13
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Soluble CD40L is associated with increased oxidative burst and neutrophil extracellular trap release in Behçet’s disease

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          Studies have suggested that soluble factors in plasma from patients with active (aBD) and inactive (iBD) Behçet’s disease (BD) stimulate neutrophil function. Soluble CD40 ligand (sCD40L) is an important mediator of inflammation in BD. Its expression and effect on neutrophil oxidative burst and neutrophil extracellular trap (NET) release have not been characterized. In this study, we sought to investigate the role of plasma and the CD40L pathway on NET release and the oxidative burst profile in patients with aBD and iBD.

          Methods

          Neutrophils and peripheral blood mononuclear cells (PBMCs) were obtained from patients with aBD ( n = 30), patients with iBD ( n = 31), and healthy control subjects (HCs; n = 30). sCD40L plasma concentration was determined in individual samples. A pool of plasma for each group was created. In some experiments, plasma pools were treated with recombinant CD40 (rhCD40-muIg) for sCD40L blockade. NET release and H 2O 2/O 2 production were determined after stimulation with phorbol 12-myristate 13-acetate, sCD40L, or plasma pool. Flow cytometric analysis was performed to evaluate the expression of (1) CD40, Mac-1, and phosphorylated NF-κB p65 on neutrophils and monocytes and (2) CD40L on activated T cells and platelets. CD40L gene expression in PBMCs was determined by qRT-PCR.

          Results

          sCD40L plasma levels were significantly higher in patients with iBD (median 17,234, range 2346–19,279 pg/ml) and patients with aBD (median 18,289, range 413–19,883 pg/ml) than in HCs (median 47.5, range 33.7–26.7 pg/ml; p < 0.001). NET release was constitutively increased in BD compared with HC. NET release and H 2O 2/O 2 were higher after stimulation with sCD40L or BD plasma and decreased after sCD40L blockade. Mac-1 expression was constitutively increased in neutrophils of patients with aBD (88.7 ± 13.2% of cells) and patients with iBD (89.2 ± 20.1% of cells) compared with HC (27.1 ± 18.8% of cells; p < 0.01). CD40 expression on phagocytes and CD40L expression on platelets were similar in the three groups. PBMCs as well as nonactivated and activated CD4 + T cells from patients with BD showed higher CD40L expression.

          Conclusions

          Plasma from patients with aBD exerts a stimulus on NET release and oxidative burst, probably induced by sCD40L.

          Electronic supplementary material

          The online version of this article (doi:10.1186/s13075-017-1443-5) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references42

          • Record: found
          • Abstract: found
          • Article: not found

          NETosis: how vital is it?

          In this review, we examine the evidence that neutrophil extracellular traps (NETs) play a critical role in innate immunity. We summarize how NETs are formed in response to various stimuli and provide evidence that NETosis is not universally a cell death pathway. Here we describe at least 2 different mechanisms by which NETs are formed, including a suicide lytic NETosis and a live cell or vital NETosis. We also evaluate the evidence for NETs in catching and killing pathogens. Finally, we examine how infections are related to the development of autoimmune and vasculitic diseases through unintended but detrimental bystander damage resulting from NET release.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Enhanced formation and disordered regulation of NETs in myeloperoxidase-ANCA-associated microscopic polyangiitis.

            Microscopic polyangiitis (MPA) is an ANCA-associated vasculitis that affects small vessels, especially renal glomeruli. We recently demonstrated that the abnormal formation and impaired degradation of neutrophil extracellular traps (NETs) may be crucially involved in the generation of myeloperoxidase (MPO)-ANCA and subsequent development of MPA. This study assessed the formation and regulation of NETs in patients with MPO-ANCA-associated MPA. Peripheral blood samples were obtained from 38 patients with MPO-ANCA-associated MPA, 23 patients with systemic lupus erythematosus (SLE), and 8 healthy controls. IgG eluted from MPO-ANCA-associated MPA sera demonstrated the highest ability to induce NETs, and this ability correlated with disease activity and paralleled ANCA affinity for MPO. Moreover, addition of recombinant human MPO to these IgG samples reduced NET induction. Additionally, MPO-ANCA-associated MPA sera exhibited lower rates of NET degradation that recovered partially upon depletion of IgG. The activity of DNase I, an important regulator of NETs, was also lower in MPO-ANCA-associated MPA and SLE sera. IgG depletion from MPO-ANCA-associated MPA sera partially restored the rate of NET degradation, and addition of DNase I synergistically enhanced this restoration. Addition of anti-MPO antibodies did not inhibit DNase I activity, and some MPO-ANCA-associated MPA sera contained anti-NET antibodies at levels not correlated with MPO-ANCA titers, suggesting the involvement of unidentified autoantibodies as well. The collective evidence suggests a vicious cycle involving MPO-ANCA and the regulation of NETs could be critically involved in the pathogenesis of MPO-ANCA-associated MPA.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Increased levels of circulating microparticles in primary Sjögren's syndrome, systemic lupus erythematosus and rheumatoid arthritis and relation with disease activity

              Introduction Cell stimulation leads to the shedding of phosphatidylserine (PS)-rich microparticles (MPs). Because autoimmune diseases (AIDs) are characterized by cell activation, we investigated level of circulating MPs as a possible biomarker in primary Sjögren's syndrome (pSS), systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Methods We measured plasma levels of total, platelet and leukocyte MPs by prothrombinase capture assay and flow cytometry in 43 patients with pSS, 20 with SLE and 24 with RA and in 44 healthy controls (HCs). Secretory phospholipase A2 (sPLA2) activity was assessed by fluorometry. Soluble CD40 ligand (sCD40L) and soluble P-selectin (sCD62P), reflecting platelet activation, were measured by ELISA. Results Patients with pSS showed increased plasma level of total MPs (mean ± SEM 8.49 ± 1.14 nM PS equivalent (Eq), P < 0.0001), as did patients with RA (7.23 ± 1.05 n PS Eq, P = 0.004) and SLE (7.3 ± 1.25 nM PS Eq, P = 0.0004), as compared with HCs (4.13 ± 0.2 nM PS Eq). Patients with AIDs all showed increased level of platelet MPs (P < 0.0001), but only those with pSS showed increased level of leukocyte MPs (P < 0.0001). Results by capture assay and flow cytometry were correlated. In patients with high disease activity according to extra-glandular complications (pSS), DAS28 (RA) or SLEDAI (SLE) compared with low-activity patients, the MP level was only slightly increased in comparison with those having a low disease activity. Platelet MP level was inversely correlated with anti-DNA antibody level in SLE (r = -0.65; P = 0.003) and serum β2 microglobulin level in pSS (r = -0.37; P < 0.03). The levels of total and platelet MPs were inversely correlated with sPLA2 activity (r = -0.37, P = 0.0007; r = -0.36, P = 0.002, respectively). sCD40L and sCD62P concentrations were significantly higher in pSS than in HC (P ≤ 0.006). Conclusions Plasma MP level is elevated in pSS, as well as in SLE and RA, and could be used as a biomarker reflecting systemic cell activation. Level of leukocyte-derived MPs is increased in pSS only. The MP level is low in case of more severe AID, probably because of high secretory phospholipase A2 (sPLA2) activity, which leads to consumption of MPs. Increase of platelet-derived MPs, sCD40L and sCD62P, highlights platelet activation in pSS.
                Bookmark

                Author and article information

                Contributors
                +55 11 5576-4239 , sperazzio@yahoo.com.br
                pereirapvs@gmail.com
                vivianycs@yahoo.com.br
                marlonvilela@yahoo.com.br
                bruno_salu@hotmail.com
                olivamlv@gmail.com
                anne.stevens@seattlechildrens.org
                alexander_wagner@uol.com.br
                hans.ochs@seattlechildrens.org
                troy.torgerson@seattlechildrens.org
                condino@icb.usp.br
                luis.andrade@unifesp.br
                Journal
                Arthritis Res Ther
                Arthritis Res. Ther
                Arthritis Research & Therapy
                BioMed Central (London )
                1478-6354
                1478-6362
                19 October 2017
                19 October 2017
                2017
                : 19
                : 235
                Affiliations
                [1 ]ISNI 0000 0001 0514 7202, GRID grid.411249.b, Division of Rheumatology, Escola Paulista de Medicina, , Federal University of Sao Paulo, ; Rua Botucatu 740, 3° Andar, 04023-062 Sao Paulo, SP Brazil
                [2 ]Fleury Group – Research and Development, Avenida General Valdomiro de Lima, 508, 04344-070 Sao Paulo, SP Brazil
                [3 ]ISNI 0000000122986657, GRID grid.34477.33, Seattle Children’s Research Institute, , University of Washington and Center for Immunity and Immunotherapies, ; 1900 9th Avenue, JMB-7, Seattle, WA 98101 USA
                [4 ]ISNI 0000 0004 1937 0722, GRID grid.11899.38, Department of Immunology, Institute of Biomedical Sciences, , University of Sao Paulo, ; Avenida Professor Lineu Prestes, 2415, 03178-200 Sao Paulo, SP Brazil
                [5 ]ISNI 0000 0001 2165 7632, GRID grid.411204.2, Department of Pathology, , Federal University of Maranhao, ; Avenida dos Portugueses, 65065-545 Sao Luiz, MA Brazil
                [6 ]ISNI 0000 0001 0514 7202, GRID grid.411249.b, Department of Biochemistry and Molecular Biology, Escola Paulista de Medicina, , Federal University of Sao Paulo, ; Rua Três de Maio, 100, 5° Andar, 04044-020 Sao Paulo, SP Brazil
                Article
                1443
                10.1186/s13075-017-1443-5
                5649058
                29052524
                1cd8cecd-08ba-4c7b-b8f2-7c139cc00db7
                © The Author(s). 2017

                Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

                History
                : 8 May 2017
                : 25 September 2017
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/501100001807, Fundação de Amparo à Pesquisa do Estado de São Paulo;
                Award ID: 2011/50292-2
                Award Recipient :
                Funded by: Research and Development from Fleury
                Award ID: 12619
                Funded by: FundRef http://dx.doi.org/10.13039/501100003593, Conselho Nacional de Desenvolvimento Científico e Tecnológico;
                Award ID: 476356/2008-3
                Award ID: 306902/2013-3
                Award Recipient :
                Categories
                Research Article
                Custom metadata
                © The Author(s) 2017

                Orthopedics
                behçet’s disease,neutrophil activation,scd40l,cd40l pathway,oxidative burst,neutrophil extracellular traps

                Comments

                Comment on this article