Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.891
Filter
1.
Sci Rep ; 14(1): 18879, 2024 08 14.
Article in English | MEDLINE | ID: mdl-39143130

ABSTRACT

Vitamin D deficiency is increasingly common in systemic lupus erythematosus (SLE) patients and is associated with the disease activity and proteinuria. Recently, alterations in metabolism have been recognized as key regulators of SLE pathogenesis. Our objective was to identify differential metabolites in the serum metabolome of SLE with vitamin D deficiency. In this study, serum samples from 31 SLE patients were collected. Levels of 25(OH)D3 were assayed by ELISA. Patients were divided into two groups according to their vitamin D level (20 ng/ml). Untargeted metabolomics were used to study the metabolite profiles in serum by high-performance liquid chromatography-tandem mass spectrometry. Subsequently, we performed metabolomics profiling analysis to identify 52 significantly altered metabolites in vitamin D deficient SLE patients. The area under the curve (AUC) from ROC analyses was calculated to assess the diagnostic potential of each candidate metabolite biomarker. Lipids accounted for 66.67% of the differential metabolites in the serum, highlighted the disruption of lipid metabolism. The 52 differential metabolites were mapped to 27 metabolic pathways, with fat digestion and absorption, as well as lipid metabolism, emerging as the most significant pathways. The AUC of (S)-Oleuropeic acid and 2-Hydroxylinolenic acid during ROC analysis were 0.867 and 0.833, respectively, indicating their promising diagnostic potential. In conclusion, our results revealed vitamin D deficiency alters SLE metabolome, impacting lipid metabolism, and thrown insights into the pathogenesis and diagnosis of SLE.


Subject(s)
Biomarkers , Lupus Erythematosus, Systemic , Metabolomics , Vitamin D Deficiency , Humans , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/metabolism , Vitamin D Deficiency/blood , Vitamin D Deficiency/complications , Female , Adult , Male , Metabolomics/methods , Biomarkers/blood , Metabolome , Middle Aged , Vitamin D/blood , Vitamin D/analogs & derivatives , Chromatography, High Pressure Liquid , Lipid Metabolism , Tandem Mass Spectrometry , ROC Curve
2.
Free Radic Biol Med ; 223: 325-340, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39111584

ABSTRACT

CX-5461, a first-in-class compound, is widely recognized as a selective inhibitor of RNA polymerase I. Recently, it has been reported to possess novel immunosuppressive properties with significant therapeutic effects in transplantation immune rejection. However, the potential use of CX-5461 for Systemic Lupus Erythematosus (SLE) treatment remains unknown. In this study, we elucidated the mechanism underlying the therapeutic efficacy of CX-5461 in lupus. Our findings demonstrated that CX-5461 selectively targets B cells and effectively reduces the proportions of B cells, germinal center B cells, and plasma cells in MRL/MPJ-Faslpr and Resiquimod (R848)-induced lupus mice. Molecular studies revealed that CX-5461 modulates CD36-Acyl-CoA Synthetase Long Chain Family Member 4 (ACSL4)-mediated glycerolipid metabolism in B cells, triggering ferroptosis through the p53- Solute Carrier Family 7 Member 11 (SLC7A11)- Arachidonate 12-Lipoxygenase (ALOX12) pathway, thereby decreasing IgG and Anti-Double-Stranded Deoxyribonucleic Acid (dsDNA) antibody levels and attenuating lupus. Collectively, these results suggest that CX-5461 holds promise as an effective candidate for targeted therapy against lupus.


Subject(s)
B-Lymphocytes , Ferroptosis , Lupus Erythematosus, Systemic , Tumor Suppressor Protein p53 , Animals , Mice , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/genetics , Ferroptosis/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 12-Lipoxygenase/genetics , Female , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Mice, Inbred MRL lpr , Disease Models, Animal , Signal Transduction/drug effects , Humans
4.
PLoS One ; 19(8): e0309303, 2024.
Article in English | MEDLINE | ID: mdl-39208266

ABSTRACT

Immunosuppressive drugs are essential for systemic lupus erythematosus (SLE) treatment, but there are concerns about their toxicity. In this study, Arthrospira platensis was used as a resource for screening of the SLE-related bioactive compounds. To discover the potential compounds, a total of 833 compounds of A. platensis C1 were retrieved from the Spirulina-Proteome Repository (SpirPro) database and by literature mining. We retrieved structures and bioassays of these compounds from PubChem database; and collected approved and potential drugs for SLE treatment from DrugBank and other databases. The result demonstrated that cytidine, desthiobiotin, agmatine, and anthranilic acid, from the alga, has Tanimoto matching scores of 100% with the following drugs: ß-arabinosylcytosine/cytarabine, d-dethiobiotin, agmatine, and anthranilic acid, respectively. The bioassay matching and disease-gene-drug-compound network analysis, using VisANT 4.0 and Cytoscape, revealed 471 SLE-related genes. Among the SLE-related genes, MDM2, TP53, and JAK2 were identified as targets of cytarabine, while PPARG and IL1B were identified as targets of d-dethiobiotin. Binding affinity between the drug ligands and the algal bioactive compound ligands with their corresponding receptors were similarly comparable scores and stable, examined by molecular docking and molecular dynamic simulations, respectively.


Subject(s)
Lupus Erythematosus, Systemic , Spirulina , Spirulina/chemistry , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/metabolism , Humans , Molecular Docking Simulation
5.
RMD Open ; 10(3)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39209371

ABSTRACT

OBJECTIVE: Previous technical limitations prevented the proof of Fcγ-receptor (FcγR)-activation by soluble immune complexes (sICs) in patients. FcγRIIIa (CD16) is a risk factor in rheumatoid arthritis (RA). We aimed at determining the presence of CD16-activating sICs in RA and control diseases. METHODS: Sera from an exploratory cohort (n=50 patients with RA) and a validation cohort (n=106 patients with RA, 20 patients with psoriasis arthritis (PsA), 22 patients with systemic lupus erythematosus (SLE) and 31 healthy controls) were analysed using a new reporter cell assay. Additionally, 26 synovial fluid samples were analysed, including paired serum/synovial samples. RESULTS: For the first time using a reliable and sensitive functional assay, the presence of sICs in RA sera was confirmed. sICs possess an intrinsic capacity to activate CD16 and can be found in both synovial fluid and in blood. In low experimental dilutions, circulating sICs were also detected in a subset of healthy people and in PsA. However, we report a significantly increased frequency of bioactive circulating sICs in RA. While the bioactivity of circulating sICs was low and did not correlate with clinical parameters, synovial sICs were highly bioactive and correlated with serum autoantibody levels. Receiver operator curves indicated that sICs bioactivity in synovial fluid could be used to discriminate immune complex-associated arthritis from non-associated forms. Finally, circulating sICs were more frequently found in SLE than in RA. The degree of CD16 bioactivity showed strong donor-dependent differences, especially in SLE. CONCLUSIONS: RA is characterised by the presence of circulating and synovial sICs that can engage and activate CD16.


Subject(s)
Antigen-Antibody Complex , Arthritis, Rheumatoid , Receptors, IgG , Synovial Fluid , Humans , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/blood , Receptors, IgG/metabolism , Antigen-Antibody Complex/metabolism , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/blood , Male , Female , Middle Aged , Synovial Fluid/immunology , Synovial Fluid/metabolism , Adult , Aged , Biomarkers , Case-Control Studies , Arthritis, Psoriatic/immunology , Arthritis, Psoriatic/metabolism , Synovial Membrane/immunology , Synovial Membrane/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/blood
6.
Nature ; 633(8029): 442-450, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39143217

ABSTRACT

Regulation of neutrophil activation is critical for disease control. Neutrophil extracellular traps (NETs), which are web-like structures composed of DNA and neutrophil-derived proteins, are formed following pro-inflammatory signals; however, if this process is uncontrolled, NETs contribute to disease pathogenesis, exacerbating inflammation and host tissue damage1,2. Here we show that myeloid inhibitory C-type lectin-like (MICL), an inhibitory C-type lectin receptor, directly recognizes DNA in NETs; this interaction is vital to regulate neutrophil activation. Loss or inhibition of MICL functionality leads to uncontrolled NET formation through the ROS-PAD4 pathway and the development of an auto-inflammatory feedback loop. We show that in the context of rheumatoid arthritis, such dysregulation leads to exacerbated pathology in both mouse models and in human patients, where autoantibodies to MICL inhibit key functions of this receptor. Of note, we also detect similarly inhibitory anti-MICL autoantibodies in patients with other diseases linked to aberrant NET formation, including lupus and severe COVID-19. By contrast, dysregulation of NET release is protective during systemic infection with the fungal pathogen Aspergillus fumigatus. Together, we show that the recognition of NETs by MICL represents a fundamental autoregulatory pathway that controls neutrophil activity and NET formation.


Subject(s)
COVID-19 , Extracellular Traps , Neutrophil Activation , Neutrophils , Protein-Arginine Deiminase Type 4 , Reactive Oxygen Species , Extracellular Traps/metabolism , Extracellular Traps/immunology , Humans , Animals , Mice , Neutrophils/immunology , Neutrophils/metabolism , Protein-Arginine Deiminase Type 4/metabolism , COVID-19/immunology , COVID-19/virology , Reactive Oxygen Species/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/metabolism , Autoantibodies/immunology , Female , Lectins, C-Type/metabolism , Lectins, C-Type/immunology , Male , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , DNA/metabolism , DNA/immunology , Aspergillus fumigatus/immunology , Aspergillus fumigatus/pathogenicity , Feedback, Physiological , Disease Models, Animal , Inflammation/immunology , Inflammation/metabolism
7.
Int Immunopharmacol ; 141: 113013, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39213866

ABSTRACT

BACKGROUND: Systemic lupus erythematosus (SLE) is an autoimmune disorder associated with the decrease and functional impairment of regulatory T cells (Tregs). In the current study, we explored the interplay of miR-155 and suppressor of cytokine signaling 1 (SOCS1) in regulating Treg function and stability in SLE. METHODS: Clinical samples from healthy subjects and SLE patients were collected, and a mouse model of SLE was established to profile the expression pattern of miR-155 and SCOS1 in Tregs. Tregs isolated from mouse spleen were stimulated by inflammatory cytokines to confirm involvement of miR-155/SOCS1 axis in dictating Treg stability and function. We also administrated synthetic miR-155 inhibitor in SLE animal model to evaluate the potential effect on rescuing Treg function and alleviating SLE progression. RESULTS: Tregs from SLE patients and SLE-induced mice exhibited a downregulation of SOCS1 and an upregulation of miR-155. In Tregs stimulated by inflammatory cytokines, Nuclear factor kappa B (NF-κB) signaling activation was required for the change of SOCS1 and miR-155 expression. miR-155 served as a negative regulator to dampen SOCS1 expression in inflammation-stimulated Tregs. The transfection of miR-155 mimic impaired the suppressive function and differentiation of Tregs through targeting SOCS1. In contrast, miR-155 inhibition improved Treg function under inflammatory stimulation and alleviated SLE conditions in the mouse model. CONCLUSION: Inflammation-induced miR-155 impairs Treg stability and function in SLE through decreasing SOCS1 expression. Targeting miR-155 might be developed as an intervention to mitigate SLE conditions.


Subject(s)
Lupus Erythematosus, Systemic , MicroRNAs , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein , T-Lymphocytes, Regulatory , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , T-Lymphocytes, Regulatory/immunology , Humans , Mice , Female , Disease Models, Animal , Cytokines/metabolism , NF-kappa B/metabolism , Cells, Cultured , Male , Adult
8.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(3): 331-348, 2024 Mar 28.
Article in English, Chinese | MEDLINE | ID: mdl-38970507

ABSTRACT

OBJECTIVES: Abnormal programmed cell death in immune cells is associated with autoimmune diseases, but the patterns of programmed cell death in systemic lupus erythematosus (SLE) and especially lupus nephritis (LN) remain unclear. This study aims to explore the association between SLE, LN, and immune cell death patterns. METHODS: Bulk RNA sequencing (bulk RNA-seq) and single-cell RNA sequencing (scRNA-seq) data were downloaded from the Gene Expression Omnibus (GEO) database. Bioinformatic analysis was conducted to explore the expression levels of genes related to 3 cell death patterns in peripheral blood mononuclear cells of SLE patients. Key cell subsets involved in the imbalance of cell death patterns were identified through scRNA-seq. Immunofluorescence was used to detect the expression levels of receptor interacting serine/threonine kinase 3 (RIPK3), mixed-lineage kinase domain-like protein (MLKL), phosphorylated MLKL (pMLKL), caspase 1 (CASP1), CD1c molecule (CD1C), C-type lectin domain containing 9A (CLEC9A), and X-C motif chemokine receptor 1 (XCR1) in dendritic cells (DC). scRNA-seq was performed on kidney tissues collected from LN patients and healthy controls (HC) at the Third Xiangya Hospital of Central South University, followed by bioinformatic analysis to identify key cell subsets involved in the imbalance of cell death patterns. Pseudotime analysis and ligand-receptor analysis were used to explore the differentiation direction and cell communication of different DC subsets. Transient transfection was used to transfect RAW264.7 cells with empty plasmid, empty plasmid+dsDNA (HSV-DNA), empty plasmid+200 µmol/L tert-butyl hydroperoxide (TBHP), stimulator of interferon genes (STING) shRNA plasmid, STING shRNA plasmid+dsDNA (HSV-DNA), and STING shRNA plasmid+200 µmol/L TBHP. Annexin V-mCherry and SYTOX Green staining were used to detect cell death in each group. Western blotting was used to detect the activation of CASP1, gasdermin D (GSDMD), RIPK3, and MLKL in each group. RESULTS: Bioinformatic analysis showed an imbalance in 3 cell death patterns in SLE and LN patients: Pro-inflammatory pyroptosis and necroptosis were activated, while anti-inflammatory apoptosis was inhibited. The key cell subsets involved were DC subsets, particularly focusing on CLEC9A+cDC1. Immunofluorescence results showed that the expression levels of RIPK3, MLKL, and CASP1 in DCs were higher in the SLE group compared to the HC group. pMLKL and CASP1 expression levels in renal cDC1 marked by CLEC9A and XCR1 were higher in the LN group than in the HC group. Pseudotime analysis and ligand-receptor analysis suggested that the CLEC9A+cDC1 subset in LN kidney tissues originated from peripheral circulation. Annexin V-mCherry and SYTOX Green staining results showed that the number of dead cells decreased in the STING shRNA transfection group compared to the empty plasmid group in RAW264.7 cells. Western blotting results showed that the activation of CASP1, GSDMD, RIPK3, and MLKL was decreased in the STING shRNA transfection group compared to the empty plasmid group. CONCLUSIONS: This study provides novel insights into the role of CLEC9A+cDC1 in the imbalance of cell death patterns in SLE and LN.


Subject(s)
Dendritic Cells , Lupus Erythematosus, Systemic , Lupus Nephritis , Receptor-Interacting Protein Serine-Threonine Kinases , Humans , Lupus Nephritis/metabolism , Lupus Nephritis/genetics , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Dendritic Cells/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Apoptosis , Protein Kinases/genetics , Protein Kinases/metabolism , Computational Biology , Leukocytes, Mononuclear/metabolism , Sequence Analysis, RNA
9.
Cell Commun Signal ; 22(1): 354, 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-38972975

ABSTRACT

BACKGROUND: Hyperactive neutrophil extracellular traps (NETs) formation plays a crucial role in active severe systemic lupus erythematosus (SLE). However, what triggers the imbalance in dysregulated NETs formation in SLE is elusive. Transfer RNA-derived small RNAs (tsRNAs) are novel non-coding RNAs, which participate in various cellular processes. We explore the role of tsRNAs on NETs formation in SLE. METHODS: We analyzed the levels of NETs DNA and platelet-derived extracellular vesicles (pEVs) from 50 SLE patients and 20 healthy control subjects. The effects of pEVs on NETs formation were evaluated by using immunofluorescence assay and myeloperoxidase-DNA PicoGreen assay. The regulatory mechanism of pEVs on NETs formation and inflammatory cytokines production were investigated using an in vitro cell-based assay. RESULTS: Increased circulating NETs DNA and pEVs were shown in SLE patients and were associated with disease activity (P < 0.005). We demonstrated that SLE patient-derived immune complexes (ICs) induced platelet activation, followed by pEVs release. ICs-triggered NETs formation was significantly enhanced in the presence of pEVs through Toll-like receptor (TLR) 8 activation. Increased levels of tRF-His-GTG-1 in pEVs and neutrophils of SLE patients were associated with disease activity. tRF-His-GTG-1 interacted with TLR8 to prime p47phox phosphorylation in neutrophils, resulting in reactive oxygen species production and NETs formation. Additionally, tRF-His-GTG-1 modulated NF-κB and IRF7 activation in neutrophils upon TLR8 engagement, resulting IL-1ß, IL-8, and interferon-α upregulation, respectively. CONCLUSIONS: The level of tRF-His-GTG-1 was positively correlated with NETs formation in SLE patients; tRF-His-GTG-1 inhibitor could efficiently suppress ICs-triggered NETs formation/hyperactivation, which may become a potential therapeutic target.


Neutrophils and platelets are key members in the immunopathogenesis of SLE. EVs play a key role in intercellular communication. Abnormal NETs formation promotes vascular complications and organ damage in SLE patients. tsRNA is a novel regulatory small non-coding RNA and participates in diverse pathological processes. Herein, we showed that SLE patient-derived ICs activates platelets directly, followed by intracellular tRF-His-GTG-1 upregulation, which is loaded into pEVs. The pEV-carried tRF-His-GTG-1 could interact with TLR8 in neutrophils, followed by activation of the downstream signaling pathway, including p47phox-NOX2-ROS, which causes NETs enhancement, while IRF7 promotes the expression of IFN-α. The tRF-His-GTG-1 inhibitor could suppress efficiently SLE ICs-induced NETs formation and pEVs primed NETs enhancement. This study offers new molecular machinery to explain the association between the platelets-derived tsRNAs, pEVs, and hyperactive NETs formation in lupus. tRF-His-GTG-1 may serve as a potential therapeutic target and help to advance our understanding of tsRNAs in SLE pathogenesis.


Subject(s)
Extracellular Traps , Extracellular Vesicles , Interferon-alpha , Lupus Erythematosus, Systemic , Adult , Female , Humans , Male , Middle Aged , Blood Platelets/metabolism , Extracellular Traps/metabolism , Extracellular Vesicles/metabolism , Interferon-alpha/metabolism , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/genetics , Neutrophils/metabolism , Toll-Like Receptor 8/metabolism , Toll-Like Receptor 8/genetics , RNA, Transfer/chemistry , RNA, Transfer/metabolism
10.
Dokl Biochem Biophys ; 517(1): 228-234, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39002011

ABSTRACT

The pathogenesis of immunoinflammatory rheumatic diseases (IRDs) is based on chronic inflammation, one of the key mechanisms of which may be abnormal activation of macrophages, leading to further disruption of the immune system. OBJECTIVE: . The objective of this study was to evaluate the proinflammatory activation of circulating monocytes in patients with IRDs. MATERIALS AND METHODS: . The study involved 149 participants (53 patients with rheumatoid arthritis (RA), 45 patients with systemic lupus erythematosus (SLE), 34 patients with systemic scleroderma (SSc), and 17 participants without IRDs) 30 to 65 years old. Basal and lipopolysaccharide (LPS)-stimulated secretion of monocytes was studied in a primary culture of monocytes obtained from blood by immunomagnetic separation. Quantitative assessment of the cytokines tumor necrosis factor α (TNF-α), interleukin 1ß (IL-1ß), as well as the chemokine monocyte chemoattractant protein-1 (MCP-1) was carried out in the culture fluid by ELISA. Proinflammatory activation of monocytes was calculated as the ratio of LPS-stimulated and basal secretions. RESULTS: . It was shown that the basal secretion of all studied cytokines was significantly increased in all groups of patients with IRDs, except for the secretion of IL-1ß in the SLE group, compared to the control. LPS-stimulated secretion of TNF-α was increased and MCP-1 was decreased in patients with IRDs compared to the control group; LPS-stimulated IL-1ß secretion only in the SSc group significantly differed from the control group. In the RA group, monocyte activation was reduced for all cytokines compared to the control; in the SLE group, for TNF-α and MCP-1; in the SSc group, for MCP-1. CONCLUSIONS: . The decrease in proinflammatory activation of monocytes in patients with IRDs is due to a high level of basal secretion of cytokines, which can lead to disruption of the adequate immune response in these diseases and is an important link in the pathogenesis of chronic inflammation.


Subject(s)
Inflammation , Monocytes , Humans , Monocytes/immunology , Monocytes/metabolism , Middle Aged , Adult , Female , Male , Inflammation/immunology , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Aged , Chemokine CCL2/metabolism , Arthritis, Rheumatoid/immunology , Rheumatic Diseases/immunology , Tumor Necrosis Factor-alpha/metabolism , Interleukin-1beta/metabolism , Scleroderma, Systemic/immunology , Scleroderma, Systemic/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Cytokines/metabolism
11.
Int J Rheum Dis ; 27(7): e15256, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38982864

ABSTRACT

The cyclic GMP-AMP synthase (cGAS), a prominent intracellular DNA sensor in mammalian cells, controls the innate immune response and the stimulator of interferon genes (STING)-mediated synthesis of pro-inflammatory cytokines, such as type-I interferon (IFN-I). For decades, IFN-I has been hypothesized to be essential in the development of systemic lupus erythematosus (SLE), a chronic multisystem autoimmunity characterized by immune complex (IC) deposition in small vessels. Recent findings revealed that the activation of the cGAS-STING pathway by self-DNA would propagate the autoimmune responses via upregulating IFN-I production in SLE. In this review, we aimed to provide a comprehensive outlook of the role of the cGAS-STING pathway in SLE pathobiology, as well as, a better understanding of current therapeutic opportunities targeting this axis.


Subject(s)
Lupus Erythematosus, Systemic , Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/drug therapy , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , Animals , Autoimmunity , Interferon Type I/metabolism , Interferon Type I/immunology , Molecular Targeted Therapy , Immunity, Innate
12.
JCI Insight ; 9(16)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39042716

ABSTRACT

Loss of NADPH oxidase (NOX2) exacerbates systemic lupus erythematosus (SLE) in mice and humans, but the mechanisms underlying this effect remain unclear. To identify the cell lineages in which NOX2 deficiency drives SLE, we employed conditional KO and chimeric approaches to delete Cybb in several hematopoietic cell lineages of MRL.Faslpr SLE-prone mice. Deletion of Cybb in macrophages/monocytes exacerbated SLE nephritis, though not to the degree observed in the Cybb global KOs. Unexpectedly, the absence of Cybb in B cells resulted in profound glomerulonephritis and interstitial nephritis, rivaling that seen with global deletion. Furthermore, we identified that NOX2 is a key regulator of TLR7, a driver of SLE pathology, both globally and specifically in B cells. This is mediated in part through suppression of TLR7-mediated NF-κB signaling in B cells. Thus, NOX2's immunomodulatory effect in SLE is orchestrated not only by its function in the myeloid compartment, but through a pivotal role in B cells by selectively inhibiting TLR7 signaling.


Subject(s)
B-Lymphocytes , Lupus Erythematosus, Systemic , Macrophages , Mice, Knockout , NADPH Oxidase 2 , Toll-Like Receptor 7 , Animals , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/genetics , Mice , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , Macrophages/immunology , Macrophages/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Signal Transduction , Female , Disease Models, Animal , NF-kappa B/metabolism , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Lupus Nephritis/metabolism , Lupus Nephritis/genetics , Membrane Glycoproteins
13.
Int J Mol Sci ; 25(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39000360

ABSTRACT

Mitochondrial dysfunction has been increasingly recognized as a trigger for systemic lupus erythematosus (SLE). Recent bioinformatics studies have suggested Fam210b as a significant candidate for the classification and therapeutic targeting of SLE. To experimentally prove the role of Fam210b in SLE, we constructed Fam210b knockout (Fam210b-/-) mice using the CRISPR-Cas9 method. We found that approximately 15.68% of Fam210b-/- mice spontaneously developed lupus-like autoimmunity, which was characterized by skin ulcerations, splenomegaly, and an increase in anti-double-stranded DNA (anti-dsDNA) IgG antibodies and anti-nuclear antibodies(ANA). Single-cell sequencing showed that Fam210b was mainly expressed in erythroid cells. Critically, the knockout of Fam210b resulted in abnormal erythrocyte differentiation and development in the spleens of mice. Concurrently, the spleens exhibited an increased number of CD71+ erythroid cells, along with elevated levels of reactive oxygen species (ROS) in the erythrocytes. The co-culture of CD71+ erythroid cells and lymphocytes resulted in lymphocyte activation and promoted dsDNA and IgG production. In summary, Fam210b knockout leads to a low probability of lupus-like symptoms in mice through the overproduction of ROS in CD71+ erythroid cells. Thus, Fam210b reduction may serve as a novel key marker that triggers the development of SLE.


Subject(s)
Lupus Erythematosus, Systemic , Mice, Knockout , Animals , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Reactive Oxygen Species/metabolism , Antibodies, Antinuclear , Mitochondrial Membranes/metabolism , Erythroid Cells/metabolism , Erythroid Cells/pathology , Disease Models, Animal , Immunoglobulin G/metabolism , Mice, Inbred C57BL , Spleen/metabolism , Spleen/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Female
14.
Arch Pharm Res ; 47(7): 632-644, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38977652

ABSTRACT

Systemic lupus erythematosus (SLE) is a common autoimmune disease, and its pathogenesis mainly involves the aberrant activation of B cells through follicular helper T (Tfh) cells to produce pathogenic antibodies, which requires more effective and safe treatment methods. Dihydroartemisinin (DHA) is the main active ingredient of artemisinin and has immunosuppressive effects. In this study, in vitro experiments confirmed that DHA inhibited Tfh cell induction and weakened its auxiliary function in B cell differentiation; furthermore, DHA directly inhibited B cell activation, differentiation, and antibody production. Furthermore, a mouse model of SLE was established, and we confirmed that DHA significantly reduced the symptoms of SLE and lupus nephritis, and decreased serum immunoglobulin (Ig)G, IgM, IgA, and anti-dsDNA levels. Moreover, DHA reduced the frequencies of total Tfh cells, activated Tfh cells, and B cell lymphoma 6, and interleukin (IL)-21 levels in Tfh cells from the spleen and lymph nodes, as well as the levels of B cells, germinal center B cells, and plasma cells in the spleen, lymph nodes, and kidneys. Additionally, DHA inhibited Tfh cells by blocking IL-2-inducible T cell kinase (ITK) signaling and its downstream nuclear factor (NF)-κB, nuclear factor of activated T cell, and activating protein-1 pathways, and directly inhibited B cells by blocking Bruton's tyrosine kinase (BTK) signaling and the downstream NF-κB and Myc pathways. Overall, our results demonstrated that DHA inhibited Tfh cells by blocking ITK signaling and also directly inhibited B cells by blocking BTK signaling. Therefore, reducing the production of pathogenic antibodies might effectively treat SLE.


Subject(s)
Artemisinins , B-Lymphocytes , Lupus Erythematosus, Systemic , Artemisinins/pharmacology , Animals , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Mice , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Female , T Follicular Helper Cells/immunology , T Follicular Helper Cells/drug effects , T Follicular Helper Cells/metabolism , Disease Models, Animal , Cell Differentiation/drug effects
15.
Chin Med J (Engl) ; 137(15): 1777-1789, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38973265

ABSTRACT

ABSTRACT: Regulated cell death (RCD) is a critical physiological process essential in maintaining skin homeostasis. Among the various forms of RCD, ferroptosis stands out due to its distinct features of iron accumulation, lipid peroxidation, and involvement of various inhibitory antioxidant systems. In recent years, an expanding body of research has solidly linked ferroptosis to the emergence of skin disorders. Therefore, understanding the mechanisms underlying ferroptosis in skin diseases is crucial for advancing therapy and prevention strategies. This review commences with a succinct elucidation of the mechanisms that underpin ferroptosis, embarks on a thorough exploration of ferroptosis's role across a spectrum of skin conditions, encompassing melanoma, psoriasis, systemic lupus erythematosus (SLE), vitiligo, and dermatological ailments precipitated by ultraviolet (UV) exposure, and scrutinizes the potential therapeutic benefits of pharmacological interventions aimed at modulating ferroptosis for the amelioration of skin diseases.


Subject(s)
Ferroptosis , Skin Diseases , Ferroptosis/physiology , Humans , Skin Diseases/metabolism , Vitiligo/metabolism , Vitiligo/therapy , Lipid Peroxidation , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/drug therapy , Iron/metabolism , Psoriasis/metabolism
16.
Int J Mol Sci ; 25(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39062968

ABSTRACT

Despite advances in understanding systemic lupus erythematosus (SLE), many challenges remain in unraveling the precise mechanisms behind the disease's development and progression. Recent evidence has questioned the role of programmed cell death protein 1 (PD-1) in suppressing autoreactive CD4+ T cells during autoimmune responses. Research has investigated the potential impacts of PD-1 on various CD4+ T-cell subpopulations, including T follicular helper (Tfh) cells, circulating Tfh (cTfh) cells, and T peripheral helper (Tph) cells, all of which exhibit substantial PD-1 expression and are closely related to several autoimmune disorders, including SLE. This review highlights the complex role of PD-1 in autoimmunity and emphasizes the imperative for further research to elucidate its functions during autoreactive T-cell responses. Additionally, we address the potential of PD-1 and its ligands as possible therapeutic targets in SLE.


Subject(s)
Autoimmunity , Lupus Erythematosus, Systemic , Programmed Cell Death 1 Receptor , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Humans , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism
17.
Mitochondrion ; 78: 101930, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39025320

ABSTRACT

BACKGROUND: Mechanisms behind multiple organ involvement in lupus, is still an enigma for researchers. Mitochondrial dysfunction and oxidative stress are known to be important aspects in lupus etiology however, their role in lupus organ manifestation is yet to be understood. The present study is based on the understanding of interplay between AMPK/PGC-1α/SIRT-1 axis, mitochondrial complexes, and anti-oxidants levels, which might be involved in lupus organ pathology. METHODOLOGY: Pristane-induced Balb/c mice lupus model (PIL) was utilised and evaluation of anti-oxidants, mitochondrial complexes, pro-inflammatory cytokines levels, biochemical parameters were performed by standard procedures. Tissues were studied by haematoxylin and eosin staining followed by immunohistochemistry. The AMPK/PGC-1α/SIRT-1 expression was analysed by using qPCR and flowcytometry. Analysis of reactive oxygen species (ROS) among WBCs was performed by using various dyes (DCFDA, Mitosox, JC-1) on flowcytometry. RESULT: Significant presence of immune complexes (Tissue sections), ANA (Serum), and pro-inflammatory cytokines (plasma), diminished anti-oxidants and altered biochemical parameters depict the altered pathology in PIL which was accompanied by dysregulated mitochondrial complex activity. Differential expression of the AMPK/PGC-1α/SIRT-1 axis was detected in tissue and correlation with mitochondrial and antioxidant activity emerged as negative in PIL group while positive in controls. Close association was observed between ROS, mitochondrial membrane potential, and AMPK/PGC-1α/SIRT-1 axis in WBCs. CONCLUSION: This study concludes that mitochondria play a dual role in lupus organ pathology, contributing to organ damage while also potentially protecting against damage through the regulation of interactions between antioxidants and the AMPK axis expression.


Subject(s)
AMP-Activated Protein Kinases , Lupus Erythematosus, Systemic , Mitochondria , Oxidation-Reduction , Animals , Female , Mice , AMP-Activated Protein Kinases/metabolism , Disease Models, Animal , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Mice, Inbred BALB C , Mitochondria/metabolism , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism , Sirtuin 1/genetics , Terpenes
18.
Nature ; 631(8022): 857-866, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38987586

ABSTRACT

Systemic lupus erythematosus (SLE) is prototypical autoimmune disease driven by pathological T cell-B cell interactions1,2. Expansion of T follicular helper (TFH) and T peripheral helper (TPH) cells, two T cell populations that provide help to B cells, is a prominent feature of SLE3,4. Human TFH and TPH cells characteristically produce high levels of the B cell chemoattractant CXCL13 (refs. 5,6), yet regulation of T cell CXCL13 production and the relationship between CXCL13+ T cells and other T cell states remains unclear. Here, we identify an imbalance in CD4+ T cell phenotypes in patients with SLE, with expansion of PD-1+/ICOS+ CXCL13+ T cells and reduction of CD96hi IL-22+ T cells. Using CRISPR screens, we identify the aryl hydrocarbon receptor (AHR) as a potent negative regulator of CXCL13 production by human CD4+ T cells. Transcriptomic, epigenetic and functional studies demonstrate that AHR coordinates with AP-1 family member JUN to prevent CXCL13+ TPH/TFH cell differentiation and promote an IL-22+ phenotype. Type I interferon, a pathogenic driver of SLE7, opposes AHR and JUN to promote T cell production of CXCL13. These results place CXCL13+ TPH/TFH cells on a polarization axis opposite from T helper 22 (TH22) cells and reveal AHR, JUN and interferon as key regulators of these divergent T cell states.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , CD4-Positive T-Lymphocytes , Chemokine CXCL13 , Interferon Type I , Lupus Erythematosus, Systemic , Proto-Oncogene Proteins c-jun , Receptors, Aryl Hydrocarbon , Female , Humans , Male , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Chemokine CXCL13/metabolism , Epigenomics , Gene Expression Profiling , Interferon Type I/immunology , Interferon Type I/metabolism , Interleukin-22/immunology , Interleukin-22/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/genetics , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Aryl Hydrocarbon/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
19.
Biochem Pharmacol ; 227: 116417, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38996931

ABSTRACT

The major role of bioactive vitamin 1,25-dihydroxyvitamin D3 (1,25(OH)2D or calcitriol) is to maintain the levels of calcium and phosphorus to achieve bone and mineral homeostasis. Dietary intake and adequate natural light exposure are the main contributors to normal vitamin D status. In addition to regulating metabolism, vitamin D exerts various immunomodulatory effects that regulate innate and adaptive immunity through immune effector cells such as monocytes, macrophages, T and B lymphocytes, and natural killer cells and nonimmune cells that express vitamin D receptors. Systemic lupus erythematosus (SLE) is an autoimmune disease with an unknown etiology, and the association between vitamin D and SLE remains incompletely understood. Given that the current treatment for SLE relies heavily on corticosteroids and that SLE patients tend to have low vitamin D status, vitamin D supplementation may help to reduce the dosage of corticosteroids and/or attenuate disease severity. In this review, we address the associations between vitamin D and several clinical aspects of SLE. In addition, the underlying immunomodulatory mechanisms accounting for the potential vitamin D-mediated therapeutic effects are discussed. Finally, several confounding factors in data interpretation and the execution of clinical trials and perspectives targeting vitamin D supplementation in patients with SLE are also addressed.


Subject(s)
Lupus Erythematosus, Systemic , Vitamin D , Humans , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Vitamin D/therapeutic use , Vitamin D/administration & dosage , Animals , Dietary Supplements , Vitamin D Deficiency/drug therapy , Vitamin D Deficiency/immunology , Vitamin D Deficiency/complications , Receptors, Calcitriol/metabolism
20.
Clin Immunol ; 265: 110285, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38880201

ABSTRACT

Effective treatment of systemic lupus erythematosus (SLE) remains an unmet need. Different subsets of macrophages play differential roles in SLE and the modulation of macrophage polarization away from M1 status is beneficial for SLE therapeutics. Given the pathogenic roles of type I interferons (IFN-I) in SLE, this study investigated the effects and mechanisms of a mitochondria localization molecule ubiquitin specific peptidase 18 (USP18) preserving anti-IFN effects and isopeptidase activity on macrophage polarization. After observing USP18 induction in monocytes from SLE patients, we studied mouse bone marrow-derived macrophages and showed that USP18 deficiency increased M1signal (LPS + IFN-γ treatment)-induced macrophage polarization, and the effects involved the induction of glycolysis and mitochondrial respiration and the expression of several glycolysis-associated enzymes and molecules, such as hypoxia-inducible factor-1α. Moreover, the effects on mitochondrial activities, such as mitochondrial DNA release and mitochondrial reactive oxygen species production were observed. In contrast, the overexpression of USP18 inhibited M1signal-mediated and enhanced interleukin-4 (IL-4)-mediated polarization of macrophages and the related cellular events. Moreover, the levels of USP18 mRNA expression showed tendency of correlation with the expression of metabolic enzymes in monocytes from patients with SLE. We thus concluded that by preserving anti-IFN effect and downregulating M1 signaling, promoting USP18 activity may serve as a useful approach for SLE therapeutics.


Subject(s)
Interleukin-4 , Lupus Erythematosus, Systemic , Macrophages , Mitochondria , Ubiquitin Thiolesterase , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Animals , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Mice , Mitochondria/metabolism , Female , Male , Adult , Glycolysis , Mice, Inbred C57BL , Signal Transduction , Reactive Oxygen Species/metabolism , Macrophage Activation/immunology , Interferon-gamma/metabolism , Interferon-gamma/immunology , Lipopolysaccharides/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL