Skip to main content
Top
Published in: BMC Immunology 1/2021

Open Access 01-12-2021 | Systemic Lupus Erythematosus | Research

The impact of neutrophil extracellular trap from patients with systemic lupus erythematosus on the viability, CD11b expression and oxidative burst of healthy neutrophils

Authors: Alimohammad Fatemi, Razieh Alipour, Hossein Khanahmad, Fereshteh Alsahebfosul, Alireza Andalib, Abbasali Pourazar

Published in: BMC Immunology | Issue 1/2021

Login to get access

Abstract

Background

NET (neutrophil extracellular trap) has been shown to directly influence inflammation; in SLE (systemic lupus erythematosus), it is reportedly a plausible cause for the broken self-tolerance that contributes to this pathology. Meanwhile, the role of NET is not easily explicable, and there is a serious discrepancy in the role of NET in SLE pathology and generally inflammation; in particular, the interactions of neutrophils with NET have been rarely inspected. This study evaluates the effect of NET on neutrophils in the context of SLE. The neutrophils were incubated by the collected NET (from SLE patients and healthy controls) and their expression of an activation marker, viability and oxidative burst ability were measured.

Results

The level of cell mortality, CD11b expression and the oxidative burst capacity were elevated in NET-treated neutrophils. Also, the elevation caused by the SLE NET was higher than that produced by the healthy NET.

Conclusion

The decreased neutrophil viability was not due to the increase in apoptosis; rather, it was because of the augmentation of other inflammatory cell-death modes. The upregulation of CD11b implies that NET causes neutrophils to more actively contribute to inflammation. The increased oxidative burst capacity of neutrophils can play a double role in inflammation. Overall, the effects induced by NET on neutrophils help prolong inflammation; accordingly, the NET collected from SLE patients is stronger than the NET from healthy individuals.
Appendix
Available only for authorised users
Literature
1.
go back to reference Moulton VR, Suarez-Fueyo A, Meidan E, Li H, Mizui M, Tsokos GC. Pathogenesis of human systemic lupus erythematosus: a cellular perspective. Trends Mol Med. 2017;23(7):615–35.PubMedPubMedCentralCrossRef Moulton VR, Suarez-Fueyo A, Meidan E, Li H, Mizui M, Tsokos GC. Pathogenesis of human systemic lupus erythematosus: a cellular perspective. Trends Mol Med. 2017;23(7):615–35.PubMedPubMedCentralCrossRef
2.
go back to reference Mistry P, Kaplan MJ. Cell death in the pathogenesis of systemic lupus erythematosus and lupus nephritis. Clin Immunol. 2017;185:59–73.PubMedCrossRef Mistry P, Kaplan MJ. Cell death in the pathogenesis of systemic lupus erythematosus and lupus nephritis. Clin Immunol. 2017;185:59–73.PubMedCrossRef
4.
go back to reference Land WG. The role of damage-associated molecular patterns in human diseases: part i-promoting inflammation and immunity. Sultan Qaboos Univ Med J. 2015;15(1):e9.PubMedPubMedCentral Land WG. The role of damage-associated molecular patterns in human diseases: part i-promoting inflammation and immunity. Sultan Qaboos Univ Med J. 2015;15(1):e9.PubMedPubMedCentral
5.
go back to reference Liew PX, Kubes P. The neutrophil’s role during health and disease. Physiol Rev. 2019;99(2):1223–48.PubMedCrossRef Liew PX, Kubes P. The neutrophil’s role during health and disease. Physiol Rev. 2019;99(2):1223–48.PubMedCrossRef
6.
go back to reference Castanheira FV, Kubes P. Neutrophils and NETs in modulating acute and chronic inflammation. Blood. 2019;133(20):2178–85.PubMedCrossRef Castanheira FV, Kubes P. Neutrophils and NETs in modulating acute and chronic inflammation. Blood. 2019;133(20):2178–85.PubMedCrossRef
7.
go back to reference Magna M, Pisetsky DS. The alarmin properties of DNA and DNA-associated nuclear proteins. Clin Ther. 2016;38(5):1029–41.PubMedCrossRef Magna M, Pisetsky DS. The alarmin properties of DNA and DNA-associated nuclear proteins. Clin Ther. 2016;38(5):1029–41.PubMedCrossRef
8.
go back to reference Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. 2013;13(3):159.CrossRefPubMed Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. 2013;13(3):159.CrossRefPubMed
9.
go back to reference Pieterse E, Hofstra J, Berden J, Herrmann M, Dieker J, Van Der Vlag J. Acetylated histones contribute to the immunostimulatory potential of neutrophil extracellular traps in systemic lupus erythematosus. Clin Exp Immunol. 2015;179(1):68–74.PubMedCrossRef Pieterse E, Hofstra J, Berden J, Herrmann M, Dieker J, Van Der Vlag J. Acetylated histones contribute to the immunostimulatory potential of neutrophil extracellular traps in systemic lupus erythematosus. Clin Exp Immunol. 2015;179(1):68–74.PubMedCrossRef
10.
go back to reference Lee KH, Kronbichler A, Park DD-Y, Park Y, Moon H, Kim H, et al. Neutrophil extracellular traps (NETs) in autoimmune diseases: a comprehensive review. Autoimmun Rev. 2017;16(11):1160–73.PubMedCrossRef Lee KH, Kronbichler A, Park DD-Y, Park Y, Moon H, Kim H, et al. Neutrophil extracellular traps (NETs) in autoimmune diseases: a comprehensive review. Autoimmun Rev. 2017;16(11):1160–73.PubMedCrossRef
11.
go back to reference Skopelja-Gardner S, Jones JD, Rigby WF. “NETtling” the host: breaking of tolerance in chronic inflammation and chronic infection. J Autoimmun. 2018;88:1–10.PubMedCrossRef Skopelja-Gardner S, Jones JD, Rigby WF. “NETtling” the host: breaking of tolerance in chronic inflammation and chronic infection. J Autoimmun. 2018;88:1–10.PubMedCrossRef
12.
go back to reference Nakazawa D, Shida H, Kusunoki Y, Miyoshi A, Nishio S, Tomaru U, et al. The responses of macrophages in interaction with neutrophils that undergo NETosis. J Autoimmun. 2016;67:19–28.PubMedCrossRef Nakazawa D, Shida H, Kusunoki Y, Miyoshi A, Nishio S, Tomaru U, et al. The responses of macrophages in interaction with neutrophils that undergo NETosis. J Autoimmun. 2016;67:19–28.PubMedCrossRef
13.
go back to reference Guimarães-Costa AB, Rochael NC, Oliveira F, Echevarria-Lima J, Saraiva EM. Neutrophil extracellular traps reprogram IL-4/GM-CSF-induced monocyte differentiation to anti-inflammatory macrophages. Front Immunol. 2017;8:523.PubMedPubMedCentralCrossRef Guimarães-Costa AB, Rochael NC, Oliveira F, Echevarria-Lima J, Saraiva EM. Neutrophil extracellular traps reprogram IL-4/GM-CSF-induced monocyte differentiation to anti-inflammatory macrophages. Front Immunol. 2017;8:523.PubMedPubMedCentralCrossRef
14.
go back to reference Tillack K, Breiden P, Martin R, Sospedra M. T lymphocyte priming by neutrophil extracellular traps links innate and adaptive immune responses. J Immunol. 2012;188(7):3150–9.PubMedCrossRef Tillack K, Breiden P, Martin R, Sospedra M. T lymphocyte priming by neutrophil extracellular traps links innate and adaptive immune responses. J Immunol. 2012;188(7):3150–9.PubMedCrossRef
15.
go back to reference Kahlenberg JM, Carmona-Rivera C, Smith CK, Kaplan MJ. Neutrophil extracellular trap–associated protein activation of the NLRP3 inflammasome is enhanced in lupus macrophages. J Immunol. 2013;190(3):1217–26.PubMedCrossRef Kahlenberg JM, Carmona-Rivera C, Smith CK, Kaplan MJ. Neutrophil extracellular trap–associated protein activation of the NLRP3 inflammasome is enhanced in lupus macrophages. J Immunol. 2013;190(3):1217–26.PubMedCrossRef
16.
go back to reference Hochberg MC. Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1997;40(9):1725.PubMedCrossRef Hochberg MC. Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1997;40(9):1725.PubMedCrossRef
17.
go back to reference Najmeh S, Cools-Lartigue J, Giannias B, Spicer J, Ferri LE. Simplified Human Neutrophil Extracellular Traps (NETs) Isolation and Handling. JoVE. 2015;(98):e52687. Najmeh S, Cools-Lartigue J, Giannias B, Spicer J, Ferri LE. Simplified Human Neutrophil Extracellular Traps (NETs) Isolation and Handling. JoVE. 2015;(98):e52687.
18.
go back to reference Saffarzadeh M, Juenemann C, Queisser MA, Lochnit G, Barreto G, Galuska SP, et al. Neutrophil extracellular traps directly induce epithelial and endothelial cell death: a predominant role of histones. PLoS One. 2012;7(2):e32366.PubMedPubMedCentralCrossRef Saffarzadeh M, Juenemann C, Queisser MA, Lochnit G, Barreto G, Galuska SP, et al. Neutrophil extracellular traps directly induce epithelial and endothelial cell death: a predominant role of histones. PLoS One. 2012;7(2):e32366.PubMedPubMedCentralCrossRef
19.
go back to reference Folco EJ, Mawson TL, Vromman A, Bernardes-Souza B, Franck G, Persson O, et al. Neutrophil extracellular traps induce endothelial cell activation and tissue factor production through interleukin-1α and cathepsin G. Arterioscler Thromb Vasc Biol. 2018;38(8):1901–12.PubMedPubMedCentralCrossRef Folco EJ, Mawson TL, Vromman A, Bernardes-Souza B, Franck G, Persson O, et al. Neutrophil extracellular traps induce endothelial cell activation and tissue factor production through interleukin-1α and cathepsin G. Arterioscler Thromb Vasc Biol. 2018;38(8):1901–12.PubMedPubMedCentralCrossRef
20.
go back to reference Schauer C, Janko C, Munoz LE, Zhao Y, Kienhöfer D, Frey B, et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat Med. 2014;20(5):511.PubMedCrossRef Schauer C, Janko C, Munoz LE, Zhao Y, Kienhöfer D, Frey B, et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat Med. 2014;20(5):511.PubMedCrossRef
21.
go back to reference Nakazawa D, Kumar SV, Marschner J, Desai J, Holderied A, Rath L, et al. Histones and neutrophil extracellular traps enhance tubular necrosis and remote organ injury in ischemic AKI. J Am Soc Nephrol. 2017;28(6):1753–68.PubMedPubMedCentralCrossRef Nakazawa D, Kumar SV, Marschner J, Desai J, Holderied A, Rath L, et al. Histones and neutrophil extracellular traps enhance tubular necrosis and remote organ injury in ischemic AKI. J Am Soc Nephrol. 2017;28(6):1753–68.PubMedPubMedCentralCrossRef
22.
go back to reference Ribon M, Seninet S, Mussard J, Sebbag M, Clavel C, Serre G, et al. Neutrophil extracellular traps exert both pro-and anti-inflammatory actions in rheumatoid arthritis that are modulated by C1q and LL-37. J Autoimmun. 2019;98:122–31.PubMedCrossRef Ribon M, Seninet S, Mussard J, Sebbag M, Clavel C, Serre G, et al. Neutrophil extracellular traps exert both pro-and anti-inflammatory actions in rheumatoid arthritis that are modulated by C1q and LL-37. J Autoimmun. 2019;98:122–31.PubMedCrossRef
23.
go back to reference Barrero CA, Perez-Leal O, Aksoy M, Moncada C, Ji R, Lopez Y, et al. Histone 3.3 participates in a self-sustaining cascade of apoptosis that contributes to the progression of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2013;188(6):673–83.PubMedPubMedCentralCrossRef Barrero CA, Perez-Leal O, Aksoy M, Moncada C, Ji R, Lopez Y, et al. Histone 3.3 participates in a self-sustaining cascade of apoptosis that contributes to the progression of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2013;188(6):673–83.PubMedPubMedCentralCrossRef
24.
go back to reference Liu L, Sun B. Neutrophil pyroptosis: new perspectives on sepsis. Cell Mol Life Sci. 2019;76(11):2031–42.PubMedCrossRef Liu L, Sun B. Neutrophil pyroptosis: new perspectives on sepsis. Cell Mol Life Sci. 2019;76(11):2031–42.PubMedCrossRef
25.
go back to reference Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018;25(3):486.PubMedPubMedCentralCrossRef Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018;25(3):486.PubMedPubMedCentralCrossRef
26.
go back to reference Urban CF, Ermert D, Schmid M, Abu-Abed U, Goosmann C, Nacken W, et al.Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans. PLoS Pathog. 2009;5(10):e1000639. Urban CF, Ermert D, Schmid M, Abu-Abed U, Goosmann C, Nacken W, et al.Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans. PLoS Pathog. 2009;5(10):e1000639.
27.
go back to reference Tadie J-M, Bae H-B, Jiang S, Park DW, Bell CP, Yang H, et al. HMGB1 promotes neutrophil extracellular trap formation through interactions with Toll-like receptor 4. Am J Phys Lung Cell Mol Phys. 2013;304(5):L342–L9. Tadie J-M, Bae H-B, Jiang S, Park DW, Bell CP, Yang H, et al. HMGB1 promotes neutrophil extracellular trap formation through interactions with Toll-like receptor 4. Am J Phys Lung Cell Mol Phys. 2013;304(5):L342–L9.
28.
go back to reference Xu J, Jiang Y, Wang J, Shi X, Liu Q, Liu Z, et al. Macrophage endocytosis of high-mobility group box 1 triggers pyroptosis. Cell Death Differ. 2014;21(8):1229.PubMedPubMedCentralCrossRef Xu J, Jiang Y, Wang J, Shi X, Liu Q, Liu Z, et al. Macrophage endocytosis of high-mobility group box 1 triggers pyroptosis. Cell Death Differ. 2014;21(8):1229.PubMedPubMedCentralCrossRef
29.
go back to reference Wei Z, Wang J, Wang Y, Wang C, Liu X, Han Z, et al. Effects of Neutrophil extracellular traps on bovine mammary epithelial cells in vitro. Front Immunol. 2019;10:1003. Wei Z, Wang J, Wang Y, Wang C, Liu X, Han Z, et al. Effects of Neutrophil extracellular traps on bovine mammary epithelial cells in vitro. Front Immunol. 2019;10:1003.
30.
go back to reference Yang J, Zhao Y, Zhang P, Li Y, Yang Y, Yang Y, et al. Hemorrhagic shock primes for lung vascular endothelial cell pyroptosis: role in pulmonary inflammation following LPS. Cell Death Dis. 2016;7(9):e2363.PubMedPubMedCentralCrossRef Yang J, Zhao Y, Zhang P, Li Y, Yang Y, Yang Y, et al. Hemorrhagic shock primes for lung vascular endothelial cell pyroptosis: role in pulmonary inflammation following LPS. Cell Death Dis. 2016;7(9):e2363.PubMedPubMedCentralCrossRef
31.
go back to reference Yang M, Yang X, Wang S, Xu L, Ke S, Ding X, et al. HMGB1-induced endothelial cell pyroptosis is involved in systemic inflammatory response syndrome following radiofrequency ablation of hepatic hemangiomas. Am J Transl Res. 2019;11(12):7555–67.PubMedPubMedCentral Yang M, Yang X, Wang S, Xu L, Ke S, Ding X, et al. HMGB1-induced endothelial cell pyroptosis is involved in systemic inflammatory response syndrome following radiofrequency ablation of hepatic hemangiomas. Am J Transl Res. 2019;11(12):7555–67.PubMedPubMedCentral
32.
go back to reference Chen L, Zhao Y, Lai D, Zhang P, Yang Y, Li Y, et al. Neutrophil extracellular traps promote macrophage pyroptosis in sepsis. Cell Death Dis. 2018;9(6):597.PubMedPubMedCentralCrossRef Chen L, Zhao Y, Lai D, Zhang P, Yang Y, Li Y, et al. Neutrophil extracellular traps promote macrophage pyroptosis in sepsis. Cell Death Dis. 2018;9(6):597.PubMedPubMedCentralCrossRef
33.
go back to reference Magna M, Pisetsky D. The role of cell death in the pathogenesis of SLE: is pyroptosis the missing link? Scand J Immunol. 2015;82(3):218–24.PubMedCrossRef Magna M, Pisetsky D. The role of cell death in the pathogenesis of SLE: is pyroptosis the missing link? Scand J Immunol. 2015;82(3):218–24.PubMedCrossRef
34.
go back to reference Decker P, Wolburg H, Rammensee HG. Nucleosomes induce lymphocyte necrosis. Eur J Immunol. 2003;33(7):1978–87.PubMedCrossRef Decker P, Wolburg H, Rammensee HG. Nucleosomes induce lymphocyte necrosis. Eur J Immunol. 2003;33(7):1978–87.PubMedCrossRef
35.
go back to reference Glennon-Alty L, Hackett AP, Chapman EA, Wright HL. Neutrophils and redox stress in the pathogenesis of autoimmune disease. Free Radic Biol Med. 2018;125:25–35.PubMedCrossRef Glennon-Alty L, Hackett AP, Chapman EA, Wright HL. Neutrophils and redox stress in the pathogenesis of autoimmune disease. Free Radic Biol Med. 2018;125:25–35.PubMedCrossRef
36.
go back to reference Lau D, Mollnau H, Eiserich JP, Freeman BA, Daiber A, Gehling UM, et al. Myeloperoxidase mediates neutrophil activation by association with CD11b/CD18 integrins. Proc Natl Acad Sci. 2005;102(2):431–6.PubMedCrossRef Lau D, Mollnau H, Eiserich JP, Freeman BA, Daiber A, Gehling UM, et al. Myeloperoxidase mediates neutrophil activation by association with CD11b/CD18 integrins. Proc Natl Acad Sci. 2005;102(2):431–6.PubMedCrossRef
37.
go back to reference Rönnefarth VM, Erbacher AI, Lamkemeyer T, Madlung J, Nordheim A, Rammensee H-G, et al. TLR2/TLR4-independent neutrophil activation and recruitment upon endocytosis of nucleosomes reveals a new pathway of innate immunity in systemic lupus erythematosus. J Immunol. 2006;177(11):7740–9.PubMedCrossRef Rönnefarth VM, Erbacher AI, Lamkemeyer T, Madlung J, Nordheim A, Rammensee H-G, et al. TLR2/TLR4-independent neutrophil activation and recruitment upon endocytosis of nucleosomes reveals a new pathway of innate immunity in systemic lupus erythematosus. J Immunol. 2006;177(11):7740–9.PubMedCrossRef
38.
go back to reference Newton RA, Hogg N. The human S100 protein MRP-14 is a novel activator of the β2 integrin Mac-1 on neutrophils. J Immunol. 1998;160(3):1427–35.PubMedCrossRef Newton RA, Hogg N. The human S100 protein MRP-14 is a novel activator of the β2 integrin Mac-1 on neutrophils. J Immunol. 1998;160(3):1427–35.PubMedCrossRef
39.
go back to reference Ribon M, Mussard J, Semerano L, Singer BB, Decker P. Extracellular Chromatin Triggers Release of Soluble CEACAM8 upon Activation of Neutrophils. Front Immunol. 2019;10:1346.PubMedPubMedCentralCrossRef Ribon M, Mussard J, Semerano L, Singer BB, Decker P. Extracellular Chromatin Triggers Release of Soluble CEACAM8 upon Activation of Neutrophils. Front Immunol. 2019;10:1346.PubMedPubMedCentralCrossRef
40.
go back to reference Lindau D. Autoimmune Inflammation in Systemic Lupus Erythematosus and Alzheimer's Disease; 2010. Lindau D. Autoimmune Inflammation in Systemic Lupus Erythematosus and Alzheimer's Disease; 2010.
41.
go back to reference Sroussi HY, Lu Y, Villines D, Sun Y. The down regulation of neutrophil oxidative metabolism by S100A8 and S100A9: implication of the protease-activated receptor-2. Mol Immunol. 2012;50(1–2):42–8.PubMedCrossRef Sroussi HY, Lu Y, Villines D, Sun Y. The down regulation of neutrophil oxidative metabolism by S100A8 and S100A9: implication of the protease-activated receptor-2. Mol Immunol. 2012;50(1–2):42–8.PubMedCrossRef
42.
go back to reference Tadié J-M, Bae H-B, Banerjee S, Zmijewski JW, Abraham E. Differential activation of RAGE by HMGB1 modulates neutrophil-associated NADPH oxidase activity and bacterial killing. Am J Phys Cell Phys. 2011;302(1):C249–C56. Tadié J-M, Bae H-B, Banerjee S, Zmijewski JW, Abraham E. Differential activation of RAGE by HMGB1 modulates neutrophil-associated NADPH oxidase activity and bacterial killing. Am J Phys Cell Phys. 2011;302(1):C249–C56.
43.
go back to reference Bruschi M, Petretto A, Santucci L, Vaglio A, Pratesi F, Migliorini P, et al. Neutrophil Extracellular Traps protein composition is specific for patients with Lupus nephritis and includes methyl-oxidized αenolase (methionine sulfoxide 93). Sci Rep. 2019;9(1):1–13.CrossRef Bruschi M, Petretto A, Santucci L, Vaglio A, Pratesi F, Migliorini P, et al. Neutrophil Extracellular Traps protein composition is specific for patients with Lupus nephritis and includes methyl-oxidized αenolase (methionine sulfoxide 93). Sci Rep. 2019;9(1):1–13.CrossRef
44.
go back to reference Berthelot F, Fattoum L, Casulli S, Gozlan J, Maréchal V, Elbim C. The effect of HMGB1, a damage-associated molecular pattern molecule, on polymorphonuclear neutrophil migration depends on its concentration. J Innate Immunity. 2012;4(1):41–58.CrossRef Berthelot F, Fattoum L, Casulli S, Gozlan J, Maréchal V, Elbim C. The effect of HMGB1, a damage-associated molecular pattern molecule, on polymorphonuclear neutrophil migration depends on its concentration. J Innate Immunity. 2012;4(1):41–58.CrossRef
45.
go back to reference Marsman G, Zeerleder S, Luken BM. Extracellular histones, cell-free DNA, or nucleosomes: differences in immunostimulation. Cell Death Dis. 2016;7(12):e2518-e.CrossRef Marsman G, Zeerleder S, Luken BM. Extracellular histones, cell-free DNA, or nucleosomes: differences in immunostimulation. Cell Death Dis. 2016;7(12):e2518-e.CrossRef
46.
go back to reference Khan SQ, Khan I, Gupta V. CD11b activity modulates pathogenesis of lupus nephritis. Front Med. 2018;5:52.CrossRef Khan SQ, Khan I, Gupta V. CD11b activity modulates pathogenesis of lupus nephritis. Front Med. 2018;5:52.CrossRef
47.
go back to reference Zeng MY, Miralda I, Armstrong CL, Uriarte SM, Bagaitkar J. The roles of NADPH oxidase in modulating neutrophil effector responses. Mol Oral Microbiol. 2019;34(2):27–38.PubMedPubMedCentralCrossRef Zeng MY, Miralda I, Armstrong CL, Uriarte SM, Bagaitkar J. The roles of NADPH oxidase in modulating neutrophil effector responses. Mol Oral Microbiol. 2019;34(2):27–38.PubMedPubMedCentralCrossRef
48.
go back to reference Alipour R, Fatemi A, Alsahebfosul F, Andalib A, Pourazar A. Autologous plasma versus fetal calf serum as a supplement for the culture of neutrophils. BMC Res Notes. 2020;13(1):1–7.CrossRef Alipour R, Fatemi A, Alsahebfosul F, Andalib A, Pourazar A. Autologous plasma versus fetal calf serum as a supplement for the culture of neutrophils. BMC Res Notes. 2020;13(1):1–7.CrossRef
Metadata
Title
The impact of neutrophil extracellular trap from patients with systemic lupus erythematosus on the viability, CD11b expression and oxidative burst of healthy neutrophils
Authors
Alimohammad Fatemi
Razieh Alipour
Hossein Khanahmad
Fereshteh Alsahebfosul
Alireza Andalib
Abbasali Pourazar
Publication date
01-12-2021
Publisher
BioMed Central
Published in
BMC Immunology / Issue 1/2021
Electronic ISSN: 1471-2172
DOI
https://doi.org/10.1186/s12865-021-00402-2

Other articles of this Issue 1/2021

BMC Immunology 1/2021 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine