Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 19.941
Filter
2.
Sci Adv ; 10(18): eadn6537, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38701219

ABSTRACT

In mammals, males and females show marked differences in immune responses. Males are globally more sensitive to infectious diseases, while females are more susceptible to systemic autoimmunity. X-chromosome inactivation (XCI), the epigenetic mechanism ensuring the silencing of one X in females, may participate in these sex biases. We perturbed the expression of the trigger of XCI, the noncoding RNA Xist, in female mice. This resulted in reactivation of genes on the inactive X, including members of the Toll-like receptor 7 (TLR7) signaling pathway, in monocyte/macrophages and dendritic and B cells. Consequently, female mice spontaneously developed inflammatory signs typical of lupus, including anti-nucleic acid autoantibodies, increased frequencies of age-associated and germinal center B cells, and expansion of monocyte/macrophages and dendritic cells. Mechanistically, TLR7 signaling is dysregulated in macrophages, leading to sustained expression of target genes upon stimulation. These findings provide a direct link between maintenance of XCI and female-biased autoimmune manifestations and highlight altered XCI as a cause of autoimmunity.


Subject(s)
Autoimmunity , Macrophages , Toll-Like Receptor 7 , X Chromosome Inactivation , Animals , Female , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Autoimmunity/genetics , Mice , Male , Macrophages/metabolism , Macrophages/immunology , RNA, Long Noncoding/genetics , Signal Transduction , Dendritic Cells/immunology , Dendritic Cells/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology
3.
Mol Biol Rep ; 51(1): 629, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38717637

ABSTRACT

It has been rediscovered in the last fifteen years that B-cells play an active role in autoimmune etiology rather than just being spectators. The clinical success of B-cell depletion therapies (BCDTs) has contributed to this. BCDTs, including those that target CD20, CD19, and BAFF, were first developed to eradicate malignant B-cells. These days, they treat autoimmune conditions like multiple sclerosis and systemic lupus erythematosus. Particular surprises have resulted from the use of BCDTs in autoimmune diseases. For example, even in cases where BCDT is used to treat the condition, its effects on antibody-secreting plasma cells and antibody levels are restricted, even though these cells are regarded to play a detrimental pathogenic role in autoimmune diseases. In this Review, we provide an update on our knowledge of the biology of B-cells, examine the outcomes of clinical studies employing BCDT for autoimmune reasons, talk about potential explanations for the drug's mode of action, and make predictions about future approaches to targeting B-cells other than depletion.


Subject(s)
Autoimmune Diseases , B-Lymphocytes , Lymphocyte Depletion , Humans , B-Lymphocytes/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Lymphocyte Depletion/methods , Antigens, CD20/immunology , Antigens, CD19/immunology , Animals , B-Cell Activating Factor/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/therapy
6.
Front Immunol ; 15: 1354348, 2024.
Article in English | MEDLINE | ID: mdl-38774864

ABSTRACT

Background: Systemic lupus erythematosus (SLE) is a multi-organ chronic autoimmune disease. Inflammatory bowel disease (IBD) is a common chronic inflammatory disease of the gastrointestinal tract. Previous studies have shown that SLE and IBD share common pathogenic pathways and genetic susceptibility, but the specific pathogenic mechanisms remain unclear. Methods: The datasets of SLE and IBD were downloaded from the Gene Expression Omnibus (GEO). Differentially expressed genes (DEGs) were identified using the Limma package. Weighted gene coexpression network analysis (WGCNA) was used to determine co-expression modules related to SLE and IBD. Pathway enrichment was performed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis for co-driver genes. Using the Least AbsoluteShrinkage and Selection Operator (Lasso) regressionand Support Vector Machine-Recursive Feature Elimination (SVM-RFE), common diagnostic markers for both diseases were further evaluated. Then, we utilizedthe CIBERSORT method to assess the abundance of immune cell infiltration. Finally,we used the single-cell analysis to obtain the location of common diagnostic markers. Results: 71 common driver genes were identified in the SLE and IBD cohorts based on the DEGs and module genes. KEGG and GO enrichment results showed that these genes were closely associated with positive regulation of programmed cell death and inflammatory responses. By using LASSO regression and SVM, five hub genes (KLRF1, GZMK, KLRB1, CD40LG, and IL-7R) were ultimately determined as common diagnostic markers for SLE and IBD. ROC curve analysis also showed good diagnostic performance. The outcomes of immune cell infiltration demonstrated that SLE and IBD shared almost identical immune infiltration patterns. Furthermore, the majority of the hub genes were commonly expressed in NK cells by single-cell analysis. Conclusion: This study demonstrates that SLE and IBD share common diagnostic markers and pathogenic pathways. In addition, SLE and IBD show similar immune cellinfiltration microenvironments which provides newperspectives for future treatment.


Subject(s)
Biomarkers , Gene Expression Profiling , Gene Regulatory Networks , Inflammatory Bowel Diseases , Lupus Erythematosus, Systemic , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/immunology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/immunology , Transcriptome , Computational Biology/methods , Gene Ontology , Databases, Genetic
7.
Front Immunol ; 15: 1380481, 2024.
Article in English | MEDLINE | ID: mdl-38774868

ABSTRACT

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.


Subject(s)
Flow Cytometry , Lectins , Lupus Erythematosus, Systemic , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Flow Cytometry/methods , Adult , Female , Male , Middle Aged , Lectins/metabolism , Lectins/immunology , Protein Binding , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Glycosylation , Galectins/metabolism , Galectins/immunology , Young Adult , Severity of Illness Index
8.
Front Immunol ; 15: 1383358, 2024.
Article in English | MEDLINE | ID: mdl-38779657

ABSTRACT

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.


Subject(s)
Gene Expression Profiling , Hematopoietic Stem Cells , Interferons , Lupus Erythematosus, Systemic , Single-Cell Analysis , Transcriptome , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Hematopoietic Stem Cells/metabolism , Interferons/metabolism , Interferons/genetics , Female , Adult , Cellular Reprogramming/genetics , Male
9.
Int J Rheum Dis ; 27(5): e15195, 2024 May.
Article in English | MEDLINE | ID: mdl-38766699

ABSTRACT

BACKGROUND/OBJECTIVE: To determine the prevalence of thyroid dysfunctions and thyroid autoantibodies in Thai systemic lupus erythematosus (SLE) patients, and compare them with age- and sex-matched healthy controls (HCs). Associations between thyroid dysfunctions and SLE disease activity, and associated factors for thyroid dysfunctions in SLE also were determined. METHOD: One hundred SLE patients, without apparent clinical thyroid disease, attended the Rheumatology Clinic between November 2021 and October 2022, were enrolled into this study. HCs were matched to SLE cases by age and sex (ratio of 1:1). Clinical manifestations, SLE disease activity and medication received were collected in all SLE patients. Thyroid function tests and thyroid autoantibodies (anti-thyroglobulin: anti-TG and anti-thyroid peroxidase: anti-TPO) were collected from all participants. RESULTS: When compared with HCs, SLE patients had higher prevalence of thyroid dysfunctions, hypothyroidism and euthyroid sick syndrome (28% vs. 7%, p < .001, and 12% vs. 2%, p = .010, and 6% vs. 0%, p = .013, respectively). Prevalence of isolated hypothyroxinemia was higher numerically in SLE patients (9% vs. 3%, p = .074). Prevalence of anti-TG or anti-TPO was no different between SLE patients and HCs (16% vs. 18%, p = .707). There was no association between SLE disease activity and abnormal thyroid functions or thyroid autoantibodies. Family history of thyroid disease and prednisolone use (>10 mg/day) were associated factors for thyroid abnormalities with adjusted OR (95% CI) of 6.13 (1.58-23.75), p = .009 and 4.00 (1.37-11.70), p = .011, respectively. CONCLUSION: Thyroid dysfunctions were more prevalent in SLE patients. Family history of thyroid disease and prednisolone use (>10 mg/day) were independent associated factors of thyroid abnormalities.


Subject(s)
Autoantibodies , Lupus Erythematosus, Systemic , Thyroid Diseases , Humans , Female , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/epidemiology , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/blood , Male , Thailand/epidemiology , Adult , Autoantibodies/blood , Prevalence , Middle Aged , Thyroid Diseases/epidemiology , Thyroid Diseases/immunology , Thyroid Diseases/diagnosis , Thyroid Diseases/blood , Case-Control Studies , Thyroid Function Tests , Biomarkers/blood , Young Adult , Risk Factors , Southeast Asian People
10.
ACS Nano ; 18(20): 13249-13265, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38720584

ABSTRACT

The therapeutic application of mesenchymal stem cells (MSCs) has good potential as a treatment strategy for systemic lupus erythematosus (SLE), but traditional MSC therapy still has limitations in effectively modulating immune cells. Herein, we present a promising strategy based on dexamethasone liposome-integrated MSCs (Dexlip-MSCs) for treating SLE via multiple immunomodulatory pathways. This therapeutic strategy prolonged the circulation time of dexamethasone liposomes in vivo, restrained CD4+T-cell proliferation, and inhibited the release of proinflammatory mediators (IFN-γ and TNF-α) by CD4+T cells. In addition, Dexlip-MSCs initiated cellular reprogramming by activating the glucocorticoid receptor (GR) signaling pathway to upregulate the expression of anti-inflammatory factors such as cysteine-rich secretory protein LCCL-containing domain 2 (CRISPLD2) and downregulate the expression of proinflammatory factors. In addition, Dexlip-MSCs synergistically increased the anti-inflammatory inhibitory effect of CD4+T cells through the release of dexamethasone liposomes or Dex-integrated MSC-derived exosomes (Dex-MSC-EXOs). Based on these synergistic biological effects, we demonstrated that Dexlip-MSCs alleviated disease progression in MRL/lpr mice more effectively than Dexlip or MSCs alone. These features indicate that our stem cell delivery strategy is a promising therapeutic approach for clinical SLE treatment.


Subject(s)
Dexamethasone , Lupus Erythematosus, Systemic , Mesenchymal Stem Cells , Animals , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Dexamethasone/pharmacology , Dexamethasone/chemistry , Lupus Erythematosus, Systemic/therapy , Lupus Erythematosus, Systemic/immunology , Mice , Liposomes/chemistry , Mesenchymal Stem Cell Transplantation , Cell Proliferation/drug effects , Female , Mice, Inbred MRL lpr , Humans , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry
11.
Front Immunol ; 15: 1371708, 2024.
Article in English | MEDLINE | ID: mdl-38756769

ABSTRACT

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.


Subject(s)
Lupus Erythematosus, Systemic , T-Lymphocytes , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/immunology , Humans , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Metabolic Networks and Pathways , Energy Metabolism , Animals , Signal Transduction , Cytokines/metabolism
12.
Front Immunol ; 15: 1373904, 2024.
Article in English | MEDLINE | ID: mdl-38715605

ABSTRACT

Having increased popularity during the Covid-19 pandemic, vitamin D3 is currently impressing thanks to the numerous researches aimed at its interactions with the body's homeostasis. At the same time, there is a peak in terms of recommendations for supplementation with it. Some of the studies focus on the link between autoimmune diseases and nutritional deficiencies, especially vitamin D3. Since the specialized literature aimed at children (patients between 0-18 years old) is far from equal to the informational diversity of the adult-centered branch, this review aims to bring up to date the relationship between the microbial and nutritional balance and the activity of pediatric systemic lupus erythematosus (pSLE). The desired practical purpose resides in a better understanding and an adequate, individualized management of the affected persons to reduce morbidity. The center of the summary is to establish the impact of hypovitaminosis D in the development and evolution of pediatric lupus erythematosus. We will address aspects related to the two entities of the impact played by vitamin D3 in the pathophysiological cascade of lupus, but also the risk of toxicity and its effects when the deficiency is over supplemented (hypervitaminosis D). We will debate the relationship of hypovitaminosis D with the modulation of immune function, the potentiation of inflammatory processes, the increase of oxidative stress, the perfusion of cognitive brain areas, the seasonal incidence of SLE and its severity. Finally, we review current knowledge, post-pandemic, regarding the hypovitaminosis D - pSLE relationship.


Subject(s)
COVID-19 , Lupus Erythematosus, Systemic , Vitamin D Deficiency , Vitamin D , Humans , Lupus Erythematosus, Systemic/immunology , COVID-19/immunology , Child , Vitamin D Deficiency/immunology , Vitamin D Deficiency/complications , Vitamin D/metabolism , SARS-CoV-2/immunology , Adolescent , Child, Preschool , Dietary Supplements
13.
J Assoc Physicians India ; 72(3): 32-35, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38736114

ABSTRACT

OBJECTIVE: Systemic lupus erythematosus (SLE) is a chronic inflammatory disease with a wide range of clinical manifestations having considerable variation in clinical features that are influenced by ethnic, sociocultural, and geographical factors. This disease primarily affects young women aged between 18 and 35 years. The aim of this present study was to delineate the clinical manifestations and immunological patterns of SLE patients from the Northeastern (NE) region of India. MATERIALS AND METHODS: The study was carried out in a tertiary care hospital from January 2016 to January 2021. Adult patients of age >18 years fulfilling systemic lupus international collaborating clinic criteria (SLICC) for classification of SLE were included in this study. Immunology such as antinuclear antibodies (ANA) and double-stranded deoxyribonucleic acid (dsDNA) were also performed followed by enzyme-linked immunosorbent assay (ELISA). RESULTS: Over a period of 5 years, 142 patients were recruited for the study, with an overall female-to-male ratio was 9.9:1, a median age at onset of 25 years (interquartile range age 21-32 years) and a mean disease duration was 15.25 months (range 2-60 months). Our study revealed that ANA was positive in 97.18% of patients while anti-dsDNA was positive in 78.68%, indicating that women from this region have higher positivity rates. CONCLUSION: Our findings support the notion that SLE is a multisystem disorder that predominantly affects young females, especially during the second and third decades of life. Hematological, mucocutaneous, and renal manifestations are common in our patients. Moreover, pulmonary, cardiovascular, and gastrointestinal (GI) manifestations were understudied in other cohorts, which is one of our study's strengths.


Subject(s)
Antibodies, Antinuclear , Lupus Erythematosus, Systemic , Humans , Lupus Erythematosus, Systemic/immunology , Adult , Female , India/epidemiology , Male , Retrospective Studies , Antibodies, Antinuclear/blood , Young Adult
16.
Arthritis Res Ther ; 26(1): 99, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741185

ABSTRACT

OBJECTIVES: This study aims to elucidate the transcriptomic signatures and dysregulated pathways in patients with Systemic Lupus Erythematosus (SLE), with a particular focus on those persisting during disease remission. METHODS: We conducted bulk RNA-sequencing of peripheral blood mononuclear cells (PBMCs) from a well-defined cohort comprising 26 remission patients meeting the Low Lupus Disease Activity State (LLDAS) criteria, 76 patients experiencing disease flares, and 15 healthy controls. To elucidate immune signature changes associated with varying disease states, we performed extensive analyses, including the identification of differentially expressed genes and pathways, as well as the construction of protein-protein interaction networks. RESULTS: Several transcriptomic features recovered during remission compared to the active disease state, including down-regulation of plasma and cell cycle signatures, as well as up-regulation of lymphocytes. However, specific innate immune response signatures, such as the interferon (IFN) signature, and gene modules involved in chromatin structure modification, persisted across different disease states. Drug repurposing analysis revealed certain drug classes that can target these persistent signatures, potentially preventing disease relapse. CONCLUSION: Our comprehensive transcriptomic study revealed gene expression signatures for SLE in both active and remission states. The discovery of gene expression modules persisting in the remission stage may shed light on the underlying mechanisms of vulnerability to relapse in these patients, providing valuable insights for their treatment.


Subject(s)
Lupus Erythematosus, Systemic , Transcriptome , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Humans , Female , Adult , Male , Middle Aged , Gene Expression Profiling/methods , Leukocytes, Mononuclear/metabolism , Protein Interaction Maps/genetics
17.
Clin Immunol ; 263: 110231, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692449

ABSTRACT

Moderate-to-severe systemic lupus erythematosus (SLE) is characterized by extensive autoantibody deposition and persistent autoinflammation. As the existing animal models are limited in accurately reproducing the pathological characteristics of human SLE, we introduced a novel animal model simulating multi-organ autoinflammation through intra-organ injections. The model closely mimicked key features of SLE, including IgG deposition, inflammation, and tissue damage. The model could be used to assess the roles of IgG, immune cells, cytokines, and Fc gamma receptor (FcγR) in the pathogenesis of autoinflammation. The results obtained from this model could be confirmed by lupus MRL/lpr mice. The review suggested that the diagnostic criteria should be reconsidered to incorporate IgG deposition in tissues and highlighted the limitations of current T-cell and B-cell-focused treatments. To summarize, the IgG deposition model can be used to investigate the pathogenesis and treatment of multi-organ tissue damage associated with SLE.


Subject(s)
Disease Models, Animal , Immunoglobulin G , Lupus Erythematosus, Systemic , Animals , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Mice , Immunoglobulin G/immunology , Humans , Mice, Inbred MRL lpr , Inflammation/immunology , Receptors, IgG/immunology , Receptors, IgG/metabolism , B-Lymphocytes/immunology
18.
Int Immunopharmacol ; 134: 112149, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38692019

ABSTRACT

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.


Subject(s)
Energy Metabolism , Lupus Erythematosus, Systemic , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/immunology , Humans , Animals , Mitochondria/metabolism , TOR Serine-Threonine Kinases/metabolism , Signal Transduction , AMP-Activated Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Adenosine Triphosphate/metabolism
19.
Cell Rep Med ; 5(5): 101569, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38744279

ABSTRACT

Systemic lupus erythematosus (SLE) displays a hallmark interferon (IFN) signature. Yet, clinical trials targeting type I IFN (IFN-I) have shown variable efficacy, and blocking IFN-II failed to treat SLE. Here, we show that IFN type levels in SLE vary significantly across clinical and transcriptional endotypes. Whereas skin involvement correlated with IFN-I alone, systemic features like nephritis associated with co-elevation of IFN-I, IFN-II, and IFN-III, indicating additive IFN effects in severe SLE. Notably, while high IFN-II/-III levels without IFN-I had a limited effect on disease activity, IFN-II was linked to IFN-I-independent transcriptional profiles (e.g., OXPHOS and CD8+GZMH+ cells), and IFN-III enhanced IFN-induced gene expression when co-elevated with IFN-I. Moreover, dysregulated IFNs do not explain the IFN signature in 64% of patients or clinical manifestations including cytopenia, serositis, and anti-phospholipid syndrome, implying IFN-independent endotypes in SLE. This study sheds light on mechanisms underlying SLE heterogeneity and the variable response to IFN-targeted therapies in clinical trials.


Subject(s)
Interferons , Lupus Erythematosus, Systemic , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Interferons/metabolism , Interferons/genetics , Female , Adult , Male , Transcriptome/genetics , Interferon Type I/metabolism , Interferon Type I/genetics , Middle Aged , Transcription, Genetic , Gene Expression Regulation
20.
Front Immunol ; 15: 1376933, 2024.
Article in English | MEDLINE | ID: mdl-38726007

ABSTRACT

Introduction: Systemic autoimmune diseases (SADs) are a significant burden on the healthcare system. Understanding the complexity of the peripheral immunophenotype in SADs may facilitate the differential diagnosis and identification of potential therapeutic targets. Methods: Single-cell mass cytometric immunophenotyping was performed on peripheral blood mononuclear cells (PBMCs) from healthy controls (HCs) and therapy-naive patients with rheumatoid arthritis (RA), progressive systemic sclerosis (SSc), and systemic lupus erythematosus (SLE). Immunophenotyping was performed on 15,387,165 CD45+ live single cells from 52 participants (13 cases/group), using an antibody panel to detect 34 markers. Results: Using the t-SNE (t-distributed stochastic neighbor embedding) algorithm, the following 17 main immune cell types were determined: CD4+/CD57- T cells, CD4+/CD57+ T cells, CD8+/CD161- T cells, CD8+/CD161+/CD28+ T cells, CD8dim T cells, CD3+/CD4-/CD8- T cells, TCRγ/δ T cells, CD4+ NKT cells, CD8+ NKT cells, classic NK cells, CD56dim/CD98dim cells, B cells, plasmablasts, monocytes, CD11cdim/CD172dim cells, myeloid dendritic cells (mDCs), and plasmacytoid dendritic cells (pDCs). Seven of the 17 main cell types exhibited statistically significant frequencies in the investigated groups. The expression levels of the 34 markers in the main populations were compared between HCs and SADs. In summary, 59 scatter plots showed significant differences in the expression intensities between at least two groups. Next, each immune cell population was divided into subpopulations (metaclusters) using the FlowSOM (self-organizing map) algorithm. Finally, 121 metaclusters (MCs) of the 10 main immune cell populations were found to have significant differences to classify diseases. The single-cell T-cell heterogeneity represented 64MCs based on the expression of 34 markers, and the frequency of 23 MCs differed significantly between at least twoconditions. The CD3- non-T-cell compartment contained 57 MCs with 17 MCs differentiating at least two investigated groups. In summary, we are the first to demonstrate the complexity of the immunophenotype of 34 markers over 15 million single cells in HCs vs. therapy-naive patients with RA, SSc, and SLE. Disease specific population frequencies or expression patterns of peripheral immune cells provide a single-cell data resource to the scientific community.


Subject(s)
Arthritis, Rheumatoid , Immunophenotyping , Lupus Erythematosus, Systemic , Scleroderma, Systemic , Single-Cell Analysis , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/diagnosis , Female , Single-Cell Analysis/methods , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/diagnosis , Middle Aged , Adult , Male , Scleroderma, Systemic/immunology , Aged , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Biomarkers
SELECTION OF CITATIONS
SEARCH DETAIL
...