1
|
Larsen ML, Nørgaard L, Linge P, Larsen JB, Langkilde HZ, Hauge EM, Thiel S, Voss A, Bengtsson A, Troldborg A. Molecular mechanisms underlying thrombosis in systemic lupus erythematosus - A Systematic review. Semin Arthritis Rheum 2025; 72:152707. [PMID: 40086157 DOI: 10.1016/j.semarthrit.2025.152707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025]
Abstract
Patients with systemic lupus erythematosus (SLE) face an approximately 30 % risk of thrombosis post-diagnosis. However, there remains significant knowledge gaps regarding causative mechanisms, and there is a lack of specific antithrombotic guidelines. This systematic review aims to examine the existing literature regarding the mechanisms contributing to thrombosis risk in SLE, focusing on five predefined procoagulant domains: autoantibodies (including antiphospholipid antibodies (aPL)), the complement system, platelets, the endothelium, and the coagulation system. The review followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statements and searched in PubMed and Embase without time restrictions. Risk of bias assessment was performed using a pre-specified evaluation tool. Out of 3,747 initially identified publications, 30 studies were included, with 28 demonstrating robust methodological quality in the risk of bias assessment. The studies were experimental, involving blood samples from cross-sectional SLE cohorts, except one animal -and one case-control study. We identified six different thrombosis mechanisms of action. Most studies concentrated on autoantibodies, predominantly aPL. Shared mechanisms between aPL and other autoantibodies may account for the increased thrombosis risk in aPL-negative SLE patients. Significant knowledge gaps remain, particularly regarding the role of the complement system in SLE-related thrombosis. Also, most research relies on cross-sectional designs, emphasizing the need for prospective cohort studies to better assess clinical factors. Finally, comprehensive studies examining the interactions between multiple procoagulant factors and their link to thrombosis are lacking. Closing these gaps in future research could improve both preventive and personalized treatment strategies for thrombosis in SLE.
Collapse
Affiliation(s)
- Mads L Larsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark.
| | - Laura Nørgaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Petrus Linge
- Department of Clinical Sciences, Rheumatology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Julie B Larsen
- Department of Clinical Biochemistry, Regional Hospital Horsens, Horsens, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Henrik Z Langkilde
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Rheumatology, Odense University Hospital, Odense, Denmark
| | - Ellen M Hauge
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Steffen Thiel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Anne Voss
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Rheumatology, Odense University Hospital, Odense, Denmark
| | - Anders Bengtsson
- Department of Clinical Sciences, Rheumatology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Anne Troldborg
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
2
|
Zheng D, Li X, Wang P, Zhu Q, Huang Z, Zhao T. Exploring the shared mechanism of fatigue between systemic lupus erythematosus and myalgic encephalomyelitis/chronic fatigue syndrome: monocytic dysregulation and drug repurposing. Front Immunol 2025; 15:1440922. [PMID: 39845969 PMCID: PMC11752880 DOI: 10.3389/fimmu.2024.1440922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025] Open
Abstract
Background SLE and ME/CFS both present significant fatigue and share immune dysregulation. The mechanisms underlying fatigue in these disorders remain unclear, and there are no standardized treatments. This study aims to explore shared mechanisms and predict potential therapeutic drugs for fatigue in SLE and ME/CFS. Methods Genes associated with SLE and ME/CFS were collected from disease target and clinical sample databases to identify overlapping genes. Bioinformatics analyses, including GO, KEGG, PPI network construction, and key target identification, were performed. ROC curve and correlation analysis of key targets, along with single-cell clustering, were conducted to validate their expression in different cell types. Additionally, an inflammation model was established using THP-1 cells to simulate monocyte activation in both diseases in vitro, and RT-qPCR was used to validate the expression of the key targets. A TF-mRNA-miRNA co-regulatory network was constructed, followed by drug prediction and molecular docking. Results Fifty-eight overlapping genes were identified, mainly involved in innate immunity and inflammation. Five key targets were identified (IL1β, CCL2, TLR2, STAT1, IFIH1). Single-cell sequencing revealed that monocytes are enriched with these targets. RT-qPCR confirmed significant upregulation of these targets in the model group. A co-regulatory network was constructed, and ten potential drugs, including suloctidil, N-Acetyl-L-cysteine, simvastatin, ACMC-20mvek, and camptothecin, were predicted. Simvastatin and camptothecin showed high affinity for the key targets. Conclusion SLE and ME/CFS share immune and inflammatory pathways. The identified key targets are predominantly enriched in monocytes at the single-cell level, suggesting that classical monocytes may be crucial in linking inflammation and fatigue. RT-qPCR confirmed upregulation in activated monocytes. The TF-mRNA-miRNA network provides a foundation for future research, and drug prediction suggests N-Acetyl-L-cysteine and camptothecin as potential therapies.
Collapse
Affiliation(s)
- Daisi Zheng
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaolong Li
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Peicheng Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Qingmiao Zhu
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhiyan Huang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Ting Zhao
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
3
|
Abernathy-Close L, Mears J, Billi AC, Sirobhushanam S, Berthier C, Lu A, Zhang Z, Hurst A, Gudjonsson JE, Kahlenberg JM. Topical Mupirocin Treatment Reduces Interferon and Myeloid Signatures in Cutaneous Lupus Erythematous Lesions Through Targeting of Staphyloccal Species. Arthritis Rheumatol 2024. [PMID: 39648343 DOI: 10.1002/art.43079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/10/2024]
Abstract
OBJECTIVE Cutaneous lupus erythematosus (CLE) is an inflammatory skin manifestation of systemic lupus erythematosus. Type I interferons (IFNs) promote inflammatory responses and are elevated in CLE lesions. We recently reported that CLE lesions are frequently colonized with Staphylococcus aureus. Here, we follow up via a proof-of-concept study to investigate whether type I IFN and inflammatory gene signatures in CLE lesions can be modulated with mupirocin, a topical antibiotic treatment against S aureus-mediated skin infections. METHODS Participants with active CLE lesions (n = 12) were recruited and randomized into a week of topical treatment with either 2% mupirocin or petroleum jelly vehicle. Paired samples were collected before and after seven days of treatment to assess microbial lesional skin responses. Microbial samples from nares and lesional skin were used to determine baseline and posttreatment Staphylococcus abundance and microbial community profiles by 16S ribosomal RNA gene sequencing. Inflammatory responses were evaluated by bulk RNA sequencing of lesional skin biopsies. RESULTS We identified 173 differentially expressed genes in CLE lesions after topical mupirocin treatment. Decreased lesional Staphylococcus burden correlated with decreased IFN pathway signaling and inflammatory gene expression and barrier dysfunction. Interestingly, mupirocin treatment lowered skin monocyte levels, and this mupirocin-associated depletion of monocytes correlated with decreased inflammatory gene expression. CONCLUSION Mupirocin treatment decreased lesional Staphylococcus, and this correlated with decreased IFN signaling and inflammatory gene expression. This study suggests a topical antibiotic could be employed to decrease lupus skin inflammation and type I IFN responses by reducing Staphylococcus colonization.
Collapse
Affiliation(s)
| | | | | | | | | | - Annie Lu
- University of Michigan, Ann Arbor
| | | | | | | | | |
Collapse
|
4
|
Zinellu A, Mangoni AA. A systematic review and meta-analysis of the association between the D-dimer and rheumatic diseases. Immun Inflamm Dis 2024; 12:e1349. [PMID: 39056561 PMCID: PMC11273555 DOI: 10.1002/iid3.1349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/30/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
INTRODUCTION There is good evidence that specific autoimmune rheumatic diseases (RDs), for example, rheumatoid arthritis and systemic lupus erythematosus (SLE), are associated with a state of hypercoagulability and an increased risk of venous thromboembolism (VTE). However, limited information regarding this association is available for other autoimmune or autoinflammatory RDs. We sought to address this issue by conducting a systematic review and meta-analysis of the association between the d-dimer, an established marker of hypercoagulability and VTE, and RDs and the possible clinical and demographic factors mediating this association. METHODS We searched the electronic databases PubMed, Web of Science, and Scopus from inception to January 31, 2024. The risk of bias and the certainty of evidence were assessed using the Joanna Briggs Institute Critical Appraisal Checklist and GRADE, respectively. RESULTS In 31 studies selected for analysis (2724 RD patients and 3437 healthy controls), RD patients had overall significantly higher d-dimer concentrations when compared to controls (standard mean difference = 0.93, 95% CI 0.76-1.10, p < .001; I2 = 86.1%, p < .001; moderate certainty of evidence). The results were stable in a sensitivity analysis. Significant associations were observed between the effect size of the between-group differences in d-dimer concentration and age, specific RD and RD category, RD duration, fibrinogen, plasminogen activator inhibitor, C-reactive protein, and erythrocyte sedimentation rate. CONCLUSIONS Overall, patients with RDs have significantly higher d-dimer concentrations when compared with healthy controls, indicating a state of hypercoagulability. The alterations in d-dimer concentrations are mediated by age, specific RD and RD category, RD duration, and markers of anticoagulation and inflammation. Further research is warranted to investigate d-dimer concentrations across the spectrum of RDs and their utility in predicting and managing VTE in these patients (PROSPERO registration number: CRD42024517712).
Collapse
Affiliation(s)
- Angelo Zinellu
- Department of Biomedical SciencesUniversity of SassariSassariItaly
| | - Arduino A. Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
- Department of Clinical PharmacologyFlinders Medical Centre, Southern Adelaide Local Health NetworkAdelaideAustralia
| |
Collapse
|
5
|
Li Z, Wang Z, Sun T, Liu S, Ding S, Sun L. Identifying key genes in CD4+ T cells of systemic lupus erythematosus by integrated bioinformatics analysis. Front Genet 2022; 13:941221. [PMID: 36046235 PMCID: PMC9420982 DOI: 10.3389/fgene.2022.941221] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by excessive activation of T and B lymphocytes and breakdown of immune tolerance to autoantigens. Despite several mechanisms including the genetic alterations and inflammatory responses have been reported, the overall signature genes in CD4+ T cells and how they affect the pathological process of SLE remain to be elucidated. This study aimed to identify the crucial genes, potential biological processes and pathways underlying SLE pathogenesis by integrated bioinformatics. The gene expression profiles of isolated peripheral CD4+ T cells from SLE patients with different disease activity and healthy controls (GSE97263) were analyzed, and 14 co-expression modules were identified using weighted gene co-expression network analysis (WGCNA). Some of these modules showed significantly positive or negative correlations with SLE disease activity, and primarily enriched in the regulation of type I interferon and immune responses. Next, combining time course sequencing (TCseq) with differentially expressed gene (DEG) analysis, crucial genes in lupus CD4+ T cells were revealed, including some interferon signature genes (ISGs). Among these genes, we identified 4 upregulated genes (PLSCR1, IFI35, BATF2 and CLDN5) and 2 downregulated genes (GDF7 and DERL3) as newfound key genes. The elevated genes showed close relationship with the SLE disease activity. In general, our study identified 6 novel biomarkers in CD4+ T cells that might contribute to the diagnosis and treatment of SLE.
Collapse
Affiliation(s)
- Zutong Li
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhilong Wang
- Department of Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Tian Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Shanshan Liu
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Shuai Ding
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- *Correspondence: Lingyun Sun, ; Shuai Ding,
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- *Correspondence: Lingyun Sun, ; Shuai Ding,
| |
Collapse
|
6
|
Dal Col J, Lamberti MJ, Nigro A, Casolaro V, Fratta E, Steffan A, Montico B. Phospholipid scramblase 1: a protein with multiple functions via multiple molecular interactors. Cell Commun Signal 2022; 20:78. [PMID: 35650588 PMCID: PMC9158361 DOI: 10.1186/s12964-022-00895-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/30/2022] [Indexed: 01/18/2023] Open
Abstract
Phospholipid scramblase 1 (PLSCR1) is the most studied protein of the scramblase family. Originally, it was identified as a membrane protein involved in maintaining plasma membrane asymmetry. However, studies conducted over the past few years have shown the involvement of PLSCR1 in several other cellular pathways. Indeed, PLSCR1 is not only embedded in the plasma membrane but is also expressed in several intracellular compartments where it interacts with a diverse repertoire of effectors, mediators, and regulators contributing to distinct cellular processes. Although most PLSCR1 interactors are thought to be cell-type specific, PLSCR1 often exerts its regulatory functions through shared mechanisms, including the trafficking of different molecules within intracellular vesicles such as endosomes, liposomes, and phagosomes. Intriguingly, besides endogenous proteins, PLSCR1 was also reported to interact with exogenous viral proteins, thereby regulating viral uptake and spread. This review aims to summarize the current knowledge about the multiple roles of PLSCR1 in distinct cellular pathways. Video Abstract
Collapse
Affiliation(s)
- Jessica Dal Col
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy.
| | - Marìa Julia Lamberti
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy.,INBIAS, CONICET-UNRC, Río Cuarto, Córdoba, Argentina
| | - Annunziata Nigro
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | - Elisabetta Fratta
- Division of Immunopathology and Cancer Biomarkers, Centro Di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Agostino Steffan
- Division of Immunopathology and Cancer Biomarkers, Centro Di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Barbara Montico
- Division of Immunopathology and Cancer Biomarkers, Centro Di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy.
| |
Collapse
|
7
|
Borchert C, Herman A, Roth M, Brooks AC, Friedenberg SG. RNA sequencing of whole blood in dogs with primary immune-mediated hemolytic anemia (IMHA) reveals novel insights into disease pathogenesis. PLoS One 2020; 15:e0240975. [PMID: 33091028 PMCID: PMC7580939 DOI: 10.1371/journal.pone.0240975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/06/2020] [Indexed: 11/29/2022] Open
Abstract
Immune-mediated hemolytic anemia (IMHA) is a life-threatening autoimmune disorder characterized by a self-mediated attack on circulating red blood cells. The disease occurs naturally in both dogs and humans, but is significantly more prevalent in dogs. Because of its shared features across species, dogs offer a naturally occurring model for studying IMHA in people. In this study, we used RNA sequencing of whole blood from treatment-naïve dogs to study transcriptome-wide changes in gene expression in newly diagnosed animals compared to healthy controls. We found many overexpressed genes in pathways related to neutrophil function, coagulation, and hematopoiesis. In particular, the most highly overexpressed gene in cases was a phospholipase scramblase, which mediates the externalization of phosphatidylserine from the inner to the outer leaflet of cell membranes. This family of genes has been shown to be critically important for programmed cell death of erythrocytes as well as the initiation of the clotting cascade. Unexpectedly, we found marked underexpression of many genes related to lymphocyte function. We also identified groups of genes that are highly associated with the inflammatory response and red blood cell regeneration in affected dogs. We did not find any genes that distinguished dogs that lived vs. those that died at 30 days following diagnosis, nor did we find any relevant genomic signatures of microbial organisms in the blood of affected animals. Future studies are warranted to validate these findings and assess their implication in developing novel therapeutic approaches for dogs and humans with IMHA.
Collapse
Affiliation(s)
- Corie Borchert
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota, United States of America
| | - Adam Herman
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Megan Roth
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota, United States of America
| | - Aimee C. Brooks
- Department of Veterinary Clinical Sciences, Purdue University College of Veterinary Medicine, West Lafayette, Indiana, United States of America
| | - Steven G. Friedenberg
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota, United States of America
| |
Collapse
|
8
|
Transient Receptor Potential Canonical 5-Scramblase Signaling Complex Mediates Neuronal Phosphatidylserine Externalization and Apoptosis. Cells 2020; 9:cells9030547. [PMID: 32110987 PMCID: PMC7140530 DOI: 10.3390/cells9030547] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 12/24/2022] Open
Abstract
Phospholipid scramblase 1 (PLSCR1), a lipid-binding and Ca2+-sensitive protein located on plasma membranes, is critically involved in phosphatidylserine (PS) externalization, an important process in cell apoptosis. Transient receptor potential canonical 5 (TRPC5), is a nonselective Ca2+ channel in neurons that interacts with many downstream molecules, participating in diverse physiological functions including temperature or mechanical sensation. The interaction between TRPC5 and PLSCR1 has never been reported. Here, we showed that PLSCR1 interacts with TRPC5 through their C-termini in HEK293 cells and mouse cortical neurons. Formation of TRPC5-PLSCR1 complex stimulates PS externalization and promotes cell apoptosis in HEK293 cells and mouse cerebral neurons. Furthermore, in vivo studies showed that PS externalization in cortical neurons induced by artificial cerebral ischemia-reperfusion was reduced in TRPC5 knockout mice compared to wild-type mice, and that the percentage of apoptotic neurons was also lower in TRPC5 knockout mice than in wild-type mice. Collectively, the present study suggested that TRPC5-PLSCR1 is a signaling complex mediating PS externalization and apoptosis in neurons and that TRPC5 plays a pathological role in cerebral-ischemia reperfusion injury.
Collapse
|
9
|
Attenuation of endothelial phosphatidylserine exposure decreases ischemia-reperfusion induced changes in microvascular permeability. J Trauma Acute Care Surg 2019. [PMID: 29538229 DOI: 10.1097/ta.0000000000001891] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Translocation of phosphatidylserine from the inner leaflet to the outer leaflet of the endothelial membrane via phospholipid scramblase-1 (PLSCR1) is an apoptotic signal responsible for the loss of endothelial barrier integrity after ischemia-reperfusion injury (IRI). We hypothesized that inhibiting phosphatidylserine expression on endothelial cells would attenuate IRI induced increases in hydraulic permeability (Lp). METHODS Mesenteric Lp was measured in rat post-capillary mesenteric venules subjected to IRI via superior mesenteric artery (SMA) occlusion (45 minutes) and release (300 minutes) in conjunction with several inhibitors of phosphatidylserine exposure as follows: (1) inhibition of PLSCR1 translocation (dithioerythritol, n = 3), (2) inhibition of PLSCR1 membrane trafficking (2-bromopalmitate [2-BP], n = 3), and (3) inhibition of ion exchange necessary for PLSCR1 function (4,4'-Diisothiocyano-2,2'-stilbenedisulfonic acid [DIDS], n = 3). Under the same IRI conditions, rats were also administered targeted inhibitors of phosphatidylserine exposure including knockdown of PLSCR1 (n = 3) using RNA interference (RNAi), and as a potential therapeutic tool Diannexin, a selective phosphatidylserine blocker (n = 3). RESULTS During IRI net Lp increased by 80% (p < 0.01). Net reductions of Lp were accomplished by 2-BP (46% reduction, p = 0.005), combined DET + 2-BP + DIDS (32% reduction, p = 0.04), RNAi (55% reduction, p = 0.002), Diannexin administered pre-SMA artery occlusion (73% reduction, p = 0.001), and post-SMA occlusion (70% reduction, p = 0.002). CONCLUSION Phosphatidylserine exposure is a key event in the pathogenesis of microvascular dysfunction during IRI. Clinically, inhibition of phosphatidylserine exposure is a promising strategy that may 1 day be used to mitigate the effects of IRI.
Collapse
|
10
|
Joseph S, George NI, Green-Knox B, Treadwell EL, Word B, Yim S, Lyn-Cook B. Epigenome-wide association study of peripheral blood mononuclear cells in systemic lupus erythematosus: Identifying DNA methylation signatures associated with interferon-related genes based on ethnicity and SLEDAI. J Autoimmun 2018; 96:147-157. [PMID: 30301579 DOI: 10.1016/j.jaut.2018.09.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/19/2018] [Accepted: 09/21/2018] [Indexed: 12/24/2022]
Abstract
Systemic lupus erythematosus (SLE or lupus) is a heterogeneous autoimmune disease characterized by the involvement of multiple organs and the production of antinuclear antibodies. DNA methylation plays an important role in the pathogenesis of lupus. We have performed an epigenome-wide DNA methylation study in lupus and healthy control (non-lupus) subjects to identify epigenetic patterns in lupus characterized ethnicity and SLE disease activity index (SLEDAI). A total of fifty-seven lupus patients (39 African American (AA) and 18 European American (EA)) and 33 healthy controls (17 AA and 16 EA) were studied. Differential DNA methylation between lupus patients and controls was assessed for approximately 485,000 CpG sites across the genome. We identified 41 differentially methylated sites (associated with 30 genes) between lupus and control s subjects, 85% of which were hypomethylated. Significant hypomethylation of differentially methylated sites was associated with several interferon-related genes, including MX1, IFI44L, PARP9, DT3XL, IFIT1, IFI44, RSAD2, PLSCR1, and IRF7. Several of these associated genes were also hypomethylated in comparisons between AA lupus and AA non-lupus subjects and between lupus patients with SLEDAI>6 and non-lupus subjects. Our analysis of gene expression data through RT-PCR confirmed these findings. Thus, the results indicate epigenetics susceptibility in lupus, which may be associated with SLEDAI score and ethnicity. In addition, our findings support the importance of the Type 1 interferon pathway in lupus pathogenesis.
Collapse
Affiliation(s)
- Stancy Joseph
- Division of Biochemical Toxicology, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, USA
| | - Nysia I George
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, USA
| | - Bridgett Green-Knox
- Division of Systems Biology, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, USA
| | | | - Beverly Word
- Division of Biochemical Toxicology, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, USA
| | - Sarah Yim
- Center for Drug Evaluation, White Oak, MD, USA
| | - Beverly Lyn-Cook
- Division of Biochemical Toxicology, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, USA.
| |
Collapse
|
11
|
Andrianova IA, Ponomareva AA, Litvinov RI. Structural Alterations of Monocytes in Systemic Lupus Erythematosus. BIONANOSCIENCE 2017. [DOI: 10.1007/s12668-017-0441-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
12
|
Tufail Y, Cook D, Fourgeaud L, Powers CJ, Merten K, Clark CL, Hoffman E, Ngo A, Sekiguchi KJ, O'Shea CC, Lemke G, Nimmerjahn A. Phosphatidylserine Exposure Controls Viral Innate Immune Responses by Microglia. Neuron 2017; 93:574-586.e8. [PMID: 28111081 DOI: 10.1016/j.neuron.2016.12.021] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/17/2016] [Accepted: 12/15/2016] [Indexed: 01/09/2023]
Abstract
Microglia are the intrinsic immune sentinels of the central nervous system. Their activation restricts tissue injury and pathogen spread, but in some settings, including viral infection, this response can contribute to cell death and disease. Identifying mechanisms that control microglial responses is therefore an important objective. Using replication-incompetent adenovirus 5 (Ad5)-based vectors as a model, we investigated the mechanisms through which microglia recognize and respond to viral uptake. Transgenic, immunohistochemical, molecular-genetic, and fluorescence imaging approaches revealed that phosphatidylserine (PtdSer) exposure on the outer leaflet of transduced cells triggers their engulfment by microglia through TAM receptor-dependent mechanisms. We show that inhibition of phospholipid scramblase 1 (PLSCR1) activity reduces intracellular calcium dysregulation, prevents PtdSer externalization, and enables months-long protection of vector-transduced, transgene-expressing cells from microglial phagocytosis. Our study identifies PLSCR1 as a potent target through which the innate immune response to viral vectors, and potentially other stimuli, may be controlled.
Collapse
Affiliation(s)
- Yusuf Tufail
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Daniela Cook
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Lawrence Fourgeaud
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Colin J Powers
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Katharina Merten
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Charles L Clark
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Elizabeth Hoffman
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Alexander Ngo
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Kohei J Sekiguchi
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Clodagh C O'Shea
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Greg Lemke
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; Immunobiology and Microbial Pathogenesis Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Axel Nimmerjahn
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
13
|
Herate C, Ramdani G, Grant NJ, Marion S, Gasman S, Niedergang F, Benichou S, Bouchet J. Phospholipid Scramblase 1 Modulates FcR-Mediated Phagocytosis in Differentiated Macrophages. PLoS One 2016; 11:e0145617. [PMID: 26745724 PMCID: PMC4712888 DOI: 10.1371/journal.pone.0145617] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 11/04/2015] [Indexed: 12/24/2022] Open
Abstract
Phospholipid Scramblase 1 (PLSCR1) was initially characterized as a type II transmembrane protein involved in bilayer movements of phospholipids across the plasma membrane leading to the cell surface exposure of phosphatidylserine, but other cellular functions have been ascribed to this protein in signaling processes and in the nucleus. In the present study, expression and functions of PLSCR1 were explored in specialized phagocytic cells of the monocyte/macrophage lineage. The expression of PLSCR1 was found to be markedly increased in monocyte-derived macrophages compared to undifferentiated primary monocytes. Surprisingly, this 3-fold increase in PLSCR1 expression correlated with an apparent modification in the membrane topology of the protein at the cell surface of differentiated macrophages. While depletion of PLSCR1 in the monocytic THP-1 cell-line with specific shRNA did not inhibit the constitutive cell surface exposure of phosphatidylserine observed in differentiated macrophages, a net increase in the FcR-mediated phagocytic activity was measured in PLSCR1-depleted THP-1 cells and in bone marrow-derived macrophages from PLSCR1 knock-out mice. Reciprocally, phagocytosis was down-regulated in cells overexpressing PLSCR1. Since endogenous PLSCR1 was recruited both in phagocytic cups and in phagosomes, our results reveal a specific role for induced PLSCR1 expression in the modulation of the phagocytic process in differentiated macrophages.
Collapse
Affiliation(s)
- Cecile Herate
- Inserm U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris-Descartes, Sorbonne Paris-Cité, Paris, France
| | - Ghania Ramdani
- Inserm U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris-Descartes, Sorbonne Paris-Cité, Paris, France
| | - Nancy J. Grant
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR3212, and Université de Strasbourg, Strasbourg, France
| | - Sabrina Marion
- Inserm U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris-Descartes, Sorbonne Paris-Cité, Paris, France
| | - Stephane Gasman
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR3212, and Université de Strasbourg, Strasbourg, France
| | - Florence Niedergang
- Inserm U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris-Descartes, Sorbonne Paris-Cité, Paris, France
| | - Serge Benichou
- Inserm U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris-Descartes, Sorbonne Paris-Cité, Paris, France
- * E-mail:
| | - Jerome Bouchet
- Inserm U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris-Descartes, Sorbonne Paris-Cité, Paris, France
| |
Collapse
|
14
|
Two c-Myc binding sites are crucial in upregulating the expression of human phospholipid scramblase 1 gene. Biochem Biophys Res Commun 2015; 469:412-7. [PMID: 26679604 DOI: 10.1016/j.bbrc.2015.11.131] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 11/29/2015] [Indexed: 11/21/2022]
Abstract
Human phospholipid scramblase 1 (hPLSCR1) is a type II endofacial membrane protein which mediates bi-directional transport of phospholipids across the plasma membrane. hPLSCR1, a multifunctional protein with variety of roles in apoptosis, tumor progression, cell signaling and anti-viral defense. The expression of such a multifunctional protein should be under tight regulation. Apart from a single report showing snail mediated down regulation of hPLSCR1, the molecular mechanisms regulating the expression of scramblases are not well elucidated. In this study we identified c-Myc as a transcriptional regulator of hPLSCR1. Transcription factor prediction tool ConSite predicted three binding sites for c-Myc. Reporter gene assays and western blot analysis revealed c-Myc mediated up regulation of hPLSCR1 expression. Deletion construct -790 lacking one c-Myc binding site showed a 27% decrease in promoter activity while deletion construct -469 lacking two c-Myc binding sites showed a 62% decrease in promoter activity. Site directed mutagenesis revealed the importance of c-Myc binding sites from -751 to -756 and -548 to -553 on the promoter of hPLSCR1in transcriptionally regulating the expression of hPLSCR1. The results were further confirmed by shRNA mediated knock down of endogenous c-Myc and in vivo interactions by ChIP assay.
Collapse
|
15
|
Kodigepalli KM, Bowers K, Sharp A, Nanjundan M. Roles and regulation of phospholipid scramblases. FEBS Lett 2014; 589:3-14. [PMID: 25479087 DOI: 10.1016/j.febslet.2014.11.036] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/22/2014] [Accepted: 11/24/2014] [Indexed: 02/06/2023]
Abstract
Phospholipid scramblase activity is involved in the collapse of phospholipid (PL) asymmetry at the plasma membrane leading to externalization of phosphatidylserine. This activity is crucial for initiation of the blood coagulation cascade and for recognition/elimination of apoptotic cells by macrophages. Efforts to identify gene products associated with this activity led to the characterization of PL scramblase (PLSCR) and XKR family members which contribute to phosphatidylserine exposure in response to apoptotic stimuli. Meanwhile, TMEM16 family members were identified to externalize phosphatidylserine in response to elevated calcium in Scott syndrome platelets, which is critical for activation of the coagulation cascade. Herein, we report their mechanisms of gene regulation, molecular functions independent of their scrambling activity, and their potential roles in pathogenic conditions.
Collapse
Affiliation(s)
- Karthik M Kodigepalli
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States
| | - Kiah Bowers
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States
| | - Arielle Sharp
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States
| | - Meera Nanjundan
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States.
| |
Collapse
|
16
|
Amengual O, Atsumi T, Oku K, Suzuki E, Horita T, Yasuda S, Koike T. Phospholipid scramblase 1 expression is enhanced in patients with antiphospholipid syndrome. Mod Rheumatol 2014. [DOI: 10.3109/s10165-012-0642-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Olga Amengual
- Department of Medicine II, Hokkaido University Graduate School of Medicine, N15 W7, Kita-ku, Sapporo 060-8638, Japan
| | - Tatsuya Atsumi
- Department of Medicine II, Hokkaido University Graduate School of Medicine, N15 W7, Kita-ku, Sapporo 060-8638, Japan
| | - Kenji Oku
- Department of Medicine II, Hokkaido University Graduate School of Medicine, N15 W7, Kita-ku, Sapporo 060-8638, Japan
| | - Eriko Suzuki
- Department of Medicine II, Hokkaido University Graduate School of Medicine, N15 W7, Kita-ku, Sapporo 060-8638, Japan
| | - Tetsuya Horita
- Department of Medicine II, Hokkaido University Graduate School of Medicine, N15 W7, Kita-ku, Sapporo 060-8638, Japan
| | - Shinsuke Yasuda
- Department of Medicine II, Hokkaido University Graduate School of Medicine, N15 W7, Kita-ku, Sapporo 060-8638, Japan
| | - Takao Koike
- Department of Medicine II, Hokkaido University Graduate School of Medicine, N15 W7, Kita-ku, Sapporo 060-8638, Japan
| |
Collapse
|
17
|
Amengual O, Atsumi T, Oku K, Suzuki E, Horita T, Yasuda S, Koike T. Phospholipid scramblase 1 expression is enhanced in patients with antiphospholipid syndrome. Mod Rheumatol 2012; 23:81-8. [PMID: 22526829 DOI: 10.1007/s10165-012-0642-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 03/22/2012] [Indexed: 10/28/2022]
Abstract
OBJECTIVE Thrombus formation is the key event of vascular manifestations in antiphospholipid syndrome (APS). Phosphatidylserine (PS) is normally sequestered in the inner leaflet of cell membranes. Externalization of PS occurs during cell activation and is essential for promoting blood coagulation and for the binding of antiphospholipid antibodies (aPL) to cells. One of the molecules involved in PS externalization is phospholipid scramblase 1 (PLSCR1). We evaluated PLSCR1 expression on monocytes from APS patients and analyzed the in vitro effect of monoclonal aPL on PLSCR1 expression. PATIENTS AND METHODS Forty patients with APS were investigated. In vitro experiments were performed in monocyte cell lines incubated with monoclonal aPL. PLSCR1 expression was determined by quantitative real-time polymerase chain reactions. PS exposure on CD14(+) cell surface was analyzed by flow cytometry. RESULTS Levels of full-length PLSCR1 messenger RNA (mRNA) were significantly increased in APS patients compared with healthy controls (2.4 ± 1.2 vs. 1.3 ± 0.4, respectively, p < 0.001). In cultured monocytes, interferon alpha enhanced tissue-factor expression mediated by β2-glycoprotein-I-dependent monoclonal anticardiolipin antibody. CONCLUSIONS Monocytes in APS patients had increased PLSCR1 mRNA expression.
Collapse
Affiliation(s)
- Olga Amengual
- Department of Medicine II, Hokkaido University Graduate School of Medicine, N15 W7, Kita-ku, Sapporo, 060-8638, Japan
| | | | | | | | | | | | | |
Collapse
|
18
|
Talukder AH, Bao M, Kim TW, Facchinetti V, Hanabuchi S, Bover L, Zal T, Liu YJ. Phospholipid scramblase 1 regulates Toll-like receptor 9-mediated type I interferon production in plasmacytoid dendritic cells. Cell Res 2012; 22:1129-39. [PMID: 22453241 DOI: 10.1038/cr.2012.45] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Toll-like receptor 9 (TLR9) senses microbial DNA in the endosomes of plasmacytoid dendritic cells (pDCs) and triggers MyD88-dependent type I interferon (IFN) responses. To better understand TLR9 biology in pDCs, we established a yeast two-hybrid library for the identification of TLR9-interacting proteins. Here, we report that an IFN-inducible protein, phospholipid scramblase 1 (PLSCR1), interacts with TLR9 in pDCs. Knockdown of PLSCR1 expression by siRNA in human pDC cell line led to a 60-70% reduction of IFN-α responses following CpG-ODN (oligodeoxynucleotide) stimulation. Primary pDCs from PLSCR1-deficient mice produced lower amount of type 1 IFN than pDCs from the wild-type mice in response to CpG-ODN, herpes simplex virus and influenza A virus. Following CpG-A stimulation, there were much lower amounts of TLR9 in the early endosomes together with CpG-A in pDCs from PLSCR1-deficient mice. Our study demonstrates that PLSCR1 is a TLR9-interacting protein that plays an important role in pDC's type 1 IFN responses by regulating TLR9 trafficking to the endosomal compartment.
Collapse
Affiliation(s)
- Amjad H Talukder
- Department of Immunology, Center for Cancer Immunology Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Yang J, Zhu X, Liu J, Ding X, Han M, Hu W, Wang X, Zhou Z, Wang S. Inhibition of Hepatitis B virus replication by phospholipid scramblase 1 in vitro and in vivo. Antiviral Res 2012; 94:9-17. [PMID: 22342889 DOI: 10.1016/j.antiviral.2012.01.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Revised: 01/20/2012] [Accepted: 01/30/2012] [Indexed: 01/03/2023]
Abstract
Human Phospholipid scramblase 1 (PLSCR1) is an α/β interferon-inducible protein that mediates antiviral activity against RNA viruses including vesicular stomatitis virus (VSV) and encephalomyocarditis virus (EMCV). In the present study, we investigated the antiviral activity of PLSCR1 protein against HBV (Hepatitis B virus). Firstly, PLSCR1 mRNA and protein expression was found to be downregulated in HepG2 cells after HBV infection. Then by performing co-transient-transfection experiments in cells and hydrodynamics-based transfection experiments in mice using a HBV expression plasmid and a PLSCR1 expression plasmid, we found that PLSCR1 inhibited HBV replication in vitro and in vivo through a significant reduction in the synthesis of viral proteins, DNA replicative intermediates and HBV RNAs. We also demonstrated that the antiviral action of PLSCR1 against HBV occurs, partly at least, by activating the Jak/Stat pathway. In conclusion, our results suggest that the expression of PLSCR1 is involved in HBV replication and that PLSCR1 has antiviral activity against HBV.
Collapse
Affiliation(s)
- Jing Yang
- Beijing Institute of Radiation Medicine, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|