1
|
Malkawi AK, Ohlund L, Rahman AMA, Sleno L, Siaj M. Co-stimulatory pathway competitive assay development using Liquid chromatography-tandem mass spectrometry (LC-MS/MS). J Pharm Biomed Anal 2024; 242:116034. [PMID: 38422671 DOI: 10.1016/j.jpba.2024.116034] [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: 08/16/2023] [Revised: 02/02/2024] [Accepted: 02/10/2024] [Indexed: 03/02/2024]
Abstract
T-cells play a significant role in the development of autoimmune diseases. The CD28-B7 costimulatory pathway is crucial for activating T-cells, and blocking this pathway is essential for treating autoimmune diseases. Therapeutic antibodies and fusion proteins that target costimulatory molecules like CD80, CD86, CTLA-4, and CD28 have been developed to explore the costimulation process and as targeted treatments. To advance our understanding of costimulation in autoimmunity and the inhibition of the costimulatory pathway, it is crucial to have an accurate, precise, and direct method for detecting and quantifying the soluble form of these molecules in body fluids and various biological systems. Herein, we developed a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for quantifying the four costimulatory proteins depending on the signature peptides derived from the soluble isoform of these proteins in multiple reaction monitoring (MRM) mode. The method was validated using the US FDA guidelines. The LOQ was determined as ∼0.5 nM for the four analytes, with quantification extended to 20 nM with a correlation coefficient of R2>0.998. The developed MRM method was used to analyze on-bead digested protein mixtures to establish a competitive assay for the CD28-B7 costimulatory pathway using CTLA4-Ig (Abatacept ™) as an FDA-approved drug for rheumatoid arthritis. The IC50 was determined to be 2.99 and 159.8 nM for sCD80 and sCD86, respectively. A straightforward MRM-based competitive assay will advance the knowledge about the costimulatory role in autoimmunity and the autoimmune therapeutic drug discovery, with the need for broad application on different in vitro and in vivo models to discover new targeted inhibitors.
Collapse
Affiliation(s)
- Abeer K Malkawi
- Department of Chemistry, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada
| | - Leanne Ohlund
- Department of Chemistry, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada
| | - Anas M Abdel Rahman
- Metabolomics Section, Department of Clinical Genomics, Center for Genome Medicine, King Faisal Specialist Hospital and Research Center (KFSHRC), Riyadh, Saudi Arabia; Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia; Department of Chemistry, Memorial University of Newfoundland, St. John's, NL AIC 5S7, Canada
| | - Lekha Sleno
- Department of Chemistry, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada
| | - Mohamed Siaj
- Department of Chemistry, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada.
| |
Collapse
|
2
|
Yi T, Zhang W, Hua Y, Xin X, Wu Z, Li Y, Wen C, Fan Y, Ji J, Xu L. Rutin alleviates lupus nephritis by inhibiting T cell oxidative stress through PPARγ. Chem Biol Interact 2024; 394:110972. [PMID: 38555047 DOI: 10.1016/j.cbi.2024.110972] [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: 01/27/2024] [Revised: 03/12/2024] [Accepted: 03/21/2024] [Indexed: 04/02/2024]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by complex clinical symptoms and multi-organ damage. One of the most prevalent complications of SLE is lupus nephritis (LN). Rutin, a natural flavonoid compound found in various plants used in traditional Chinese medicine, has shown promising anti-inflammatory, antioxidant, and renal protective effects. In our study, we treated MRL/lpr mice, a model known for spontaneously developing LN, with Rutin. Our findings reveal that Rutin markedly reduced serum cytokine and autoantibody levels and decreased inflammatory cell infiltration in renal tissues, thereby ameliorating kidney pathology. In vitro experiments indicated that Rutin's therapeutic effect on LN is linked to its significant reduction of oxidative stress in T cells. Further investigations suggest that Rutin enhances oxidative stress management through the modulation of Peroxisome proliferator-activated receptor gamma (PPARγ). We observed that Rutin modulates PPARγ activity, leading to reduced transcriptional activity of NF-κB and STAT3, which in turn inhibits the secretion of inflammatory cytokines such as IL-6, TNF-α, and IL-17. In summary, Rutin can exert an antioxidant effect by regulating PPARγ and shows therapeutic action against LN.
Collapse
Affiliation(s)
- Tongtong Yi
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China
| | - Wei Zhang
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China
| | - Ying Hua
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China
| | - Xingpan Xin
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China
| | - Zhenyu Wu
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China
| | - Ying Li
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China
| | - Chengping Wen
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China
| | - Yongsheng Fan
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China; Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinjun Ji
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China.
| | - Li Xu
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China.
| |
Collapse
|
3
|
Houssaini H, Bouallegui E, Abida O, Tahri S, Elloumi N, Hachicha H, Marzouk S, Bahloul Z, Masmoudi H, Fakhfakh R. ICOS gene polymorphisms in systemic lupus erythematosus: A case-control study. Int J Immunogenet 2023; 50:194-205. [PMID: 37338463 DOI: 10.1111/iji.12625] [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: 02/06/2023] [Revised: 05/23/2023] [Accepted: 06/11/2023] [Indexed: 06/21/2023]
Abstract
The inducible T-cell costimulator (ICOS) may play an important role in adaptive immunity by regulating the interaction between T cells and antigen-presenting cells. Disruption of this molecule can lead to autoimmune diseases, in particular systemic lupus erythematosus (SLE). In this study, we aimed to explore the possible association between ICOS gene polymorphisms and SLE as well as their influence on disease susceptibility and clinical outcomes. A further objective was to assess the potential impact of these polymorphisms on RNA expression. A case-control study, including 151 patients with SLE, and 291 unrelated healthy controls (HC) matched in gender, and geographical origin, was performed to genotype two polymorphisms located in the ICOS gene: rs11889031 (-693 G/A) and rs10932029 (IVS1 + 173 T/C); using the polymerase chain reaction (PCR)-restriction fragment length polymorphism method. The different genotypes were validated by direct sequencing. The expression level of ICOS mRNA was assessed by quantitative PCR in peripheral blood mononuclear cells of SLE patients and HC. The results were analysed using Shesis and spss.20. Our results revealed a significant association between ICOS gene rs11889031 > CC genotype and SLE disease (codominant genetic model 1, (C/C vs. C/T), p = .001, odds ratio [OR] = 2.18 IC [1.36-3.49]); codominant genetic model 2, (C/C vs. T/T) p = .007, OR = 15.29 IC [1.97-118.5]); dominant genetic model, (C/C vs. C/T + T/T) p = .0001, OR = 2.44 IC [1.53-3.9]). Besides, there was a marginal association between rs11889031 > TT genotype and T allele with a protective role from SLE (recessive genetic model, p = .016, OR = 0.08 IC [0.01-0.63] and p = 7.6904E - 05, OR = 0.43 IC = [0.28-0.66], respectively). Moreover, statistical analysis indicated that the rs11889031 > CC genotype was linked with clinical and serological manifestations of SLE, including blood pressure, and anti-SSA antibodies production in SLE patients. However, the ICOS gene rs10932029 polymorphism was not associated with susceptibility to SLE. On the other side, we did not note any effect of the two selected polymorphisms on the level of ICOS mRNA gene expression. The study showed a significant predisposing association of the ICOS rs11889031 > CC genotype with SLE, in contrast to a protective effect of rs11889031 > TT genotype in Tunisian patients. Our results suggest that ICOS rs11889031 may act as a risk factor for SLE and could be used as a genetic susceptibility biomarker.
Collapse
Affiliation(s)
- Hana Houssaini
- Autoimmunity, Cancer, and Immunogenetics Research Laboratory, LR18SP12, Habib Bourguiba University Hospital of Sfax, University of Sfax, Sfax, Tunisia
| | - Emna Bouallegui
- Autoimmunity, Cancer, and Immunogenetics Research Laboratory, LR18SP12, Habib Bourguiba University Hospital of Sfax, University of Sfax, Sfax, Tunisia
| | - Olfa Abida
- Autoimmunity, Cancer, and Immunogenetics Research Laboratory, LR18SP12, Habib Bourguiba University Hospital of Sfax, University of Sfax, Sfax, Tunisia
| | - Safa Tahri
- Autoimmunity, Cancer, and Immunogenetics Research Laboratory, LR18SP12, Habib Bourguiba University Hospital of Sfax, University of Sfax, Sfax, Tunisia
| | - Nesrine Elloumi
- Autoimmunity, Cancer, and Immunogenetics Research Laboratory, LR18SP12, Habib Bourguiba University Hospital of Sfax, University of Sfax, Sfax, Tunisia
| | - Hend Hachicha
- Autoimmunity, Cancer, and Immunogenetics Research Laboratory, LR18SP12, Habib Bourguiba University Hospital of Sfax, University of Sfax, Sfax, Tunisia
- Department of Immunology, Habib Bourguiba University Hospital, University of Sfax, Sfax, Tunisia
| | - Sameh Marzouk
- Internal Medicine Department, HediChaker University Hospital, University of Sfax, Sfax, Tunisia
| | - Zouhir Bahloul
- Internal Medicine Department, HediChaker University Hospital, University of Sfax, Sfax, Tunisia
| | - Hatem Masmoudi
- Autoimmunity, Cancer, and Immunogenetics Research Laboratory, LR18SP12, Habib Bourguiba University Hospital of Sfax, University of Sfax, Sfax, Tunisia
- Department of Immunology, Habib Bourguiba University Hospital, University of Sfax, Sfax, Tunisia
| | - Raouia Fakhfakh
- Autoimmunity, Cancer, and Immunogenetics Research Laboratory, LR18SP12, Habib Bourguiba University Hospital of Sfax, University of Sfax, Sfax, Tunisia
| |
Collapse
|
4
|
New biologics and targeted therapies in systemic lupus: From new molecular targets to new indications. A systematic review. Joint Bone Spine 2023; 90:105523. [PMID: 36623799 DOI: 10.1016/j.jbspin.2023.105523] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/02/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Despite available therapies, persistently active and corticosteroid-dependent Systemic Lupus Erythematosus (SLE) represent a significant therapeutic challenge. The purpose of this systematic review was to provide an updated view of targeted therapies currently in clinical development in SLE, with a special focus on the most promising ones. METHODS We performed a systematic review of targeted therapies in clinical development in SLE in clinicaltrials.gov (search date: 28th of August 2022). Targeted therapies (defined as drugs specifically designed to block certain molecules, receptors, or pathways involved in the development of SLE) were extracted. For each investigational drug, we considered only the study at the most advanced stage of clinical development. RESULTS The systematic review yielded a total of 92 targeted therapies (58 biological DMARDs [bDMARDs] and 34 targeted synthetic [ts]DMARDs) assessed in a total of 203 clinical trials. The candidate drugs reached phase I (n=20), Ia/IIb (n=6), phase II (n=51), phase II/III (n=1), phase III (n=13) and phase IV (n=1). These trials were reported as recruiting (n=31), active but not recruiting (n=8), not yet recruiting (n=4), enrolling by invitation (n=2), completed (n=31), prematurely terminated (n=12) and withdrawn in 1 (status unknown in 3). The main investigational drugs for SLE target inflammatory cytokines, chemokines or their receptors (n=19), intracellular signaling pathways (n=18), B cells (n=14) or plasma cells (n=7),T/B cells co-stimulation molecules (n=10), complement molecules (n=5),T lymphocytes (n=2), plasmacytoid dendritic cells (n=2), as well as various other immune targets (n=15). CONCLUSION The pipeline of investigational drugs in SLE is highly diversified and will hopefully enable more optimal Treat-To-Target with the goal of disease modification. Companion biomarkers will be needed to better characterized SLE heterogeneity and optimize treatment selection at the individual-patient level.
Collapse
|
5
|
Ceccarelli F, Natalucci F, Di Filippo A, Olivieri G, Napoletano C, Rughetti A, Nuti M, Zizzari IG, Conti F. Membrane and Soluble CD137 in Systemic Lupus Erythematosus: Role as Biomarkers for Disease Activity. J Immunol Res 2023; 2023:2344239. [PMID: 37114204 PMCID: PMC10129430 DOI: 10.1155/2023/2344239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 03/05/2023] [Accepted: 03/24/2023] [Indexed: 04/29/2023] Open
Abstract
Objective The role of T cells in the pathogenesis of systemic lupus erythematosus (SLE) has recently gained attention. Costimulatory molecules are membrane proteins strictly associated with T-cell receptor (TCR), acting by activating or inhibiting T cells and antigen-presenting cells (APC) through direct and reverse signaling, thus becoming responsible for the development of effector T cells or regulatory T cells. The primary objective of the present case-control study was to evaluate the cell membrane expression of CD137 on T cells and the serum concentration of CD137 (sCD137) in a SLE cohort. Materials We enrolled SLE patients and sex/age-matched healthy subjects (HS). Disease activity was assessed by SLEDAI-2K. By application of flow cytometry, we evaluated the expression of CD137 on CD4+ and CD8+ lymphocytes. ELISA test was performed to evaluate serum levels of sCD137. Results Twenty-one SLE patients (M/F 1/20; median age 48 years (IQR 17); median disease duration 144 months (IQR 204)) were evaluated. SLE patients showed %CD3+CD137+ cells significantly higher compared to HS (median 5.32 (IQR 6.11) versus 3.3 (IQR 1.8), p = 0.001). In SLE patients, %CD4+CD137+ cells positively correlated with SLEDAI-2K (p = 0.0082, r = 0.58, CI (0.15-0.82); indeed, %CD4+CD137+ cells were significantly lower in SLE patients with a remission status compared to those not reaching this condition (median 1.07 (IQR 0.91) versus 1.58 (IQR 2.42), p = 0.013). Accordingly, sCD137 levels were significantly lower in remission status (31.30 pg/mL (IQR 102.2 versus median 122.8 pg/mL (IQR 536); p = 0.03) and correlated with %CD4+CD137+ cells (p = 0.012, r = 0.60, CI (0.15-0.84)). Conclusion Our results suggest a possible involvement of CD137-CD137L axis in SLE pathogenesis, as demonstrated by higher expression of CD137 on CD4+ cells in SLE compared with HS. Furthermore, the positive correlation between SLEDAI-2K and membrane CD137 expression on CD4+ cells, as well as soluble CD137, indicates a possible use as biomarkers for disease activity.
Collapse
Affiliation(s)
- Fulvia Ceccarelli
- Lupus Clinic, Rheumatology Unit, Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesco Natalucci
- Lupus Clinic, Rheumatology Unit, Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Alessandra Di Filippo
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Giulio Olivieri
- Lupus Clinic, Rheumatology Unit, Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Chiara Napoletano
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Aurelia Rughetti
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Marianna Nuti
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Ilaria Grazia Zizzari
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Fabrizio Conti
- Lupus Clinic, Rheumatology Unit, Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
6
|
Gerasimova EV, Tabakov DV, Gerasimova DA, Popkova TV. Activation Markers on B and T Cells and Immune Checkpoints in Autoimmune Rheumatic Diseases. Int J Mol Sci 2022; 23:ijms23158656. [PMID: 35955790 PMCID: PMC9368764 DOI: 10.3390/ijms23158656] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022] Open
Abstract
In addition to identifying the major B- and T-cell subpopulations involved in autoimmune rheumatic diseases (ARDs), in recent years special attention has been paid to studying the expression of their activation markers and immune checkpoints (ICPs). The activation markers on B and T cells are a consequence of the immune response, and these molecules are considered as sensitive specific markers of ARD activity and as promising targets for immunotherapy. ICPs regulate the activation of the immune response by preventing the initiation of autoimmune processes, and they modulate it by reducing immune cell-induced organ and tissue damage. The article considers the possible correlation of ICPs with the activity of ARDs, the efficacy of specific ARD treatments, and the prospects for the use of activation molecules and activation/blocking ICPs for the treatment of ARDs.
Collapse
Affiliation(s)
- Elena V. Gerasimova
- Department of Systemic Rheumatic Diseases, V.A. Nasonova Research Institute of Rheumatology, Kashirskoe Shosse, 115522 Moscow, Russia
- Correspondence: ; Tel.: +7-905-538-0399
| | - Dmitry V. Tabakov
- Department of Systemic Rheumatic Diseases, V.A. Nasonova Research Institute of Rheumatology, Kashirskoe Shosse, 115522 Moscow, Russia
| | - Daria A. Gerasimova
- Department of Organization and Economy of Pharmacy, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2, Trubetskaya St., 119526 Moscow, Russia
| | - Tatiana V. Popkova
- Department of Systemic Rheumatic Diseases, V.A. Nasonova Research Institute of Rheumatology, Kashirskoe Shosse, 115522 Moscow, Russia
| |
Collapse
|
7
|
Li M, Zhang YJ, Liu DX, Liu Z, Fu M, Yang QR, Sun HS. Expression of caveolin family proteins in serum of patients with systemic lupus erythematosus. Lupus 2021; 30:1819-1828. [PMID: 34569384 DOI: 10.1177/09612033211035508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVES Caveolin family proteins, including caveolin-1 (Cav-1), caveolin-2 (Cav-2), and caveolin-3 (Cav-3), are identified as the principal protein components of caveolae in mammalian cells. Circulating form of caveolin family proteins can be used as a good potential biomarker for predicting disease. METHODS To investigate the clinical significance of the serological levels of caveolin family proteins in patients with systemic lupus erythematosus (SLE), we evaluated the soluble serum levels of caveolin family proteins in patients with SLE by enzyme-linked immunosorbent assay (ELISA) and assessed their associations with various known clinical variables. RESULTS The major findings of our study are as follows: Cav-2 was not detected in serum of SLE patients and normal controls (NCs). Serum Cav-1 and Cav-3 levels were higher in SLE patients compared with NCs. There were no significant correlations between serum Cav-1 and Cav-3 levels and SLE disease activity. Further analysis showed that serum Cav-3 may be more valuable as a marker than serum Cav-1 in SLE patients. CONCLUSION Serum levels of Cav-1 and Cav-3 might have a diagnostic role in patients with SLE. However, their predictive and prognostic value was not determined. Further studies are necessary to determine the potential clinical significance of these assays in SLE.
Collapse
Affiliation(s)
- Ming Li
- Department of Rheumatology and Immunology, 34708Shandong Provincial Hospital Affiliated to Shandong First Medical University (Shandong Provincial Hospital), Jinan, P.R. China
| | - Yi-Jing Zhang
- Department of Geriatric Gastroenterology, 34708Shandong Provincial Hospital Affiliated to Shandong First Medical University (Shandong Provincial Hospital), Jinan, P.R. China
| | - Dong-Xia Liu
- Department of Rheumatology and Immunology, 34708Shandong Provincial Hospital Affiliated to Shandong First Medical University (Shandong Provincial Hospital), Jinan, P.R. China
| | - Zhi Liu
- Department of Clinical Laboratory, 34708Shandong Provincial Hospital Affiliated to Shandong First Medical University (Shandong Provincial Hospital), Jinan, P.R. China
| | - Min Fu
- Department of Rheumatology and Immunology, 34708Shandong Provincial Hospital Affiliated to Shandong First Medical University (Shandong Provincial Hospital), Jinan, P.R. China
| | - Qing-Rui Yang
- Department of Rheumatology and Immunology, 34708Shandong Provincial Hospital Affiliated to Shandong First Medical University (Shandong Provincial Hospital), Jinan, P.R. China
| | - Hong-Sheng Sun
- Department of Rheumatology and Immunology, 34708Shandong Provincial Hospital Affiliated to Shandong First Medical University (Shandong Provincial Hospital), Jinan, P.R. China
| |
Collapse
|
8
|
Huang Y, Pan C, Liu Y, Lin S, Zhan Y, Zhang Y, Zhan F. Immune Function and Mechanism of Costimulating Molecules PD-1 and OX40 in Rheumatoid Arthritis. J Interferon Cytokine Res 2021; 40:530-539. [PMID: 33201766 DOI: 10.1089/jir.2020.0010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Rheumatoid arthritis (RA) is a T lymphocyte-mediated autoimmune disease, although its immune mechanism has not been fully studied. In this study, healthy controls (HC), osteoarthritis patients (OA), and RA patients were enrolled, and mice were evenly divided into control, collagen-induced arthritis (CIA), PD-1 Fc/CIA (PD-1 Fc membrane fusion protein administered to CIA mice), OX40 Fc/CIA (OX40 Fc membrane fusion protein administered to CIA mice), and PD-1 Fc + OX40 Fc/CIA groups. The expressions of programmed death-1 (PD-1) and OX40 in CD4+ T lymphocytes and the levels of sPD-1, immunoglobulin, and proinflammatory factors in patients and mice were measured. The results showed that the expression levels of PD-1 and OX40 in CD4+ T lymphocytes separated from the peripheral blood and synovial fluid of RA patients and the spleen of CIA mice were observably elevated. The levels of soluble PD-1, interleukin (IL)-2, IL-4, IL-5, IL-17, and interferon-γ (IFN-γ) in RA patients obviously increased. In animal experiments, PD-1 Fc not only increased the serum levels of immunoglobulin G (IgG), IgG1, and IgG2a in CIA mice, but also increased the levels of IL-4, IL-2, IL-5, IL-17, and IFN-γ in mouse spleen cells and joint tissues, which, however, were reversed by OX40 Fc. In conclusion, OX40 inhibition could reverse the progression of RA caused by PD-1 blocking, and PD-1 might be a potential target for RA. Clinical Trials.gov ID: HGH2018012203.
Collapse
Affiliation(s)
- Yanyan Huang
- Department of Rheumatology and Immunology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Chuying Pan
- Department of Rheumatology and Immunology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Ying Liu
- Department of Rheumatology and Immunology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Shudian Lin
- Department of Rheumatology and Immunology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Yuwei Zhan
- Department of Rheumatology and Immunology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Yan Zhang
- Department of Rheumatology and Immunology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Feng Zhan
- Department of Rheumatology and Immunology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| |
Collapse
|
9
|
Sui S, Sun L, Zhang W, Li J, Han J, Zheng J, Xin H. LncRNA MEG8 Attenuates Cerebral Ischemia After Ischemic Stroke Through Targeting miR-130a-5p/VEGFA Signaling. Cell Mol Neurobiol 2021; 41:1311-1324. [PMID: 32627090 PMCID: PMC11448590 DOI: 10.1007/s10571-020-00904-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023]
Abstract
MEG8 is involved in ischemia stroke, however, its role in ischemia stroke remains unknown. The current research aimed to investigate the effects and mechanisms of MEG8 in ischemic stroke. Mouse brain microvascular endothelial cells (BMECs) were treated by oxygen-glucose deprivation (OGD). Then, the expressions of MEG8 and miR-130a-5p were detected by quantitative reverse transcription-polymerase chain reaction (q-PCR). Cell counting kit-8 (CCK-8), wound-healing, tube formation, Western blot, and q-PCR assays were performed to detect the effects of MEG8 and miR-130a-5p on cell viability, migration, and angiogenesis and VEGFA expression. Bioinformatics, dual-luciferase reporter assay, and RNA immunoprecipitation analysis were carried out to investigate the targeting relationship between MEG8 and miR-130a-5p, and between miR-130a-5p and VEGFA. Then, rat middle cerebral artery occlusion (MCAO) model and MEG8 overexpression MCAO model were established, and neurological deficit and infarct volume of the model rats were evaluated. Finally, Western blot and q-PCR were carried out to detect the expressions of MEG8, miR-130a-5p, and VEGFA. MEG8 was upregulated and miR-130a-5p was downregulated in OGD-treated BMECs. MiR-130a-5p was found to be a target of MEG8, and VEGFA was predicted to be a potential target of miR-130a-5p. Downregulation of MEG8 inhibited the cell viability, migration, and angiogenesis and the expression of VEGFA via negatively regulating miR-130a-5p of BMECs treated by OGD/non-OGD. In addition, MEG8 reduced cerebral ischemia, neurological score and miR-130a-5p expression, and increased VEGFA expression of MCAO rat. Our findings proved that MEG8 regulates angiogenesis and attenuates cerebral ischemia after ischemic stroke via miR-130a-5p/VEGFA signaling.
Collapse
Affiliation(s)
- Shihua Sui
- Department of Neurology, People's Hospital of Rizhao, No. 126, Donggang District, Tai'an Road, Rizhao, 276826, Shandong, China
| | - Lei Sun
- Department of Neurology, People's Hospital of Rizhao, No. 126, Donggang District, Tai'an Road, Rizhao, 276826, Shandong, China
| | - Wenjing Zhang
- Department of Neurology, People's Hospital of Rizhao, No. 126, Donggang District, Tai'an Road, Rizhao, 276826, Shandong, China
| | - Jiamei Li
- Department of Neurology, People's Hospital of Rizhao, No. 126, Donggang District, Tai'an Road, Rizhao, 276826, Shandong, China
| | - Jingcui Han
- Evoked Potential Room, People's Hospital of Rizhao, Rizhao, Shandong, China
| | - Jiaping Zheng
- Department of Neurology, People's Hospital of Rizhao, No. 126, Donggang District, Tai'an Road, Rizhao, 276826, Shandong, China
| | - Hua Xin
- Department of Neurology, People's Hospital of Rizhao, No. 126, Donggang District, Tai'an Road, Rizhao, 276826, Shandong, China.
| |
Collapse
|
10
|
Zhang J, Chen Y, Chen T, Miao B, Tang Z, Hu X, Luo Y, Zheng T, Na N. Single-cell transcriptomics provides new insights into the role of fibroblasts during peritoneal fibrosis. Clin Transl Med 2021; 11:e321. [PMID: 33784014 PMCID: PMC7908046 DOI: 10.1002/ctm2.321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/25/2021] [Accepted: 01/25/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The contributions of various types of cell populations in dialysis-related peritoneal fibrosis are poorly understood. Single-cell RNA sequencing brings single-cell level resolution to the analysis of cellular transcriptomics, which provides a new way to further characterize the distinct roles and functional states of each cell population during peritoneal fibrosis. METHODS Single-cell transcriptomics from normal peritoneal tissues of six patients, from effluent of patients with short-term peritoneal dialysis (less than 2 weeks, n = 6), and from long-term peritoneal dialysis patients (more than 6 years, n = 4) were analyzed. RESULTS We identified a distinct cell component between samples among different groups. Functional analysis of the differentially expressed genes identified cell type specific biological processes relevant to different fibrosis stages. Well-known key molecular mechanisms participating in the pathophysiology of peritoneal fibrosis were vitrified, and some of them were found to be restricted to specific cell types. Gradually growing enrichment of PI3K/AKT/mTOR pathway and impairment of oxidative phosphorylation in mesothelial cells and fibroblasts were found from healthy control, short-term dialysis, to long-term dialysis, respectively. The fibroblasts' population obtained from the patients, who received peritoneal dialysis, showed a functional characteristic of immune-chemotaxis and immune response, which was characterized by broadly significant increase in the expression of interleukins, chemokines, cytokines, and human leukocyte antigens. Furthermore, we described the intercellular crosstalk networks based on receptor-ligand interactions, and highlighted a central role of fibroblasts in regulating the key mechanisms of peritoneal fibrosis through crosstalk with other cells. CONCLUSIONS In summary, despite describing information for fibrogenic molecular mechanisms in the resolution level of individual cell populations, this work identifies the significant functional evolution of fibroblasts during peritoneal fibrosis. This study also reveals the intercellular receptor-ligand interactions in which the fibroblasts serve as a major node, eventually providing new insights into the role of fibroblasts during disease pathogenesis.
Collapse
Affiliation(s)
- Jinhua Zhang
- Department of Kidney TransplantationThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Yuxian Chen
- Department of Joint SurgeryThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Tufeng Chen
- Department of Gastrointestinal SurgeryThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Bin Miao
- Department of Kidney TransplantationThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Zuofu Tang
- Department of Kidney TransplantationThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Xiao Hu
- Department of Kidney TransplantationThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - You Luo
- Department of Kidney TransplantationThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Tong Zheng
- Department of Kidney TransplantationThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Ning Na
- Department of Kidney TransplantationThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
11
|
An updated advance of autoantibodies in autoimmune diseases. Autoimmun Rev 2020; 20:102743. [PMID: 33333232 DOI: 10.1016/j.autrev.2020.102743] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 08/06/2020] [Indexed: 12/18/2022]
Abstract
Autoantibodies are abnormal antibodies which are generated by pathogenic B cells when targeting an individual's own tissue. Autoantibodies have been identified as a symbol of autoimmune disorders and are frequently considered a clinical marker of these disorders. Autoimmune diseases, including system lupus erythematosus and rheumatoid arthritis, consist of a series of disorders that share some similarities and differences. They are characterized by chronic, systemic, excessive immune activation and inflammation and involve in almost all body tissues. Autoimmune diseases occur more frequently in women than men due to hormonal impacts. In this review we systemically introduce and summarize the latest advances of various autoantibodies in multiple autoimmune diseases.
Collapse
|
12
|
Xiao ZX, Hu X, Jarjour W, Zheng SG. The role of B7 family members in the generation of Immunoglobulin. J Leukoc Biol 2020; 109:377-382. [PMID: 33118237 DOI: 10.1002/jlb.1mr0420-003rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 04/08/2020] [Accepted: 04/15/2020] [Indexed: 10/23/2022] Open
Abstract
Ig is a Y-shaped protein produced by plasma cells and exerts multiple functions in humoral immunity. There are five groups of Igs including IgA, IgD, IgE, IgG, and IgM, which differ in their heavy chain class. The primary function of Igs includes the neutralization of extrinsic pathogens, agglutination of foreign cells for phagocytosis, precipitation of soluble antigens in serum, and complement fixation. The B cells activated by antigen(s) can differentiate into antibody-producing cells that are called plasma cells and usually matured in the germinal center (GC). Follicular T helper (Tfh) cells crosstalk with antigen-presenting cells and play a crucial role in the development of the GC. Moreover, Tfh cells regulate trafficking through the GC to allow formative interaction with GC B cells that ultimately results in affinity maturation, B-cell memory, and Ig class switching. The B7 family is a series of number of structurally related membrane proteins that bind with a specific receptor to deliver costimulatory or co-inhibitory signals that regulate the activation of T cells in GC. Here, we review and summarize the recent advance of the effects of B7 family members on Ig production and relative diseases.
Collapse
Affiliation(s)
- Ze Xiu Xiao
- Institute of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaojiang Hu
- Institute of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wael Jarjour
- Department of Internal Medicine, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
| | - Song Guo Zheng
- Department of Internal Medicine, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
13
|
Radmanesh F, Mahmoudi M, Yazdanpanah E, Keyvani V, Kia N, Nikpoor AR, Zafari P, Esmaeili SA. The immunomodulatory effects of mesenchymal stromal cell-based therapy in human and animal models of systemic lupus erythematosus. IUBMB Life 2020; 72:2366-2381. [PMID: 33006813 DOI: 10.1002/iub.2387] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/11/2020] [Accepted: 09/12/2020] [Indexed: 12/17/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic systemic autoimmune inflammatory disease with no absolute cure. Although the exact etiopathogenesis of SLE is still enigmatic, it has been well demonstrated that a combination of genetic predisposition and environmental factors trigger a disturbance in immune responses and thereby participate in the development of this condition. Almost all available therapeutic strategies in SLE are primarily based on the administration of immunosuppressive drugs and are not curative. Mesenchymal stromal cells (MSCs) are a subset of non-hematopoietic adult stem cells that can be isolated from many adult tissues and are increasingly recognized as immune response modulating agents. MSC-mediated inhibition of immune responses is a complex mechanism that involves almost every aspect of the immune response. MSCs suppress the maturation of antigen-presenting cells (DC and MQ), proliferation of T cells (Th1, T17, and Th2), proliferation and immunoglobulin production of B cells, the cytotoxic activity of CTL and NK cells in addition to increasing regulatory cytokines (TGF-β and IL10), and decreasing inflammatory cytokines (IL17, INF-ϒ, TNF-α, and IL12) levels. MSCs have shown encouraging results in the treatment of several autoimmune diseases, in particular SLE. This report aims to review the beneficial and therapeutic properties of MSCs; it also focuses on the results of animal model studies, preclinical studies, and clinical trials of MSC therapy in SLE from the immunoregulatory aspect.
Collapse
Affiliation(s)
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Esmaeil Yazdanpanah
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahideh Keyvani
- Molecular Genetics, Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Nadia Kia
- Skin Cancer Prevention Research Center, Torvergata University of Medical Sciences, Rome, Italy
| | - Amin Reza Nikpoor
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Parisa Zafari
- Department of Immunology, School of Medicine, Mazandaran University of Medical Science, Sari, Iran.,Student Research Committee, Mazandaran University of Medical Science, Sari, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
14
|
Wu Q, Cao F, Tao J, Li X, Zheng SG, Pan HF. Pentraxin 3: A promising therapeutic target for autoimmune diseases. Autoimmun Rev 2020; 19:102584. [PMID: 32534154 DOI: 10.1016/j.autrev.2020.102584] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 02/20/2020] [Indexed: 12/15/2022]
Abstract
Pentraxin 3 (PTX3) is a prototypic humoral soluble pattern recognition molecule that exerts a pivotal role in innate immune response and inflammation, as well as in tissue damage and remodeling. Recently, emerging evidence has revealed that PTX3 is involved in the occurrence and development of various autoimmune diseases, such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), ankylosing spondylitis (AS), systemic sclerosis (SSc), inflammatory bowel disease (IBD), multiple sclerosis (MS) and psoriasis, etc. In this review, we have succinctly summarized the complex immunological functions of PTX3 and mostly focused on recent findings of the pleiotropic activities played by PTX3 in the pathogenesis of autoimmune diseases, aiming at hopefully offering possible future therapeutic alternatives.
Collapse
Affiliation(s)
- Qian Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, 81 Meishan Road, Hefei, Anhui, China
| | - Fan Cao
- Department of Clinical Medicine, The second School of Clinical Medicine, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China
| | - Jinhui Tao
- Department of Rheumatology and Immunology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaomei Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui, China
| | - Song Guo Zheng
- Department of Internal Medicine, Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH 43210, USA.
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, 81 Meishan Road, Hefei, Anhui, China.
| |
Collapse
|
15
|
Huang G, Zhang Y, Wei X, Yu Z, Lai J, Shen Q, Chen X, Tan G, Chen C, Luo W, Li Y, Zhou M, Li Y, Li B. CD8+GITR+ T cells may negatively regulate T cell overactivation in aplastic anemia. Immunol Invest 2020; 50:406-415. [PMID: 32462957 DOI: 10.1080/08820139.2020.1770785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Guixuan Huang
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Yuping Zhang
- Department of Hematology, Guangzhou First People’s Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, China
| | - Xiaolei Wei
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhi Yu
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jing Lai
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Qi Shen
- Department of Hematology, Second Clinical Medical College of Jinan University (Shenzhen People’s Hospital), Shengzhen, China
| | - Xiaohui Chen
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Guangxiao Tan
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Cunte Chen
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | | | - Yumiao Li
- Department of Hematology, Guangzhou First People’s Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, China
| | - Ming Zhou
- Department of Hematology, Guangzhou First People’s Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, China
| | - Yangqiu Li
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Bo Li
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
16
|
Research Progress on Regulatory B Cells in Systemic Lupus Erythematosus. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7948687. [PMID: 31240224 PMCID: PMC6556307 DOI: 10.1155/2019/7948687] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/05/2019] [Indexed: 11/25/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic, systemic, autoimmune inflammatory disease characterized by the production of numerous autoantibodies and cytokines, as well as multiple organ damage. Specific B cell subsets negatively regulate immune responses and have been termed regulatory B cells (Bregs). Bregs are characterized by the production of the immunoregulatory cytokines interleukin (IL)-10, IL-35, and transforming growth factor (TGF)-β. Bregs suppress other immune cells through the secretion of these immunosuppressive cytokines and have thus been studied extensively for their potential role in the treatment of various autoimmune diseases. The progress of the research on Bregs and SLE in recent years is reviewed in this paper.
Collapse
|