51
|
Protein and functional isoform levels and genetic variants of the BAFF and APRIL pathway components in systemic lupus erythematosus. Sci Rep 2022; 12:11219. [PMID: 35780200 PMCID: PMC9250527 DOI: 10.1038/s41598-022-15549-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/24/2022] [Indexed: 11/11/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is the prototype of an autoimmune disease. Belimumab, a monoclonal antibody targets BAFF, is the only biologic approved for SLE and active lupus nephritis. BAFF is a cytokine with a key-regulatory role in the B cell homeostasis, which acts by binding to three receptors: BAFF-R, TACI and BCMA. TACI and BCMA also bind APRIL. Many studies reported elevated soluble BAFF and APRIL levels in the sera of SLE patients, but other questions about the role of this system in the disease remain open. The study aimed to investigate the utility of the cytokine levels in serum and urine as biomarkers, the role of non-functional isoforms, and the association of gene variants with the disease. This case–control study includes a cohort (women, 18–60 years old) of 100 patients (48% with nephritis) and 100 healthy controls. We used ELISA assays to measure the cytokine concentrations in serum (sBAFF and sAPRIL) and urine (uBAFF and uAPRIL); TaqMan Gene Expression Assays to quantify the relative mRNA expression of ΔBAFF, βAPRIL, and εAPRIL, and next-generation sequencing to genotype the cytokine (TNFSF13 and TNFSF13B) and receptor (TNFRSF13B, TNFRSF17 and TNFRSF13C) genes. The statistical tests used were: Kruskal–Wallis (qualitative variables), the Spearman Rho coefficient (correlations), the Chi-square and SKAT (association of common and rare genetic variants, respectively). As expected, sBAFF and sAPRIL levels were higher in patients than in controls (p ≤ 0.001) but found differences between patient subgroups. sBAFF and sAPRIL significantly correlated only in patients with nephritis (rs = 0.67, p ≤ 0.001) and βAPRIL levels were lower in patients with nephritis (p = 0.04), and ΔBAFF levels were lower in patients with dsDNA antibodies (p = 0.04). Rare variants of TNFSF13 and TNFRSF13B and TNFSF13 p.Gly67Arg and TNFRSF13B p.Val220Ala were associated with SLE. Our study supports differences among SLE patient subgroups with diverse clinical features in the BAFF/APRIL pathway. In addition, it suggests the involvement of genetic variants in the susceptibility to the disease.
Collapse
|
52
|
Shen Z, Ma J, Peng R, Hu B, Zhao Y, Liu S, Hong J. Biomarkers in Ocular Graft-Versus-Host Disease: Implications for the Involvement of B Cells. Transplant Cell Ther 2022; 28:749.e1-749.e7. [DOI: 10.1016/j.jtct.2022.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 10/16/2022]
|
53
|
Person T, King RG, Rizk DV, Novak J, Green TJ, Reily C. Cytokines and Production of Aberrantly O-Glycosylated IgA1, the Main Autoantigen in IgA Nephropathy. J Interferon Cytokine Res 2022; 42:301-315. [PMID: 35793525 PMCID: PMC9536348 DOI: 10.1089/jir.2022.0039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/16/2022] [Indexed: 01/09/2023] Open
Abstract
Immunoglobulin A (IgA) nephropathy is the most common primary glomerulonephritis worldwide, with no disease-specific treatment and up to 40% of patients progressing to kidney failure. IgA nephropathy (IgAN), characterized by IgA1-containing immunodeposits in the glomeruli, is considered to be an autoimmune disease in which the kidneys are injured as innocent bystanders. Glomerular immunodeposits are thought to originate from the circulating immune complexes that contain aberrantly O-glycosylated IgA1, the main autoantigen in IgAN, bound by IgG autoantibodies. A common clinical manifestation associated with IgAN includes synpharyngitic hematuria at disease onset or during disease activity. This observation suggests a connection of disease pathogenesis with an activated mucosal immune system of the upper-respiratory and/or gastrointestinal tract and IgA1 glycosylation. In fact, some cytokines can enhance production of aberrantly O-glycosylated IgA1. This process involves abnormal cytokine signaling in IgA1-producing cells from patients with IgAN. In this article, we present our view of pathogenesis of IgAN and review how some cytokines can contribute to the disease process by enhancing production of aberrantly glycosylated IgA1. We also review current clinical trials of IgAN based on cytokine-targeting therapeutic approaches.
Collapse
Affiliation(s)
- Taylor Person
- Department of Microbiology and Birmingham, Alabama, USA
| | - R. Glenn King
- Department of Microbiology and Birmingham, Alabama, USA
| | - Dana V. Rizk
- Department of Medicine University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jan Novak
- Department of Microbiology and Birmingham, Alabama, USA
| | - Todd J. Green
- Department of Microbiology and Birmingham, Alabama, USA
| | - Colin Reily
- Department of Microbiology and Birmingham, Alabama, USA
- Department of Medicine University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
54
|
Marsman C, Verhoeven D, Koers J, Rispens T, ten Brinke A, van Ham SM, Kuijpers TW. Optimized Protocols for In-Vitro T-Cell-Dependent and T-Cell-Independent Activation for B-Cell Differentiation Studies Using Limited Cells. Front Immunol 2022; 13:815449. [PMID: 35844625 PMCID: PMC9278277 DOI: 10.3389/fimmu.2022.815449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background/Methods For mechanistic studies, in-vitro human B-cell differentiation and generation of plasma cells are invaluable techniques. However, the heterogeneity of both T-cell-dependent (TD) and T-cell-independent (TI) stimuli and the disparity of culture conditions used in existing protocols make the interpretation of results challenging. The aim of the present study was to achieve the most optimal B-cell differentiation conditions using isolated CD19+ B cells and peripheral blood mononuclear cell (PBMC) cultures. We addressed multiple seeding densities, different durations of culturing, and various combinations of TD and TI stimuli including B-cell receptor (BCR) triggering. B-cell expansion, proliferation, and differentiation were analyzed after 6 and 9 days by measuring B-cell proliferation and expansion, plasmablast and plasma cell formation, and immunoglobulin (Ig) secretion. In addition, these conditions were extrapolated using cryopreserved cells and differentiation potential was compared. Results This study demonstrates improved differentiation efficiency after 9 days of culturing for both B-cells and PBMC cultures using CD40L and IL-21 as TD stimuli and 6 days for CpG and IL-2 as TI stimuli. We arrived at optimized protocols requiring 2,500 and 25,000 B–cells per culture well for the TD and TI assays, respectively. The results of the PBMC cultures were highly comparable to the B-cell cultures, which allows dismissal of additional B-cell isolation steps prior to culturing. In these optimized TD conditions, the addition of anti-BCR showed a little effect on phenotypic B-cell differentiation; however, it interferes with Ig secretion measurements. The addition of IL-4 to the TD stimuli showed significantly lower Ig secretion. The addition of BAFF to optimized TI conditions showed enhanced B-cell differentiation and Ig secretion in B-cell but not in PBMC cultures. With this approach, efficient B-cell differentiation and Ig secretion were accomplished when starting from fresh or cryopreserved samples. Conclusion Our methodology demonstrates optimized TD and TI stimulation protocols for more in-depth analysis of B-cell differentiation in primary human B-cell and PBMC cultures while requiring low amounts of B cells, making them ideally suited for future clinical and research studies on B-cell differentiation of patient samples from different cohorts of B-cell-mediated diseases.
Collapse
Affiliation(s)
- Casper Marsman
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Dorit Verhoeven
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Jana Koers
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Anja ten Brinke
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - S. Marieke van Ham
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Taco W. Kuijpers, ; S. Marieke van Ham,
| | - Taco W. Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Taco W. Kuijpers, ; S. Marieke van Ham,
| |
Collapse
|
55
|
Sevdali E, Block V, Lataretu M, Li H, Smulski CR, Briem JS, Heitz Y, Fischer B, Ramirez NJ, Grimbacher B, Jäck HM, Voll RE, Hölzer M, Schneider P, Eibel H. BAFFR activates PI3K/AKT signaling in human naive but not in switched memory B cells through direct interactions with B cell antigen receptors. Cell Rep 2022; 39:111019. [PMID: 35767961 DOI: 10.1016/j.celrep.2022.111019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/27/2022] [Accepted: 06/08/2022] [Indexed: 12/28/2022] Open
Abstract
Binding of BAFF to BAFFR activates in mature B cells PI3K/AKT signaling regulating protein synthesis, metabolic fitness, and survival. In humans, naive and memory B cells express the same levels of BAFFR, but only memory B cells seem to survive without BAFF. Here, we show that BAFF activates PI3K/AKT only in naive B cells and changes the expression of genes regulating migration, proliferation, growth, and survival. BAFF-induced PI3K/AKT activation requires direct interactions between BAFFR and the B cell antigen receptor (BCR) components CD79A and CD79B and is enhanced by the AKT coactivator TCL1A. Compared to memory B cells, naive B cells express more surface BCRs, which interact better with BAFFR than IgG or IgA, thus allowing stronger responses to BAFF. As ablation of BAFFR in naive and memory B cells causes cell death independent of BAFF-induced signaling, BAFFR seems to act also as an intrinsic factor for B cell survival.
Collapse
Affiliation(s)
- Eirini Sevdali
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Violeta Block
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Marie Lataretu
- RNA Bioinformatics and High-Throughput Analysis, Faculty of Mathematics and Computer Science, University of Jena, Leutragraben 1, 07743 Jena, Germany
| | - Huiying Li
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Cristian R Smulski
- Medical Physics Department, Centro Atómico Bariloche, Comisión Nacional de Energía Atómica (CNEA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Avenida E-Bustillo 9500, R8402AGP Río Negro, San Carlos de Bariloche, Argentina
| | - Jana-Susann Briem
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Yannic Heitz
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Beate Fischer
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Neftali-Jose Ramirez
- Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany; Institute for Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Bodo Grimbacher
- Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany; Institute for Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Hans-Martin Jäck
- Department of Medicine, Division of Immunology, University of Erlangen, Glückstraße 6, 91054 Erlangen, Germany
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Martin Hölzer
- Methodology and Research Infrastructure, MF1 Bioinformatics, Robert Koch Institute, Nordufer 20, 13353 Berlin, Germany
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Ch. des Boveresses 155, 1066 Epalinges, Switzerland
| | - Hermann Eibel
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany.
| |
Collapse
|
56
|
Chawla S, Barman P, Tyagi R, Jindal AK, Sharma S, Rawat A, Singh S. Autoimmune Cytopenias in Common Variable Immunodeficiency Are a Diagnostic and Therapeutic Conundrum: An Update. Front Immunol 2022; 13:869466. [PMID: 35795667 PMCID: PMC9251126 DOI: 10.3389/fimmu.2022.869466] [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: 02/04/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Common variable immunodeficiency (CVID) is the most common symptomatic primary immunodeficiency (PID). CVID is a heterogenous condition and clinical manifestations may vary from increased susceptibility to infections to autoimmune manifestations, granulomatous disease, polyclonal lymphoproliferation, and increased risk of malignancy. Autoimmune manifestations may, at times, be the first and only clinical presentation of CVID, resulting in diagnostic dilemma for the treating physician.Autoimmune cytopenias (autoimmune haemolytic anaemia and/or thrombocytopenia) are the most common autoimmune complications seen in patients with CVID. Laboratory investigations such as antinuclear antibodies, direct Coomb’s test and anti-platelet antibodies may not be useful in patients with CVID because of lack of specific antibody response. Moreover, presence of autoimmune cytopenias may pose a significant therapeutic challenge as use of immunosuppressive agents can be contentious in these circumstances. It has been suggested that serum immunoglobulins must be checked in all patients presenting with autoimmune cytopenia such as immune thrombocytopenia or autoimmune haemolytic anaemia.It has been observed that patients with CVID and autoimmune cytopenias have a different clinical and immunological profile as compared to patients with CVID who do not have an autoimmune footprint. Monogenic defects have been identified in 10-50% of all patients with CVID depending upon the population studied. Monogenic defects are more likely to be identified in patients with CVID with autoimmune complications. Common genetic defects that may lead to CVID with an autoimmune phenotype include nuclear factor kappa B subunit 1 (NF-kB1), Lipopolysaccharide (LPS)-responsive beige-like anchor protein (LRBA), cytotoxic T lymphocyte antigen 4 (CTLA4), Phosphoinositide 3-kinase (PI3K), inducible T-cell costimulatory (ICOS), IKAROS and interferon regulatory factor-2 binding protein 2 (IRF2BP2).In this review, we update on recent advances in pathophysiology and management of CVID with autoimmune cytopenias.
Collapse
|
57
|
Yao Y, Zhu J, Qin S, Zhou Z, Zeng Q, Long R, Mao Z, Dong X, Zhao R, Zhang R, Zhang S, Huang S, Chen L. Resveratrol induces autophagy impeding BAFF-stimulated B-cell proliferation and survival by inhibiting the Akt/mTOR pathway. Biochem Pharmacol 2022; 202:115139. [PMID: 35697119 PMCID: PMC9283307 DOI: 10.1016/j.bcp.2022.115139] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 11/02/2022]
Abstract
Therapeutically targeting B cells has received great attention in the treatment of B-cell malignancies and autoimmune diseases. The B-cell activating factor (BAFF) is critical to the survival of normal and neoplastic B cells, and excess production of BAFF contributes to autoimmune diseases. Resveratrol, a natural polyphenolic compound, has a positive effect on the treatment of autoimmune diseases. However, how resveratrol affects BAFF-stimulated B-cell proliferation and survival is poorly understood. Here, we show that resveratrol increased autophagosome formation and ATG5/LC3-II levels and decreased p62 level, promoting autophagic flux/autophagy and thereby suppressing the basal or human soluble BAFF (hsBAFF)-stimulated proliferation and survival of normal and B-lymphoid (Raji) cells. This is supported by the findings that inhibition of autophagy with 3-methyladenine (3-MA, an inhibitor of Vps34) or ATG5 shRNA attenuates resveratrol-induced autophagy and -reduced proliferation/viability in B-cells. Inhibition of mTOR with rapamycin or knockdown of mTOR potentiated resveratrol-induced autophagy and inhibition of hsBAFF-stimulated B-cell proliferation/viability, while overexpression of wild-type mTOR conferred resistance to the actions of resveratrol. Similarly, inhibition of Akt with Akt inhibitor X or ectopic expression of dominant negative Akt reinforced resveratrol-induced autophagy and inhibition of hsBAFF-stimulated B-cell proliferation/viability, whereas expression of constitutively active Akt conferred resistance to the actions of resveratrol. Taken together, these results indicate that resveratrol induces autophagy impeding BAFF-stimulated proliferation and survival via blocking the Akt/mTOR signaling pathway in normal and neoplastic B cells. Our findings highlight that resveratrol has a great potential for prevention and treatment of excessive BAFF-elicited aggressive B-cell disorders and autoimmune diseases.
Collapse
Affiliation(s)
- Yajie Yao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Jiawei Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Shanshan Qin
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Zhihan Zhou
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Qingyu Zeng
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Ruyu Long
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Zun Mao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Xiaoqing Dong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Rui Zhao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Ruijie Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Shuangquan Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Shreveport, LA 71130-3932, USA; Department of Hematology and Oncology, Shreveport, LA 71130-3932, USA; Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA.
| | - Long Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China.
| |
Collapse
|
58
|
Santillán-López E, Muñoz-Valle JF, Oregon-Romero E, Espinoza-García N, Treviño-Talavera BA, Salazar-Camarena DC, Marín-Rosales M, Cruz A, Alvarez-Gómez JA, Sagrero-Fabela N, Cerpa-Cruz S, Palafox-Sánchez CA. Analysis of TNFSF13B polymorphisms and BAFF expression in rheumatoid arthritis and primary Sjögren's syndrome patients. Mol Genet Genomic Med 2022; 10:e1950. [PMID: 35411715 PMCID: PMC9184664 DOI: 10.1002/mgg3.1950] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/17/2022] [Accepted: 04/01/2022] [Indexed: 11/11/2022] Open
Abstract
Background The increased expression of B cell‐activating factor (BAFF) has been linked to autoantibody production in autoimmune diseases (ADs). The aim of this study was to investigate the association among TNFSF13B gene (OMIM: 603969) single nucleotide polymorphisms (SNPs), TNFSF13B mRNA, and soluble BAFF (sBAFF) expression in patients with rheumatoid arthritis (RA) and primary Sjögren's syndrome (pSS). The diagnostic value of sBAFF also was evaluated by the area under the curve (AUC) of receiver operating characteristic or receptor (ROC) curves. Methods Genotypes of the TNFSF13B rs9514827 (−2841 T > C), rs1041569 (−2701 A > T) and rs9514828 (−871 C > T) SNPs were determined by PCR‐RFLP assay. TNFSF13B mRNA and sBAFF expression were performed by RT‐qPCR and ELISA, respectively. The study included 320 RA patients, 101 pSS patients, and 309 healthy subjects (HS). Results The rs9514828 T allele and the TAT haplotype were associated with an increased risk to develop RA. In both ADs, the TNFSF13B mRNA levels were increased in comparison with HS. The rs9514828 (−871 C > T) polymorphism was associated with increased gene expression in RA patients. Also, sBAFF levels were higher in both ADs, however pSS patients showed the highest sBAFF levels. sBAFF showed higher diagnostic performance for pSS with an AUC of 0.968, with a similar accuracy of anti‐SSA/Ro antibody diagnosis (AUC = 0.974). Conclusions Our findings demonstrate that the TNFSF13B rs9514828 (−871 C > T) polymorphism is a risk factor for RA in the western Mexican population. sBAFF levels may be a potential diagnosis biomarker in pSS.
Collapse
Affiliation(s)
- Enrique Santillán-López
- Doctorado en Ciencias Biomédicas (DCB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - José Francisco Muñoz-Valle
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Edith Oregon-Romero
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Noemí Espinoza-García
- Doctorado en Ciencias en Biología Molecular en Medicina (DCBMM), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | | | - Diana Celeste Salazar-Camarena
- Grupo de Inmunología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Miguel Marín-Rosales
- Grupo de Inmunología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico.,Hospital General de Occidente, Secretaría de Salud Jalisco, Guadalajara, Mexico
| | - Alvaro Cruz
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Jhonatan Antonio Alvarez-Gómez
- Doctorado en Ciencias en Biología Molecular en Medicina (DCBMM), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Nefertari Sagrero-Fabela
- Doctorado en Ciencias Biomédicas (DCB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Sergio Cerpa-Cruz
- Servicio de Reumatología, O.P.D. Hospital Civil de Guadalajara "Fray Antonio Alcalde", Guadalajara, Mexico
| | - Claudia Azucena Palafox-Sánchez
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico.,Grupo de Inmunología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| |
Collapse
|
59
|
Xu Y, Xu JW, Wang YJ, Tung TH, Chien CW. Belimumab combined with standard therapy does not increase adverse effects compared with a control treatment: A systematic review and meta-analysis of randomised controlled trials. Int Immunopharmacol 2022; 109:108811. [PMID: 35512563 DOI: 10.1016/j.intimp.2022.108811] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/04/2022] [Accepted: 04/25/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND The increasing administration of belimumab has demonstrated its biological benefits. Prior meta-analyses have examined the overall adverse events (AEs) associated with belimumab, but such knowledge needs to be updated with a high volume of new trials. However, little is known about the occurrence of AEs associated with different underlying diseases. This study aimed to address the safety of the intravenous (IV) administration of belimumab combined with standard of care (SoC) therapy in Systemic lupus erythematosus (SLE) patients. METHODS We used PubMed, Embase, and the Cochrane Library to systematically search for randomised controlled trials (RCTs) reporting AEs and specific AEs in SLE patients receiving belimumab and SoC therapy before 30 November 2021. We extracted the data of the eligible studies and calculated pooled risk ratios (RRs) and their 95% confidence intervals (CIs) in SLE patients receiving belimumab and SoC therapy and experiencing various AEs. The main outcomes were as follows: (1) any AEs, any serious AEs (SAEs), and any severe AEs; (2) serious organ specific adverse events; (3) adverse events of special interest (AESIs). RESULTS Of the 1,621 studies identified, nine RCTs involving 7,974 patients were eligible for the meta-analysis. There were no significant differences between the experimental and control groups in terms of the incident of AEs: AEs (RR = 0.99, 95% CI: 0.97-1.02, P = 0.68), SAEs (RR = 0.91, 95% CI: 0.81-1.02, P = 0.09), and severe AEs (RR = 0.92, 95% CI: 0.75-1.14, P = 0.46). The pooled data also showed that there was no significant correlation between five types of SAEs grouped by organ class and the IV belimumab (10 mg/kg) intervention, except for 'infections and infestations' (RR = 0.82, 95% CI: 0.70-0.97, P = 0.02) and 'musculoskeletal and connective tissue disorders' (RR = 0.46, 95% CI: 0.32-0.67, P < 0.0001). In addition, we found no significant association between AESIs and the IV administration of belimumab (10 mg/kg) (all malignancies: RR = 1.53, 95% CI: 0.69-3.36, P = 0.3; all post-infusion systemic reactions: RR = 1.05, 95% CI: 0.85-1.30, P = 0.63; depression: RR = 1.42, 95% CI: 0.92-2.20, P = 0.11; serious depression: RR = 2.60, 95% CI: 0.85-7.93, P = 0.09; suicide or self-injury: RR = 0.97, 95% CI: 0.48-1.96, P = 0.92; serious suicide or self-injury: RR = 1.26, 95% CI: 0.59-2.70, P = 0.56). CONCLUSIONS According to the results of the meta-analysis, SLE patients did not have significantly increased risk of experiencing any type of AEs when receiving SoC therapy. Special caution should be exercised during close follow-ups and individual clinical management before drug prescription.
Collapse
Affiliation(s)
- Ying Xu
- Institute for Hospital Management, Tsing Hua University, Shenzhen Campus, Shenzhen 518055, Guangdong Province, China
| | - Jia-Wen Xu
- Institute for Hospital Management, Tsing Hua University, Shenzhen Campus, Shenzhen 518055, Guangdong Province, China
| | - Yan-Jiao Wang
- Institute for Hospital Management, Tsing Hua University, Shenzhen Campus, Shenzhen 518055, Guangdong Province, China
| | - Tao-Hsin Tung
- Evidence-based Medicine Centre, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Ching-Wen Chien
- Institute for Hospital Management, Tsing Hua University, Shenzhen Campus, Shenzhen 518055, Guangdong Province, China.
| |
Collapse
|
60
|
Stathopoulos P, Dalakas MC. Evolution of Anti-B Cell Therapeutics in Autoimmune Neurological Diseases. Neurotherapeutics 2022; 19:691-710. [PMID: 35182380 PMCID: PMC9294112 DOI: 10.1007/s13311-022-01196-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2022] [Indexed: 02/08/2023] Open
Abstract
B cells have an ever-increasing role in the etiopathology of a number of autoimmune neurological disorders, acting as antigen-presenting cells facilitating antibody production but also as sensors, coordinators, and regulators of the immune response. In particular, B cells can regulate the T cell activation process through their participation in antigen presentation, production of proinflammatory cytokines (bystander activation or suppression), and contribution to ectopic lymphoid aggregates. Such an important interplay between B and T cells makes therapeutic depletion of B cells an attractive treatment strategy. The last decade, anti-B cell therapies using monoclonal antibodies against B cell surface molecules have evolved into a rational approach for successfully treating autoimmune neurological disorders, even when T cells seem to be the main effector cells. The paper summarizes basic aspects of B cell biology, discusses the roles of B cells in neurological autoimmunities, and highlights how the currently available or under development anti-B cell therapeutics exert their action in the wide spectrum and immunologically diverse neurological disorders. The efficacy of the various anti-B cell therapies and practical issues on induction and maintenance therapy is specifically detailed for the treatment of patients with multiple sclerosis, neuromyelitis-spectrum disorders, autoimmune encephalitis and hyperexcitability CNS disorders, autoimmune neuropathies, myasthenia gravis, and inflammatory myopathies. The success of anti-B cell therapies in inducing long-term remission in IgG4 neuroautoimmunities is also highlighted pointing out potential biomarkers for follow-up infusions.
Collapse
Affiliation(s)
- Panos Stathopoulos
- 1st Department of Neurology, National and Kapodistrian University of Athens, Athens, Greece
| | - Marinos C Dalakas
- Thomas Jefferson University, Philadelphia, PA, USA.
- Neuroimmunology Unit, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
61
|
Antiviral CD19 +CD27 + Memory B Cells Are Associated with Protection from Recurrent Asymptomatic Ocular Herpesvirus Infection. J Virol 2022; 96:e0205721. [PMID: 34985998 DOI: 10.1128/jvi.02057-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Reactivation of herpes simplex virus 1 (HSV-1) from latently infected neurons of the trigeminal ganglia (TG) leads to blinding recurrent herpetic disease in symptomatic (SYMP) individuals. Although the role of T cells in herpes immunity seen in asymptomatic (ASYMP) individuals is heavily explored, the role of B cells is less investigated. In the present study, we evaluated whether B cells are associated with protective immunity against recurrent ocular herpes. The frequencies of circulating HSV-specific memory B cells and of memory follicular helper T cells (CD4+ Tfh cells), which help B cells produce antibodies, were compared between HSV-1-infected SYMP and ASYMP individuals. The levels of IgG/IgA and neutralizing antibodies were compared in SYMP and ASYMP individuals. We found that (i) the ASYMP individuals had increased frequencies of HSV-specific CD19+CD27+ memory B cells, and (ii) high frequencies of HSV-specific switched IgG+CD19+CD27+ memory B cells detected in ASYMP individuals were directly proportional to high frequencies of CD45R0+CXCR5+CD4+ memory Tfh cells. However, no differences were detected in the level of HSV-specific IgG/IgA antibodies in SYMP and ASYMP individuals. Using the UV-B-induced HSV-1 reactivation mouse model, we found increased frequencies of HSV-specific antibody-secreting plasma HSV-1 gD+CD138+ B cells within the TG and circulation of ASYMP mice compared to those of SYMP mice. In contrast, no significant differences in the frequencies of B cells were found in the cornea, spleen, and bone-marrow. Our findings suggest that circulating antibody-producing HSV-specific memory B cells recruited locally to the TG may contribute to protection from symptomatic recurrent ocular herpes. IMPORTANCE Reactivation of herpes simplex virus 1 (HSV-1) from latently infected neurons of the trigeminal ganglia (TG) leads to blinding recurrent herpetic disease in symptomatic (SYMP) individuals. Although the role of T cells in herpes immunity against blinding recurrent herpetic disease is heavily explored, the role of B cells is less investigated. In the present study, we found that in both asymptomatic (ASYMP) individuals and ASYMP mice, there were increased frequencies of HSV-specific memory B cells that were directly proportional to high frequencies of memory Tfh cells. Moreover, following UV-B-induced reactivation, we found increased frequencies of HSV-specific antibody-secreting plasma B cells within the TG and circulation of ASYMP mice compared to those of SYMP mice. Our findings suggest that circulating antibody-producing HSV-specific memory B cells recruited locally to the TG may contribute to protection from recurrent ocular herpes.
Collapse
|
62
|
Sari S, Cinar S, Yalcinkaya Y, Artim-Esen B, Ozluk Y, Gul A, Ocal L, Deniz G, Inanc M. The relationship between serum A proliferation-inducing ligand and B-cell activating factor levels with disease activity and organ involvement in systemic lupus erythematosus. Lupus 2022; 31:555-564. [DOI: 10.1177/09612033221086123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Objectives We aim to investigate the association between serum B-cell activating factor (BAFF) and A proliferation-inducing ligand (APRIL) levels with disease activity and clinical findings in SLE patients. Methods Seventy-nine patients with SLE and 27 healthy controls were included into the study. Serum BAFF and APRIL levels were measured by using ELISA. In 19 patients with active disease at the time of the assessment, BAFF/APRIL levels were reassessed after 6 months of follow-up and disease activity was evaluated by using SLEDAI-2K. The relationship between renal histopathology index scores and lupus nephritis (LN) classes with serum BAFF/APRIL levels was examined in 16 patients who had recent renal involvement and underwent biopsy during the study. Results Although both BAFF/APRIL levels were higher in patients with SLE compared to the control group ( p < 0.001), no correlation was found between BAFF/APRIL levels and SLEDAI scores. Serum BAFF levels were higher in patients with renal disease activity ( p = 0.01), and there was a significant correlation between APRIL levels and proteinuria (r = 0.42, p = 0.02). A weak inverse correlation was observed between BAFF and C3 levels (r = 0.25, p = 0.02). No correlation was found between BAFF/APRIL levels and renal SLEDAI scores, renal histopathology, activity, and chronicity index scores. In the active disease group after treatment, there was no significant change in serum BAFF levels, but a significant increase in serum APRIL levels was observed. Conclusion These results suggest that both cytokines are involved in the pathogenesis of SLE and that serum BAFF can be valuable as a biomarker in SLE especially in patients with renal activity.
Collapse
Affiliation(s)
- Selma Sari
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Suzan Cinar
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Yasemin Yalcinkaya
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Bahar Artim-Esen
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Yasemin Ozluk
- Department of Pathology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ahmet Gul
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Lale Ocal
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Günnur Deniz
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Murat Inanc
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
63
|
Nobari ST, Nojadeh JN, Talebi M. B-cell maturation antigen targeting strategies in multiple myeloma treatment, advantages and disadvantages. J Transl Med 2022; 20:82. [PMID: 35144648 PMCID: PMC8832753 DOI: 10.1186/s12967-022-03285-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/29/2022] [Indexed: 01/02/2023] Open
Abstract
B cell maturation antigen (BCMA), a transmembrane glycoprotein member of the tumor necrosis factor receptor superfamily 17 (TNFRSF17), highly expressed on the plasma cells of Multiple myeloma (MM) patients, as well as the normal population. BCMA is used as a biomarker for MM. Two members of the TNF superfamily proteins, including B-cell activating factor (BAFF) and A proliferation-inducing ligand (APRIL), are closely related to BCMA and play an important role in plasma cell survival and progression of MM. Despite the maximum specificity of the monoclonal antibody technologies, introducing the tumor-specific antigen(s) is not applicable for all malignancies, such as MM that there plenty of relatively specific antigens such as GPCR5D, MUC1, SLAMF7 and etc., but higher expression of BCMA on these cells in comparison with normal ones can be regarded as a relatively exclusive marker. Currently, different monoclonal antibody (mAb) technologies applied in anti-MM therapies such as daratuzumab, SAR650984, GSK2857916, and CAR-T cell therapies are some of these tools that are reviewed in the present manuscript. By the way, the structure, function, and signaling of the BCMA and related molecule(s) role in normal plasma cells and MM development, evaluated as well as the potential side effects of its targeting by different CAR-T cells generations. In conclusion, BCMA can be regarded as an ideal molecule to be targeted in immunotherapeutic methods, regarding lower potential systemic and local side effects.
Collapse
Affiliation(s)
- Shirin Teymouri Nobari
- Department of Medical Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Jafar Nouri Nojadeh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Talebi
- Department of Applied Cells Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
64
|
Fleming A, Castro‐Dopico T, Clatworthy MR. B cell class switching in intestinal immunity in health and disease. Scand J Immunol 2022; 95:e13139. [PMID: 34978077 PMCID: PMC9285483 DOI: 10.1111/sji.13139] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 12/31/2021] [Accepted: 12/31/2021] [Indexed: 12/12/2022]
Abstract
The gastrointestinal tract is colonized by trillions of commensal microorganisms that collectively form the microbiome and make essential contributions to organism homeostasis. The intestinal immune system must tolerate these beneficial commensals, whilst preventing pathogenic organisms from systemic spread. Humoral immunity plays a key role in this process, with large quantities of immunoglobulin (Ig)A secreted into the lumen on a daily basis, regulating the microbiome and preventing bacteria from encroaching on the epithelium. However, there is an increasing appreciation of the role of IgG antibodies in intestinal immunity, including beneficial effects in neonatal immune development, pathogen and tumour resistance, but also of pathological effects in driving chronic inflammation in inflammatory bowel disease (IBD). These antibody isotypes differ in effector function, with IgG exhibiting more proinflammatory capabilities compared with IgA. Therefore, the process that leads to the generation of different antibody isotypes, class-switch recombination (CSR), requires careful regulation and is orchestrated by the immunological cues generated by the prevalent local challenge. In general, an initiating signal such as CD40 ligation on B cells leads to the induction of activation-induced cytidine deaminase (AID), but a second cytokine-mediated signal determines which Ig heavy chain is expressed. Whilst the cytokines driving intestinal IgA responses are well-studied, there is less clarity on how IgG responses are generated in the intestine, and how these cues might become dysfunctional in IBD. Here, we review the key mechanisms regulating class switching to IgA vs IgG in the intestine, processes that could be therapeutically manipulated in infection and IBD.
Collapse
Affiliation(s)
- Aaron Fleming
- Molecular Immunity UnitDepartment of MedicineCambridge Institute of Therapeutic Immunology and Infectious DiseasesUniversity of CambridgeCambridgeUK
| | - Tomas Castro‐Dopico
- Molecular Immunity UnitDepartment of MedicineCambridge Institute of Therapeutic Immunology and Infectious DiseasesUniversity of CambridgeCambridgeUK
- The Francis Crick InstituteLondonUK
| | - Menna R. Clatworthy
- Molecular Immunity UnitDepartment of MedicineCambridge Institute of Therapeutic Immunology and Infectious DiseasesUniversity of CambridgeCambridgeUK
- Cellular GeneticsWellcome Trust Sanger InstituteHinxtonUK
- NIHR Cambridge Biomedical Research CentreCambridgeUK
| |
Collapse
|
65
|
Shim JA, Jo Y, Hwang H, Lee SE, Ha D, Lee JH, Kim J, Song P, Lee D, Hong C. Defects in aminoacyl-tRNA synthetase cause partial B and T cell immunodeficiency. Cell Mol Life Sci 2022; 79:87. [PMID: 35067747 PMCID: PMC11071942 DOI: 10.1007/s00018-021-04122-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 11/29/2022]
Abstract
Aminoacyl-tRNA synthetases (ARSs) are emerging as important regulators in various immune diseases; however, their roles in immune cells remain unclear. In this study, using alanyl-tRNA synthetase (AARS) mutant (sti) mice with neurodegenerative disorder, we investigated the effect of translational fidelity in immune cells. Dysfunctional AARS caused disorders in immune cell responses and cellularity. The impairment was caused by dampened TCR signaling than cytokine signaling. Therefore, sti mutant inhibits TCR signaling, impeding T cell survival and responses. B cell numbers were decreased in sti mice. Despite low B cell cellularity, serum IgM, IgA, and IgE levels were higher in sti mice than in wild-type mice. Misacylation of ARS and the consequent translational infidelity induce disturbances in signaling pathways critical for immune cell survival and responses. Our findings provide a novel mechanism by which translational fidelity might play a critical role in cellular and humoral immune responses.
Collapse
Affiliation(s)
- Ju A Shim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Yuna Jo
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Hyunju Hwang
- Asan Institute for Life Sciences and Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - So Eun Lee
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Dahyeon Ha
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Jun Hwa Lee
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Jayoung Kim
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Parkyong Song
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Changwan Hong
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea.
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea.
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea.
| |
Collapse
|
66
|
Potential therapeutic strategies in chronic inflammatory demyelinating polyradiculoneuropathy. Clin Exp Rheumatol 2022; 21:103032. [PMID: 34999243 DOI: 10.1016/j.autrev.2022.103032] [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: 12/06/2021] [Accepted: 01/01/2022] [Indexed: 11/23/2022]
Abstract
Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is an autoimmune neuropathy involving peripheral nerve and nerve roots. The pathological hallmark of CIDP is macrophage-induced demyelination. Antibodies against nerve fibers, complement decomposition, abnormalities in plasma and cerebrospinal fluid cytokine profile, and changes of peripheral blood cell proportion were also reported in CIDP patients. These findings in immunopathology provide support for the introduction of potential therapeutic options for the treatment of CIDP. In this review, we systematically listed the potential therapeutic strategies targeting different components of the immune system by comparing the treatment of other autoimmune inflammatory diseases of the nervous system. Several ongoing clinical trials will assess the efficacy and safety of potential CIDP treatments.
Collapse
|
67
|
Shater H, Fawzy M, Farid A, El-Amir A, Fouad S, Madbouly N. The potential use of serum interleukin-21 as biomarker for lupus nephritis activity compared to cytokines of the tumor necrosis factor (TNF) family. Lupus 2022; 31:55-64. [PMID: 34978958 DOI: 10.1177/09612033211063794] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Lupus nephritis (LN) is a significant consequence of systemic lupus erythematosus (SLE). To the best of our knowledge, this is the first work that focuses on evaluation of serum interleukin (IL-) 21 as a diagnostic biomarker of LN activity, compared to B lymphocyte stimulator (BlyS), tumor necrosis factor ligand superfamily member 13 (TNF-SF13), and traditional techniques of active LN attempting to compare their diagnostic usefulness. METHODS Serum levels of IL-21, BlyS, and TNF-SF13 during LN were investigated. Twenty-five biopsy-proven, active LN female patients and 15 SLE patients without active LN and 20 healthy controls (HCs) joined this work. RESULTS Serum IL-21 level was significantly higher in active LN group than in inactive LN group. Correlation analysis showed that serum IL-21 levels were significantly correlated with total SLEDAI (r = 0.41, p = 0.03), renal-SLEDAI (r = 0.48, p = 0.04), renal activity index (AI) (r = 0.93; p < 0.001), and 24-h proteinuria (r = 0.51; p > 0.008). Receiver operating characteristic curve (ROC) revealed the ability of serum IL-21 to discriminate between active and inactive LN with 70% sensitivity at >240 pg/ml cutoff point (AUC 0.809). CONCLUSION For Egyptian SLE patients, serum levels of IL-21 were superior to TNF-SF13 and BlyS and correlated significantly with the activity indexes of LN, indicating a promising role as a potential biomarker of active LN.
Collapse
Affiliation(s)
- Hend Shater
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Mary Fawzy
- Department of Internal Medicine, Faculty of Medicine (Kasr Al-Ainy School of Medicine), 98876Cairo University, Cairo, Egypt
| | - Alyaa Farid
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Azza El-Amir
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Salwa Fouad
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Neveen Madbouly
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| |
Collapse
|
68
|
Kneiber D, Kowalski EH, Amber KT. The Immunogenetics of Autoimmune Blistering Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1367:173-212. [DOI: 10.1007/978-3-030-92616-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
|
69
|
Merino-Vico A, van Hamburg JP, Tas SW. B Lineage Cells in ANCA-Associated Vasculitis. Int J Mol Sci 2021; 23:387. [PMID: 35008813 PMCID: PMC8745114 DOI: 10.3390/ijms23010387] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/21/2021] [Accepted: 12/28/2021] [Indexed: 12/18/2022] Open
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a systemic autoimmune disease that affects small sized blood vessels and can lead to serious complications in the lungs and kidneys. The prominent presence of ANCA autoantibodies in this disease implicates B cells in its pathogenesis, as these are the precursors of the ANCA-producing plasma cells (PCs). Further evidence supporting the potential role of B lineage cells in vasculitis are the increased B cell cytokine levels and the dysregulated B cell populations in patients. Confirmation of the contribution of B cells to pathology arose from the beneficial effect of anti-CD20 therapy (i.e., rituximab) in AAV patients. These anti-CD20 antibodies deplete circulating B cells, which results in amelioration of disease. However, not all patients respond completely, and this treatment does not target PCs, which can maintain ANCA production. Hence, it is important to develop more specific therapies for AAV patients. Intracellular signalling pathways may be potential therapeutic targets as they can show (disease-specific) alterations in certain B lineage cells, including pathogenic B cells, and contribute to differentiation and survival of PCs. Preliminary data on the inhibition of certain signalling molecules downstream of receptors specific for B lineage cells show promising therapeutic effects. In this narrative review, B cell specific receptors and their downstream signalling molecules that may contribute to pathology in AAV are discussed, including the potential to therapeutically target these pathways.
Collapse
Affiliation(s)
- Ana Merino-Vico
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.-V.); (J.P.v.H.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Jan Piet van Hamburg
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.-V.); (J.P.v.H.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Sander W. Tas
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.-V.); (J.P.v.H.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
70
|
Role of B-Cell Activating Factor (BAFF) in Inflammatory Bowel Disease. Diagnostics (Basel) 2021; 12:diagnostics12010045. [PMID: 35054212 PMCID: PMC8774757 DOI: 10.3390/diagnostics12010045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
As early commencement of inflammatory bowel disease (IBD) treatment has been shown to substantially improve outcomes, it is of utmost importance to make a timely diagnosis of this disease. Despite undisputed sensitivity of fecal calprotectin, the most widely accepted IBD biomarker, in discriminating between irritable bowel syndrome (IBS) and IBD, as well as recognized role in monitoring disease activity and response to therapy, perhaps the biggest setback of calprotectin use in IBD is lack of specificity. Therefore, an additional biomarker in IBD is warranted. B-cell activating factor (BAFF), a member of the tumor necrosis factor (TNF) superfamily, recently emerged as a viable candidate for this role. So far, overproduction of BAFF has been observed in various autoimmune diseases, most notably in systemic lupus erythematosus, where BAFF-inhibitor belimumab was approved for treatment. As BAFF levels were also shown to correlate with indices of IBD, in this review we aimed to summarize the current evidence with respect to the role of BAFF in diagnosis and assessing the activity of IBD, as well as putative therapeutic implications that may arise from exploring of this relation.
Collapse
|
71
|
Spinosa MD, Montgomery WG, Lempicki M, Srikakulapu P, Johnsrude MJ, McNamara CA, Upchurch GR, Ailawadi G, Leitinger N, Meher AK. B Cell-Activating Factor Antagonism Attenuates the Growth of Experimental Abdominal Aortic Aneurysm. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:2231-2244. [PMID: 34509440 PMCID: PMC8647430 DOI: 10.1016/j.ajpath.2021.08.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/02/2021] [Accepted: 08/20/2021] [Indexed: 10/20/2022]
Abstract
B cell-activating factor (BAFF), part of a tumor necrosis factor family of cytokines, was recently identified as a regulator of atherosclerosis; however, its role in aortic aneurysm has not been determined. Here, the study examined the effect of selective BAFF antagonism using an anti-BAFF antibody (blocks binding of BAFF to receptors BAFF receptor 3, transmembrane activator and CAML interactor, and B-cell maturation antigen) and mBaffR-mFc (blocks binding of BAFF to BAFF receptor 3) on a murine model of abdominal aortic aneurysm (AAA). In a prevention strategy, the antagonists were injected before the induction of AAA, and in an intervention strategy, the antagonists were injected after the induction of AAA. Both strategies attenuated the formation of AAA. In the intervention group, BAFF antagonism depleted most of the mature B-cell subsets in spleen and circulation, leading to enhanced resolution of inflammation in AAA as indicated by decreased infiltration of B cells and proinflammatory macrophages and a reduced number of apoptotic cells. In AAA tissues, B cells and macrophages were found in close contact. In vitro, B cells, irrespective of treatment with BAFF, impaired the efferocytosis activity of macrophages, suggesting a direct innate role of B cells on macrophage function. Altogether, BAFF antagonism affects survival of the mature B cells, promotes resolution of inflammation in the aorta, and attenuates the growth of AAA in mice.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/immunology
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/therapy
- B-Cell Activating Factor/antagonists & inhibitors
- B-Cell Activating Factor/genetics
- B-Cell Activating Factor/immunology
- B-Cell Activating Factor/physiology
- B-Lymphocyte Subsets/pathology
- Cell Count
- Cells, Cultured
- Chemotaxis, Leukocyte/physiology
- Disease Models, Animal
- Disease Progression
- Humans
- Immunoglobulin Fc Fragments/pharmacology
- Immunoglobulin Fc Fragments/therapeutic use
- Macrophages/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
Collapse
Affiliation(s)
- Michael D Spinosa
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | | | - Melissa Lempicki
- Department of Microbiology and Immunology, East Carolina University, Greenville, North Carolina
| | - Prasad Srikakulapu
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia
| | - Matthew J Johnsrude
- Department of Microbiology and Immunology, East Carolina University, Greenville, North Carolina
| | - Coleen A McNamara
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia
| | - Gilbert R Upchurch
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Gorav Ailawadi
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Akshaya K Meher
- Department of Microbiology and Immunology, East Carolina University, Greenville, North Carolina; Department of Pharmacology, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
72
|
Wei M, Guo H, Liu S, Xu F, Zhang Y, Shi J, Xu Z, Chen Y. Combined immune score predicts the prognosis of newly diagnosed multiple myeloma patients in the bortezomib-based therapy era. Medicine (Baltimore) 2021; 100:e27521. [PMID: 34731142 PMCID: PMC8519192 DOI: 10.1097/md.0000000000027521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 09/27/2021] [Indexed: 01/05/2023] Open
Abstract
To investigate the effect of a combined immune score including the lymphocyte-to-monocyte ratio (LMR) and uninvolved immunoglobulin (u-Ig) levels on the prognosis of newly diagnosed multiple myeloma (NDMM) patients treated with bortezomib.Clinical data of 201 NDMM patients were retrospectively analyzed. Patients with LMR ≥ 3.6 and LMR < 3.6 were scored 0 and 1, respectively. Patients with preserved u-Ig levels, suppression of 1 u-Ig, and suppression of at least 2 u-Igs were scored 0, 1, and 2, respectively. The immune score, established from these individual scores, was used to separate patients into good (0-1 points), intermediate (2 points), and poor (3 points) risk groups. The baseline data, objective remission rate (ORR), whether receive maintenance treatment regularly and overall survival of patients before treatment were analyzed.The ORR of the good-risk group was significantly higher than that of the intermediate-risk group (75.6% vs 57.7%, P = .044) and the poor-risk group (75.6% vs 48.2%, P = .007). The multivariate analysis results showed that age ≥ 65 years, International Staging System stage III, platelet count ≤ 100 × 109/L, lactate dehydrogenase (LDH) > 250 U/L, serum calcium > 2.75 mmol/L, no receipt of regular maintenance treatment, LMR < 3.6, suppressed u-Igs = 1, suppressed u-Igs ≥ 2, intermediate-risk group and poor-risk group were independent predictors of poor overall survival.In the bortezomib era, the LMR, u-Ig levels, and the immune score play an important role in the prognosis of NDMM patients. Among them, the immune score showed the strongest prognostic value, and it could be a beneficial supplement for the early identification of high-risk patients.
Collapse
Affiliation(s)
- Min Wei
- Department of Hematology, Henan University People's Hospital, School of Clinical Medicine, Henan University, Zhengzhou, Henan, People's Republic of China
| | - Honggang Guo
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| | - Siwei Liu
- Department of Hematology, Peking University First Hospital, Peking, People's Republic of China
| | - Fangfang Xu
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| | - Yin Zhang
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| | - Jie Shi
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| | - Zhiwei Xu
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| | - Yuqing Chen
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
73
|
Czaja AJ. Review article: targeting the B cell activation system in autoimmune hepatitis. Aliment Pharmacol Ther 2021; 54:902-922. [PMID: 34506662 DOI: 10.1111/apt.16574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/30/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND The B cell activation system, consisting of B cell activating factor and a proliferation-inducing ligand, may have pathogenic effects in autoimmune hepatitis. AIMS To describe the biological actions of the B cell activation system, indicate its possible role in autoimmune diseases, and evaluate its prospects as a therapeutic target in autoimmune hepatitis METHODS: English abstracts were identified in PubMed by multiple search terms. Full length articles were selected for review, and secondary and tertiary bibliographies were developed. RESULTS The B cell activating factor is crucial for the maturation and survival of B cells, and it can co-stimulate T cell activation, proliferation, and survival. It can also modulate the immune response by inducing interleukin 10 production by regulatory B cells. A proliferation-inducing ligand modulates and diversifies the antibody response by inducing class-switch recombination in B cells. It can also increase the proliferation, survival, and antigen activation of T cells. These immune stimulatory actions can be modulated by inducing proliferation of regulatory T cells. The B cell activation system has been implicated in diverse autoimmune diseases, and therapeutic blockade is a management strategy now being evaluated in autoimmune hepatitis. CONCLUSIONS The B cell activation system has profound effects on B and T cell function in autoimmune diseases. Blockade therapy is being actively evaluated in autoimmune hepatitis. Clarification of the critical pathogenic components of the B cell activation system will improve the targeting, efficacy, and safety of blockade therapy in this disease.
Collapse
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| |
Collapse
|
74
|
Yamamoto Y, Matsui N, Uzawa A, Ozawa Y, Kanai T, Oda F, Kondo H, Ohigashi I, Takizawa H, Kondo K, Sugano M, Kitaichi T, Hata H, Kaji R, Kuwabara S, Yamamura T, Izumi Y. Intrathymic Plasmablasts Are Affected in Patients With Myasthenia Gravis With Active Disease. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/6/e1087. [PMID: 34561276 PMCID: PMC8474506 DOI: 10.1212/nxi.0000000000001087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 08/04/2021] [Indexed: 12/31/2022]
Abstract
Background and Objectives To investigate intrathymic B lymphopoiesis in patients with myasthenia gravis (MG) and explore thymus pathology associated with clinical impact. Methods Thymic lymphocytes from 15 young patients without MG, 22 adult patients without MG, 14 patients with MG without thymoma, and 11 patients with MG with thymoma were subjected to flow cytometry analysis of T follicular helper (Tfh), naive B, memory B, plasmablasts, CD19+B220high thymic B cells, B-cell activating factor receptor, and C-X-C chemokine receptor 5 (CXCR5). Peripheral blood mononuclear cells of 16 healthy subjects and 21 untreated patients with MG were also analyzed. Immunologic values were compared, and correlations between relevant values and clinical parameters were evaluated. Results The frequencies of circulating and intrathymic plasmablasts were significantly higher in patients with MG than controls. On the other hand, the frequency of CD19+B220high thymic B cells was not increased in MG thymus. We observed a significant increase in CXCR5 expression on plasmablasts in MG thymus and an increased frequency of intrathymic plasmablasts that was correlated with preoperative disease activity. The frequency of intrathymic Tfh cells was significantly lower in patients who received immunosuppressive (IS) therapy than those without IS therapy. However, there was no significant difference in the frequency of intrathymic plasmablasts irrespective of IS therapy. Discussion Our findings confirmed a correlation between increased frequency of intrathymic plasmablasts and disease activity before thymectomy. We postulate that activated intrathymic plasmablasts endow pathogenic capacity in MG.
Collapse
Affiliation(s)
- Yohei Yamamoto
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Naoko Matsui
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan.
| | - Akiyuki Uzawa
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Yukiko Ozawa
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Tetsuya Kanai
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Fumiko Oda
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Hiroyuki Kondo
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Izumi Ohigashi
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Hiromitsu Takizawa
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Kazuya Kondo
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Mikio Sugano
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Takashi Kitaichi
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Hiroki Hata
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Ryuji Kaji
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Satoshi Kuwabara
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Takashi Yamamura
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Yuishin Izumi
- From the Department of Neurology (Y.Y., N.M., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (A.U., Y.O., T. Kanai, F.O., S.K.), Graduate School of Medicine, Chiba University; Division of Experimental Immunology (H.K., I.O.), Institute of Advanced Medical Sciences, Tokushima University; Department of Thoracic, Endocrine Surgery and Oncology (H.T.), Tokushima University Graduate School of Biomedical Sciences; Department of Oncological Medical Services (K.K.), Tokushima University Graduate School of Biomedical Sciences; Department of Cardiovascular Surgery (M.S., T. Kitaichi, H.H.), Tokushima University Graduate School of Biomedical Sciences; Department of Neurology (R.K.), National Hospital Organization Utano Hospital, Kyoto; and Department of Immunology (T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| |
Collapse
|
75
|
Shi F, Xue R, Zhou X, Shen P, Wang S, Yang Y. Telitacicept as a BLyS/APRIL dual inhibitor for autoimmune disease. Immunopharmacol Immunotoxicol 2021; 43:666-673. [PMID: 34519594 DOI: 10.1080/08923973.2021.1973493] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The pathogenic roles for B cells in autoimmunity include produce pathogenic autoantibodies and modulate immune responses via the production of cytokines and chemokines. The B lymphocyte stimulator BLyS (also known as B-cell-activating factor, BAFF) and APRIL (a proliferation-inducing ligand) are critical factors in the maintenance of the B-cell pool and humoral immunity, namely BLyS modulates the differentiation and maturation of immature B cell, while APRIL modulates the function and survival of long-lived plasma cell, which plays a prominent role in the pathogenesis of autoimmune diseases. Telitacicept is a novel recombinant fusion protein of both the ligand-binding domain of the TACI receptor and the Fc component of human IgG and which is a BLyS/APRIL dual inhibitor. Moreover, telitacicept was developed by Remegen Co., Ltd. in China and is approved to treat systemic lupus erythematosus in China. We review the rationale, clinical evidence, and future perspectives of telitacicept for the treatment of autoimmune disease.HighlightThe B lymphocyte stimulator BLyS (also known as B-cell-activating factor, BAFF) and APRIL (a proliferation-inducing ligand), members of tumor necrosis factor (TNF) family, and which are critical factors in the maintenance of the B-cell pool and humoral immunity.BAFF and APRIL are implicated in the pathogenesis of several human autoimmune diseases with autoreactive B-cell involvement, and targeting both is beneficial for the treatment of autoimmune diseases.Telitacicept is a novel recombinant fusion protein of both the ligand-binding domain of the TACI receptor and the Fc component of human IgG, as a BLyS/APRIL dual inhibitor and which has been approved by National Medical Products Administration (MNPA) for the treatment of patients with SLE in China.With more clinical trials underway, telitacicept may also be approved for the treatment of other autoimmune diseases in the future.
Collapse
Affiliation(s)
- Fan Shi
- School of Stomatology of Qingdao University, Qingdao, China.,Department of Oral and Maxillofacial Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Ran Xue
- Department of Pharmacy, The First Affiliated Hospital of Xi 'an Jiaotong University, Chang'an District Hospital, Xi 'an, China
| | - Xuexiao Zhou
- School of Stomatology of Qingdao University, Qingdao, China.,Department of Oral and Maxillofacial Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Pei Shen
- School of Stomatology of Qingdao University, Qingdao, China.,Department of Oral and Maxillofacial Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Shengzhi Wang
- Department of Oral and Maxillofacial Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yun Yang
- Department of Oral and Maxillofacial Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| |
Collapse
|
76
|
Engh JA, Ueland T, Agartz I, Andreou D, Aukrust P, Boye B, Bøen E, Drange OK, Elvsåshagen T, Hope S, Høegh MC, Joa I, Johnsen E, Kroken RA, Lagerberg TV, Lekva T, Malt UF, Melle I, Morken G, Nærland T, Steen VM, Wedervang-Resell K, Weibell MA, Westlye LT, Djurovic S, Steen NE, Andreassen OA. Plasma Levels of the Cytokines B Cell-Activating Factor (BAFF) and A Proliferation-Inducing Ligand (APRIL) in Schizophrenia, Bipolar, and Major Depressive Disorder: A Cross Sectional, Multisite Study. Schizophr Bull 2021; 48:37-46. [PMID: 34499169 PMCID: PMC8781325 DOI: 10.1093/schbul/sbab106] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Immune dysfunction has been implicated in the pathogenesis of schizophrenia and other nonaffective psychosis (SCZ), bipolar spectrum disorder (BIP) and major depressive disorder (MDD). The cytokines B cell-activating factor (BAFF) and A proliferation-inducing ligand (APRIL) belong to the tumor necrosis factor (TNF) super family and are essential in orchestrating immune responses. Abnormal levels of BAFF and APRIL have been found in autoimmune diseases with CNS affection. METHODS We investigated if plasma levels of BAFF and APRIL differed between patients with SCZ, BIP, and MDD with psychotic symptoms (n = 2009) and healthy control subjects (HC, n = 1212), and tested for associations with psychotic symptom load, controlling for sociodemographic status, antipsychotic and other psychotropic medication, smoking, body-mass-index, and high sensitivity CRP. RESULTS Plasma APRIL level was significantly lower across all patient groups compared to HC (P < .001; Cohen's d = 0.33), and in SCZ compared to HC (P < .001; d = 0.28) and in BIP compared to HC (P < .001; d = 0.37). Lower plasma APRIL was associated with higher psychotic symptom load with nominal significance (P = .017), but not with any other clinical characteristics. Plasma BAFF was not significantly different across patient groups vs HC, but significantly higher in BIP compared to HC (P = .040; d = 0.12) and SCZ (P = .027; d = 0.10). CONCLUSIONS These results show aberrant levels of BAFF and APRIL and association with psychotic symptoms in patients with SCZ and BIP. This suggest that dysregulation of the TNF system, mediated by BAFF and APRIL, is involved in the pathophysiology of psychotic disorders.
Collapse
Affiliation(s)
- John Abel Engh
- Norwegian Centre for Mental Disorders Research, NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Vestfold Hospital Trust, Division of Mental health and Addiction, Tønsberg, Norway,To whom correspondence should be addressed; Norwegian Centre for Mental Disorders Research, NORMENT, Oslo, Norway; tel: 023-02-73-50 (022-11-78-43 dir), fax: 023-02-73-33,
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway,K.G. Jebsen Thrombosis Research and Expertise Center, University of Troms, Tromsø, Norway
| | - Ingrid Agartz
- Norwegian Centre for Mental Disorders Research, NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden,Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Dimitrios Andreou
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Birgitte Boye
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway,Psychosomatic and Consultation-liason Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Erlend Bøen
- Psychosomatic and Consultation-liason Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ole Kristian Drange
- Department of Mental Health, Norwegian University of Science and Technology, NTNU, Trondheim, Norway,Department of Østmarka, Division of Mental Health, St. Olavs University Hospital, Trondheim, Norway,Department of Psychiatry, St Olav University Hospital, Trondheim, Norway
| | - Torbjørn Elvsåshagen
- Norwegian Centre for Mental Disorders Research, NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Sigrun Hope
- Norwegian Centre for Mental Disorders Research, NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Department of Neuro Habilitation, Oslo University Hospital Ullevål, Oslo, Norway
| | - Margrethe Collier Høegh
- Norwegian Centre for Mental Disorders Research, NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Inge Joa
- TIPS, Network for Clinical Research in Psychosis, Stavanger University Hospital, Stavanger, Norway,Network for Medical Sciences, Faculty of Health, University of Stavanger, Stavanger, Norway
| | - Erik Johnsen
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway,University of Bergen, Bergen, Norway,Norwegian Centre for Mental Disorders Research, NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Rune Andreas Kroken
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway,University of Bergen, Bergen, Norway,Norwegian Centre for Mental Disorders Research, NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Trine Vik Lagerberg
- Norwegian Centre for Mental Disorders Research, NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | | | - Ingrid Melle
- Norwegian Centre for Mental Disorders Research, NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gunnar Morken
- Department of Mental Health, Norwegian University of Science and Technology, NTNU, Trondheim, Norway,Department of Psychiatry, St Olav University Hospital, Trondheim, Norway
| | - Terje Nærland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway,K.G. Jebsen Center for Neurodevelopmental Disorders, Oslo, Norway,Department of Rare Disorders and Disabilities, Oslo University Hospital, Oslo, Norway
| | - Vidar Martin Steen
- University of Bergen, Bergen, Norway,Norwegian Centre for Mental Disorders Research, NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway,Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Kirsten Wedervang-Resell
- Norwegian Centre for Mental Disorders Research, NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Melissa Auten Weibell
- TIPS, Network for Clinical Research in Psychosis, Stavanger University Hospital, Stavanger, Norway,Network for Medical Sciences, Faculty of Health, University of Stavanger, Stavanger, Norway
| | - Lars Tjelta Westlye
- Norwegian Centre for Mental Disorders Research, NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Department of Psychology, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- Norwegian Centre for Mental Disorders Research, NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway,Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Nils Eiel Steen
- Norwegian Centre for Mental Disorders Research, NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole Andreas Andreassen
- Norwegian Centre for Mental Disorders Research, NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
77
|
Wang L, Zhang T, Zhang Z, Wang Z, Zhou YJ, Wang Z. B cell activating factor regulates periodontitis development by suppressing inflammatory responses in macrophages. BMC Oral Health 2021; 21:426. [PMID: 34481478 PMCID: PMC8418735 DOI: 10.1186/s12903-021-01788-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/28/2021] [Indexed: 01/02/2023] Open
Abstract
Background B cell activating factor (BAFF) is a member of the tumor necrosis factor (TNF) superfamily with immunomodulatory effects on both innate and adaptive immune responses. Periodontitis is an inflammatory disease characterized by periodontal soft tissue inflammation and the progressive loss of periodontal ligament and alveolar bone. Macrophages are closely related to periodontitis progression. However, the role of BAFF in periodontitis development and macrophage polarization and the underlying mechanism remain unknown. Methods In vivo, a ligation-induced mouse model of periodontitis for BAFF blockade was established to investigate the expression of inducible nitric oxide synthase (iNOS) through real-time PCR (RT-PCR) and immunohistochemistry. In addition, the level of TNF-α in the periodontium, the number of osteoclasts, and alveolar bone resorption were observed. In vitro, RAW 264.7 macrophage cells were treated with 100 ng/mL Porphyromonas gingivalis lipopolysaccharide (P. gingivalis LPS) in either the presence or absence of 50 nM small interfering RNA (siRNA) targeting BAFF, followed by further incubation for 24 h. These cells and supernatants were collected and stored for RT-PCR, enzyme-linked immunosorbent assay, western blotting and immunofluorescence microscopy. Results In vivo, BAFF blockade decreased the levels of TNF-α in the periodontium in a ligature-induced mouse periodontitis model. Reduced osteoclast formation and lower alveolar bone loss were also observed. In addition, BAFF blockade was related to the expression of polarization signature molecules in macrophages. In vitro, BAFF knockdown notably suppressed the production of TNF-α in RAW 264.7 cells stimulated by P. gingivalis LPS. Moreover, BAFF knockdown attenuated the polarization of RAW 264.7 cells into classically activated macrophages (M1), with reduced expression of iNOS. Conclusions Based on our limited evidence, we showed BAFF blockade exhibits potent anti-inflammatory properties in mice experimental periodontitis in vivo and in P. gingivalis LPS-treated RAW 264.7 cells in vitro, and macrophage polarization may be responsible for this effect. Supplementary Information The online version contains supplementary material available at 10.1186/s12903-021-01788-6.
Collapse
Affiliation(s)
- Lixia Wang
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8th Gongti South Road, Beijing, 100020, China.,International Medical Center, Tianjin Stomatological Hospital, School Medicine, Nankai University, 75th Dagu North Road, Tianjin, 300041, China.,Tianjin Key Laboratory of Oral Maxillofacial Function Reconstruction, 75th Dagu North Road, Tianjin, 300041, China
| | - Tianyi Zhang
- Department of Stomatology, School of Stomatology, Shanxi Medical University, 56 Xinjian South Road, Yingze, Taiyuan, 030001, Shaanxi, China
| | - Zheng Zhang
- International Medical Center, Tianjin Stomatological Hospital, School Medicine, Nankai University, 75th Dagu North Road, Tianjin, 300041, China.,Tianjin Key Laboratory of Oral Maxillofacial Function Reconstruction, 75th Dagu North Road, Tianjin, 300041, China
| | - Zihan Wang
- Department of Immunology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Beijing Key Laboratory for Cancer Invasion and Metastasis, Department of Oncology, Capital Medical University, Beijing, China
| | - Yu-Jie Zhou
- Department of Immunology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Beijing Key Laboratory for Cancer Invasion and Metastasis, Department of Oncology, Capital Medical University, Beijing, China.
| | - Zuomin Wang
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8th Gongti South Road, Beijing, 100020, China.
| |
Collapse
|
78
|
Olaso D, Manook M, Moris D, Knechtle S, Kwun J. Optimal Immunosuppression Strategy in the Sensitized Kidney Transplant Recipient. J Clin Med 2021; 10:3656. [PMID: 34441950 PMCID: PMC8396983 DOI: 10.3390/jcm10163656] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/09/2021] [Accepted: 08/15/2021] [Indexed: 01/10/2023] Open
Abstract
Patients with previous sensitization events against anti-human leukocyte antigens (HLA) often have circulating anti-HLA antibodies. Following organ transplantation, sensitized patients have higher rates of antibody-mediated rejection (AMR) compared to those who are non-sensitized. More stringent donor matching is required for these patients, which results in a reduced donor pool and increased time on the waitlist. Current approaches for sensitized patients focus on reducing preformed antibodies that preclude transplantation; however, this type of desensitization does not modulate the primed immune response in sensitized patients. Thus, an optimized maintenance immunosuppressive regimen is necessary for highly sensitized patients, which may be distinct from non-sensitized patients. In this review, we will discuss the currently available therapeutic options for induction, maintenance, and adjuvant immunosuppression for sensitized patients.
Collapse
Affiliation(s)
| | | | | | - Stuart Knechtle
- Duke Transplant Center, Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA; (D.O.); (M.M.); (D.M.)
| | - Jean Kwun
- Duke Transplant Center, Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA; (D.O.); (M.M.); (D.M.)
| |
Collapse
|
79
|
Lower BAFF Levels in Myasthenic Patients Treated with Glucocorticoids. Arch Immunol Ther Exp (Warsz) 2021; 69:22. [PMID: 34338918 PMCID: PMC8328853 DOI: 10.1007/s00005-021-00626-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/06/2021] [Indexed: 10/26/2022]
Abstract
B-cell activating factor (BAFF), a member of tumor necrosis factor family, activates B cells, promotes their survival and proliferation. BAFF is considered to have an influence on development of autoimmune diseases including myasthenia gravis (MG). We aimed to evaluate BAFF serum levels in MG patients, their potential connection with therapy and course of MG. Cross-sectional study. Two hundred eighteen adult patients with MG (67% women, age: 18-89 years, 82.6% AChR antibody seropositive (AChRAb(+)). Serum BAFF levels, their relationship with severity of clinical symptoms, therapy conducted, clinical and demographic features and other factors were analyzed. Patients with AChRAb(+) MG demonstrated significantly higher BAFF levels than MuSK-MG patients (831.2 ± 285.4 pg/ml vs. 745.6 ± 633.4 pg/ml, respectively; p = 0.030). Serum BAFF levels in women were significantly higher than in men (855.9 ± 302.5 vs. 756.6 ± 289.4, respectively; p = 0.017). Mean serum BAFF level was significantly decreased in patients who were ever treated with corticosteroids (CS) (770.4 ± 327.8 pg/ml vs. 891.3 ± 246.1 pg/ml, respectively; p = 0.001). Thymoma-MG patients demonstrated significantly lower BAFF levels (671.2 ± 244.9 vs. 833.5 ± 302.4, respectively; p = 0.044). Thymectomized patients did not differ in BAFF levels from the MG patients who had not undergone thymectomy. In multiple linear regression model, recent CS therapy and male sex were found to be independent predictors of lower BAFF levels. Serum BAFF level is decreased in patients treated with CS, which may suggest inhibiting influence of CS on BAFF-a potential mechanism contributing to the effectiveness of such therapy.
Collapse
|
80
|
Isolation of primary human B lymphocytes from tonsils compared to blood as alternative source for ex vivo application. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1179:122853. [PMID: 34325309 DOI: 10.1016/j.jchromb.2021.122853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 01/18/2023]
Abstract
B lymphocytes ('B cells') are components of the human immune system with obvious potential for medical and biotechnological applications. Here, we discuss the isolation of primary human B cells from both juvenile and adult tonsillar material using a two-step procedure based on gradient centrifugation followed by separation on a nylon wool column as alternative to the current gold standard, i.e., negative immunosorting from buffy coats by antibody-coated magnetic beads. We show that the nylon wool separation is a low-cost method well suited to the isolation of large amounts of primary B cells reaching purities ≥ 80%. More importantly, this method allows the preservation of all B cell subsets, while MACS sorting seems to be biased against a certain B cell subtype, namely the CD27+ B cells. Importantly, compared to blood, the excellent recovery yield during purification of tonsillar B cells provides high number of cells, hence increases the number of subsequent experiments feasible with identical cell material, consequently improving comparability of results. The cultivability of the isolated B cells was demonstrated using pokeweed mitogen (PWM) as a stimulatory substance. Our results showed for the first time that the proliferative response of tonsillar B cells to mitogens declines with the age of the donor. Furthermore, we observed that PWM treatment stimulates the proliferation of a dedicated subpopulation and induces some terminal differentiation with ASCs signatures. Taken together this indicates that the proposed isolation procedure preserves the proliferative capability as well as the differentiation capacity of the B cells.
Collapse
|
81
|
Eslami M, Schneider P. Function, occurrence and inhibition of different forms of BAFF. Curr Opin Immunol 2021; 71:75-80. [PMID: 34182216 DOI: 10.1016/j.coi.2021.06.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/26/2021] [Accepted: 06/06/2021] [Indexed: 01/27/2023]
Abstract
B cell activating factor (BAFF or BLyS), an important cytokine for B cell survival and humoral immune responses, is targeted in the clinic for the treatment of systemic lupus erythematosus. This review focuses on the structure, function and inhibition profiles of membrane-bound BAFF, soluble BAFF 3-mer and soluble BAFF 60-mer, all of which have distinct properties. BAFF contains a loop region not required for receptor binding but essential for receptor activation via promotion of BAFF-to-BAFF contacts. This loop region additionally allows formation of BAFF 60-mer, in which epitopes of the BAFF inhibitor belimumab are inaccessible. If 60-mer forms in humans, it is predicted to be short-lived and to act locally because adult serum contains a BAFF 60-mer dissociating activity. Cord blood contains elevated levels of BAFF, part of which displays attributes of 60-mer, suggesting a role for this form of BAFF in the development of foetal or neonate B cells.
Collapse
Affiliation(s)
- Mahya Eslami
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
82
|
Atisha-Fregoso Y, Toz B, Diamond B. Meant to B: B cells as a therapeutic target in systemic lupus erythematosus. J Clin Invest 2021; 131:149095. [PMID: 34128474 PMCID: PMC8203443 DOI: 10.1172/jci149095] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
B cells have a prominent role in the pathogenesis of systemic lupus erythematosus (SLE). They are mediators of inflammation through the production of pathogenic antibodies that augment inflammation and cause direct tissue and cell damage. Multiple therapeutic agents targeting B cells have been successfully used in mouse models of SLE; however, these preclinical studies have led to approval of only one new agent to treat patients with SLE: belimumab, a monoclonal antibody targeting B cell-activating factor (BAFF). Integrating the experience acquired from previous clinical trials with the knowledge generated by new studies about mechanisms of B cell contributions to SLE in specific groups of patients is critical to the development of new treatment strategies that will help to improve outcomes in patients with SLE. In particular, a sharper focus on B cell differentiation to plasma cells is warranted.
Collapse
Affiliation(s)
- Yemil Atisha-Fregoso
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, New York, USA
| | - Bahtiyar Toz
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Queens Hospital Center, New York, New York, USA
| | - Betty Diamond
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| |
Collapse
|
83
|
Chen X, Ma J, Yao Y, Zhu J, Zhou Z, Zhao R, Dong X, Gao W, Zhang S, Huang S, Chen L. Metformin prevents BAFF activation of Erk1/2 from B-cell proliferation and survival by impeding mTOR-PTEN/Akt signaling pathway. Int Immunopharmacol 2021; 96:107771. [PMID: 34004440 DOI: 10.1016/j.intimp.2021.107771] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/15/2022]
Abstract
B-cell activating factor (BAFF) is an essential cytokine for B-cell maturation, differentiation and survival, and excess BAFF induces aggressive or neoplastic B-cell disorders and contributes to development of autoimmune diseases. Metformin, an anti-diabetic drug, has recently garnered a great attention due to its anti-proliferative and immune-modulatory features. However, little is known regarding the effect of metformin on BAFF-stimulated B cells. Here, we show that metformin attenuated human soluble BAFF (hsBAFF)-induced cell proliferation and survival by blocking the Erk1/2 pathway in normal and B-lymphoid (Raji) cells. Pretreatment with U0126, knockdown of Erk1/2, or expression of dominant negative MKK1 strengthened metformin's inhibition of hsBAFF-activated Erk1/2 and B-cell proliferation/viability, whereas expression of constitutively active MKK1 rendered high resistance to metformin. Further investigation found that overexpression of wild type PTEN or ectopic expression of dominant negative Akt potentiated metformin's suppression of hsBAFF-induced Erk1/2 activation and proliferation/viability in Raji cells, implying a PTEN/Akt-dependent mechanism involved. Furthermore, we noticed that metformin hindered hsBAFF-activated mTOR pathway in B cells. Inhibition of mTOR with rapamycin or knockdown of mTOR enhanced metformin's suppression of hsBAFF-induced phosphorylation of S6K1, PTEN, Akt, and Erk1/2, as well as B-cell proliferation/viability. These results indicate that metformin prevents BAFF activation of Erk1/2 from cell proliferation and survival by impeding mTOR-PTEN/Akt signaling pathway in normal and neoplastic B-lymphoid cells. Our findings support that metformin has a great potential for prevention of excessive BAFF-induced aggressive B-cell malignancies and autoimmune diseases.
Collapse
Affiliation(s)
- Xiaoling Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Jing Ma
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Yajie Yao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Jiawei Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Zhihan Zhou
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Rui Zhao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Xiaoqing Dong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Wei Gao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Shuangquan Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA; Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA.
| | - Long Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, PR China.
| |
Collapse
|
84
|
Wang J, Yang C, Hou X, Xu J, Yun Y, Qin L, Yang P. Rapamycin Modulates the Proinflammatory Memory-Like Response of Microglia Induced by BAFF. Front Immunol 2021; 12:639049. [PMID: 34054807 PMCID: PMC8158300 DOI: 10.3389/fimmu.2021.639049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/21/2021] [Indexed: 11/21/2022] Open
Abstract
Background Recently trained immunity of microglia provided an opportunity to study the chronic effect of microglial activation and its metabolic rewiring in neuroimmunological diseases. Since elevated levels of B cell-activating factor (BAFF) have been proved to be associated with some chronic neuroimmunological disorders. Here, we used the trained innate immunity model to analyze the effect of BAFF, a vital regulator of the adaptive immune system, on long-term microglial activation and metabolic reprogramming in vitro and in vivo. Methods and results In vitro, BV2 cells and mouse primary microglial cells were incubated with BAFF for 24 h (BAFF priming). After 5 days of resting, microglia were restimulated with LPS (LPS restimulation) or BAFF (BAFF restimulation). BAFF priming induced a pro-inflammatory trained immunity-phenotype of both BV2 cells and primary microglial cells, which was indicated by morphological change, secretion of pro-inflammatory cytokine and chemokine upon LPS restimulation or BAFF restimulation. The production of lactate and NAD+/NADH ratio were elevated 5 days after BAFF priming. The activation of the Akt/mTOR/HIF-1α pathway was induced by BAFF priming and lasted for 5 days. Pretreating the BV2 cells or mouse primary microglial cells with rapamycin blocked mTOR/HIF-1α activation and cellular metabolic reprogramming induced by BAFF training. Consistently, rapamycin efficiently suppressed the trained immunity-like responses of microglia triggered by BAFF. In vivo, adult male mice were treated with BAFF by intracerebroventricular injection for priming and 7 days later with BAFF for restimulation. BAFF training activated microglia in the cortex and hippocampus. The production of proinflammatory cytokines and chemokines was elevated after BAFF training. Conclusion Our current data, for the first time, demonstrate that BAFF priming induces a proinflammatory memory-like response of microglia not only to LPS but also to BAFF itself. Rapamycin inhibits microglial priming triggered by BAFF through targeting the mTOR/HIF-1α signaling pathway. Our data reveal a novel role of BAFF in trained immunity and that rapamycin may be a potential therapeutic target of neuroimmunological diseases.
Collapse
Affiliation(s)
- Jianing Wang
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Chunshu Yang
- Department of 1st Cancer Institute, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiaoyu Hou
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Jingyi Xu
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Yang Yun
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ling Qin
- Department of Physiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Pingting Yang
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
85
|
Takatani A, Nakamura H, Furukawa K, Endo Y, Umeda M, Shimizu T, Nishihata S, Kitaoka K, Nakamura T, Kawakami A. Inhibitory effect of HTLV-1 infection on the production of B-cell activating factors in established follicular dendritic cell-like cells. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:777-791. [PMID: 33943020 PMCID: PMC8342235 DOI: 10.1002/iid3.432] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/01/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022]
Abstract
Introduction The low frequency of ectopic germinal center in labial salivary glands of patients with human T‐cell leukemia virus type 1 (HTLV‐1) antibody‐positive Sjögren's syndrome (SS) suggests that HTLV‐1 has some effects on follicular dendritic cells (FDCs). Methods We used flow cytometry, immunofluorescence, and enzyme‐linked immunosorbent assays (ELISAs) to investigate the direct effect of HTLV‐1 on B‐cell activating factors produced by established FDC like cells obtained from excised human tonsils. We then measured the serum B‐cell activating factor (BAFF) and C‐X‐C motif ligand (CXCL) 13 concentrations of the HTLV‐1‐seropositive SS patients and the HTLV‐1‐seronegative SS patients by ELISA. Results Among the 31 isolated specimens, 22 showed morphological characteristics of FDCs. Day 2‐cultured specimens showed expressions of CD14, CD23, CD40, intracellular adhesion molecule‐1 (ICAM‐1), and vascular cell adhesion molecule‐1. After 2 weeks, 12 of these specimens expressed ICAM‐1, FDC, and fibroblast cell marker. Intracellular BAFF and CXCL13 were constitutively expressed regardless of stimulation. After direct coculture with the HTLV‐1‐infected T‐cell line HCT‐5 or MT‐2, the BAFF and CXCL13 expressions on the FDC‐like cells were decreased in accord with the increased number of HCT‐5 and MT‐2 cells with styliform change and without HTLV‐1 Gag protein expression. Interferons upregulated the concentration of BAFF (but not CXCL13) in the culture supernatant, which showed a declining trend under the presence of HCT‐5 or MT‐2. The serum concentrations of BAFF and CXCL13 in the HTLV‐1‐seropositive SS patients were lower than those of the HTLV‐1 seronegative SS patients. Conclusions HTLV‐1 partially inhibited the BAFF and CXCL13 expressions of established FDC‐like cells.
Collapse
Affiliation(s)
- Ayuko Takatani
- Division of Advanced Preventive Medical Sciences, Department of Immunology and RheumatologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Hideki Nakamura
- Division of Advanced Preventive Medical Sciences, Department of Immunology and RheumatologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Kaori Furukawa
- Division of Advanced Preventive Medical Sciences, Department of Immunology and RheumatologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Yushiro Endo
- Division of Advanced Preventive Medical Sciences, Department of Immunology and RheumatologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Masataka Umeda
- Division of Advanced Preventive Medical Sciences, Department of Immunology and RheumatologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Toshimasa Shimizu
- Division of Advanced Preventive Medical Sciences, Department of Immunology and RheumatologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Shin‐ya Nishihata
- Division of Advanced Preventive Medical Sciences, Department of Immunology and RheumatologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Kyoko Kitaoka
- Department of Otolaryngology—Head and Neck SurgeryNagasaki University HospitalNagasakiJapan
| | - Tatsufumi Nakamura
- Department of Social Work, Faculty of Human and Social StudiesNagasaki International UniversitySaseboJapan
| | - Atsushi Kawakami
- Division of Advanced Preventive Medical Sciences, Department of Immunology and RheumatologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| |
Collapse
|
86
|
Stohl HE, Yu N, Stohl W. First-trimester serum BAFF:sFlt-1 ratio as a candidate early biomarker of spontaneous abortion. Am J Reprod Immunol 2021; 86:e13428. [PMID: 33837577 DOI: 10.1111/aji.13428] [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: 11/23/2020] [Revised: 03/16/2021] [Accepted: 04/06/2021] [Indexed: 11/30/2022] Open
Abstract
PROBLEM Immunologic, angiogenic, and anti-angiogenic factors are associated with spontaneous abortion (SAB). B cell-activating factor (BAFF), a proliferation-inducing ligand (APRIL), placental growth factor (PlGF), and soluble fms-like tyrosine kinase-1 (sFlt-1) may play a role in SAB and may serve singly or in combination as an early biomarker of SAB. METHOD OF STUDY In this prospective observational study, serum sFlt-1, PIGF, BAFF, and APRIL levels were measured in the first trimester of pregnancy in a medically diverse group of women and in non-pregnant controls. Associations and discriminative values of first-trimester sFlt-1, PIGF, BAFF, and APRIL levels and the corresponding APRIL:BAFF, BAFF:sFlt-1, and sFlt-1:PlGF ratios with development of SAB were tested. RESULTS Median serum BAFF level was lower (p = .007) and median serum sFlt-1 level was higher (p < .001), in the first trimester of pregnancy than in non-pregnant controls. SAB developed in 27 of the pregnant women (11.3%), and first-trimester levels of BAFF (but not APRIL) and sFlt-1 (but not PIGF) were associated with SAB. Using optimal cutoffs determined through receiver operating characteristics curves, the best discriminator of SAB was the serum BAFF:sFlt-1 ratio, specifically among non-nulliparous women and women with prior SAB. CONCLUSION First-trimester serum BAFF:sFlt-1 ratio is a candidate indicator/predictor of SAB among non-nulliparous women and women with prior SAB. If validated through additional studies, then early identification of pregnant women at high risk for SAB through this simple blood test would assist in counseling and facilitate clinical trials of therapeutic interventions.
Collapse
Affiliation(s)
- Hindi E Stohl
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ning Yu
- Division of Rheumatology, Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - William Stohl
- Division of Rheumatology, Department of Medicine, Los Angeles County + University of Southern California Medical Center, Los Angeles, CA, USA.,Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
87
|
Zhang J, Li Y, Wu X, Zhong R, Wei J, Wang Z, Zhang X. Molecular structure, expression, and function analysis of BAFF gene in Chinese sucker, Myxocyprinus asiaticus. FISH PHYSIOLOGY AND BIOCHEMISTRY 2021; 47:225-238. [PMID: 33405065 DOI: 10.1007/s10695-020-00906-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 11/23/2020] [Indexed: 06/12/2023]
Abstract
B cell activating factor (BAFF), belonging to the tumor necrosis factor superfamily (TNFSF), is a critical cytokine for B cell survival and immunoglobulin secretion. Here, the BAFF gene of Chinese sucker (Myxocyprinus asiaticus) (MaBAFF) was cloned using RT-PCR and RACE (rapid amplification of cDNA end) techniques. The open reading frame (ORF) of MaBAFF encodes a 272-amino acid protein containing a transmembrane domain, a TNF family signature, and a putative furin protease cleavage site as seen in BAFFs from other species. Tissue expression profiles of MaBAFF determined by absolute and relative quantification of real-time quantitative PCR (qPCR) showed that MaBAFF is widely distributed in various tissues, with the highest expression in spleen. MaBAFF can be detected during fertilized egg period by RT-PCR. Upon induction by A. hydrophila, the expression of MaBAFF was up-regulated in spleen from 48 to 72 h, and the expression of BAFF and IgM all reached a peak at 48 h in head kidney. The soluble BAFF gene (MasBAFF) had been cloned into pET30a. SDS-PAGE and Western blotting analysis confirmed that the His-MasBAFF was efficiently expressed in Escherichia coli Rosset (DE3). CCK-8 assay indicated that the MasBAFF recombinant protein (200 ng/ml) could prolong the survival of peripheral blood leukocytes. Based on ELISA screening and Western blotting, monoclonal antibody 1-F2A3 against recombinant MasBAFF was selected and used for immunohistochemistry, which showed that BAFF-positive cells were detected in spleen and head kidney. Our results raise the possibility that MaBAFF may be useful to enhance immune efficacy in Chinese sucker disease defense.
Collapse
Affiliation(s)
- Jiaojiao Zhang
- Key laboratory of Eco-environment in the Three Gorges Reservoir Region of Ministry of Education, School of Life Sciences, Southwest University, No. 1 Tiansheng Road, Beibei District, Chongqing, 400715, China
| | - Yujin Li
- Key laboratory of Eco-environment in the Three Gorges Reservoir Region of Ministry of Education, School of Life Sciences, Southwest University, No. 1 Tiansheng Road, Beibei District, Chongqing, 400715, China
| | - Xia Wu
- Key laboratory of Eco-environment in the Three Gorges Reservoir Region of Ministry of Education, School of Life Sciences, Southwest University, No. 1 Tiansheng Road, Beibei District, Chongqing, 400715, China
| | - Ruonan Zhong
- Key laboratory of Eco-environment in the Three Gorges Reservoir Region of Ministry of Education, School of Life Sciences, Southwest University, No. 1 Tiansheng Road, Beibei District, Chongqing, 400715, China
| | - Jing Wei
- Key laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Zhijian Wang
- Key laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Xiaoping Zhang
- Key laboratory of Eco-environment in the Three Gorges Reservoir Region of Ministry of Education, School of Life Sciences, Southwest University, No. 1 Tiansheng Road, Beibei District, Chongqing, 400715, China.
| |
Collapse
|
88
|
Ding J, Cai Y, Deng Y, Jiang X, Gao M, Lin Y, Zhao N, Wang Z, Yu H, Lv W, Zhang Y, Hao Y, Guan Y. Telitacicept Following Plasma Exchange in the Treatment of Subjects With Recurrent NMOSD: Study Protocol for a Single-Center, Single-Arm, Open-Label Study. Front Neurol 2021; 12:596791. [PMID: 33868140 PMCID: PMC8044936 DOI: 10.3389/fneur.2021.596791] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/19/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune demyelinating disease that recurrently relapses and leads to severe disability. The available choices for disease prevention are few or intolerable. Previous studies suggested that telitacicept may provide a promising therapeutic strategy for autoimmune diseases involving B cells. Therefore, this study aims to assess the effectiveness and safety of telitacicept for recurrent NMOSD. Methods: We will perform a single-arm, single-center, open-label, specialist study with a total enrollment of eight participants. The treatment regimen includes plasma exchange three times and subcutaneous injection of telitacicept for 46 cycles, with a total period of 48 weeks. The primary endpoint is the time to first recurrence after enrollment. Secondary endpoints are Expanded Disability Status Scale (EDSS) score, Opticospinal Impairment Scale (OSIS) score, Hauser Ambulation Index, number of lesions on MRI, and changes in visual evoked potential (VEP), optical coherence tomography (OCT) and immunologic status. All adverse events after medication will be documented and investigated. Discussion: This study will explore the safety and effectiveness of telitacicept following plasma exchange regarding the time to recurrence in neuromyelitis optica spectrum disorder (NMOSD) for the first time. Clinical Trial Registration:Chictr.org.cn, identifier ChiCTR1800019427
Collapse
Affiliation(s)
- Jie Ding
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yu Cai
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ye Deng
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xianguo Jiang
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Meichun Gao
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yan Lin
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Nan Zhao
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ze Wang
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Haojun Yu
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wenwen Lv
- Clinical Research Center, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ying Zhang
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yong Hao
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yangtai Guan
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
89
|
Depascale R, Gatto M, Zen M, Saccon F, Larosa M, Zanatta E, Bindoli S, Doria A, Iaccarino L. Belimumab: a step forward in the treatment of systemic lupus erythematosus. Expert Opin Biol Ther 2021; 21:563-573. [PMID: 33630721 DOI: 10.1080/14712598.2021.1895744] [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: 01/10/2023]
Abstract
Introduction: Systemic Lupus Erythematosus (SLE) is a chronic B cell-mediated autoimmune disease which can potentially involve several organs and systems. The development of SLE is associated with a complexity of genetic, hormonal and environmental factors leading to immune deregulation and production of autoantibodies. Therefore, novel therapies have focused on B cells as key effectors of SLE pathogenesis. Belimumab is a fully humanized monoclonal antibody that antagonizes B-lymphocyte stimulator (BLyS); it is the first and the only biological drug approved for SLE in over 50 years.Areas covered: In this review we discuss the pharmacological properties of belimumab, new recommendations for its use in clinical practice and its evidence of efficacy and safety based on clinical trial and real-life data.Expert opinion: Efficacy and safety of belimumab in clinical practice have been well established. To date, it is known that early introduction of belimumab in SLE can maximize the efficacy of the drug. A number of questions are still open, such as the timing of belimumab discontinuation and its possible association with other biological drugs, which need to be assessed in future studies.
Collapse
Affiliation(s)
| | - Mariele Gatto
- Division of Rheumatology, University of Padova, Padova, Italy
| | - Margherita Zen
- Division of Rheumatology, University of Padova, Padova, Italy
| | | | | | | | - Sara Bindoli
- Division of Rheumatology, University of Padova, Padova, Italy
| | - Andrea Doria
- Division of Rheumatology, University of Padova, Padova, Italy
| | - Luca Iaccarino
- Division of Rheumatology, University of Padova, Padova, Italy
| |
Collapse
|
90
|
Liossis SN, Staveri C. What's New in the Treatment of Systemic Lupus Erythematosus. Front Med (Lausanne) 2021; 8:655100. [PMID: 33748165 PMCID: PMC7973110 DOI: 10.3389/fmed.2021.655100] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/12/2021] [Indexed: 01/12/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune multisystem disease with a variable presentation and manifestations ranging from mild to severe or even life-threatening. There is an ongoing and unmet need for novel, disease-specific, effective and safe treatment modalities. The aim of this review is to summarize data on SLE treatment that have emerged over the last 3 years. We will put emphasis on studies evaluating potential treatments on severe lupus manifestations such as lupus nephritis. Despite the existence of several therapeutic agents in SLE, the disease keeps causing significant morbidity. It is encouraging that a variety of therapeutic options are currently under investigation, although there are occasional trial failures.
Collapse
Affiliation(s)
- Stamatis Nick Liossis
- Division of Rheumatology, Department of Internal Medicine, Patras University Hospital, Patras, Greece.,Division of Rheumatology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
| | - Chrysanthi Staveri
- Division of Rheumatology, Department of Internal Medicine, Patras University Hospital, Patras, Greece
| |
Collapse
|
91
|
Kumar G, Maria Z, Kohli U, Agasing A, Quinn JL, Ko RM, Zamvil SS, Axtell RC. CNS Autoimmune Responses in BCMA-Deficient Mice Provide Insight for the Failure of Atacicept in MS. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/3/e973. [PMID: 33649164 PMCID: PMC7954465 DOI: 10.1212/nxi.0000000000000973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/21/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVE B cells have emerged as a therapeutic target for MS. Anti-CD20 antibodies, which deplete B cells, are effective therapies for MS. However, atacicept (TACI-Fc), which blocks BAFF and APRIL and reduces B cells, unexpectedly exacerbates MS. We tested the hypothesis that B cell maturation antigen (BCMA), a receptor for BAFF and APRIL, plays a role in the paradoxical effects of anti-CD20 antibody and TACI-Fc using experimental autoimmune encephalomyelitis (EAE). METHODS EAE was induced in wild-type (BCMA+/+) and BCMA-deficient (BCMA-/-) mice with an immunization of rodent myelin oligodendrocyte glycoprotein (MOG)35-55 peptide. Treatment with anti-CD20 antibody, TACI-Fc, and isotype controls was administered by intraperitoneal injections. CNS infiltration was evaluated by histology; immune cell phenotypes were evaluated by flow cytometry; MOG-specific antibodies were determined by ELISA. Mixed bone marrow chimeras and cell culture assays were used to identify the specific subsets of immune cells affected by BCMA deficiency. RESULTS First, we found that BCMA-/- mice had more severe EAE compared with BCMA+/+ mice and the increased disease was associated with elevated anti-MOG B-cell responses. Second, we found that anti-CD20 therapy attenuated EAE in BCMA-/- mice but not in BCMA+/+ mice. Third, TACI-Fc attenuated EAE in BCMA+/+ mice but not in BCMA-/- mice. Mixed bone marrow chimeric and cell culture experiments demonstrated that BCMA deficiency elevates inflammatory B-cell responses but inhibits inflammatory responses in macrophages. CONCLUSIONS BCMA has multifaceted roles during inflammation that affects therapeutic efficacies of anti-CD20 and TACI-Fc in EAE. Our results from BCMA-deficient mice provide insights into the failure of atacicept in MS.
Collapse
Affiliation(s)
- Gaurav Kumar
- From the ACI (G.K., Z.M., U.K., A.A., J.L.Q., R.M.K., R.C.A.), Oklahoma Medical Research Foundation; and Department of Neurology and Program in Immunology, (S.S.Z.), University of California San Francisco
| | - Zahra Maria
- From the ACI (G.K., Z.M., U.K., A.A., J.L.Q., R.M.K., R.C.A.), Oklahoma Medical Research Foundation; and Department of Neurology and Program in Immunology, (S.S.Z.), University of California San Francisco
| | - Uday Kohli
- From the ACI (G.K., Z.M., U.K., A.A., J.L.Q., R.M.K., R.C.A.), Oklahoma Medical Research Foundation; and Department of Neurology and Program in Immunology, (S.S.Z.), University of California San Francisco
| | - Agnieshka Agasing
- From the ACI (G.K., Z.M., U.K., A.A., J.L.Q., R.M.K., R.C.A.), Oklahoma Medical Research Foundation; and Department of Neurology and Program in Immunology, (S.S.Z.), University of California San Francisco
| | - James L Quinn
- From the ACI (G.K., Z.M., U.K., A.A., J.L.Q., R.M.K., R.C.A.), Oklahoma Medical Research Foundation; and Department of Neurology and Program in Immunology, (S.S.Z.), University of California San Francisco
| | - Rose M Ko
- From the ACI (G.K., Z.M., U.K., A.A., J.L.Q., R.M.K., R.C.A.), Oklahoma Medical Research Foundation; and Department of Neurology and Program in Immunology, (S.S.Z.), University of California San Francisco
| | - Scott S Zamvil
- From the ACI (G.K., Z.M., U.K., A.A., J.L.Q., R.M.K., R.C.A.), Oklahoma Medical Research Foundation; and Department of Neurology and Program in Immunology, (S.S.Z.), University of California San Francisco
| | - Robert C Axtell
- From the ACI (G.K., Z.M., U.K., A.A., J.L.Q., R.M.K., R.C.A.), Oklahoma Medical Research Foundation; and Department of Neurology and Program in Immunology, (S.S.Z.), University of California San Francisco.
| |
Collapse
|
92
|
B Cell Aberrance in Lupus: the Ringleader and the Solution. Clin Rev Allergy Immunol 2021; 62:301-323. [PMID: 33534064 DOI: 10.1007/s12016-020-08820-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2020] [Indexed: 12/18/2022]
Abstract
Systemic lupus erythematosus (SLE) is a prototypical autoimmune disease with high heterogeneity but the common characterization of numerous autoantibodies and systemic inflammation which lead to the damage of multiple organs. Aberrance of B cells plays a pivotal role in the immunopathogenesis of SLE via both antibody-dependent and antibody-independent manners. Escape of autoreactive B cells from the central and peripheral tolerance checkpoints, over-activation of B cells and their excessive cytokines release which drive T cells and dendritic cells stimulation, and dysregulated surface molecules, as well as intracellular signal pathways involved in B cell biology, are all contributing to B cell aberrance and participating in the pathogenesis of SLE. Based on that rationale, targeting aberrance of B cells and relevant molecules and pathways is expected to be a promising strategy for lupus control. Multiple approaches targeting B cells through different mechanisms have been attempted, including B-cell depletion via monoclonal antibodies against B-cell-specific molecules, blockade of B-cell survival and activation factors, suppressing T-B crosstalk by interrupting costimulatory molecules and inhibiting intracellular activation signaling cascade by targeting pathway molecules in B cells. Though most attempts ended in failure, the efficacy of B-cell targeting has been encouraged by the FDA approval of belimumab that blocks B cell-activating factor (BAFF) and the recommended use of anti-CD20 as a remedial therapy in refractory lupus. Still, quantities of clinical trials targeting B cells or relevant molecules are ongoing and some of them have displayed promising preliminary results. Additionally, advances in multi-omics studies help deepen our understandings of B cell biology in lupus and may promote the discovery of novel potential therapeutic targets. The combination of real-world data with basic research achievements may pave the road to conquering lupus.
Collapse
|
93
|
Felföldi B, Bódi I, Minkó K, Benyeda Z, Nagy N, Magyar A, Oláh I. Infection of bursal disease virus abrogates the extracellular glycoprotein in the follicular medulla. Poult Sci 2021; 100:101000. [PMID: 33690054 PMCID: PMC7938241 DOI: 10.1016/j.psj.2021.01.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/25/2020] [Accepted: 01/02/2021] [Indexed: 11/11/2022] Open
Abstract
In the medulla of bursal follicle, only the secretory dendritic cell (BSDC) is furnished with secretory machinery. The granular discharge of BSDC appears in membrane-bound and solubilized forms. Movat pentachrome staining proves that the solubilized form is a glycoprotein, which fills up the extracellular space of follicular medulla. The glycoprotein contributes to bursal microenvironment and may be attached to the surface of medullary lymphocytes. The secretory granules of BSDC may be fused, resulting in large, irregular dense bodies, which are the first sign of BSDC transformation to macrophage-like cells (Mal). To determine the effect of infectious bursal disease virus (IBDV) infection on the extracellular glycoprotein and BSDC, SPF chickens were experimentally infected with IBDV. On the surface of BSDC, the secretory substance is in high concentration, which may contribute to primary binding of IBDV to BSDC. The early distribution of IBDV infected cells is in consent with that BSDC. The IBDV infected BSDC rapidly transforms to Mal in which the glycoprotein staining appears. In the dense bodies, the packed virus particles inhibit the virus particles preventing the granular discharge, which may represent the first, early phase of virus replication cycle. The absence of extracellular glycoprotein results in alteration in the medullary microenvironment and subsequently B cell apoptosis. On the surface of medullary B cells, the solubilized secretory substance can be in much lower concentration, which results in secondary binding of IBDV to B cells. In secondary, late phase of virus replication cycle, the virus particles are not packed in electron dense substance which results in cytolytic lymphocytes and presence of virus in extracellular space. The Mal emigrates into the cortex, where induces inflammation, recruiting heterophil granulocyte and monocyte.
Collapse
Affiliation(s)
- Balázs Felföldi
- Scientific Support and Investigation Unit, Ceva-Phylaxia Co. Ltd., Ceva Animal Health, 1107 Budapest, Hungary
| | - Ildikó Bódi
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, 1094, Budapest, Hungary
| | - Krisztina Minkó
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, 1094, Budapest, Hungary
| | | | - Nándor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, 1094, Budapest, Hungary
| | - Attila Magyar
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, 1094, Budapest, Hungary
| | - Imre Oláh
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, 1094, Budapest, Hungary.
| |
Collapse
|
94
|
Varrica C, Dias HS, Reis C, Carvalheiro M, Simões S. Targeted delivery in scleroderma fibrosis. Autoimmun Rev 2020; 20:102730. [PMID: 33338593 DOI: 10.1016/j.autrev.2020.102730] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 10/02/2020] [Indexed: 12/29/2022]
Abstract
Systemic sclerosis (SSc) is considered one of the most challenging and difficult to treat among rheumatic disorders, due to its severity, multiorgan manifestation and different outcomes. It manifests fibrosis in different organs, mostly in skin and lungs. The skin fibrosis expression is considered the first sign of the disease and usually it is followed by internal organ fibrosis. An aberrant immune system activation seems to relate to the expression of the disease, but even environmental influences and dysregulation of many molecules signalling pathways are involved in the development of the disease. Current therapies are limited and characterized by multiple side effects: systemic route is the elective administration route, which decreases patient adherence to the therapy, as they are often already bothered by pain and disfigurement. Treatments available are organ-based, originally indicated for other conditions and there is no therapy available to reduce the fibroblast population size within existing fibrotic lesions. Disease-modifying therapies or immunomodulatory agents that are highly effective in other rheumatic diseases have shown disappointing results in SSc. There are thus no standardized and effective treatments for this disease, and there are even unanswered questions related to the insurgence of the pathology and all the mechanisms involved. An ideal approach could be considered "targeted therapy" that will be an increasingly attainable objective insofar as our understanding of the disease improves. The advantages in identifying the molecule and the signalling pathways involved in the pathology have helped to find some novel compounds for the therapy of scleroderma fibrosis or following innovative uses for already-approved drugs, corroborated by many clinical studies.
Collapse
Affiliation(s)
- Carla Varrica
- University of Pavia, Corso Strada Nuova, 65, 27100 Pavia, Italy
| | - Helena Sofia Dias
- Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Catarina Reis
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal; IBEB, Biophysics and Biomedical Engineering, Faculty of Sciences, Universidade de Lisboa, Campo Grande, 1649-016 Lisboa, Portugal
| | - Manuela Carvalheiro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Sandra Simões
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|
95
|
Möckel T, Basta F, Weinmann-Menke J, Schwarting A. B cell activating factor (BAFF): Structure, functions, autoimmunity and clinical implications in Systemic Lupus Erythematosus (SLE). Autoimmun Rev 2020; 20:102736. [PMID: 33333233 DOI: 10.1016/j.autrev.2020.102736] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 08/28/2020] [Indexed: 12/30/2022]
Abstract
The B cell activating factor (BAFF), or B lymphocyte stimulator (BLyS), is a B cell survival factor which supports autoreactive B cells and prevents their deletion. BAFF expression is closely linked with autoimmunity and is enhanced by genetic alterations and viral infections. Furthermore, BAFF seems to be involved in adipogenesis, atherosclerosis, neuro-inflammatory processes and ischemia reperfusion (I/R) injury. BAFF is commonly overexpressed in Systemic Lupus Erythematosus (SLE) and strongly involved in the pathogenesis of the disease. The relationship between BAFF levels, disease activity and damage accrual in SLE is controversial, but growing evidence is emerging on its role in renal involvement. Belimumab, a biologic BAFF inhibitor, has been the first biologic agent licensed for SLE therapy so far. As Rituximab (RTX) has been shown to increase BAFF levels following B cell depletion, the combination therapy of RTX plus belimumab (being evaluated in two RCT) seems to be a valuable option for several clinical scenarios. In this review we will highlight the growing body of evidence of immune and non-immune related BAFF expression in experimental and clinical settings.
Collapse
Affiliation(s)
- Tamara Möckel
- Department of Internal Medicine I, Division of Rheumatology and Clinical Immunology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | - Fabio Basta
- Acura Rheumatology Center Rhineland Palatinate, Bad Kreuznach, Germany
| | - Julia Weinmann-Menke
- Department of Internal Medicine I, Division of Nephrology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Andreas Schwarting
- Department of Internal Medicine I, Division of Rheumatology and Clinical Immunology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany; Acura Rheumatology Center Rhineland Palatinate, Bad Kreuznach, Germany
| |
Collapse
|
96
|
Demel I, Bago JR, Hajek R, Jelinek T. Focus on monoclonal antibodies targeting B-cell maturation antigen (BCMA) in multiple myeloma: update 2021. Br J Haematol 2020; 193:705-722. [PMID: 33216972 DOI: 10.1111/bjh.17235] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/01/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022]
Abstract
Remarkable advances have been achieved in the treatment of multiple myeloma (MM) in the last decade, which saw targeted immunotherapy, represented by anti-CD38 monoclonal antibodies, successfully incorporated across indications. However, myeloma is still considered curable in only a small subset of patients, and the majority of them eventually relapse. B-cell maturation antigen (BCMA) is expressed exclusively in mature B lymphocytes and plasma cells, and represents an ideal new target for immunotherapy, presented by bispecific antibody (bsAb) constructs, antibody-drug conjugates (ADCs) and chimeric antigen receptor T (CAR-T) cells. Each of them has proved its efficacy with the potential for deep and long-lasting responses as a single agent therapy in heavily pretreated patients. As a result, belantamab mafodotin was approved by the United States Food and Drug Administration for the treatment of relapsed/refractory MM, as the first anti-BCMA agent. In the present review, we focus on monoclonal antibodies targeting BCMA - bsAbs and ADCs. The data from preclinical studies as well as first-in-human clinical trials will be reviewed, together with the coverage of their constructs and mechanisms of action. The present results have laid the groundwork for the ongoing or upcoming clinical trials with combinatory regimens, which have always been a cornerstone in the treatment of MM.
Collapse
Affiliation(s)
- Ivo Demel
- Department of Haemato-oncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Julio Rodriguez Bago
- Department of Haemato-oncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Roman Hajek
- Department of Haemato-oncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Tomas Jelinek
- Department of Haemato-oncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| |
Collapse
|
97
|
Bajda S, Blazquez-Navarro A, Samans B, Wehler P, Kaliszczyk S, Amini L, Schmueck-Henneresse M, Witzke O, Dittmer U, Westhoff TH, Viebahn R, Reinke P, Thomusch O, Hugo C, Olek S, Roch T, Babel N. The role of soluble mediators in the clinical course of EBV infection and B cell homeostasis after kidney transplantation. Sci Rep 2020; 10:19594. [PMID: 33177622 PMCID: PMC7658229 DOI: 10.1038/s41598-020-76607-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 09/24/2020] [Indexed: 01/13/2023] Open
Abstract
Epstein-Barr virus (EBV) reactivation can lead to serious complications in kidney transplant patients, including post-transplant lymphoproliferative disorder (PTLD). Here, we have assessed the impact of EBV on B cell homeostasis at cellular and humoral level. In a multicenter study monitoring 540 kidney transplant patients during the first post-transplant year, EBV reactivation was detected in 109 patients. Thirteen soluble factors and B cell counts were analyzed in an EBV+ sub-cohort (N = 54) before, at peak and after EBV clearance and compared to a control group (N = 50). The B cell activating factor (BAFF) was significantly elevated among EBV+ patients. No additional soluble factors were associated with EBV. Importantly, in vitro experiments confirmed the proliferative effect of BAFF on EBV-infected B cells, simultaneously promoting EBV production. In contrast, elevated levels of BAFF in EBV+ patients did not lead to B cell expansion in vivo. Moreover, diminished positive inter-correlations of soluble factors and alterations of the bi-directional interplay between B cell and soluble factors were observed in EBV+ patients at peak and after clearance. Our data suggest that such alterations may counteract the proliferative effect of BAFF, preventing B cell expansion. The role of these alterations in lymphoma development should be analyzed in future studies.
Collapse
Affiliation(s)
- Sharon Bajda
- Berlin Institute of Health Center for Regenerative Therapies (BCRT): Berlin-Brandenburger Centrum für Regenerative Therapien, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Arturo Blazquez-Navarro
- Berlin Institute of Health Center for Regenerative Therapies (BCRT): Berlin-Brandenburger Centrum für Regenerative Therapien, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Systems Immunology Lab, Department of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- Medical Department I, Center for Translational Medicine, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Björn Samans
- Ivana Türbachova Laboratory for Epigenetics, Epiontis GmbH, Precision for Medicine Group, Berlin, Germany
| | - Patrizia Wehler
- Berlin Institute of Health Center for Regenerative Therapies (BCRT): Berlin-Brandenburger Centrum für Regenerative Therapien, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Medical Department I, Center for Translational Medicine, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Sviatlana Kaliszczyk
- Berlin Institute of Health Center for Regenerative Therapies (BCRT): Berlin-Brandenburger Centrum für Regenerative Therapien, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Medical Department I, Center for Translational Medicine, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Leila Amini
- Berlin Institute of Health Center for Regenerative Therapies (BCRT): Berlin-Brandenburger Centrum für Regenerative Therapien, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Schmueck-Henneresse
- Berlin Institute of Health Center for Regenerative Therapies (BCRT): Berlin-Brandenburger Centrum für Regenerative Therapien, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulf Dittmer
- Department of Infectious Diseases, Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Timm H Westhoff
- Medical Department I, Center for Translational Medicine, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Richard Viebahn
- Chirurgical University Hospital, University Hospital Knappschaftskrankenhaus Bochum, University Hospital of the Ruhr-University Bochum, Bochum, Germany
| | - Petra Reinke
- Berlin Institute of Health Center for Regenerative Therapies (BCRT): Berlin-Brandenburger Centrum für Regenerative Therapien, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Oliver Thomusch
- Department of General Surgery, University Hospital Freiburg, Freiburg, Germany
| | - Christian Hugo
- Medical Clinic 3 - Nephrology Unit, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Sven Olek
- Ivana Türbachova Laboratory for Epigenetics, Epiontis GmbH, Precision for Medicine Group, Berlin, Germany
| | - Toralf Roch
- Berlin Institute of Health Center for Regenerative Therapies (BCRT): Berlin-Brandenburger Centrum für Regenerative Therapien, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Medical Department I, Center for Translational Medicine, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Nina Babel
- Berlin Institute of Health Center for Regenerative Therapies (BCRT): Berlin-Brandenburger Centrum für Regenerative Therapien, Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Medical Department I, Center for Translational Medicine, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany.
| |
Collapse
|
98
|
Abdelhamid L, Cabana-Puig X, Mu Q, Moarefian M, Swartwout B, Eden K, Das P, Seguin RP, Xu L, Lowen S, Lavani M, Hrubec TC, Jones CN, Luo XM. Quaternary Ammonium Compound Disinfectants Reduce Lupus-Associated Splenomegaly by Targeting Neutrophil Migration and T-Cell Fate. Front Immunol 2020; 11:575179. [PMID: 33193366 PMCID: PMC7609861 DOI: 10.3389/fimmu.2020.575179] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/30/2020] [Indexed: 12/13/2022] Open
Abstract
Hypersensitivity reactions and immune dysregulation have been reported with the use of quaternary ammonium compound disinfectants (QACs). We hypothesized that QAC exposure would exacerbate autoimmunity associated with systemic lupus erythematosus (lupus). Surprisingly, however, we found that compared to QAC-free mice, ambient exposure of lupus-prone mice to QACs led to smaller spleens with no change in circulating autoantibodies or the severity of glomerulonephritis. This suggests that QACs may have immunosuppressive effects on lupus. Using a microfluidic device, we showed that ambient exposure to QACs reduced directional migration of bone marrow-derived neutrophils toward an inflammatory chemoattractant ex vivo. Consistent with this, we found decreased infiltration of neutrophils into the spleen. While bone marrow-derived neutrophils appeared to exhibit a pro-inflammatory profile, upregulated expression of PD-L1 was observed on neutrophils that infiltrated the spleen, which in turn interacted with PD-1 on T cells and modulated their fate. Specifically, QAC exposure hindered activation of splenic T cells and increased apoptosis of effector T-cell populations. Collectively, these results suggest that ambient QAC exposure decreases lupus-associated splenomegaly likely through neutrophil-mediated toning of T-cell activation and/or apoptosis. However, our findings also indicate that even ambient exposure could alter immune cell phenotypes, functions, and their fate. Further investigations on how QACs affect immunity under steady-state conditions are warranted.
Collapse
Affiliation(s)
- Leila Abdelhamid
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Xavier Cabana-Puig
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Qinghui Mu
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
- School of Medicine, Stanford University, Stanford, CA, United States
| | - Maryam Moarefian
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, United States
| | - Brianna Swartwout
- Translational Biology, Medicine and Health Graduate Program, Virginia Tech, Roanoke, VA, United States
| | - Kristin Eden
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Prerna Das
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Ryan P. Seguin
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - Libin Xu
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - Sarah Lowen
- Department of Anatomical Sciences, Edward Via College of Osteopathic Medicine-Virginia Campus, Blacksburg, VA, United States
| | - Mital Lavani
- Department of Anatomical Sciences, Edward Via College of Osteopathic Medicine-Virginia Campus, Blacksburg, VA, United States
| | - Terry C. Hrubec
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
- Department of Anatomical Sciences, Edward Via College of Osteopathic Medicine-Virginia Campus, Blacksburg, VA, United States
| | - Caroline N. Jones
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
- Department of Bioengineering, University of Texas, Dallas, TX, United States
| | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
99
|
Magliozzi R, Marastoni D, Calabrese M. The BAFF / APRIL system as therapeutic target in multiple sclerosis. Expert Opin Ther Targets 2020; 24:1135-1145. [PMID: 32900236 DOI: 10.1080/14728222.2020.1821647] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The complex system of BAFF (B-cell-activating factor of the TNF family) and APRIL (A proliferation-inducing ligand) has been studied in animal models of autoimmune diseases such as those resembling human systemic lupus erythematosus and Sjogren's syndrome and multiple sclerosis (MS). Accumulating evidence suggests that BAFF and APRIL have a physiological role in B cell immunity regulation, however inappropriate production of these factors may represent a key event which disrupts immune tolerance which is associated with systemic autoimmune diseases. AREAS COVERED We provide an update on the latest studies of the BAFF/APRIL system in multiple sclerosis, as well as on related clinical trials. EXPERT OPINION Experimental and clinical evidence suggests that increased BAFF levels may interfere directly and indirectly with B cell immunity; this can lead to breakdown of immune tolerance, the production of autoantibodies and continuous local intracerebral inflammation and brain tissue destruction. A more comprehensive understanding of the cell/molecular mechanism immune reactions specifically regulated by BAFF/APRIL in MS would better elucidate the specific cell phenotype targeted by actual anti-BAFF/APRIL therapies; this may enable the identification of either specific biomarkers of MS subgroups that would benefit of anti-BAFF/APRIL treatments or new targets of MS-specific anti-BAFF/APRIL therapies.
Collapse
Affiliation(s)
- Roberta Magliozzi
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona , Verona, Italy
| | - Damiano Marastoni
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona , Verona, Italy
| | - Massimiliano Calabrese
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona , Verona, Italy
| |
Collapse
|
100
|
Abstract
Therapeutic targeting of immune checkpoints has garnered significant attention in the area of cancer immunotherapy, in which efforts have focused in particular on cytotoxic T lymphocyte antigen 4 (CTLA4) and PD1, both of which are members of the CD28 family. In autoimmunity, these same pathways can be targeted to opposite effect: to curb the over-exuberant immune response. The CTLA4 checkpoint serves as an exemplar, whereby CTLA4 activity is blocked by antibodies in cancer immunotherapy and augmented by the provision of soluble CTLA4 in autoimmunity. Here, we review the targeting of co-stimulatory molecules in autoimmune diseases, focusing in particular on agents directed at members of the CD28 or tumour necrosis factor receptor families. We present the state of the art in co-stimulatory blockade approaches, including rational combinations of immune inhibitory agents, and discuss the future opportunities and challenges in this field.
Collapse
|