were massively differentiated after C. albicans infection, and these cells were superior to MHC class II-F4/80+ macrophages that were preferentially differentiated in wild-type mouse kidneys in killing of extracellular hyphal C. albicans Taken together, our results identify IL-33 as critical early regulator controlling a serial downstream signaling events of innate defense to C. albicans infection.


Subject(s)
Candida albicans/immunology , Candidiasis/immunology , Immunity, Innate/immunology , Interleukin-10/metabolism , Interleukin-23 Subunit p19/metabolism , Interleukin-33/immunology , Animals , Candidiasis/microbiology , Dendritic Cells/immunology , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Histocompatibility Antigens Class II/immunology , Immunocompromised Host/immunology , Interleukin-10/genetics , Interleukin-33/genetics , Killer Cells, Natural/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Phagocytosis/immunology , Signal Transduction/immunology
15.
Andrologia ; 54(5): e14387, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35092070

ABSTRACT

Benign prostatic hyperplasia (BPH) is known to be associated with sleep disturbance and inflammation. The objective of the study was to assess the levels of serum MMP-9 and its inhibitor (TIMP-1), interleukin-23 and pentraxin-3 and their association with sleep quality index and prostate size in BPH patients. Eighty-eight BPH patients were recruited based on clinical and ultrasound findings. MMP-9 and its inhibitor (TIMP-1), interleukin-23 and pentraxin-3 were estimated in all the subjects. Sleep quality was assessed using Pittsburgh Sleep Quality Index (PQSI). Interleukin-23 was significantly correlated with prostate size (p = 0.031), TIMP-1 (p = 0.035), MMP-9 (p = 0.004) and PSQI score (p = 0.020). TIMP-1 was significantly correlated with MMP-9 (p = 0.006) and prostate size (p = 0.016). Pentraxin-3 was positively correlated with PSQI score (p = 0.047). Multivariate analysis shows that interleukin -23 (p = 0.006) predicts prostate enlargement in BPH patients. Interleukin-23 was significantly increased in BPH patients with PSQI score >9 compared to those with PSQI <9. We conclude that poor sleep quality contributes to inflammation in BPH. Inflammation leads to prostate enlargement in patients with BPH.


Subject(s)
C-Reactive Protein/metabolism , Interleukin-23 Subunit p19/metabolism , Matrix Metalloproteinase 9/metabolism , Prostatic Hyperplasia , Prostatic Neoplasms , Serum Amyloid P-Component/metabolism , Humans , Inflammation/complications , Interleukin-23 , Male , Prostate , Prostatic Hyperplasia/complications , Sleep Quality , Tissue Inhibitor of Metalloproteinase-1
16.
Int J Mol Sci ; 22(23)2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34884474

ABSTRACT

Interleukin (IL) 23 (p19/p40) plays a critical role in the pathogenesis of psoriasis and is upregulated in psoriasis skin lesions. In clinical practice, anti-IL-23Ap19 antibodies are highly effective against psoriasis. IL-39 (p19/ Epstein-Barr virus-induced (EBI) 3), a newly discovered cytokine in 2015, shares the p19 subunit with IL-23. Anti-IL-23Ap19 antibodies may bind to IL-39; also, the cytokine may contribute to the pathogenesis of psoriasis. To investigate IL23Ap19- and/or EBI3-including cytokines in psoriatic keratinocytes, we analyzed IL-23Ap19 and EBI3 expressions in psoriasis skin lesions, using immunohistochemistry and normal human epidermal keratinocytes (NHEKs) stimulated with inflammatory cytokines, using quantitative real-time polymerase chain reaction (RT-PCR), enzyme-linked immunosorbent assay (ELISA), and liquid chromatography-electrospray tandem mass spectrometry (LC-Ms/Ms). Immunohistochemical analysis showed that IL-23Ap19 and EBI3 expressions were upregulated in the psoriasis skin lesions. In vitro, these expressions were synergistically induced by the triple combination of tumor necrosis factor (TNF)-α, IL-17A, and interferon (IFN)-γ, and suppressed by dexamethasone, vitamin D3, and acitretin. In ELISA and LC-Ms/Ms analyses, keratinocyte-derived IL-23Ap19 and EBI3, but not heterodimeric forms, were detected with humanized anti-IL-23Ap19 monoclonal antibodies, tildrakizumab, and anti-EBI3 antibodies, respectively. Psoriatic keratinocytes may express IL-23Ap19 and EBI3 proteins in a monomer or homopolymer, such as homodimer or homotrimer.


Subject(s)
Interleukin-23 Subunit p19/metabolism , Interleukins/metabolism , Minor Histocompatibility Antigens/metabolism , Psoriasis/immunology , Up-Regulation , Antibodies, Monoclonal, Humanized/pharmacology , Cell Line , Chromatography, Liquid , Cytokines/genetics , Cytokines/metabolism , Humans , Interleukin-23 Subunit p19/genetics , Interleukins/genetics , Keratinocytes/immunology , Minor Histocompatibility Antigens/genetics , Psoriasis/genetics , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Up-Regulation/drug effects
17.
Int J Mol Sci ; 22(21)2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34769069

ABSTRACT

C-reactive protein (CRP) is an acute-phase protein in humans that is produced in high quantities by the liver upon infection and under inflammatory conditions. Although CRP is commonly used as a marker of inflammation, CRP can also directly contribute to inflammation by eliciting pro-inflammatory cytokine production by immune cells. Since CRP is highly elevated in serum under inflammatory conditions, we have studied the CRP-induced cytokine profile of human monocytes, one of the main innate immune cell populations in blood. We identified that CRP is relatively unique in its capacity to induce production of the pro-inflammatory cytokine IL-23, which was in stark contrast to a wide panel of pattern recognition receptor (PRR) ligands. We show that CRP-induced IL-23 production was mediated at the level of gene transcription, since CRP particularly promoted gene transcription of IL23A (encoding IL-23p19) instead of IL12A (encoding IL-12p35), while PRR ligands induce the opposite response. Interestingly, when CRP stimulation was combined with PRR ligand stimulation, as for example, occurs in the context of sepsis, IL-23 production by monocytes was strongly reduced. Combined, these data identify CRP as a unique individual ligand to induce IL-23 production by monocytes, which may contribute to shaping systemic immune responses under inflammatory conditions.


Subject(s)
C-Reactive Protein/metabolism , Interleukin-23 Subunit p19/metabolism , Monocytes/metabolism , Cells, Cultured , Humans , Interleukin-23 Subunit p19/genetics , RNA, Messenger/genetics , Transcriptional Activation
18.
J Exp Med ; 218(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34279540

ABSTRACT

Inflammatory skin diseases including atopic dermatitis (AD) and psoriasis (PSO) are underpinned by dendritic cell (DC)-mediated T cell responses. Currently, the heterogeneous human cutaneous DC population is incompletely characterized, and its contribution to these diseases remains unclear. Here, we performed index-sorted single-cell flow cytometry and RNA sequencing of lesional and nonlesional AD and PSO skin to identify macrophages and all DC subsets, including the newly described mature LAMP3+BIRC3+ DCs enriched in immunoregulatory molecules (mregDC) and CD14+ DC3. By integrating our indexed data with published skin datasets, we generated a myeloid cell universe of DC and macrophage subsets in healthy and diseased skin. Importantly, we found that CD14+ DC3s increased in PSO lesional skin and co-produced IL1B and IL23A, which are pathological in PSO. Our study comprehensively describes the molecular characteristics of macrophages and DC subsets in AD and PSO at single-cell resolution, and identifies CD14+ DC3s as potential promoters of inflammation in PSO.


Subject(s)
Dermatitis, Atopic/pathology , Interleukin-1beta/metabolism , Interleukin-23 Subunit p19/metabolism , Langerhans Cells/pathology , Psoriasis/pathology , Dermatitis, Atopic/metabolism , Gene Expression , Gene Regulatory Networks , Humans , Interleukin-15/metabolism , Langerhans Cells/metabolism , Lipopolysaccharide Receptors/metabolism , Macrophages/cytology , Psoriasis/metabolism , Single-Cell Analysis
19.
Front Immunol ; 12: 622770, 2021.
Article in English | MEDLINE | ID: mdl-34149686

ABSTRACT

The term spondyloarthritis (SpA) encompasses a heterogeneous group of inflammatory musculoskeletal diseases with several common genetic background and clinical features, including the possible involvement of the axial skeleton with peripheral mono- or oligo- arthritis and frequently coexisting skin, eye and intestinal manifestations. When the sacroiliac joints or other parts of the spine or thoracic wall are predominantly affected at magnetic resonance or X-ray imaging with inflammatory back pain, the disease is classified as axial SpA and the therapeutic choices are significantly different compared to cases of peripheral arthritis. Moving from the narrow effectiveness and safety profiles of non-steroidal anti-inflammatory drugs, there has been a significant research effort aimed at identifying new treatments based on our better understanding of the pathogenesis of SpA. Indeed, in parallel with the solid data demonstrating that IL-17 and IL-23 are key cytokines in the development of enthesitis and spondylitis, monoclonal antibodies interfering with this pathway have been developed for the treatment of axial SpA. Furthermore, the IL-17/IL-23 axis is key to extra-articular manifestations such as inflammatory bowel disease, uveitis, and psoriasis which are frequent comorbidities of SpA. Currently available drugs act through these mechanisms recognizing IL-23 and targeting IL-17, such as secukinumab and ixekizumab. These therapeutic approaches are now envisioned in the international treatment recommendations for psoriatic arthritis with an axial phenotype as well as for ankylosing spondylitis (AS). We will provide herein a concise comprehensive overview of the clinical evidence supporting the use of these and other drugs acting on IL-23 and IL-17 in axial SpA.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Clinical Trials as Topic , Interleukin-17/antagonists & inhibitors , Interleukin-23 Subunit p19/antagonists & inhibitors , Spondylarthritis/drug therapy , Anti-Inflammatory Agents/adverse effects , Antibodies, Monoclonal/adverse effects , Evidence-Based Medicine , Humans , Interleukin-17/metabolism , Interleukin-23 Subunit p19/metabolism , Signal Transduction , Spondylarthritis/diagnosis , Spondylarthritis/immunology , Spondylarthritis/metabolism , Treatment Outcome
20.
Cell Prolif ; 54(5): e13024, 2021 May.
Article in English | MEDLINE | ID: mdl-33751722

ABSTRACT

OBJECTIVES: Guillain-Barré syndrome (GBS) results from autoimmune attack on the peripheral nerves, causing sensory, motor and autonomic abnormalities. Emerging evidence suggests that there might be an association between COVID-19 and GBS. Nevertheless, the underlying pathophysiological mechanism remains unclear. MATERIALS AND METHODS: We performed bioinformatic analyses to delineate the potential genetic crosstalk between COVID-19 and GBS. RESULTS: COVID-19 and GBS were associated with a similar subset of immune/inflammation regulatory genes, including TNF, CSF2, IL2RA, IL1B, IL4, IL6 and IL10. Protein-protein interaction network analysis revealed that the combined gene set showed an increased connectivity as compared to COVID-19 or GBS alone, particularly the potentiated interactions with CD86, IL23A, IL27, ISG20, PTGS2, HLA-DRB1, HLA-DQB1 and ITGAM, and these genes are related to Th17 cell differentiation. Transcriptome analysis of peripheral blood mononuclear cells from patients with COVID-19 and GBS further demonstrated the activation of interleukin-17 signalling in both conditions. CONCLUSIONS: Augmented Th17 cell differentiation and cytokine response was identified in both COVID-19 and GBS. PBMC transcriptome analysis also suggested the pivotal involvement of Th17 signalling pathway. In conclusion, our data suggested aberrant Th17 cell differentiation as a possible mechanism by which COVID-19 can increase the risk of GBS.


Subject(s)
COVID-19/pathology , Computational Biology/methods , Cytokines/metabolism , Guillain-Barre Syndrome/pathology , Th17 Cells/immunology , B7-2 Antigen/metabolism , COVID-19/complications , COVID-19/virology , Cell Differentiation , Gene Regulatory Networks , Guillain-Barre Syndrome/etiology , Guillain-Barre Syndrome/metabolism , Humans , Interleukin-23 Subunit p19/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Protein Interaction Maps , SARS-CoV-2/isolation & purification , Signal Transduction , Th17 Cells/cytology , Th17 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...