Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.864
Filtrer
1.
Cell Commun Signal ; 22(1): 354, 2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-38972975

RÉSUMÉ

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.


Sujet(s)
Pièges extracellulaires , Vésicules extracellulaires , Interféron alpha , Lupus érythémateux disséminé , Humains , Lupus érythémateux disséminé/métabolisme , Lupus érythémateux disséminé/génétique , Pièges extracellulaires/métabolisme , Vésicules extracellulaires/métabolisme , Femelle , Adulte , Mâle , Interféron alpha/métabolisme , Granulocytes neutrophiles/métabolisme , Adulte d'âge moyen , Récepteur de type Toll-8/métabolisme , Récepteur de type Toll-8/génétique , Plaquettes/métabolisme
2.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(3): 331-348, 2024 Mar 28.
Article de Anglais, Chinois | MEDLINE | ID: mdl-38970507

RÉSUMÉ

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.


Sujet(s)
Cellules dendritiques , Lupus érythémateux disséminé , Glomérulonéphrite lupique , Receptor-Interacting Protein Serine-Threonine Kinases , Humains , Glomérulonéphrite lupique/métabolisme , Glomérulonéphrite lupique/génétique , Lupus érythémateux disséminé/génétique , Lupus érythémateux disséminé/métabolisme , Cellules dendritiques/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Apoptose , Protein kinases/génétique , Protein kinases/métabolisme , Biologie informatique , Agranulocytes/métabolisme , Analyse de séquence d'ARN
3.
Elife ; 132024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38860651

RÉSUMÉ

The autoimmune disease lupus erythematosus (lupus) is characterized by photosensitivity, where even ambient ultraviolet radiation (UVR) exposure can lead to development of inflammatory skin lesions. We have previously shown that Langerhans cells (LCs) limit keratinocyte apoptosis and photosensitivity via a disintegrin and metalloprotease 17 (ADAM17)-mediated release of epidermal growth factor receptor (EGFR) ligands and that LC ADAM17 sheddase activity is reduced in lupus. Here, we sought to understand how the lupus skin environment contributes to LC ADAM17 dysfunction and, in the process, differentiate between effects on LC ADAM17 sheddase function, LC ADAM17 expression, and LC numbers. We show through transcriptomic analysis a shared IFN-rich environment in non-lesional skin across human lupus and three murine models: MRL/lpr, B6.Sle1yaa, and imiquimod (IMQ) mice. IFN-I inhibits LC ADAM17 sheddase activity in murine and human LCs, and IFNAR blockade in lupus model mice restores LC ADAM17 sheddase activity, all without consistent effects on LC ADAM17 protein expression or LC numbers. Anti-IFNAR-mediated LC ADAM17 sheddase function restoration is associated with reduced photosensitive responses that are dependent on EGFR signaling and LC ADAM17. Reactive oxygen species (ROS) is a known mediator of ADAM17 activity; we show that UVR-induced LC ROS production is reduced in lupus model mice, restored by anti-IFNAR, and is cytoplasmic in origin. Our findings suggest that IFN-I promotes photosensitivity at least in part by inhibiting UVR-induced LC ADAM17 sheddase function and raise the possibility that anifrolumab ameliorates lupus skin disease in part by restoring this function. This work provides insight into IFN-I-mediated disease mechanisms, LC regulation, and a potential mechanism of action for anifrolumab in lupus.


Sujet(s)
Protéine ADAM17 , Cellules de Langerhans , Lupus érythémateux disséminé , Peau , Protéine ADAM17/métabolisme , Protéine ADAM17/génétique , Animaux , Humains , Cellules de Langerhans/métabolisme , Souris , Peau/métabolisme , Peau/anatomopathologie , Peau/effets des radiations , Lupus érythémateux disséminé/métabolisme , Rayons ultraviolets/effets indésirables , Femelle , Modèles animaux de maladie humaine , Photodermatoses/métabolisme , Interférons/métabolisme , Souris de lignée MRL lpr
4.
Front Immunol ; 15: 1339680, 2024.
Article de Anglais | MEDLINE | ID: mdl-38881906

RÉSUMÉ

Background: SLE is a complex autoimmune disease with deleterious effects on various organs. Accumulating evidence has shown abnormal vitamin B12 and one-carbon flux contribute to immune dysfunction. Transcobalamin II (TCN2) belongs to the vitamin B12-binding protein family responsible for the cellular uptake of vitamin B12. The role of TCN2 in SLE is still unclear. Methods: We collected clinical information and blood from 51 patients with SLE and 28 healthy controls. RNA sequencing analysis, qPCR, and western blot confirmed the alteration of TCN2 in disease monocytes. The correlation between TCN2 expression and clinical features and serological abnormalities was analyzed. TCN2 heterozygous knockout THP1 cells were used to explore the effects of TCN2 dysfunction on monocytes. CCK-8 assay and EdU staining were used to detect cell proliferation. ELISA was conducted to assess vitamin B12, glutathione, and cytokines changes. UHPLC-MRM-MS/MS was used to detect changes in the intermediates of the one-carbon cycle. Flow cytometry is used to detect cell cycle, ROS, mitoROS, and CD14 changes. Results: Elevated TCN2 in monocytes was correlated positively with disease progression and specific tissue injuries. Using CD14+ monocytes and TCN2 genetically modified THP1 cell lines, we found that the TCN2 was induced by LPS in serum from SLE patients. TCN2 heterozygous knockout inhibited cellular vitamin B12 uptake and one-carbon metabolism, leading to cell proliferation arrest and decreased Toll-like receptor 4 (TLR4)-mediated CCL2 release. Methionine cycle metabolites, s-adenosylmethionine and homocysteine, rescued these effects, whereas folate treatment proved to be ineffective. Folate deficiency also failed to replicate the impact of TCN2 downregulation on THP1 inflammatory response. Conclusion: Our study elucidated the unique involvement of TCN2-driven one-carbon flux on SLE-associated monocyte behavior. Increased TCN2 may promote disease progression and tissue damage by enhancing one-carbon flux, fostering monocyte proliferation, and exacerbating TLR4 mediated inflammatory responses. The inhibition of TCN2 may be a promising therapeutic approach to ameliorate SLE.


Sujet(s)
Prolifération cellulaire , Acide folique , Lupus érythémateux disséminé , Monocytes , Récepteur de type Toll-4 , Transcobalamines , Humains , Récepteur de type Toll-4/métabolisme , Récepteur de type Toll-4/génétique , Lupus érythémateux disséminé/métabolisme , Lupus érythémateux disséminé/immunologie , Monocytes/métabolisme , Monocytes/immunologie , Transcobalamines/métabolisme , Transcobalamines/génétique , Femelle , Acide folique/métabolisme , Mâle , Adulte , Inflammation/métabolisme , Inflammation/immunologie , Adulte d'âge moyen , Cellules THP-1 , Carbone/métabolisme , Vitamine B12/métabolisme , Études cas-témoins
5.
Int J Mol Sci ; 25(11)2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38891798

RÉSUMÉ

Systemic lupus erythematosus (SLE) is a multifactorial autoimmune disease characterized by self-immune tolerance breakdown and the production of autoantibodies, causing the deposition of immune complexes and triggering inflammation and immune-mediated damage. SLE pathogenesis involves genetic predisposition and a combination of environmental factors. Clinical manifestations are variable, making an early diagnosis challenging. Heat shock proteins (Hsps), belonging to the chaperone system, interact with the immune system, acting as pro-inflammatory factors, autoantigens, as well as immune tolerance promoters. Increased levels of some Hsps and the production of autoantibodies against them are correlated with SLE onset and progression. The production of these autoantibodies has been attributed to molecular mimicry, occurring upon viral and bacterial infections, since they are evolutionary highly conserved. Gut microbiota dysbiosis has been associated with the occurrence and severity of SLE. Numerous findings suggest that proteins and metabolites of commensal bacteria can mimic autoantigens, inducing autoimmunity, because of molecular mimicry. Here, we propose that shared epitopes between human Hsps and those of gut commensal bacteria cause the production of anti-Hsp autoantibodies that cross-react with human molecules, contributing to SLE pathogenesis. Thus, the involvement of the chaperone system, gut microbiota dysbiosis, and molecular mimicry in SLE ought to be coordinately studied.


Sujet(s)
Dysbiose , Microbiome gastro-intestinal , Lupus érythémateux disséminé , Mimétisme moléculaire , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/microbiologie , Lupus érythémateux disséminé/métabolisme , Humains , Mimétisme moléculaire/immunologie , Dysbiose/immunologie , Microbiome gastro-intestinal/immunologie , Chaperons moléculaires/métabolisme , Chaperons moléculaires/immunologie , Protéines du choc thermique/immunologie , Protéines du choc thermique/métabolisme , Autoanticorps/immunologie , Animaux , Autoantigènes/immunologie , Autoantigènes/métabolisme , Auto-immunité
6.
Proc Natl Acad Sci U S A ; 121(24): e2322009121, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38843187

RÉSUMÉ

Follicular helper T (TFH) cells mediate germinal center reactions to generate high affinity antibodies against specific pathogens, and their excessive production is associated with the pathogenesis of systemic autoimmune diseases such as systemic lupus erythematosus (SLE). ETV5, a member of the ETS transcription factor family, promotes TFH cell differentiation in mice. In this study, we examined the role of ETV5 in the pathogenesis of lupus in mice and humans. T cell-specific deletion of Etv5 alleles ameliorated TFH cell differentiation and autoimmune phenotypes in lupus mouse models. Further, we identified SPP1 as an ETV5 target that promotes TFH cell differentiation in both mice and humans. Notably, extracellular osteopontin (OPN) encoded by SPP1 enhances TFH cell differentiation by activating the CD44-AKT signaling pathway. Furthermore, ETV5 and SPP1 levels were increased in CD4+ T cells from patients with SLE and were positively correlated with disease activity. Taken together, our findings demonstrate that ETV5 is a lupus-promoting transcription factor, and secreted OPN promotes TFH cell differentiation.


Sujet(s)
Différenciation cellulaire , Lupus érythémateux disséminé , Ostéopontine , Facteurs de transcription , Animaux , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/métabolisme , Lupus érythémateux disséminé/génétique , Lupus érythémateux disséminé/anatomopathologie , Ostéopontine/métabolisme , Ostéopontine/génétique , Souris , Humains , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Lymphocytes T auxiliaires/immunologie , Lymphocytes T auxiliaires/métabolisme , Lymphocytes T auxiliaires folliculaires/immunologie , Lymphocytes T auxiliaires folliculaires/métabolisme , Femelle , Modèles animaux de maladie humaine , Souris knockout
7.
Int J Mol Sci ; 25(11)2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38892349

RÉSUMÉ

Systemic lupus erythematosus (SLE) is an autoimmune disease affecting mostly women of child-bearing age. Immune dysfunction in SLE results from disrupted apoptosis which lead to an unregulated interferon (IFN) stimulation and the production of autoantibodies, leading to immune complex formation, complement activation, and organ damage. Lupus nephritis (LN) is a common and severe complication of SLE, impacting approximately 30% to 40% of SLE patients. Recent studies have demonstrated an alteration in mitochondrial homeostasis in SLE patients. Mitochondrial dysfunction contributes significantly to SLE pathogenesis by enhancing type 1 IFN production through various pathways involving neutrophils, platelets, and T cells. Defective mitophagy, the process of clearing damaged mitochondria, exacerbates this cycle, leading to increased immune dysregulation. In this review, we aim to detail the physiopathological link between mitochondrial dysfunction and disease activity in SLE. Additionally, we will explore the potential role of mitochondria as biomarkers and therapeutic targets in SLE, with a specific focus on LN. In LN, mitochondrial abnormalities are observed in renal cells, correlating with disease progression and renal fibrosis. Studies exploring cell-free mitochondrial DNA as a biomarker in SLE and LN have shown promising but preliminary results, necessitating further validation and standardization. Therapeutically targeting mitochondrial dysfunction in SLE, using drugs like metformin or mTOR inhibitors, shows potential in modulating immune responses and improving clinical outcomes. The interplay between mitochondria, immune dysregulation, and renal involvement in SLE and LN underscores the need for comprehensive research and innovative therapeutic strategies. Understanding mitochondrial dynamics and their impact on immune responses offers promising avenues for developing personalized treatments and non-invasive biomarkers, ultimately improving outcomes for LN patients.


Sujet(s)
Lupus érythémateux disséminé , Glomérulonéphrite lupique , Mitochondries , Humains , Glomérulonéphrite lupique/métabolisme , Glomérulonéphrite lupique/anatomopathologie , Glomérulonéphrite lupique/immunologie , Glomérulonéphrite lupique/étiologie , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Lupus érythémateux disséminé/métabolisme , Lupus érythémateux disséminé/anatomopathologie , Lupus érythémateux disséminé/immunologie , ADN mitochondrial/métabolisme , Animaux , Marqueurs biologiques , Mitophagie
8.
J Autoimmun ; 146: 103245, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38754236

RÉSUMÉ

B cell responses to nucleic acid-containing self-antigens that involve intracellular nucleic acid sensors play a crucial role in autoantibody production in SLE. CD72 is an inhibitory B cell co-receptor that down-regulates BCR signaling, and prevents the development of SLE. We previously showed that CD72 recognizes the RNA-containing self-antigen Sm/RNP, a target of SLE-specific autoantibodies, and induces B cell tolerance to Sm/RNP by specifically inhibiting B cell response to this self-antigen. Here, we address whether CD72 inhibits B cell response to ribosomes because the ribosome is an RNA-containing self-antigen and is a target of SLE-specific autoantibodies as well as Sm/RNP. We demonstrate that CD72 recognizes ribosomes as a ligand, and specifically inhibits BCR signaling induced by ribosomes. Although conventional protein antigens by themselves do not induce proliferation of specific B cells, ribosomes induce proliferation of B cells reactive to ribosomes in a manner dependent on RNA. This proliferative response is down-regulated by CD72. These results suggest that ribosomes activate B cells by inducing dual signaling through BCR and intracellular RNA sensors and that CD72 inhibits B cell response to ribosomes. Moreover, CD72-/- but not CD72+/+ mice spontaneously produce anti-ribosome autoantibodies. Taken together, CD72 induces B cell self-tolerance to ribosomes by recognizing ribosomes and inhibiting RNA-dependent B cell response to this self-antigen. CD72 appears to prevent development of SLE by inhibiting autoimmune B cell responses to multiple RNA-containing self-antigens. Because these self-antigens but not protein self-antigens induce RNA-dependent B cell activation, self-tolerance to RNA-containing self-antigens may require a distinct tolerance mechanism mediated by CD72.


Sujet(s)
Antigènes CD , Antigènes de différenciation des lymphocytes B , Autoanticorps , Autoantigènes , Lymphocytes B , Lupus érythémateux disséminé , Récepteurs pour l'antigène des lymphocytes B , Ribosomes , Transduction du signal , Animaux , Ribosomes/métabolisme , Ribosomes/immunologie , Souris , Récepteurs pour l'antigène des lymphocytes B/métabolisme , Récepteurs pour l'antigène des lymphocytes B/immunologie , Autoanticorps/immunologie , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/métabolisme , Antigènes de différenciation des lymphocytes B/immunologie , Antigènes de différenciation des lymphocytes B/métabolisme , Antigènes CD/métabolisme , Antigènes CD/immunologie , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Transduction du signal/immunologie , Autoantigènes/immunologie , Souris knockout , Activation des lymphocytes/immunologie , Prolifération cellulaire , Tolérance immunitaire , Humains
9.
Front Immunol ; 15: 1371708, 2024.
Article de Anglais | MEDLINE | ID: mdl-38756769

RÉSUMÉ

Impaired metabolism is recognized as an important contributor to pathogenicity of T cells in Systemic Lupus Erythematosus (SLE). Over the last two decades, we have acquired significant knowledge about the signaling and transcriptomic programs related to metabolic rewiring in healthy and SLE T cells. However, our understanding of metabolic network activity derives largely from studying metabolic pathways in isolation. Here, we argue that enzymatic activities are necessarily coupled through mass and energy balance constraints with in-built network-wide dependencies and compensation mechanisms. Therefore, metabolic rewiring of T cells in SLE must be understood in the context of the entire network, including changes in metabolic demands such as shifts in biomass composition and cytokine secretion rates as well as changes in uptake/excretion rates of multiple nutrients and waste products. As a way forward, we suggest cell physiology experiments and integration of orthogonal metabolic measurements through computational modeling towards a comprehensive understanding of T cell metabolism in lupus.


Sujet(s)
Lupus érythémateux disséminé , Lymphocytes T , Lupus érythémateux disséminé/métabolisme , Lupus érythémateux disséminé/immunologie , Humains , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Voies et réseaux métaboliques , Métabolisme énergétique , Animaux , Transduction du signal , Cytokines/métabolisme
10.
Clin Immunol ; 264: 110243, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38735509

RÉSUMÉ

OBJECTIVE: To link changes in the B-cell transcriptome from systemic lupus erythematosus (SLE) patients with those in their macroenvironment, including cellular and fluidic components. METHODS: Analysis was performed on 363 patients and 508 controls, encompassing transcriptomics, metabolomics, and clinical data. B-cell and whole-blood transcriptomes were analysed using DESeq and GSEA. Plasma and urine metabolomics peak changes were quantified and annotated using Ceu Mass Mediator database. Common sources of variation were identified using MOFA integration analysis. RESULTS: Cellular macroenvironment was enriched in cytokines, stress responses, lipidic synthesis/mobility pathways and nucleotide degradation. B cells shared these pathways, except nucleotide degradation diverted to nucleotide salvage pathway, and distinct glycosylation, LPA receptors and Schlafen proteins. CONCLUSIONS: B cells showed metabolic changes shared with their macroenvironment and unique changes directly or indirectly induced by IFN-α signalling. This study underscores the importance of understanding the interplay between B cells and their macroenvironment in SLE pathology.


Sujet(s)
Lymphocytes B , Lupus érythémateux disséminé , Métabolomique , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/métabolisme , Humains , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Femelle , Adulte , Mâle , Transcriptome , Adulte d'âge moyen , Analyse de profil d'expression de gènes , Multi-omique
11.
Front Immunol ; 15: 1380481, 2024.
Article de Anglais | MEDLINE | ID: mdl-38774868

RÉSUMÉ

Objectives: Cell surface glycosylation can influence protein-protein interactions with particular relevance to changes in core fucosylation and terminal sialylation. Glycans are ligands for immune regulatory lectin families like galectins (Gals) or sialic acid immunoglobulin-like lectins (Siglecs). This study delves into the glycan alterations within immune subsets of systemic lupus erythematosus (SLE). Methods: Evaluation of binding affinities of Galectin-1, Galectin-3, Siglec-1, Aleuria aurantia lectin (AAL, recognizing core fucosylation), and Sambucus nigra agglutinin (SNA, specific for α-2,6-sialylation) was conducted on various immune subsets in peripheral blood mononuclear cells (PBMCs) from control and SLE subjects. Lectin binding was measured by multi-parameter flow cytometry in 18 manually gated subsets of T-cells, NK-cells, NKT-cells, B-cells, and monocytes in unstimulated resting state and also after 3-day activation. Stimulated pre-gated populations were subsequently clustered by FlowSOM algorithm based on lectin binding and activation markers, CD25 or HLA-DR. Results: Elevated AAL, SNA and CD25+/CD25- SNA binding ratio in certain stimulated SLE T-cell subsets correlated with SLE Disease Activity Index 2000 (SLEDAI-2K) scores. The significantly increased frequencies of activated AALlow Siglec-1low NK metaclusters in SLE also correlated with SLEDAI-2K indices. In SLE, activated double negative NKTs displayed significantly lower core fucosylation and CD25+/CD25- Siglec-1 binding ratio, negatively correlating with disease activity. The significantly enhanced AAL binding in resting SLE plasmablasts positively correlated with SLEDAI-2K scores. Conclusion: Alterations in the glycosylation of immune cells in SLE correlate with disease severity, which might represent potential implications in the pathogenesis of SLE.


Sujet(s)
Cytométrie en flux , Lectines , Lupus érythémateux disséminé , Humains , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/métabolisme , Cytométrie en flux/méthodes , Adulte , Femelle , Mâle , Adulte d'âge moyen , Lectines/métabolisme , Lectines/immunologie , Liaison aux protéines , Agranulocytes/immunologie , Agranulocytes/métabolisme , Glycosylation , Galectines/métabolisme , Galectines/immunologie , Jeune adulte , Indice de gravité de la maladie
12.
Front Immunol ; 15: 1383358, 2024.
Article de Anglais | MEDLINE | ID: mdl-38779657

RÉSUMÉ

Introduction: Immune cells that contribute to the pathogenesis of systemic lupus erythematosus (SLE) derive from adult hematopoietic stem and progenitor cells (HSPCs) within the bone marrow (BM). For this reason, we reasoned that fundamental abnormalities in SLE can be traced to a BM-derived HSPC inflammatory signature. Methods: BM samples from four SLE patients, six healthy controls, and two umbilical cord blood (CB) samples were used. CD34+ cells were isolated from BM and CB samples, and single-cell RNA-sequencing was performed. Results: A total of 426 cells and 24,473 genes were used in the analysis. Clustering analysis resulted in seven distinct clusters of cell types. Mutually exclusive markers, which were characteristic of each cell type, were identified. We identified three HSPC subpopulations, one of which consisted of proliferating cells (MKI67 expressing cells), one T-like, one B-like, and two myeloid-like progenitor subpopulations. Differential expression analysis revealed i) cell cycle-associated signatures, in healthy BM of HSPC clusters 3 and 4 when compared with CB, and ii) interferon (IFN) signatures in SLE BM of HSPC clusters 3 and 4 and myeloid-like progenitor cluster 5 when compared with healthy controls. The IFN signature in SLE appeared to be deregulated following TF regulatory network analysis and differential alternative splicing analysis between SLE and healthy controls in HSPC subpopulations. Discussion: This study revealed both quantitative-as evidenced by decreased numbers of non-proliferating early progenitors-and qualitative differences-characterized by an IFN signature in SLE, which is known to drive loss of function and depletion of HSPCs. Chronic IFN exposure affects early hematopoietic progenitors in SLE, which may account for the immune aberrancies and the cytopenias in SLE.


Sujet(s)
Analyse de profil d'expression de gènes , Cellules souches hématopoïétiques , Interférons , Lupus érythémateux disséminé , Analyse sur cellule unique , Transcriptome , Humains , Lupus érythémateux disséminé/génétique , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/métabolisme , Cellules souches hématopoïétiques/métabolisme , Interférons/métabolisme , Interférons/génétique , Femelle , Adulte , Reprogrammation cellulaire/génétique , Mâle
13.
Int J Mol Sci ; 25(10)2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38791389

RÉSUMÉ

The pathogenesis of systemic lupus erythematosus (SLE) is linked to the differential roles of toll-like receptors (TLRs), particularly TLR7, TLR8, and TLR9. TLR7 overexpression or gene duplication, as seen with the Y-linked autoimmune accelerator (Yaa) locus or TLR7 agonist imiquimod, correlates with increased SLE severity, and specific TLR7 polymorphisms and gain-of-function variants are associated with enhanced SLE susceptibility and severity. In addition, the X-chromosome location of TLR7 and its escape from X-chromosome inactivation provide a genetic basis for female predominance in SLE. The absence of TLR8 and TLR9 have been shown to exacerbate the detrimental effects of TLR7, leading to upregulated TLR7 activity and increased disease severity in mouse models of SLE. The regulatory functions of TLR8 and TLR9 have been proposed to involve competition for the endosomal trafficking chaperone UNC93B1. However, recent evidence implies more direct, regulatory functions of TLR9 on TLR7 activity. The association between age-associated B cells (ABCs) and autoantibody production positions these cells as potential targets for treatment in SLE, but the lack of specific markers necessitates further research for precise therapeutic intervention. Therapeutically, targeting TLRs is a promising strategy for SLE treatment, with drugs like hydroxychloroquine already in clinical use.


Sujet(s)
Lupus érythémateux disséminé , Récepteur de type Toll-7 , Lupus érythémateux disséminé/génétique , Lupus érythémateux disséminé/métabolisme , Animaux , Humains , Souris , Récepteur de type Toll-7/métabolisme , Récepteur de type Toll-7/génétique , Récepteurs de type Toll/métabolisme , Récepteur de type Toll-8/métabolisme , Récepteur de type Toll-8/génétique , Modèles animaux de maladie humaine , Prédisposition génétique à une maladie
14.
Int Immunopharmacol ; 134: 112149, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38692019

RÉSUMÉ

Systemic lupus erythematosus (SLE) is a severe autoimmune disease with significant socioeconomic impact worldwide. Orderly energy metabolism is essential for normal immune function, and disordered energy metabolism is increasingly recognized as an important contributor to the pathogenesis of SLE. Disorders of energy metabolism are characterized by increased reactive oxygen species, ATP deficiency, and abnormal metabolic pathways. Oxygen and mitochondria are critical for the production of ATP, and both mitochondrial dysfunction and hypoxia affect the energy production processes. In addition, several signaling pathways, including mammalian target of rapamycin (mTOR)/adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) signaling and the hypoxia-inducible factor (HIF) pathway also play important regulatory roles in energy metabolism. Furthermore, drugs with clear clinical effects on SLE, such as sirolimus, metformin, and tacrolimus, have been proven to improve the disordered energy metabolism of immune cells, suggesting the potential of targeting energy metabolism for the treatment of SLE. Moreover, several metabolic modulators under investigation are expected to have potential therapeutic effects in SLE. This review aimed to gain insights into the role and mechanism of abnormal energy metabolism in the pathogenesis of SLE, and summarizes the progression of metabolic modulator in the treatment of SLE.


Sujet(s)
Métabolisme énergétique , Lupus érythémateux disséminé , Lupus érythémateux disséminé/métabolisme , Lupus érythémateux disséminé/immunologie , Humains , Animaux , Mitochondries/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Transduction du signal , AMP-Activated Protein Kinases/métabolisme , Espèces réactives de l'oxygène/métabolisme , Adénosine triphosphate/métabolisme
15.
Cell Rep ; 43(6): 114248, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38795350

RÉSUMÉ

Cyclic GMP-AMP synthase (cGAS) undergoes liquid-liquid phase separation (LLPS) to trigger downstream signaling upon double-stranded DNA (dsDNA) stimulation, and the condensed cGAS colocalizes with stress granules (SGs). However, the molecular mechanism underlying the modulation of cGAS activation by SGs remains elusive. In this study, we show that USP8 is localized to SGs upon dsDNA stimulation and potentiates cGAS-stimulator of interferon genes (STING) signaling. A USP8 inhibitor ameliorates pathological inflammation in Trex1-/- mice. Systemic lupus erythematosus (SLE) databases indicate a positive correlation between USP8 expression and SLE. Mechanistic study shows that the SG protein DDX3X promotes cGAS phase separation and activation in a manner dependent on its intrinsic LLPS. USP8 cleaves K27-linked ubiquitin chains from the intrinsically disordered region (IDR) of DDX3X to enhance its condensation. In conclusion, we demonstrate that USP8 catalyzes the deubiquitination of DDX3X to facilitate cGAS condensation and activation and that inhibiting USP8 is a promising strategy for alleviating cGAS-mediated autoimmune diseases.


Sujet(s)
DEAD-box RNA helicases , Interféron de type I , Nucleotidyltransferases , Granules de stress , Ubiquitin thiolesterase , Ubiquitination , Humains , Animaux , Nucleotidyltransferases/métabolisme , Ubiquitin thiolesterase/métabolisme , Souris , DEAD-box RNA helicases/métabolisme , Interféron de type I/métabolisme , Granules de stress/métabolisme , Lupus érythémateux disséminé/métabolisme , Lupus érythémateux disséminé/anatomopathologie , Transduction du signal , Souris de lignée C57BL , Cellules HEK293 , Protéines membranaires/métabolisme , Souris knockout , Exodeoxyribonucleases/métabolisme , Endopeptidases , Phosphoprotéines , Complexes de tri endosomique requis pour le transport
16.
J Autoimmun ; 146: 103221, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38643728

RÉSUMÉ

Inflammatory T cells contribute to the pathogenesis of autoimmune diseases such as systemic lupus erythematosus (SLE). Analysis of the T-cell transcriptomics data of two independent SLE patient cohorts by three machine learning models revealed the pseudogene UHRF1P as a novel SLE biomarker. The pseudogene-encoded UHRF1P protein was overexpressed in peripheral blood T cells of SLE patients. The UHRF1P protein lacks the amino-terminus of its parental UHRF1 protein, resulting in missing the proteasome-binding ubiquitin-like (Ubl) domain of UHRF1. T-cell-specific UHRF1P transgenic mice manifested the induction of IL-17A and autoimmune inflammation. Mechanistically, UHFR1P prevented UHRF1-induced Lys48-linked ubiquitination and degradation of MAP4K3 (GLK), which is a kinase known to induce IL-17A. Consistently, IL-17A induction and autoimmune phenotypes of UHRF1P transgenic mice were obliterated by MAP4K3 knockout. Collectively, UHRF1P overexpression in T cells inhibits the E3 ligase function of its parental UHRF1 and induces autoimmune diseases.


Sujet(s)
Protéines liant les séquences stimulatrices de type CCAAT , Interleukine-17 , Lupus érythémateux disséminé , Souris transgéniques , Protein-Serine-Threonine Kinases , Ubiquitin-protein ligases , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/génétique , Lupus érythémateux disséminé/métabolisme , Animaux , Interleukine-17/métabolisme , Interleukine-17/génétique , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Humains , Souris , Protéines liant les séquences stimulatrices de type CCAAT/métabolisme , Protéines liant les séquences stimulatrices de type CCAAT/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Ubiquitination , Souris knockout , Modèles animaux de maladie humaine , Transduction du signal , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Auto-immunité , Femelle
17.
Adv Sci (Weinh) ; 11(22): e2400446, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38639386

RÉSUMÉ

Despite accumulating evidence linking defective lysosome function with autoimmune diseases, how the catabolic machinery is regulated to maintain immune homeostasis remains unknown. Late endosomal/lysosomal adaptor, MAPK and mTOR activator 5 (Lamtor5) is a subunit of the Ragulator mediating mechanistic target of rapamycin complex 1 (mTORC1) activation in response to amino acids, but its action mode and physiological role are still unclear. Here it is demonstrated that Lamtor5 level is markedly decreased in peripheral blood mononuclear cells (PBMCs) of patients with systemic lupus erythematosus (SLE). In parallel, the mice with myeloid Lamtor5 ablation developed SLE-like manifestation. Impaired lysosomal function and aberrant activation of mTORC1 are evidenced in Lamtor5 deficient macrophages and PBMCs of SLE patients, accompanied by blunted autolysosomal pathway and undesirable inflammatory responses. Mechanistically, it is shown that Lamtor5 is physically associated with ATP6V1A, an essential subunit of vacuolar H+-ATPase (v-ATPase), and promoted the V0/V1 holoenzyme assembly to facilitate lysosome acidification. The binding of Lamtor5 to v-ATPase affected the lysosomal tethering of Rag GTPase and weakened its interaction with mTORC1 for activation. Overall, Lamtor5 is identified as a critical factor for immune homeostasis by intergrading v-ATPase activity, lysosome function, and mTOR pathway. The findings provide a potential therapeutic target for SLE and/or other autoimmune diseases.


Sujet(s)
Auto-immunité , Lupus érythémateux disséminé , Lysosomes , Vacuolar Proton-Translocating ATPases , Animaux , Femelle , Humains , Souris , Auto-immunité/immunologie , Auto-immunité/génétique , Modèles animaux de maladie humaine , Agranulocytes/métabolisme , Agranulocytes/immunologie , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/génétique , Lupus érythémateux disséminé/métabolisme , Lysosomes/métabolisme , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexe-1 cible mécanistique de la rapamycine/génétique , Vacuolar Proton-Translocating ATPases/métabolisme , Vacuolar Proton-Translocating ATPases/génétique , Vacuolar Proton-Translocating ATPases/immunologie
18.
Clin Immunol ; 263: 110224, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38648959

RÉSUMÉ

The pathophysiology of systemic lupus erythematosus (SLE) is multifactorial and involves alterations in metabolic pathways, including glycolysis, lipid metabolism, amino acid metabolism, and mitochondrial dysfunction. Increased glycolysis in SLE T cells, which is associated with elevated glucose transporter 1 expression, suggests targeting glucose transporters and hexokinase as potential treatments. Abnormalities in lipid metabolism, particularly in lipid rafts and enzymes, present new therapeutic targets. This review discusses how changes in glutaminolysis and tryptophan metabolism affect T-cell function, suggesting new therapeutic interventions, as well as mitochondrial dysfunction in SLE, which increases reactive oxygen species. The review also emphasizes that modulating metabolic pathways in immune cells is a promising approach for SLE treatment, and can facilitate personalized therapies based on individual metabolic profiles of patients with SLE. The review provides novel insights into strategies for managing SLE.


Sujet(s)
Lupus érythémateux disséminé , Voies et réseaux métaboliques , Humains , Lupus érythémateux disséminé/métabolisme , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/traitement médicamenteux , Voies et réseaux métaboliques/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Métabolisme lipidique/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Tryptophane/métabolisme , Animaux , Glycolyse/effets des médicaments et des substances chimiques
19.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(3): 465-473, 2024 Mar 20.
Article de Chinois | MEDLINE | ID: mdl-38597437

RÉSUMÉ

OBJECTIVE: To explore the therapeutic mechanism of Jianpi Zishen (JPZS) granules for systemic lupus erythematosus(SLE) in light of podocyte autophagy regulation. METHODS: TCMSP, GeneCards, OMIM, and TTD databases were used to obtain the targets of JPZS granules, SLE, and podocyte autophagy. The protein-protein interaction network was constructed using Cytoscape, and the key active ingredients and targets were screened for molecular docking. In the clinical study, 46 patients with SLE were randomized into two groups to receive baseline treatment with prednisone acetate and mycophenolate mofetil (control group) and additional treatment with JPZS granules (observation group) for 12 weeks, with 10 healthy volunteers as the healthy control group. Urinary levels of nephrin and synaptopodin of the patients were detected with ELISA. Western blotting was performed to determine peripheral blood levels of p-JAK1/JAK1, p-STAT1/STAT1, LC3II/LC3I, and p62 proteins of the participants. RESULTS: Four key active ingredients and 5 core target genes (STAT1, PIK3CG, MAPK1, PRKCA, and CJA1) were obtained, and enrichment analysis identified the potentially involved signaling pathways including AGE-RAGE, JAK/STAT, EGFR, and PI3K/Akt. Molecular docking analysis showed that STAT1 was the most promising target protein with the highest binding activity, suggesting its role as an important mediator for signal transduction after JPZS granule treatment. In the 43 SLE patients available for analysis, treatment with JPZS granule significantly reduced serum levels of p-JAK1/JAK1, p-STAT1/STAT1, and LC3II/LC3I (P < 0.05 or 0.01), increased the protein level of P62 (P < 0.05), and reduced urinary levels of nephrin and synaptopodin (P < 0.05). CONCLUSION: The therapeutic effect of JPZS granules on SLE is mediated probably by coordinated actions of quercetin, kaempferol, ß-sitosterol, and isorhamnetin on their target gene STAT1 to inhibit the JAK/STAT pathway, thus suppressing autophagy and alleviating podocyte injuries in SLE.


Sujet(s)
Médicaments issus de plantes chinoises , Lupus érythémateux disséminé , Podocytes , Humains , Autophagie , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/usage thérapeutique , Janus kinases/métabolisme , Lupus érythémateux disséminé/traitement médicamenteux , Lupus érythémateux disséminé/métabolisme , Simulation de docking moléculaire , Pharmacologie des réseaux , Phosphatidylinositol 3-kinases/métabolisme , Podocytes/métabolisme , Transduction du signal , Facteurs de transcription STAT/métabolisme
20.
Sci Rep ; 14(1): 9555, 2024 04 25.
Article de Anglais | MEDLINE | ID: mdl-38664528

RÉSUMÉ

Systemic lupus erythematosus (SLE) is an autoimmune and multisystem disease with a high public health impact. Lupus nephritis (LN), commonly known as renal involvement in SLE, is associated with a poorer prognosis and increased rates of morbidity and mortality in patients with SLE. Identifying new urinary biomarkers that can be used for LN prognosis or diagnosis is essential and is part of current active research. In this study, we applied an untargeted metabolomics approach involving liquid and gas chromatography coupled with mass spectrometry to urine samples collected from 17 individuals with SLE and no kidney damage, 23 individuals with LN, and 10 clinically healthy controls (HCs) to identify differential metabolic profiles for SLE and LN. The data analysis revealed a differentially abundant metabolite expression profile for each study group, and those metabolites may act as potential differential biomarkers of SLE and LN. The differential metabolic pathways found between the LN and SLE patients with no kidney involvement included primary bile acid biosynthesis, branched-chain amino acid synthesis and degradation, pantothenate and coenzyme A biosynthesis, lysine degradation, and tryptophan metabolism. Receiver operating characteristic curve analysis revealed that monopalmitin, glycolic acid, and glutamic acid allowed for the differentiation of individuals with SLE and no kidney involvement and individuals with LN considering high confidence levels. While the results offer promise, it is important to recognize the significant influence of medications and other external factors on metabolomics studies. This impact has the potential to obscure differences in metabolic profiles, presenting a considerable challenge in the identification of disease biomarkers. Therefore, experimental validation should be conducted with a larger sample size to explore the diagnostic potential of the metabolites found as well as to examine how treatment and disease activity influence the identified chemical compounds. This will be crucial for refining the accuracy and effectiveness of using urine metabolomics for diagnosing and monitoring lupus and lupus nephritis.


Sujet(s)
Marqueurs biologiques , Lupus érythémateux disséminé , Glomérulonéphrite lupique , Métabolomique , Humains , Femelle , Lupus érythémateux disséminé/urine , Lupus érythémateux disséminé/métabolisme , Adulte , Métabolomique/méthodes , Marqueurs biologiques/urine , Mâle , Colombie , Glomérulonéphrite lupique/urine , Glomérulonéphrite lupique/diagnostic , Glomérulonéphrite lupique/métabolisme , Métabolome , Adulte d'âge moyen , Études de cohortes , Études cas-témoins , Chromatographie gazeuse-spectrométrie de masse , Jeune adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...