Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters

Publication year range
1.
Rheumatology (Oxford) ; 63(2): 551-562, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37341646

ABSTRACT

OBJECTIVES: Platelets and low-density neutrophils (LDNs) are major players in the immunopathogenesis of SLE. Despite evidence showing the importance of platelet-neutrophil complexes (PNCs) in inflammation, little is known about the relationship between LDNs and platelets in SLE. We sought to characterize the role of LDNs and Toll-like receptor 7 (TLR7) in clinical disease. METHODS: Flow cytometry was used to immunophenotype LDNs from SLE patients and controls. The association of LDNs with organ damage was investigated in a cohort of 290 SLE patients. TLR7 mRNA expression was assessed in LDNs and high-density neutrophils (HDNs) using publicly available mRNA sequencing datasets and our own cohort using RT-PCR. The role of TLR7 in platelet binding was evaluated in platelet-HDN mixing studies using TLR7-deficient mice and Klinefelter syndrome patients. RESULTS: SLE patients with active disease have more LDNs, which are heterogeneous and more immature in patients with evidence of kidney dysfunction. LDNs are platelet bound, in contrast to HDNs. LDNs settle in the peripheral blood mononuclear cell (PBMC) layer due to the increased buoyancy and neutrophil degranulation from platelet binding. Mixing studies demonstrated that this PNC formation was dependent on platelet-TLR7 and that the association results in increased NETosis. The neutrophil:platelet ratio is a useful clinical correlate for LDNs, and a higher NPR is associated with past and current flares of LN. CONCLUSIONS: LDNs sediment in the upper PBMC fraction due to PNC formation, which is dependent on the expression of TLR7 in platelets. Collectively, our results reveal a novel TLR7-dependent crosstalk between platelets and neutrophils that may be an important therapeutic opportunity for LN.


Subject(s)
Lupus Nephritis , Neutrophils , Animals , Humans , Mice , Leukocytes, Mononuclear , Lupus Nephritis/pathology , Neutrophils/metabolism , RNA, Messenger/metabolism , Toll-Like Receptor 7/genetics
2.
Rheumatology (Oxford) ; 60(8): 3936-3944, 2021 08 02.
Article in English | MEDLINE | ID: mdl-33369679

ABSTRACT

OBJECTIVES: To identify and quantify the level of CD34+CD133+CD309+ circulating angiogenic cells (CAC) and explore factors associated with the level of CAC in patients with SLE. METHODS: The peripheral blood mononuclear cells of consecutive SLE patients and demographically matched healthy controls (HC) were extracted and identified, enumerated and compared for CAC levels by multi-colour flow cytometry based on the EULAR Scleroderma Trials and Research (EUSTAR) recommendation. Meta-analyses were performed by combining the current and previous case-control studies, aiming to increase the statistical power to discern the difference in CAC level between SLE patients and HC. Mixed-model meta-regression was conducted to explore potential demographic and clinical factors that were associated with CAC level. RESULTS: A lower level of CAC was found in 29 SLE patients compared with 24 HC [mean (s.d.) 10.76 (13.9) vs 24.58 (25.4) cells/ml, P = 0.015]. Random-effects meta-analyses of the current and six previously published case-control studies involving 401 SLE patients and 228 HC revealed a lower CAC level compared with HC (standardized mean difference = -2.439, P = 0.001). Meta-regression analysis demonstrated that HCQ use was associated with a more discrepant CAC level between both groups (P = 0.01115). CONCLUSION: SLE patients had a significantly lower CD34+CD133+CD309+ CAC level than HC, and HCQ use was associated with a more discrepant CAC level between SLE patients and HC. This study triggers further observational, interventional and mechanistic studies to address the beneficial impact of HCQ on the functionality of CAC in SLE patients.


Subject(s)
Endothelial Progenitor Cells/cytology , Lupus Erythematosus, Systemic/blood , AC133 Antigen/metabolism , Adult , Antigens, CD34/metabolism , Antirheumatic Agents/therapeutic use , Case-Control Studies , Endothelial Progenitor Cells/metabolism , Female , Flow Cytometry , Humans , Hydroxychloroquine/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Male , Middle Aged , Regression Analysis , Vascular Endothelial Growth Factor Receptor-2/metabolism , Young Adult
3.
Int J Mol Sci ; 21(23)2020 Nov 24.
Article in English | MEDLINE | ID: mdl-33255483

ABSTRACT

In eukaryotes, microRNAs (miRNAs) have roles in development, homeostasis, disease and the immune response. Recent work has shown that plant and mammalian miRNAs also mediate cross-kingdom and cross-domain communications. However, these studies remain controversial and are lacking critical mechanistic explanations. Bacteria do not produce miRNAs themselves, and therefore it is unclear how these eukaryotic RNA molecules could function in the bacterial recipient. In this review, we compare and contrast the biogenesis and functions of regulatory RNAs in eukaryotes and bacteria. As a result, we discovered several conserved features and homologous components in these distinct pathways. These findings enabled us to propose novel mechanisms to explain how eukaryotic miRNAs could function in bacteria. Further understanding in this area is necessary to validate the findings of existing studies and could facilitate the use of miRNAs as novel tools for the directed remodelling of the human microbiota.


Subject(s)
Bacteria/genetics , Eukaryota/genetics , MicroRNAs/genetics , RNA/genetics , Humans , Microbiota/genetics
4.
Cytometry A ; 95(3): 268-278, 2019 03.
Article in English | MEDLINE | ID: mdl-30549398

ABSTRACT

Neutrophil extracellular traps (NETs) are web-like structures composed of decondensed chromatin and antimicrobial proteins that are released into the extracellular space during microbial infections. This active cell death program is known as NETosis. To date, florescence microscopy is the widely accepted method for visualization and quantification of NETs. However, this method is subjective, time consuming and yields low numbers of analyzed polymorphonuclear cells (PMNs) per sample. Increasing interest has emerged on the identification of NETs using flow cytometry techniques. However, flow cytometry analysis of NETs requires particular precautions for sample preparation to obtain reproducible data. Herein, we describe a flow cytometry-based assay for high-throughput detection and quantification of NETosis in mixed cell populations. We used fluorescent-labeled antibodies against cell markers on PMNs together with a combination of nucleic acid stains to measure NETosis in whole blood (WB) and purified PMNs. Using plasma membrane-impermeable DNA-binding dye, SYTOX Orange (SO), we found that cell-appendant DNA of NETting PMNs were positive for SO and DAPI. The combination of optimally diluted antibody and nucleic acid dyes required no washing and yielded low background fluorescence. Significant correlations were found for NETosis from WB and purified PMNs. We then validated the assay by comparing with time-lapse live cell fluorescence microscopy and determined very good intraassay and interassay variances. The assay was then applied to a disease associated with NETosis, systemic lupus erythematosus (SLE). We examined PMA-induced NETosis in peripheral PMNs from SLE patients and controls and in bone marrow PMNs from multiple murine models. In summary, this assay is observer-independent and allows for rapid assessment of a large number of PMNs per sample. Use of this assay does not require sophisticated microscopic equipment like imaging flow cytometers and may be a starting point to analyze extracellular trap formation from immune cells other than PMNs. © 2018 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.


Subject(s)
Extracellular Traps/metabolism , Flow Cytometry/methods , Fluorescent Antibody Technique/methods , Microscopy, Fluorescence/methods , Neutrophils/metabolism , Animals , Bone Marrow Cells/metabolism , DNA/analysis , DNA/chemistry , Disease Models, Animal , Extracellular Traps/chemistry , Female , High-Throughput Screening Assays , Humans , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/metabolism , Mice , Mice, Inbred C57BL , Neutrophils/cytology , Neutrophils/drug effects , Regulated Cell Death/drug effects , Regulated Cell Death/genetics
5.
Proc Natl Acad Sci U S A ; 112(45): E6195-204, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26512111

ABSTRACT

Glomerulonephritis is a common and debilitating feature of systemic lupus erythematosus (SLE). The precise immune mechanisms that drive the progression from benign autoimmunity to glomerulonephritis are largely unknown. Previous investigations have shown that a moderate increase of the innate Toll-like receptor 7 (TLR7) is sufficient for the development of nephritis. In these systems normalization of B-cell TLR7 expression or temporal depletion of plasmacytoid dendritic cells (pDCs) slow progression; however, the critical cell that is responsible for driving full immunopathology remains unidentified. In this investigation we have shown that conventional DC expression of TLR7 is essential for severe autoimmunity in the Sle1Tg7 model of SLE. We show that a novel expanding CD11b(+) conventional DC subpopulation dominates the infiltrating renal inflammatory milieu, localizing to the glomeruli. Moreover, exposure of human myeloid DCs to IFN-α or Flu increases TLR7 expression, suggesting they may have a role in self-RNA recognition pathways in clinical disease. To our knowledge, this study is the first to highlight the importance of conventional DC-TLR7 expression for kidney pathogenesis in a murine model of SLE.


Subject(s)
Dendritic Cells/metabolism , Lupus Nephritis/physiopathology , Toll-Like Receptor 7/metabolism , Up-Regulation , Analysis of Variance , Animals , Base Sequence , CD11b Antigen/metabolism , DNA Primers/genetics , Flow Cytometry , Gene Expression Profiling/methods , Humans , Image Processing, Computer-Assisted , Kidney Glomerulus/cytology , Kidney Glomerulus/pathology , Lupus Nephritis/metabolism , Mice , Microscopy, Confocal , Molecular Sequence Data , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Statistics, Nonparametric
6.
Rheumatology (Oxford) ; 56(suppl_1): i88-i99, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28013204

ABSTRACT

Mouse models of SLE have been indispensable tools to study disease pathogenesis, to identify genetic susceptibility loci and targets for drug development, and for preclinical testing of novel therapeutics. Recent insights into immunological mechanisms of disease progression have boosted a revival in SLE drug development. Despite promising results in mouse studies, many novel drugs have failed to meet clinical end points. This is probably because of the complexity of the disease, which is driven by polygenic predisposition and diverse environmental factors, resulting in a heterogeneous clinical presentation. Each mouse model recapitulates limited aspects of lupus, especially in terms of the mechanism underlying disease progression. The main mouse models have been fairly successful for the evaluation of broad-acting immunosuppressants. However, the advent of targeted therapeutics calls for a selection of the most appropriate model(s) for testing and, ultimately, identification of patients who will be most likely to respond.


Subject(s)
Disease Models, Animal , Lupus Erythematosus, Systemic/immunology , Mice , Animals , Humans , Lupus Erythematosus, Systemic/genetics
7.
Rheumatology (Oxford) ; 56(6): 1025-1030, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28137763

ABSTRACT

Objective: Siglecs are sialic acid-binding immunoglobulin-like lectins expressed on the surface of immune cells, which participate in the discrimination of self and non-self. We investigated myeloid CD33-related Siglec expression in a cohort of patients with SLE. Methods: Cell surface expression of Siglec-5/14, Siglec-9 and Siglec-10 on peripheral myeloid subsets were analysed from 39 SLE patients using flow cytometry. Genotyping of the Siglec-5/14 locus was also performed. Clinical markers of SLE disease activity, including SLEDAI, serum complement concentrations and serum autoantibodies, were assessed and correlated with Siglec levels. Results: Siglec-14 expression on SLE monocytes (median = 518, interquartile range: 411) was significantly higher when compared with healthy controls (median = 427, interquartile range: 289.3; P < 0.05) and correlated positively with SLEDAI scoring and anti-Sm and anti-SSB autoantibodies (P < 0.05). A negative correlation was determined with patient serum C3 concentrations (P < 0.005). Genotyping of the Siglec-5/14 locus revealed a high frequency of the Siglec-14 null allele across both groups, reflecting the incidence in Asian populations. Conclusion: Our data suggest that the Siglec immunomodulatory molecules, in particular Siglec-14 expression on monocytes, may play an important role in the inflammatory events of SLE. No bias was found with regard to SIGLEC14 genotype in our patient group compared with healthy controls. Larger comparisons of mixed ethnicity might, however, reveal an important role for Siglecs in the pathogenesis of autoimmune disease.


Subject(s)
Lectins/metabolism , Lupus Erythematosus, Systemic/metabolism , Receptors, Cell Surface/metabolism , Adult , Anti-Inflammatory Agents/therapeutic use , Case-Control Studies , Female , Genotype , Heterozygote , Humans , Lectins/genetics , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/ethnology , Male , Middle Aged , Polymorphism, Genetic , Prednisolone/therapeutic use , Receptors, Cell Surface/genetics , Up-Regulation/physiology , Young Adult
8.
Rheumatology (Oxford) ; 56(suppl_1): i55-i66, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28375453

ABSTRACT

SLE is a chronic autoimmune disease caused by perturbations of the immune system. The clinical presentation is heterogeneous, largely because of the multiple genetic and environmental factors that contribute to disease initiation and progression. Over the last 60 years, there have been a number of significant leaps in our understanding of the immunological mechanisms driving disease processes. We now know that multiple leucocyte subsets, together with inflammatory cytokines, chemokines and regulatory mediators that are normally involved in host protection from invading pathogens, contribute to the inflammatory events leading to tissue destruction and organ failure. In this broad overview, we discuss the main pathways involved in SLE and highlight new findings. We describe the immunological changes that characterize this form of autoimmunity. The major leucocytes that are essential for disease progression are discussed, together with key mediators that propagate the immune response and drive the inflammatory response in SLE.


Subject(s)
Autoimmunity/immunology , B-Lymphocytes/immunology , Cytokines/immunology , Environment , Inflammation/immunology , Lupus Erythematosus, Systemic/immunology , Autoimmunity/genetics , Genetic Predisposition to Disease , Humans , Inflammation/genetics , Lupus Erythematosus, Systemic/genetics , Self Tolerance/genetics , Self Tolerance/immunology
9.
Int Immunol ; 28(5): 223-32, 2016 05.
Article in English | MEDLINE | ID: mdl-26567289

ABSTRACT

The toll-like receptors (TLRs) are important innate receptors recognizing potentially pathogenic material. However, they also play a significant role in the development of Alzheimer's disease, cancer, autoimmunity and the susceptibility to viral infections. Macrophages are essential for an effective immune response to foreign material and the resolution of inflammation. In these studies, we examined the impact of different TLR ligands on macrophage cell function. We demonstrate that stimulation of all TLRs tested increases the phagocytosis of apoptotic cells by macrophages. TLR7 and TLR9 ligation decreased the levels of the surface co-expression molecules CD86 and MHCII, which was associated with a concomitant reduction in antigen presentation and proliferation of T cells. This down-regulation in macrophage function was not due to an increase in cell death. In fact, exposure to TLR7 or TLR9 ligands promoted cell viability for up to 9 days, in contrast to TLR3 or TLR4. Additionally, macrophages exposed to TLR7/TLR9 ligands had a significantly lower ratio of Il-12/Il-10 mRNA expression compared with those treated with the TLR4 ligand, LPS. Taken together, these data demonstrate that TLR7/TLR9 ligands push the macrophage into a phagocytic long-lived cell, with a decreased capacity of antigen presentation and reminiscent of the M2 polarized state.


Subject(s)
Antigen Presentation , Macrophages/immunology , Membrane Glycoproteins/agonists , Membrane Glycoproteins/immunology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/immunology , Animals , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Ligands , Lipopolysaccharides/toxicity , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Phagocytosis/drug effects , Phagocytosis/genetics , Toll-Like Receptor 7/genetics , Toll-Like Receptor 9/genetics
10.
J Immunol ; 194(11): 5174-86, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25917094

ABSTRACT

Human dendritic cells (DCs) regulate the balance between immunity and tolerance through selective activation by environmental and pathogen-derived triggers. To characterize the rapid changes that occur during this process, we analyzed the underlying metabolic activity across a spectrum of functional DC activation states, from immunogenic to tolerogenic. We found that in contrast to the pronounced proinflammatory program of mature DCs, tolerogenic DCs displayed a markedly augmented catabolic pathway, related to oxidative phosphorylation, fatty acid metabolism, and glycolysis. Functionally, tolerogenic DCs demonstrated the highest mitochondrial oxidative activity, production of reactive oxygen species, superoxide, and increased spare respiratory capacity. Furthermore, assembled, electron transport chain complexes were significantly more abundant in tolerogenic DCs. At the level of glycolysis, tolerogenic and mature DCs showed similar glycolytic rates, but glycolytic capacity and reserve were more pronounced in tolerogenic DCs. The enhanced glycolytic reserve and respiratory capacity observed in these DCs were reflected in a higher metabolic plasticity to maintain intracellular ATP content. Interestingly, tolerogenic and mature DCs manifested substantially different expression of proteins involved in the fatty acid oxidation (FAO) pathway, and FAO activity was significantly higher in tolerogenic DCs. Inhibition of FAO prevented the function of tolerogenic DCs and partially restored T cell stimulatory capacity, demonstrating their dependence on this pathway. Overall, tolerogenic DCs show metabolic signatures of increased oxidative phosphorylation programing, a shift in redox state, and high plasticity for metabolic adaptation. These observations point to a mechanism for rapid genome-wide reprograming by modulation of underlying cellular metabolism during DC differentiation.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Immune Tolerance/immunology , Mitochondria/metabolism , Oxygen Consumption , 3-Hydroxyacyl CoA Dehydrogenases/antagonists & inhibitors , 3-Hydroxyacyl CoA Dehydrogenases/genetics , Acetyl-CoA C-Acyltransferase/antagonists & inhibitors , Acetyl-CoA C-Acyltransferase/genetics , Carbon-Carbon Double Bond Isomerases/antagonists & inhibitors , Carbon-Carbon Double Bond Isomerases/genetics , Cell Differentiation , Cells, Cultured , Electron Transport Chain Complex Proteins/biosynthesis , Electron Transport Chain Complex Proteins/metabolism , Enoyl-CoA Hydratase/antagonists & inhibitors , Enoyl-CoA Hydratase/genetics , Fatty Acids/metabolism , Glycolysis , Humans , Leukocytes, Mononuclear/immunology , Oxidation-Reduction , Oxidative Phosphorylation , Racemases and Epimerases/antagonists & inhibitors , Racemases and Epimerases/genetics , Superoxides/metabolism , T-Lymphocytes/immunology
11.
J Immunol ; 191(12): 6128-35, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24218451

ABSTRACT

The three-prime repair exonuclease 1 (TREX1) is the most abundant exonuclease in mammalian cells. Mutations in Trex1 gene are being linked to the development of Aicardi-Goutières syndrome, an inflammatory disease of the brain, and systemic lupus erythematosus. In clinical cases and in a Trex1-deficient murine model, chronic production of type I IFN plays a pathogenic role. In this study, we demonstrate that Trex1(-/-) mice present inflammatory signatures in many different organs, including the brain. Trex1 is highly induced in macrophages in response to proinflammatory stimuli, including TLR7 and TLR9 ligands. Our findings show that, in the absence of Trex1, macrophages displayed an exacerbated proinflammatory response. More specifically, following proinflammatory stimulation, Trex1(-/-) macrophages exhibited an increased TNF-α and IFN-α production, higher levels of CD86, and increased Ag presentation to CD4(+) T cells, as well as an impaired apoptotic T cell clearance. These results evidence an unrevealed function of the Trex1 as a negative regulator of macrophage inflammatory activation and demonstrate that macrophages play a major role in diseases associated with Trex1 mutations, which contributes to the understanding of inflammatory signature in these diseases.


Subject(s)
Exodeoxyribonucleases/physiology , Inflammation/immunology , Macrophage Activation/physiology , Phosphoproteins/physiology , Animals , Antigen Presentation , Apoptosis , B7-2 Antigen/biosynthesis , B7-2 Antigen/genetics , Brain Chemistry , Exodeoxyribonucleases/deficiency , Exodeoxyribonucleases/immunology , Gene Expression Regulation/immunology , Humans , Inflammation/metabolism , Interferon-alpha/biosynthesis , Interferon-alpha/genetics , Jurkat Cells , L Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phagocytosis , Phosphoproteins/deficiency , Phosphoproteins/immunology , Recombinant Proteins/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Toll-Like Receptor 9/physiology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
12.
J Immunol ; 191(8): 4375-82, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24048896

ABSTRACT

TLRs play a pivotal role in the recognition of bacteria and viruses. Members of the family recognize specific pathogen sequences to trigger both MyD88 and TRIF-dependent pathways to stimulate a plethora of cells. Aberrant activation of these pathways is known to play a critical role in the development of autoimmunity and cancer. However, how these pathways are entirely regulated is not fully understood. In these studies, we have identified Annexin-A1 (ANXA1) as a novel regulator of TLR-induced IFN-ß and CXCL10 production. We demonstrate that in the absence of ANXA1, mice produce significantly less IFN-ß and CXCL10, and macrophages and plasmacytoid dendritic cells have a deficiency in activation following polyinosinic:polycytidylic acid administration in vivo. Furthermore, a deficiency in activation is observed in macrophages after LPS and polyinosinic:polycytidylic acid in vitro. In keeping with these findings, overexpression of ANXA1 resulted in enhanced IFN-ß and IFN-stimulated responsive element promoter activity, whereas silencing of ANXA1 impaired TLR3- and TLR4-induced IFN-ß and IFN-stimulated responsive element activation. In addition, we show that the C terminus of ANXA1 directly associates with TANK-binding kinase 1 to regulate IFN regulatory factor 3 translocation and phosphorylation. Our findings demonstrate that ANXA1 plays an important role in TLR activation, leading to an augmentation in the type 1 IFN antiviral cytokine response.


Subject(s)
Annexin A1/metabolism , Interferon-beta/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Active Transport, Cell Nucleus , Animals , Annexin A1/biosynthesis , Annexin A1/genetics , Cell Line , Chemokine CXCL10/biosynthesis , Dendritic Cells/metabolism , Enzyme Activation , HEK293 Cells , Humans , Interferon Regulatory Factor-3/metabolism , Lipopolysaccharides , Macrophage Activation/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Phosphorylation , Poly I-C/pharmacology , Signal Transduction/immunology
13.
J Immunol ; 189(12): 5786-96, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23150717

ABSTRACT

Systemic lupus erythematosus (SLE) is a chronic systemic autoimmune disease characterized by the production of antinuclear autoantibodies. Antinuclear autoantibody development is recognized as one of the initial stages of disease that often results in systemic inflammation, kidney disease, and death. The etiology is complex, but it is clear that innate pathways may play an important role in disease progression. Recent data have highlighted an important role for the TLR family, particularly TLR7, in both human disease and murine models. In this study, we have presented a low copy conditional TLR7 transgenic (Tg7) mouse strain that does not develop spontaneous autoimmunity. When we combine Tg7 with the Sle1 lupus susceptibility locus, the mice develop severe disease. Using the CD19(Cre) recombinase system, we normalized expression of TLR7 solely within the B cells. Using this method we demonstrated that overexpression of TLR7 within the B cell compartment reduces the marginal zone B cell compartment and increases B and T cell activation but not T follicular helper cell development. Moreover, this enhanced B cell TLR7 expression permits the specific development of Abs to RNA/protein complexes and exacerbates SLE disease.


Subject(s)
Autoantibodies/biosynthesis , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Gene Expression Regulation, Developmental/immunology , Genetic Predisposition to Disease , Lupus Erythematosus, Systemic/immunology , Membrane Glycoproteins/genetics , Toll-Like Receptor 7/genetics , Animals , Autoantibodies/adverse effects , B-Lymphocyte Subsets/metabolism , Disease Progression , Epistasis, Genetic/immunology , Female , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Male , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Toll-Like Receptor 7/biosynthesis , Toll-Like Receptor 7/physiology , Transgenes/immunology
15.
Front Immunol ; 13: 807050, 2022.
Article in English | MEDLINE | ID: mdl-35154124

ABSTRACT

Cytokine release syndrome (CRS) is a phenomenon of immune hyperactivation described in the setting of immunotherapy. Unlike other immune-related adverse events, CRS triggered by immune checkpoint inhibitors (ICIs) is not well described. The clinical characteristics and course of 25 patients with ICI-induced CRS from 2 tertiary hospitals were abstracted retrospectively from the medical records and analyzed. CRS events were confirmed by 2 independent reviewers and graded using the Lee et al. scale. The median duration of CRS was 15.0 days (Q1; Q3 6.3; 29.8) and 10 (40.0%) had multiple episodes of CRS flares. Comparing the clinical factors and biomarkers in Grades 1-2 and 3-5 CRS, we found that patients with Grades 3-5 CRS had following: (i) had longer time to fever onset [25.0 days (Q1; Q3 13.0; 136.5) vs. 3.0 days (Q1; Q3 0.0; 18.0), p=0.027]; (ii) more cardiovascular (p=0.002), neurologic (p=0.001), pulmonary (p=0.044) and rheumatic (p=0.037) involvement; (iii) lower platelet count (p=0.041) and higher urea (p=0.041) at presentation compared to patients with Grades 1-2 CRS. 7 patients (28.0%) with Grades 1-2 CRS were rechallenged using ICIs without event. 9 patients (36.0%) were treated with pulse methylprednisolone and 6 patients (24.0%) were treated with tocilizumab. Despite this, 3 patients (50%) who received tocilizumab had fatal (Grade 5) outcomes from ICI-induced CRS. Longer time to fever onset, lower platelet count and higher urea at presentation were associated with Grade 3-5 CRS. These parameters may be used to predict which patients are likely to develop severe CRS.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Cytokine Release Syndrome/chemically induced , Cytokine Release Syndrome/drug therapy , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy/adverse effects , Methylprednisolone/administration & dosage , Neoplasms/therapy , Severity of Illness Index , Aged , Biomarkers/blood , Cytokine Release Syndrome/blood , Fatal Outcome , Female , Humans , Male , Middle Aged , Pulse Therapy, Drug/methods , Retrospective Studies , Tertiary Care Centers , Treatment Outcome
16.
Arthritis Rheum ; 62(11): 3436-46, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20722038

ABSTRACT

OBJECTIVE: CXCR4 is a chemokine with multiple effects on the immune system. In murine lupus models, we demonstrated that monocytes, neutrophils, and B cells overexpressed CXCR4 and that its ligand, CXCL12, was up-regulated in diseased kidneys. We undertook this study to determine whether CXCR4 expression was increased in peripheral blood leukocytes from patients with systemic lupus erythematosus (SLE) and whether CXCL12 expression was increased in kidneys from patients with SLE. METHODS: Peripheral blood leukocytes from 31 SLE patients, 8 normal controls, and 9 patients with rheumatoid arthritis were prospectively analyzed by flow cytometry for CXCR4 expression. Biopsy samples (n = 14) from patients with lupus nephritis (LN) were immunostained with anti-CXCL12 antibody. RESULTS: CD19+ B cells and CD4+ T cells from SLE patients displayed a >2-fold increase (P = 0.0001) and >3-fold increase (P < 0.0001), respectively, in median CXCR4 expression compared with that in controls (n = 7-8). Moreover, CXCR4 expression on B cells was 1.61-fold higher in patients with SLE Disease Activity Index (SLEDAI) scores >10 (n = 8) than in patients with SLEDAI scores ≤10 (n = 16) (P = 0.0008), 1.71-fold higher in patients with class IV LN (n = 5) than in patients with other classes of LN (n = 7) (P = 0.02), and 1.40-fold higher in patients with active neuropsychiatric SLE (NPSLE) (n = 6) than in patients with inactive NPSLE (n = 18) (P = 0.01). CXCL12 was significantly up-regulated in the tubules and glomeruli of kidneys in patients with LN (n = 14), with the percentage of positive cells correlating positively with the severity of LN. CONCLUSION: CXCR4 appears to be up-regulated in multiple leukocyte subsets in SLE patients. The heightened expression of CXCR4 on B cells in active NPSLE and of CXCL12 in nephritic kidneys suggests that the CXCR4/CXCL12 axis might be a potential therapeutic target for SLE patients with kidney and/or central nervous system involvement.


Subject(s)
Chemokine CXCL12/metabolism , Lupus Erythematosus, Systemic/metabolism , Receptors, CXCR4/metabolism , Adolescent , Adult , Aged , Chemokine CXCL12/immunology , Female , Flow Cytometry , Humans , Immunohistochemistry , Leukocytes/immunology , Leukocytes/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/immunology , Lupus Nephritis/metabolism , Male , Middle Aged , Receptors, CXCR4/immunology , Up-Regulation/immunology
17.
J Immunol ; 182(5): 2700-7, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234164

ABSTRACT

Preterm birth occurs at a rate of 12.7% in the U.S. and is the primary cause of fetal morbidity in the first year of life as well as the cause of later health problems. Elucidation of mechanisms controlling cervical remodeling is critical for development of therapies to reduce the incidence of prematurity. The cervical extracellular matrix must be disorganized during labor to allow birth, followed by a rapid repair postpartum. Leukocytes infiltrate the cervix before and after birth and are proposed to regulate matrix remodeling during cervical ripening via release of proteolytic enzymes. In the current study, flow cytometry and cell sorting were used to determine the role of immune cells in cervical matrix remodeling before, during, and after parturition. Markers of myeloid cell differentiation and activation were assessed to define phenotype and function. Tissue monocytes and eosinophils increased in the cervix before birth in a progesterone-regulated fashion, whereas macrophage numbers were unchanged. Neutrophils increased in the postpartum period. Increased mRNA expression of Csfr1 and markers of alternatively activated M2 macrophages during labor or shortly postpartum suggest a function of M2 macrophages in postpartum tissue repair. Changes in cervical myeloid cell numbers are not reflected in the peripheral blood. These data along with our previous studies suggest that myeloid-derived cells do not orchestrate processes required for initiation of cervical ripening before birth. Additionally, macrophages with diverse phenotypes (M1 and M2) are present in the cervix and are most likely involved in the postpartum repair of tissue.


Subject(s)
Cervix Uteri/cytology , Cervix Uteri/immunology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Parturition/immunology , Parturition/metabolism , Animals , Animals, Newborn , Cell Movement/immunology , Cervix Uteri/metabolism , Eosinophils/immunology , Eosinophils/metabolism , Female , Immunophenotyping , Inflammation Mediators/metabolism , Leukocyte Count , Macrophage Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Myeloid Cells/cytology , Pregnancy , Time Factors , Wound Healing/immunology
18.
J Immunol ; 182(7): 4448-58, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19299746

ABSTRACT

Among various surface molecules screened, CXCR4 was significantly up-regulated on monocytes, neutrophils, B cell subsets, and plasma cells in multiple murine models of lupus with active nephritis, including B6.Sle1Yaa, BXSB, and MRL.lpr. TLR-mediated signaling and inflammatory cytokines accounted in part for this increase. Increased CXCR4 expression was associated with functional consequences, including increased migration and enhanced B cell survival. Simultaneously, the ligand for CXCR4, CXCL12, was significantly up-regulated in the nephritic kidneys. Treatment with a peptide antagonist of CXCR4 prolonged survival and reduced serum autoantibodies, splenomegaly, intrarenal leukocyte trafficking, and end organ disease in a murine model of lupus. These findings underscore the pathogenic role of CXCR4/CXCL12 in lymphoproliferative lupus and lupus nephritis and highlight this axis as a promising therapeutic target in this disease.


Subject(s)
Chemokine CXCL12/biosynthesis , Leukocytes/immunology , Lupus Erythematosus, Systemic/immunology , Receptors, CXCR4/biosynthesis , Animals , Chemokine CXCL12/immunology , Chemotaxis, Leukocyte/immunology , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunohistochemistry , Inflammation/etiology , Inflammation/immunology , Inflammation/metabolism , Leukocytes/metabolism , Lupus Erythematosus, Systemic/complications , Lupus Nephritis/etiology , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Male , Mice , Receptors, CXCR4/immunology , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Up-Regulation
19.
J Immunol ; 183(10): 6831-8, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19864599

ABSTRACT

Increased Type I IFNs or IFN-I have been associated with human systemic lupus erythematosus. Interestingly augmenting or negating IFN-I activity in murine lupus not only modulates systemic autoimmunity, but also impacts lupus nephritis, suggesting that IFN-I may be acting at the level of the end-organ. We find resident renal cells to be a dominant source of IFN-I in an experimental model of autoantibody-induced nephritis. In this model, augmenting IFN-I amplified antibody-triggered nephritis, whereas ablating IFN-I activity ameliorated disease. One mechanism through which increased IFN-I drives immune-mediated nephritis might be operative through increased recruitment of inflammatory monocytes and neutrophils, though this hypothesis needs further validation. Collectively, these studies indicate that an important contribution of IFN-I toward the disease pathology seen in systemic autoimmunity may be exercised at the level of the end-organ.


Subject(s)
Cytokines/immunology , Glomerulonephritis/immunology , Interferon Type I/immunology , Kidney Failure, Chronic/immunology , Receptor, Interferon alpha-beta/immunology , Animals , Antiviral Agents/immunology , Antiviral Agents/pharmacology , Autoantibodies/blood , Autoantibodies/drug effects , Cytokines/drug effects , Cytokines/metabolism , Disease Models, Animal , Glomerulonephritis/metabolism , Humans , Interferon Type I/pharmacology , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Kidney Failure, Chronic/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Recombinant Proteins
20.
Curr Protoc ; 1(11): e272, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34748281

ABSTRACT

Systemic Lupus Erythematosus (SLE) is a complex and heterogenous autoimmune disease, where genetics, immunology, and environmental factors all play a role. Murine models have contributed critical information on mechanisms of disease and prospective therapeutics. The key features that have been used to study the disease include the development of anti-nuclear autoantibodies (ANAs), splenomegaly, and kidney disease. The loss of tolerance and subsequent autoimmune features, and the progression to severe disease, are all dependent on immune dysregulation. In this article, we will describe the methods used to evaluate the underlying immunological features of the disease, as a more sensitive strategy to understand the disease itself and the mechanisms of potential novel therapeutics. © 2021 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: End study protocols for tissue harvesting Basic Protocol 2: End study protocols for tissue processing Basic Protocol 3: Immunophenotyping using flow cytometry protocols Support Protocol: Tissue processing for cold storage Basic Protocol 4: Additional tissue processing for later analyses Basic Protocol 5: Analysis of serum auto-antibodies by ELISAs (ANAs, snRNP, and dsDNA).


Subject(s)
Autoimmune Diseases , Lupus Erythematosus, Discoid , Lupus Erythematosus, Systemic , Animals , Autoantibodies , Disease Models, Animal , Mice
SELECTION OF CITATIONS
SEARCH DETAIL