Skip to main content
Top
Published in: Seminars in Immunopathology 4/2013

01-07-2013 | Review

Neutrophil extracellular chromatin traps connect innate immune response to autoimmunity

Authors: Marko Radic, Tony N. Marion

Published in: Seminars in Immunopathology | Issue 4/2013

Login to get access

Abstract

Autoantibodies to DNA and histones (chromatin) are the defining antigen specificity in systemic lupus erythematosus (SLE) and related musculoskeletal disorders but the mechanisms responsible for their induction remain mysterious. That situation rapidly changed once neutrophil extracellular chromatin traps (NETs) were discovered and observed to play a conserved role in innate immune responses to a broad variety of microbial pathogens. At the center of an infectious process, neutrophils exert various antimicrobial defenses, including the release of nuclear chromatin into the extracellular space. The externalized NETs, a complex meshwork of nuclear chromatin and antimicrobial proteins, serve to immobilize and degrade microbial pathogens. Here, we critically evaluate the evidence supporting NETs versus apoptotic bodies as a source for nuclear antigens in autoimmunity. We also discuss the possibility that NET chromatin forms an essential component of immune deposits in the pathogenesis of glomerulonephritis in SLE and other autoimmune immune complex diseases.
Literature
1.
go back to reference Park H, Bourla AB, Kastner DL, Colbert RA, Siegel RM (2012) Lighting the fires within: the cell biology of autoinflammatory diseases. Nat Rev Immunol 12(8):570–580. doi:10.1038/nri3261 PubMedCrossRef Park H, Bourla AB, Kastner DL, Colbert RA, Siegel RM (2012) Lighting the fires within: the cell biology of autoinflammatory diseases. Nat Rev Immunol 12(8):570–580. doi:10.​1038/​nri3261 PubMedCrossRef
6.
go back to reference Hoffman IE, Peene I, Meheus L, Huizinga TW, Cebecauer L, Isenberg D, De Bosschere K, Hulstaert F, Veys EM, De Keyser F (2004) Specific antinuclear antibodies are associated with clinical features in systemic lupus erythematosus. Ann Rheum Dis 63(9):1155–1158. doi:10.1136/ard.2003.013417 PubMedCrossRef Hoffman IE, Peene I, Meheus L, Huizinga TW, Cebecauer L, Isenberg D, De Bosschere K, Hulstaert F, Veys EM, De Keyser F (2004) Specific antinuclear antibodies are associated with clinical features in systemic lupus erythematosus. Ann Rheum Dis 63(9):1155–1158. doi:10.​1136/​ard.​2003.​013417 PubMedCrossRef
11.
go back to reference Gabriel SE (2001) The epidemiology of rheumatoid arthritis. Rheum Dis Clin North Am 27(2):269–281PubMedCrossRef Gabriel SE (2001) The epidemiology of rheumatoid arthritis. Rheum Dis Clin North Am 27(2):269–281PubMedCrossRef
13.
go back to reference Nandakumar KS, Holmdahl R (2006) Antibody-induced arthritis: disease mechanisms and genes involved at the effector phase of arthritis. Arthritis Res Ther 8(6):223. doi:10.1186/ar2089 PubMedCrossRef Nandakumar KS, Holmdahl R (2006) Antibody-induced arthritis: disease mechanisms and genes involved at the effector phase of arthritis. Arthritis Res Ther 8(6):223. doi:10.​1186/​ar2089 PubMedCrossRef
15.
go back to reference Shlomchik MJ, Marshak-Rothstein A, Wolfowicz CB, Rothstein TL, Weigert MG (1987) The role of clonal selection and somatic mutation in autoimmunity. Nature 328(6133):805–811. doi:10.1038/328805a0 PubMedCrossRef Shlomchik MJ, Marshak-Rothstein A, Wolfowicz CB, Rothstein TL, Weigert MG (1987) The role of clonal selection and somatic mutation in autoimmunity. Nature 328(6133):805–811. doi:10.​1038/​328805a0 PubMedCrossRef
16.
go back to reference Schellekens GA, de Jong BA, van den Hoogen FH, van de Putte LB, van Venrooij WJ (1998) Citrulline is an essential constituent of antigenic determinants recognized by rheumatoid arthritis-specific autoantibodies. J Clin Invest 101(1):273–281. doi:10.1172/JCI1316 PubMedCrossRef Schellekens GA, de Jong BA, van den Hoogen FH, van de Putte LB, van Venrooij WJ (1998) Citrulline is an essential constituent of antigenic determinants recognized by rheumatoid arthritis-specific autoantibodies. J Clin Invest 101(1):273–281. doi:10.​1172/​JCI1316 PubMedCrossRef
19.
go back to reference Hermansson M, Artemenko K, Ossipova E, Eriksson H, Lengqvist J, Makrygiannakis D, Catrina AI, Nicholas AP, Klareskog L, Savitski M, Zubarev RA, Jakobsson PJ (2010) MS analysis of rheumatoid arthritic synovial tissue identifies specific citrullination sites on fibrinogen. Proteomics Clin Appl 4(5):511–518. doi:10.1002/prca.200900088 PubMed Hermansson M, Artemenko K, Ossipova E, Eriksson H, Lengqvist J, Makrygiannakis D, Catrina AI, Nicholas AP, Klareskog L, Savitski M, Zubarev RA, Jakobsson PJ (2010) MS analysis of rheumatoid arthritic synovial tissue identifies specific citrullination sites on fibrinogen. Proteomics Clin Appl 4(5):511–518. doi:10.​1002/​prca.​200900088 PubMed
20.
go back to reference Suzuki A, Yamada R, Chang X, Tokuhiro S, Sawada T, Suzuki M, Nagasaki M, Nakayama-Hamada M, Kawaida R, Ono M, Ohtsuki M, Furukawa H, Yoshino S, Yukioka M, Tohma S, Matsubara T, Wakitani S, Teshima R, Nishioka Y, Sekine A, Iida A, Takahashi A, Tsunoda T, Nakamura Y, Yamamoto K (2003) Functional haplotypes of PADI4, encoding citrullinating enzyme peptidylarginine deiminase 4, are associated with rheumatoid arthritis. Nat Genet 34(4):395–402. doi:10.1038/ng1206 PubMedCrossRef Suzuki A, Yamada R, Chang X, Tokuhiro S, Sawada T, Suzuki M, Nagasaki M, Nakayama-Hamada M, Kawaida R, Ono M, Ohtsuki M, Furukawa H, Yoshino S, Yukioka M, Tohma S, Matsubara T, Wakitani S, Teshima R, Nishioka Y, Sekine A, Iida A, Takahashi A, Tsunoda T, Nakamura Y, Yamamoto K (2003) Functional haplotypes of PADI4, encoding citrullinating enzyme peptidylarginine deiminase 4, are associated with rheumatoid arthritis. Nat Genet 34(4):395–402. doi:10.​1038/​ng1206 PubMedCrossRef
21.
go back to reference Nakashima K, Hagiwara T, Ishigami A, Nagata S, Asaga H, Kuramoto M, Senshu T, Yamada M (1999) Molecular characterization of peptidylarginine deiminase in HL-60 cells induced by retinoic acid and 1alpha,25-dihydroxyvitamin D(3). J Biol Chem 274(39):27786–27792PubMedCrossRef Nakashima K, Hagiwara T, Ishigami A, Nagata S, Asaga H, Kuramoto M, Senshu T, Yamada M (1999) Molecular characterization of peptidylarginine deiminase in HL-60 cells induced by retinoic acid and 1alpha,25-dihydroxyvitamin D(3). J Biol Chem 274(39):27786–27792PubMedCrossRef
22.
go back to reference Dwivedi N, Upadhyay J, Neeli I, Khan S, Pattanaik D, Myers L, Kirou KA, Hellmich B, Knuckley B, Thompson PR, Crow MK, Mikuls TR, Csernok E, Radic M (2012) Felty’s syndrome autoantibodies bind to deiminated histones and neutrophil extracellular chromatin traps. Arthritis Rheum 64(4):982–992. doi:10.1002/art.33432 PubMedCrossRef Dwivedi N, Upadhyay J, Neeli I, Khan S, Pattanaik D, Myers L, Kirou KA, Hellmich B, Knuckley B, Thompson PR, Crow MK, Mikuls TR, Csernok E, Radic M (2012) Felty’s syndrome autoantibodies bind to deiminated histones and neutrophil extracellular chromatin traps. Arthritis Rheum 64(4):982–992. doi:10.​1002/​art.​33432 PubMedCrossRef
23.
go back to reference Holman HR, Kunkel HG (1957) Affinity between the lupus erythematosus serum factor and cell nuclei and nucleoprotein. Science 126(3265):162–163PubMedCrossRef Holman HR, Kunkel HG (1957) Affinity between the lupus erythematosus serum factor and cell nuclei and nucleoprotein. Science 126(3265):162–163PubMedCrossRef
24.
go back to reference Ceppellini R, Polli E, Celada F (1957) A DNA-reacting factor in serum of a patient with lupus erythematosus diffusus. Proc Soc Exp Biol Med 96(3):572–574PubMed Ceppellini R, Polli E, Celada F (1957) A DNA-reacting factor in serum of a patient with lupus erythematosus diffusus. Proc Soc Exp Biol Med 96(3):572–574PubMed
25.
go back to reference Miescher P (1957) The antigenic constituents of the neutrophilic leukocyte with special reference to the L. E. phenomenon. Vox Sang 2(3):145–158PubMedCrossRef Miescher P (1957) The antigenic constituents of the neutrophilic leukocyte with special reference to the L. E. phenomenon. Vox Sang 2(3):145–158PubMedCrossRef
26.
go back to reference Koffler D, Schur PH, Kunkel HG (1967) Immunological studies concerning the nephritis of systemic lupus erythematosus. J Exp Med 126(4):607–624PubMedCrossRef Koffler D, Schur PH, Kunkel HG (1967) Immunological studies concerning the nephritis of systemic lupus erythematosus. J Exp Med 126(4):607–624PubMedCrossRef
27.
go back to reference Leadbetter EA, Rifkin IR, Hohlbaum AM, Beaudette BC, Shlomchik MJ, Marshak-Rothstein A (2002) Chromatin-IgG complexes activate B cells by dual engagement of IgM and toll-like receptors. Nature 416(6881):603–607. doi:10.1038/416603a PubMedCrossRef Leadbetter EA, Rifkin IR, Hohlbaum AM, Beaudette BC, Shlomchik MJ, Marshak-Rothstein A (2002) Chromatin-IgG complexes activate B cells by dual engagement of IgM and toll-like receptors. Nature 416(6881):603–607. doi:10.​1038/​416603a PubMedCrossRef
28.
29.
go back to reference Pisitkun P, Deane JA, Difilippantonio MJ, Tarasenko T, Satterthwaite AB, Bolland S (2006) Autoreactive B cell responses to RNA-related antigens due to TLR7 gene duplication. Science 312(5780):1669–1672. doi:10.1126/science.1124978 PubMedCrossRef Pisitkun P, Deane JA, Difilippantonio MJ, Tarasenko T, Satterthwaite AB, Bolland S (2006) Autoreactive B cell responses to RNA-related antigens due to TLR7 gene duplication. Science 312(5780):1669–1672. doi:10.​1126/​science.​1124978 PubMedCrossRef
30.
go back to reference Subramanian S, Tus K, Li QZ, Wang A, Tian XH, Zhou J, Liang C, Bartov G, McDaniel LD, Zhou XJ, Schultz RA, Wakeland EK (2006) A Tlr7 translocation accelerates systemic autoimmunity in murine lupus. Proc Natl Acad Sci U S A 103(26):9970–9975. doi:10.1073/pnas.0603912103 PubMedCrossRef Subramanian S, Tus K, Li QZ, Wang A, Tian XH, Zhou J, Liang C, Bartov G, McDaniel LD, Zhou XJ, Schultz RA, Wakeland EK (2006) A Tlr7 translocation accelerates systemic autoimmunity in murine lupus. Proc Natl Acad Sci U S A 103(26):9970–9975. doi:10.​1073/​pnas.​0603912103 PubMedCrossRef
31.
go back to reference Santiago-Raber ML, Kikuchi S, Borel P, Uematsu S, Akira S, Kotzin BL, Izui S (2008) Evidence for genes in addition to Tlr7 in the Yaa translocation linked with acceleration of systemic lupus erythematosus. J Immunol 181(2):1556–1562PubMed Santiago-Raber ML, Kikuchi S, Borel P, Uematsu S, Akira S, Kotzin BL, Izui S (2008) Evidence for genes in addition to Tlr7 in the Yaa translocation linked with acceleration of systemic lupus erythematosus. J Immunol 181(2):1556–1562PubMed
32.
go back to reference Nickerson KM, Christensen SR, Shupe J, Kashgarian M, Kim D, Elkon K, Shlomchik MJ (2010) TLR9 regulates TLR7- and MyD88-dependent autoantibody production and disease in a murine model of lupus. J Immunol 184(4):1840–1848. doi:10.4049/jimmunol.0902592 PubMedCrossRef Nickerson KM, Christensen SR, Shupe J, Kashgarian M, Kim D, Elkon K, Shlomchik MJ (2010) TLR9 regulates TLR7- and MyD88-dependent autoantibody production and disease in a murine model of lupus. J Immunol 184(4):1840–1848. doi:10.​4049/​jimmunol.​0902592 PubMedCrossRef
34.
go back to reference Murakami A, Takahashi Y, Nishimura M, Shimizu T, Azuma T (2010) The amino acid residue at position 95 and the third CDR region in the H chain determine the ceiling affinity and the maturation pathway of an anti-(4-hydroxy-3-nitrophenyl)acetyl antibody. Mol Immunol 48(1–3):48–58. doi:10.1016/j.molimm.2010.09.013 PubMedCrossRef Murakami A, Takahashi Y, Nishimura M, Shimizu T, Azuma T (2010) The amino acid residue at position 95 and the third CDR region in the H chain determine the ceiling affinity and the maturation pathway of an anti-(4-hydroxy-3-nitrophenyl)acetyl antibody. Mol Immunol 48(1–3):48–58. doi:10.​1016/​j.​molimm.​2010.​09.​013 PubMedCrossRef
35.
go back to reference Shlomchik MJ, Aucoin AH, Pisetsky DS, Weigert MG (1987) Structure and function of anti-DNA autoantibodies derived from a single autoimmune mouse. Proc Natl Acad Sci U S A 84(24):9150–9154PubMedCrossRef Shlomchik MJ, Aucoin AH, Pisetsky DS, Weigert MG (1987) Structure and function of anti-DNA autoantibodies derived from a single autoimmune mouse. Proc Natl Acad Sci U S A 84(24):9150–9154PubMedCrossRef
36.
go back to reference Marion TN, Bothwell AL, Briles DE, Janeway CA Jr (1989) IgG anti-DNA autoantibodies within an individual autoimmune mouse are the products of clonal selection. J Immunol 142(12):4269–4274PubMed Marion TN, Bothwell AL, Briles DE, Janeway CA Jr (1989) IgG anti-DNA autoantibodies within an individual autoimmune mouse are the products of clonal selection. J Immunol 142(12):4269–4274PubMed
38.
go back to reference Marion TN, Tillman DM, Jou NT, Hill RJ (1992) Selection of immunoglobulin variable regions in autoimmunity to DNA. Immunol Rev 128:123–149PubMedCrossRef Marion TN, Tillman DM, Jou NT, Hill RJ (1992) Selection of immunoglobulin variable regions in autoimmunity to DNA. Immunol Rev 128:123–149PubMedCrossRef
39.
go back to reference Tillman DM, Jou NT, Hill RJ, Marion TN (1992) Both IgM and IgG anti-DNA antibodies are the products of clonally selective B cell stimulation in (NZB × NZW)F1 mice. J Exp Med 176(3):761–779PubMedCrossRef Tillman DM, Jou NT, Hill RJ, Marion TN (1992) Both IgM and IgG anti-DNA antibodies are the products of clonally selective B cell stimulation in (NZB × NZW)F1 mice. J Exp Med 176(3):761–779PubMedCrossRef
40.
go back to reference Radic MZ, Mackle J, Erikson J, Mol C, Anderson WF, Weigert M (1993) Residues that mediate DNA binding of autoimmune antibodies. J Immunol 150(11):4966–4977PubMed Radic MZ, Mackle J, Erikson J, Mol C, Anderson WF, Weigert M (1993) Residues that mediate DNA binding of autoimmune antibodies. J Immunol 150(11):4966–4977PubMed
42.
go back to reference Madaio MP, Hodder S, Schwartz RS, Stollar BD (1984) Responsiveness of autoimmune and normal mice to nucleic acid antigens. J Immunol 132(2):872–876PubMed Madaio MP, Hodder S, Schwartz RS, Stollar BD (1984) Responsiveness of autoimmune and normal mice to nucleic acid antigens. J Immunol 132(2):872–876PubMed
43.
go back to reference Desai DD, Marion TN (2000) Induction of anti-DNA antibody with DNA-peptide complexes. Int Immunol 12(11):1569–1578PubMedCrossRef Desai DD, Marion TN (2000) Induction of anti-DNA antibody with DNA-peptide complexes. Int Immunol 12(11):1569–1578PubMedCrossRef
44.
46.
go back to reference Radic M, Marion T, Monestier M (2004) Nucleosomes are exposed at the cell surface in apoptosis. J Immunol 172(11):6692–6700PubMed Radic M, Marion T, Monestier M (2004) Nucleosomes are exposed at the cell surface in apoptosis. J Immunol 172(11):6692–6700PubMed
47.
go back to reference Casciola-Rosen LA, Anhalt G, Rosen A (1994) Autoantigens targeted in systemic lupus erythematosus are clustered in two populations of surface structures on apoptotic keratinocytes. J Exp Med 179(4):1317–1330PubMedCrossRef Casciola-Rosen LA, Anhalt G, Rosen A (1994) Autoantigens targeted in systemic lupus erythematosus are clustered in two populations of surface structures on apoptotic keratinocytes. J Exp Med 179(4):1317–1330PubMedCrossRef
49.
go back to reference Radic MZ, Shah K, Zhang W, Lu Q, Lemke G, Hilliard GM (2006) Heterogeneous nuclear ribonucleoprotein P2 is an autoantibody target in mice deficient for Mer, Axl, and Tyro3 receptor tyrosine kinases. J Immunol 176(1):68–74PubMed Radic MZ, Shah K, Zhang W, Lu Q, Lemke G, Hilliard GM (2006) Heterogeneous nuclear ribonucleoprotein P2 is an autoantibody target in mice deficient for Mer, Axl, and Tyro3 receptor tyrosine kinases. J Immunol 176(1):68–74PubMed
50.
go back to reference Kerr JF, Wyllie AH, Currie AR (1972) Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 26(4):239–257PubMedCrossRef Kerr JF, Wyllie AH, Currie AR (1972) Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 26(4):239–257PubMedCrossRef
51.
go back to reference Fadok VA, Bratton DL, Guthrie L, Henson PM (2001) Differential effects of apoptotic versus lysed cells on macrophage production of cytokines: role of proteases. J Immunol 166(11):6847–6854PubMed Fadok VA, Bratton DL, Guthrie L, Henson PM (2001) Differential effects of apoptotic versus lysed cells on macrophage production of cytokines: role of proteases. J Immunol 166(11):6847–6854PubMed
52.
go back to reference Xu H, Li H, Suri-Payer E, Hardy RR, Weigert M (1998) Regulation of anti-DNA B cells in recombination-activating gene-deficient mice. J Exp Med 188(7):1247–1254PubMedCrossRef Xu H, Li H, Suri-Payer E, Hardy RR, Weigert M (1998) Regulation of anti-DNA B cells in recombination-activating gene-deficient mice. J Exp Med 188(7):1247–1254PubMedCrossRef
53.
go back to reference Kawane K, Fukuyama H, Yoshida H, Nagase H, Ohsawa Y, Uchiyama Y, Okada K, Iida T, Nagata S (2003) Impaired thymic development in mouse embryos deficient in apoptotic DNA degradation. Nat Immunol 4(2):138–144. doi:10.1038/ni881 PubMedCrossRef Kawane K, Fukuyama H, Yoshida H, Nagase H, Ohsawa Y, Uchiyama Y, Okada K, Iida T, Nagata S (2003) Impaired thymic development in mouse embryos deficient in apoptotic DNA degradation. Nat Immunol 4(2):138–144. doi:10.​1038/​ni881 PubMedCrossRef
54.
go back to reference Mevorach D, Zhou JL, Song X, Elkon KB (1998) Systemic exposure to irradiated apoptotic cells induces autoantibody production. J Exp Med 188(2):387–392PubMedCrossRef Mevorach D, Zhou JL, Song X, Elkon KB (1998) Systemic exposure to irradiated apoptotic cells induces autoantibody production. J Exp Med 188(2):387–392PubMedCrossRef
58.
go back to reference Wakeland EK, Liu K, Graham RR, Behrens TW (2001) Delineating the genetic basis of systemic lupus erythematosus. Immunity 15(3):397–408PubMedCrossRef Wakeland EK, Liu K, Graham RR, Behrens TW (2001) Delineating the genetic basis of systemic lupus erythematosus. Immunity 15(3):397–408PubMedCrossRef
60.
go back to reference Harper L, Ren Y, Savill J, Adu D, Savage CO (2000) Antineutrophil cytoplasmic antibodies induce reactive oxygen-dependent dysregulation of primed neutrophil apoptosis and clearance by macrophages. Am J Pathol 157(1):211–220. doi:10.1016/S0002-9440(10)64532-4 PubMedCrossRef Harper L, Ren Y, Savill J, Adu D, Savage CO (2000) Antineutrophil cytoplasmic antibodies induce reactive oxygen-dependent dysregulation of primed neutrophil apoptosis and clearance by macrophages. Am J Pathol 157(1):211–220. doi:10.​1016/​S0002-9440(10)64532-4 PubMedCrossRef
63.
go back to reference Kim HS, Yoon H, Minn I, Park CB, Lee WT, Zasloff M, Kim SC (2000) Pepsin-mediated processing of the cytoplasmic histone H2A to strong antimicrobial peptide buforin I. J Immunol 165(6):3268–3274PubMed Kim HS, Yoon H, Minn I, Park CB, Lee WT, Zasloff M, Kim SC (2000) Pepsin-mediated processing of the cytoplasmic histone H2A to strong antimicrobial peptide buforin I. J Immunol 165(6):3268–3274PubMed
66.
go back to reference Garcia-Romo GS, Caielli S, Vega B, Connolly J, Allantaz F, Xu Z, Punaro M, Baisch J, Guiducci C, Coffman RL, Barrat FJ, Banchereau J, Pascual V (2011) Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci Transl Med 3(73):73ra20. doi:10.1126/scitranslmed.3001201 PubMedCrossRef Garcia-Romo GS, Caielli S, Vega B, Connolly J, Allantaz F, Xu Z, Punaro M, Baisch J, Guiducci C, Coffman RL, Barrat FJ, Banchereau J, Pascual V (2011) Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci Transl Med 3(73):73ra20. doi:10.​1126/​scitranslmed.​3001201 PubMedCrossRef
67.
go back to reference Hakkim A, Fuchs TA, Martinez NE, Hess S, Prinz H, Zychlinsky A, Waldmann H (2011) Activation of the Raf-MEK-ERK pathway is required for neutrophil extracellular trap formation. Nat Chem Biol 7(2):75–77. doi:10.1038/nchembio.496 PubMedCrossRef Hakkim A, Fuchs TA, Martinez NE, Hess S, Prinz H, Zychlinsky A, Waldmann H (2011) Activation of the Raf-MEK-ERK pathway is required for neutrophil extracellular trap formation. Nat Chem Biol 7(2):75–77. doi:10.​1038/​nchembio.​496 PubMedCrossRef
69.
go back to reference Fuchs TA, Abed U, Goosmann C, Hurwitz R, Schulze I, Wahn V, Weinrauch Y, Brinkmann V, Zychlinsky A (2007) Novel cell death program leads to neutrophil extracellular traps. J Cell Biol 176(2):231–241. doi:10.1083/jcb.200606027 PubMedCrossRef Fuchs TA, Abed U, Goosmann C, Hurwitz R, Schulze I, Wahn V, Weinrauch Y, Brinkmann V, Zychlinsky A (2007) Novel cell death program leads to neutrophil extracellular traps. J Cell Biol 176(2):231–241. doi:10.​1083/​jcb.​200606027 PubMedCrossRef
73.
go back to reference Metzler KD, Fuchs TA, Nauseef WM, Reumaux D, Roesler J, Schulze I, Wahn V, Papayannopoulos V, Zychlinsky A (2011) Myeloperoxidase is required for neutrophil extracellular trap formation: implications for innate immunity. Blood 117(3):953–959. doi:10.1182/blood-2010-06-290171 PubMedCrossRef Metzler KD, Fuchs TA, Nauseef WM, Reumaux D, Roesler J, Schulze I, Wahn V, Papayannopoulos V, Zychlinsky A (2011) Myeloperoxidase is required for neutrophil extracellular trap formation: implications for innate immunity. Blood 117(3):953–959. doi:10.​1182/​blood-2010-06-290171 PubMedCrossRef
74.
76.
go back to reference Neeli I, Khan SN, Radic M (2008) Histone deimination as a response to inflammatory stimuli in neutrophils. J Immunol 180(3):1895–1902PubMed Neeli I, Khan SN, Radic M (2008) Histone deimination as a response to inflammatory stimuli in neutrophils. J Immunol 180(3):1895–1902PubMed
79.
go back to reference McKee AS, Munks MW, MacLeod MK, Fleenor CJ, Van Rooijen N, Kappler JW, Marrack P (2009) Alum induces innate immune responses through macrophage and mast cell sensors, but these sensors are not required for alum to act as an adjuvant for specific immunity. J Immunol 183(7):4403–4414. doi:10.4049/jimmunol.0900164 PubMedCrossRef McKee AS, Munks MW, MacLeod MK, Fleenor CJ, Van Rooijen N, Kappler JW, Marrack P (2009) Alum induces innate immune responses through macrophage and mast cell sensors, but these sensors are not required for alum to act as an adjuvant for specific immunity. J Immunol 183(7):4403–4414. doi:10.​4049/​jimmunol.​0900164 PubMedCrossRef
80.
81.
go back to reference Leshner M, Wang S, Lewis C, Zheng H, Chen XA, Santy L, Wang Y (2012) PAD4 Mediated histone hypercitrullination induces heterochromatin decondensation and chromatin unfolding to form neutrophil extracellular trap-like structures. Front Immunol 3:307. doi:10.3389/fimmu.2012.00307 PubMedCrossRef Leshner M, Wang S, Lewis C, Zheng H, Chen XA, Santy L, Wang Y (2012) PAD4 Mediated histone hypercitrullination induces heterochromatin decondensation and chromatin unfolding to form neutrophil extracellular trap-like structures. Front Immunol 3:307. doi:10.​3389/​fimmu.​2012.​00307 PubMedCrossRef
82.
go back to reference Kessenbrock K, Krumbholz M, Schonermarck U, Back W, Gross WL, Werb Z, Grone HJ, Brinkmann V, Jenne DE (2009) Netting neutrophils in autoimmune small-vessel vasculitis. Nat Med 15(6):623–625. doi:10.1038/nm.1959 PubMedCrossRef Kessenbrock K, Krumbholz M, Schonermarck U, Back W, Gross WL, Werb Z, Grone HJ, Brinkmann V, Jenne DE (2009) Netting neutrophils in autoimmune small-vessel vasculitis. Nat Med 15(6):623–625. doi:10.​1038/​nm.​1959 PubMedCrossRef
83.
go back to reference Sangaletti S, Tripodo C, Chiodoni C, Guarnotta C, Cappetti B, Casalini P, Piconese S, Parenza M, Guiducci C, Vitali C, Colombo MP (2012) Neutrophil extracellular traps mediate transfer of cytoplasmic neutrophil antigens to myeloid dendritic cells toward ANCA induction and associated autoimmunity. Blood 120(15):3007–3018. doi:10.1182/blood-2012-03-416156 PubMedCrossRef Sangaletti S, Tripodo C, Chiodoni C, Guarnotta C, Cappetti B, Casalini P, Piconese S, Parenza M, Guiducci C, Vitali C, Colombo MP (2012) Neutrophil extracellular traps mediate transfer of cytoplasmic neutrophil antigens to myeloid dendritic cells toward ANCA induction and associated autoimmunity. Blood 120(15):3007–3018. doi:10.​1182/​blood-2012-03-416156 PubMedCrossRef
86.
go back to reference Courtney PA, Crockard AD, Williamson K, Irvine AE, Kennedy RJ, Bell AL (1999) Increased apoptotic peripheral blood neutrophils in systemic lupus erythematosus: relations with disease activity, antibodies to double stranded DNA, and neutropenia. Ann Rheum Dis 58(5):309–314PubMedCrossRef Courtney PA, Crockard AD, Williamson K, Irvine AE, Kennedy RJ, Bell AL (1999) Increased apoptotic peripheral blood neutrophils in systemic lupus erythematosus: relations with disease activity, antibodies to double stranded DNA, and neutropenia. Ann Rheum Dis 58(5):309–314PubMedCrossRef
87.
go back to reference Hashimoto Y, Nakano K, Yoshinoya S, Tanimoto K, Itoh K (1992) Endothelial cell destruction by polymorphonuclear leukocytes incubated with sera from patients with systemic lupus erythematosus (SLE). Scand J Rheumatol 21(5):209–214PubMedCrossRef Hashimoto Y, Nakano K, Yoshinoya S, Tanimoto K, Itoh K (1992) Endothelial cell destruction by polymorphonuclear leukocytes incubated with sera from patients with systemic lupus erythematosus (SLE). Scand J Rheumatol 21(5):209–214PubMedCrossRef
88.
go back to reference Jonsson H, Sturfelt G (1990) A novel assay for neutrophil clustering activity of human sera: relation to disease activity and neutropenia in systemic lupus erythematosus. Ann Rheum Dis 49(1):46–50PubMedCrossRef Jonsson H, Sturfelt G (1990) A novel assay for neutrophil clustering activity of human sera: relation to disease activity and neutropenia in systemic lupus erythematosus. Ann Rheum Dis 49(1):46–50PubMedCrossRef
89.
go back to reference Lande R, Ganguly D, Facchinetti V, Frasca L, Conrad C, Gregorio J, Meller S, Chamilos G, Sebasigari R, Riccieri V, Bassett R, Amuro H, Fukuhara S, Ito T, Liu YJ, Gilliet M (2011) Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus. Sci Transl Med 3(73):73ra19. doi:10.1126/scitranslmed.3001180 PubMedCrossRef Lande R, Ganguly D, Facchinetti V, Frasca L, Conrad C, Gregorio J, Meller S, Chamilos G, Sebasigari R, Riccieri V, Bassett R, Amuro H, Fukuhara S, Ito T, Liu YJ, Gilliet M (2011) Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus. Sci Transl Med 3(73):73ra19. doi:10.​1126/​scitranslmed.​3001180 PubMedCrossRef
90.
go back to reference Villanueva E, Yalavarthi S, Berthier CC, Hodgin JB, Khandpur R, Lin AM, Rubin CJ, Zhao W, Olsen SH, Klinker M, Shealy D, Denny MF, Plumas J, Chaperot L, Kretzler M, Bruce AT, Kaplan MJ (2011) Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immunostimulatory molecules in systemic lupus erythematosus. J Immunol 187(1):538–552. doi:10.4049/jimmunol.1100450 PubMedCrossRef Villanueva E, Yalavarthi S, Berthier CC, Hodgin JB, Khandpur R, Lin AM, Rubin CJ, Zhao W, Olsen SH, Klinker M, Shealy D, Denny MF, Plumas J, Chaperot L, Kretzler M, Bruce AT, Kaplan MJ (2011) Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immunostimulatory molecules in systemic lupus erythematosus. J Immunol 187(1):538–552. doi:10.​4049/​jimmunol.​1100450 PubMedCrossRef
91.
go back to reference Hakkim A, Furnrohr BG, Amann K, Laube B, Abed UA, Brinkmann V, Herrmann M, Voll RE, Zychlinsky A (2010) Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc Natl Acad Sci U S A 107(21):9813–9818. doi:10.1073/pnas.0909927107 PubMedCrossRef Hakkim A, Furnrohr BG, Amann K, Laube B, Abed UA, Brinkmann V, Herrmann M, Voll RE, Zychlinsky A (2010) Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc Natl Acad Sci U S A 107(21):9813–9818. doi:10.​1073/​pnas.​0909927107 PubMedCrossRef
92.
go back to reference Zykova SN, Tveita AA, Rekvig OP (2010) Renal Dnase1 enzyme activity and protein expression is selectively shut down in murine and human membranoproliferative lupus nephritis. PLoS One 5(8). doi:10.1371/journal.pone.0012096 Zykova SN, Tveita AA, Rekvig OP (2010) Renal Dnase1 enzyme activity and protein expression is selectively shut down in murine and human membranoproliferative lupus nephritis. PLoS One 5(8). doi:10.​1371/​journal.​pone.​0012096
93.
go back to reference Leffler J, Martin M, Gullstrand B, Tyden H, Lood C, Truedsson L, Bengtsson AA, Blom AM (2012) Neutrophil extracellular traps that are not degraded in systemic lupus erythematosus activate complement exacerbating the disease. J Immunol 188(7):3522–3531. doi:10.4049/jimmunol.1102404 PubMedCrossRef Leffler J, Martin M, Gullstrand B, Tyden H, Lood C, Truedsson L, Bengtsson AA, Blom AM (2012) Neutrophil extracellular traps that are not degraded in systemic lupus erythematosus activate complement exacerbating the disease. J Immunol 188(7):3522–3531. doi:10.​4049/​jimmunol.​1102404 PubMedCrossRef
94.
go back to reference Urban CF, Ermert D, Schmid M, Abu-Abed U, Goosmann C, Nacken W, Brinkmann V, Jungblut PR, Zychlinsky A (2009) Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against candida albicans. PLoS Pathog 5(10):e1000639. doi:10.1371/journal.ppat.1000639 PubMedCrossRef Urban CF, Ermert D, Schmid M, Abu-Abed U, Goosmann C, Nacken W, Brinkmann V, Jungblut PR, Zychlinsky A (2009) Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against candida albicans. PLoS Pathog 5(10):e1000639. doi:10.​1371/​journal.​ppat.​1000639 PubMedCrossRef
95.
go back to reference Wang Y, Li M, Stadler S, Correll S, Li P, Wang D, Hayama R, Leonelli L, Han H, Grigoryev SA, Allis CD, Coonrod SA (2009) Histone hypercitrullination mediates chromatin decondensation and neutrophil extracellular trap formation. J Cell Biol 184(2):205–213. doi:10.1083/jcb.200806072 PubMedCrossRef Wang Y, Li M, Stadler S, Correll S, Li P, Wang D, Hayama R, Leonelli L, Han H, Grigoryev SA, Allis CD, Coonrod SA (2009) Histone hypercitrullination mediates chromatin decondensation and neutrophil extracellular trap formation. J Cell Biol 184(2):205–213. doi:10.​1083/​jcb.​200806072 PubMedCrossRef
97.
go back to reference Heinecke JW (1999) Mass spectrometric quantification of amino acid oxidation products in proteins: insights into pathways that promote LDL oxidation in the human artery wall. FASEB J 13(10):1113–1120PubMed Heinecke JW (1999) Mass spectrometric quantification of amino acid oxidation products in proteins: insights into pathways that promote LDL oxidation in the human artery wall. FASEB J 13(10):1113–1120PubMed
99.
go back to reference Dwivedi N, Radic M (2012) Neutrophil activation and B-cell stimulation in the pathogenesis of Felty’s syndrome. Pol Arch Med Wewn 122(7–8):374–379PubMed Dwivedi N, Radic M (2012) Neutrophil activation and B-cell stimulation in the pathogenesis of Felty’s syndrome. Pol Arch Med Wewn 122(7–8):374–379PubMed
100.
go back to reference Lande R, Gregorio J, Facchinetti V, Chatterjee B, Wang YH, Homey B, Cao W, Wang YH, Su B, Nestle FO, Zal T, Mellman I, Schroder JM, Liu YJ, Gilliet M (2007) Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide. Nature 449(7162):564–569. doi:10.1038/nature06116 PubMedCrossRef Lande R, Gregorio J, Facchinetti V, Chatterjee B, Wang YH, Homey B, Cao W, Wang YH, Su B, Nestle FO, Zal T, Mellman I, Schroder JM, Liu YJ, Gilliet M (2007) Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide. Nature 449(7162):564–569. doi:10.​1038/​nature06116 PubMedCrossRef
101.
go back to reference Andrews BS, Eisenberg RA, Theofilopoulos AN, Izui S, Wilson CB, McConahey PJ, Murphy ED, Roths JB, Dixon FJ (1978) Spontaneous murine lupus-like syndromes. Clinical and immunopathological manifestations in several strains. J Exp Med 148:1198–1215PubMedCrossRef Andrews BS, Eisenberg RA, Theofilopoulos AN, Izui S, Wilson CB, McConahey PJ, Murphy ED, Roths JB, Dixon FJ (1978) Spontaneous murine lupus-like syndromes. Clinical and immunopathological manifestations in several strains. J Exp Med 148:1198–1215PubMedCrossRef
102.
go back to reference Channing AA, Kasuga T, Horowitz RE, Dubois EL, Demopoulos HB (1965) An ultrastructural study of spontaneous lupus nephritis in the NZB-BL-NZW mouse. Am J Pathol 47(4):677–694PubMed Channing AA, Kasuga T, Horowitz RE, Dubois EL, Demopoulos HB (1965) An ultrastructural study of spontaneous lupus nephritis in the NZB-BL-NZW mouse. Am J Pathol 47(4):677–694PubMed
103.
go back to reference Farquhar MG, Vernier RL, Good RA (1957) An electron microscope study of the glomerulus in nephrosis, glomerulonephritis, and lupus erythematosus. J Exp Med 106(5):649–660PubMedCrossRef Farquhar MG, Vernier RL, Good RA (1957) An electron microscope study of the glomerulus in nephrosis, glomerulonephritis, and lupus erythematosus. J Exp Med 106(5):649–660PubMedCrossRef
104.
go back to reference Kalaaji M, Mortensen E, Jorgensen L, Olsen R, Rekvig OP (2006) Nephritogenic lupus antibodies recognize glomerular basement membrane-associated chromatin fragments released from apoptotic intraglomerular cells. Am J Pathol 168(6):1779–1792. doi:10.2353/ajpath.2006.051329 PubMedCrossRef Kalaaji M, Mortensen E, Jorgensen L, Olsen R, Rekvig OP (2006) Nephritogenic lupus antibodies recognize glomerular basement membrane-associated chromatin fragments released from apoptotic intraglomerular cells. Am J Pathol 168(6):1779–1792. doi:10.​2353/​ajpath.​2006.​051329 PubMedCrossRef
106.
go back to reference Chen K, Nishi H, Travers R, Tsuboi N, Martinod K, Wagner DD, Stan R, Croce K, Mayadas TN (2012) Endocytosis of soluble immune complexes leads to their clearance by FcgammaRIIIB but induces neutrophil extracellular traps via FcgammaRIIA in vivo. Blood 120(22):4421–4431. doi:10.1182/blood-2011-12-401133 PubMedCrossRef Chen K, Nishi H, Travers R, Tsuboi N, Martinod K, Wagner DD, Stan R, Croce K, Mayadas TN (2012) Endocytosis of soluble immune complexes leads to their clearance by FcgammaRIIIB but induces neutrophil extracellular traps via FcgammaRIIA in vivo. Blood 120(22):4421–4431. doi:10.​1182/​blood-2011-12-401133 PubMedCrossRef
108.
go back to reference Isenberg DA (1997) Autoantibodies: markers of disease or pathogenic? Ann N Y Acad Sci 823:256–262PubMedCrossRef Isenberg DA (1997) Autoantibodies: markers of disease or pathogenic? Ann N Y Acad Sci 823:256–262PubMedCrossRef
109.
go back to reference Manson JJ, Ma A, Rogers P, Mason LJ, Berden JH, van der Vlag J, D’Cruz DP, Isenberg DA, Rahman A (2009) Relationship between anti-dsDNA, anti-nucleosome and anti-alpha-actinin antibodies and markers of renal disease in patients with lupus nephritis: a prospective longitudinal study. Arthritis Res Ther 11(5):R154. doi:10.1186/ar2831 PubMedCrossRef Manson JJ, Ma A, Rogers P, Mason LJ, Berden JH, van der Vlag J, D’Cruz DP, Isenberg DA, Rahman A (2009) Relationship between anti-dsDNA, anti-nucleosome and anti-alpha-actinin antibodies and markers of renal disease in patients with lupus nephritis: a prospective longitudinal study. Arthritis Res Ther 11(5):R154. doi:10.​1186/​ar2831 PubMedCrossRef
110.
go back to reference Wu T, Xie C, Wang HW, Zhou XJ, Schwartz N, Calixto S, Mackay M, Aranow C, Putterman C, Mohan C (2007) Elevated urinary VCAM-1, P-selectin, soluble TNF receptor-1, and CXC chemokine ligand 16 in multiple murine lupus strains and human lupus nephritis. J Immunol 179(10):7166–7175PubMed Wu T, Xie C, Wang HW, Zhou XJ, Schwartz N, Calixto S, Mackay M, Aranow C, Putterman C, Mohan C (2007) Elevated urinary VCAM-1, P-selectin, soluble TNF receptor-1, and CXC chemokine ligand 16 in multiple murine lupus strains and human lupus nephritis. J Immunol 179(10):7166–7175PubMed
111.
113.
go back to reference Madaio MP (2003) Lupus autoantibodies 101: one size does not fit all; however, specificity influences pathogenicity. Clin Exp Immunol 131(3):396–397PubMedCrossRef Madaio MP (2003) Lupus autoantibodies 101: one size does not fit all; however, specificity influences pathogenicity. Clin Exp Immunol 131(3):396–397PubMedCrossRef
114.
go back to reference Dixon FJ, Oldstone MB, Tonietti G (1971) Pathogenesis of immune complex glomerulonephritis of new zealand mice. J Exp Med 134(3):65–71PubMed Dixon FJ, Oldstone MB, Tonietti G (1971) Pathogenesis of immune complex glomerulonephritis of new zealand mice. J Exp Med 134(3):65–71PubMed
116.
go back to reference Kramers C, Hylkema MN, van Bruggen MC, van de Lagemaat R, Dijkman HB, Assmann KJ, Smeenk RJ, Berden JH (1994) Anti-nucleosome antibodies complexed to nucleosomal antigens show anti-DNA reactivity and bind to rat glomerular basement membrane in vivo. J Clin Invest 94(2):568–577. doi:10.1172/JCI117371 PubMedCrossRef Kramers C, Hylkema MN, van Bruggen MC, van de Lagemaat R, Dijkman HB, Assmann KJ, Smeenk RJ, Berden JH (1994) Anti-nucleosome antibodies complexed to nucleosomal antigens show anti-DNA reactivity and bind to rat glomerular basement membrane in vivo. J Clin Invest 94(2):568–577. doi:10.​1172/​JCI117371 PubMedCrossRef
117.
go back to reference van Bruggen MC, Walgreen B, Rijke TP, Tamboer W, Kramers K, Smeenk RJ, Monestier M, Fournie GJ, Berden JH (1997) Antigen specificity of anti-nuclear antibodies complexed to nucleosomes determines glomerular basement membrane binding in vivo. Eur J Immunol 27(6):1564–1569. doi:10.1002/eji.1830270636 PubMedCrossRef van Bruggen MC, Walgreen B, Rijke TP, Tamboer W, Kramers K, Smeenk RJ, Monestier M, Fournie GJ, Berden JH (1997) Antigen specificity of anti-nuclear antibodies complexed to nucleosomes determines glomerular basement membrane binding in vivo. Eur J Immunol 27(6):1564–1569. doi:10.​1002/​eji.​1830270636 PubMedCrossRef
119.
go back to reference Kalaaji M, Fenton KA, Mortensen ES, Olsen R, Sturfelt G, Alm P, Rekvig OP (2007) Glomerular apoptotic nucleosomes are central target structures for nephritogenic antibodies in human SLE nephritis. Kidney Int 71(7):664–672. doi:10.1038/sj.ki.5002133 PubMedCrossRef Kalaaji M, Fenton KA, Mortensen ES, Olsen R, Sturfelt G, Alm P, Rekvig OP (2007) Glomerular apoptotic nucleosomes are central target structures for nephritogenic antibodies in human SLE nephritis. Kidney Int 71(7):664–672. doi:10.​1038/​sj.​ki.​5002133 PubMedCrossRef
120.
121.
go back to reference Mjelle JE, Rekvig OP, Fenton KA (2007) Nucleosomes possess a high affinity for glomerular laminin and collagen IV and bind nephritogenic antibodies in murine lupus-like nephritis. Ann Rheum Dis 66(12):1661–1668. doi:10.1136/ard.2007.070482 PubMedCrossRef Mjelle JE, Rekvig OP, Fenton KA (2007) Nucleosomes possess a high affinity for glomerular laminin and collagen IV and bind nephritogenic antibodies in murine lupus-like nephritis. Ann Rheum Dis 66(12):1661–1668. doi:10.​1136/​ard.​2007.​070482 PubMedCrossRef
122.
123.
go back to reference Termaat RM, Assmann KJ, Dijkman HB, van Gompel F, Smeenk RJ, Berden JH (1992) Anti-DNA antibodies can bind to the glomerulus via two distinct mechanisms. Kidney Int 42(6):1363–1371PubMedCrossRef Termaat RM, Assmann KJ, Dijkman HB, van Gompel F, Smeenk RJ, Berden JH (1992) Anti-DNA antibodies can bind to the glomerulus via two distinct mechanisms. Kidney Int 42(6):1363–1371PubMedCrossRef
124.
go back to reference Faaber P, Rijke TP, van de Putte LB, Capel PJ, Berden JH (1986) Cross-reactivity of human and murine anti-DNA antibodies with heparan sulfate. The major glycosaminoglycan in glomerular basement membranes. J Clin Invest 77(6):1824–1830. doi:10.1172/JCI112508 PubMedCrossRef Faaber P, Rijke TP, van de Putte LB, Capel PJ, Berden JH (1986) Cross-reactivity of human and murine anti-DNA antibodies with heparan sulfate. The major glycosaminoglycan in glomerular basement membranes. J Clin Invest 77(6):1824–1830. doi:10.​1172/​JCI112508 PubMedCrossRef
125.
go back to reference Madaio MP, Carlson J, Cataldo J, Ucci A, Migliorini P, Pankewycz O (1987) Murine monoclonal anti-DNA antibodies bind directly to glomerular antigens and form immune deposits. J Immunol 138(9):2883–2889PubMed Madaio MP, Carlson J, Cataldo J, Ucci A, Migliorini P, Pankewycz O (1987) Murine monoclonal anti-DNA antibodies bind directly to glomerular antigens and form immune deposits. J Immunol 138(9):2883–2889PubMed
127.
go back to reference Vlahakos DV, Foster MH, Adams S, Katz M, Ucci AA, Barrett KJ, Datta SK, Madaio MP (1992) Anti-DNA antibodies form immune deposits at distinct glomerular and vascular sites. Kidney Int 41(6):1690–1700. doi:10.1038/ki.1992.242 PubMedCrossRef Vlahakos DV, Foster MH, Adams S, Katz M, Ucci AA, Barrett KJ, Datta SK, Madaio MP (1992) Anti-DNA antibodies form immune deposits at distinct glomerular and vascular sites. Kidney Int 41(6):1690–1700. doi:10.​1038/​ki.​1992.​242 PubMedCrossRef
128.
go back to reference D’Andrea DM, Coupaye-Gerard B, Kleyman TR, Foster MH, Madaio MP (1996) Lupus autoantibodies interact directly with distinct glomerular and vascular cell surface antigens. Kidney Int 49(5):1214–1221. doi:10.1038/ki.1996.176 PubMedCrossRef D’Andrea DM, Coupaye-Gerard B, Kleyman TR, Foster MH, Madaio MP (1996) Lupus autoantibodies interact directly with distinct glomerular and vascular cell surface antigens. Kidney Int 49(5):1214–1221. doi:10.​1038/​ki.​1996.​176 PubMedCrossRef
129.
go back to reference Sabbaga J, Line SR, Potocnjak P, Madaio MP (1989) A murine nephritogenic monoclonal anti-DNA autoantibody binds directly to mouse laminin, the major non-collagenous protein component of the glomerular basement membrane. Eur J Immunol 19(1):137–143. doi:10.1002/eji.1830190122 PubMedCrossRef Sabbaga J, Line SR, Potocnjak P, Madaio MP (1989) A murine nephritogenic monoclonal anti-DNA autoantibody binds directly to mouse laminin, the major non-collagenous protein component of the glomerular basement membrane. Eur J Immunol 19(1):137–143. doi:10.​1002/​eji.​1830190122 PubMedCrossRef
130.
go back to reference Chan TM, Frampton G, Staines NA, Hobby P, Perry GJ, Cameron JS (1992) Different mechanisms by which anti-DNA MoAbs bind to human endothelial cells and glomerular mesangial cells. Clin Exp Immunol 88(1):68–74PubMedCrossRef Chan TM, Frampton G, Staines NA, Hobby P, Perry GJ, Cameron JS (1992) Different mechanisms by which anti-DNA MoAbs bind to human endothelial cells and glomerular mesangial cells. Clin Exp Immunol 88(1):68–74PubMedCrossRef
131.
go back to reference Deocharan B, Qing X, Lichauco J, Putterman C (2002) Alpha-actinin is a cross-reactive renal target for pathogenic anti-DNA antibodies. J Immunol 168(6):3072–3078PubMed Deocharan B, Qing X, Lichauco J, Putterman C (2002) Alpha-actinin is a cross-reactive renal target for pathogenic anti-DNA antibodies. J Immunol 168(6):3072–3078PubMed
132.
go back to reference Hasselaar P, Derksen RH, Blokzijl L, de Groot PG (1990) Crossreactivity of antibodies directed against cardiolipin, DNA, endothelial cells and blood platelets. Thromb Haemost 63(2):169–173PubMed Hasselaar P, Derksen RH, Blokzijl L, de Groot PG (1990) Crossreactivity of antibodies directed against cardiolipin, DNA, endothelial cells and blood platelets. Thromb Haemost 63(2):169–173PubMed
133.
go back to reference Moscato S, Pratesi F, Bongiorni F, Scavuzzo MC, Chimenti D, Bombardieri S, Migliorini P (2002) Endothelial cell binding by systemic lupus antibodies: functional properties and relationship with anti-DNA activity. J Autoimmun 18(3):231–238PubMedCrossRef Moscato S, Pratesi F, Bongiorni F, Scavuzzo MC, Chimenti D, Bombardieri S, Migliorini P (2002) Endothelial cell binding by systemic lupus antibodies: functional properties and relationship with anti-DNA activity. J Autoimmun 18(3):231–238PubMedCrossRef
134.
go back to reference Qing X, Zavadil J, Crosby MB, Hogarth MP, Hahn BH, Mohan C, Gilkeson GS, Bottinger EP, Putterman C (2006) Nephritogenic anti-DNA antibodies regulate gene expression in MRL/lpr mouse glomerular mesangial cells. Arthritis Rheum 54(7):2198–2210. doi:10.1002/art.21934 PubMedCrossRef Qing X, Zavadil J, Crosby MB, Hogarth MP, Hahn BH, Mohan C, Gilkeson GS, Bottinger EP, Putterman C (2006) Nephritogenic anti-DNA antibodies regulate gene expression in MRL/lpr mouse glomerular mesangial cells. Arthritis Rheum 54(7):2198–2210. doi:10.​1002/​art.​21934 PubMedCrossRef
136.
go back to reference Ben Chetrit E, Dunsky EH, Wollner S, Eilat D (1985) In vivo clearance and tissue uptake of an anti-DNA monoclonal antibody and its complexes with DNA. Clin Exp Immunol 60(1):159–168PubMed Ben Chetrit E, Dunsky EH, Wollner S, Eilat D (1985) In vivo clearance and tissue uptake of an anti-DNA monoclonal antibody and its complexes with DNA. Clin Exp Immunol 60(1):159–168PubMed
137.
go back to reference Emlen W, Mannik M (1982) Clearance of circulating DNA-anti-DNA immune complexes in mice. J Exp Med 155(4):1210–1215PubMedCrossRef Emlen W, Mannik M (1982) Clearance of circulating DNA-anti-DNA immune complexes in mice. J Exp Med 155(4):1210–1215PubMedCrossRef
138.
go back to reference Gauthier VJ, Mannik M, Striker GE (1982) Effect of cationized antibodies in performed immune complexes on deposition and persistence in renal glomeruli. J Exp Med 156(3):766–777PubMedCrossRef Gauthier VJ, Mannik M, Striker GE (1982) Effect of cationized antibodies in performed immune complexes on deposition and persistence in renal glomeruli. J Exp Med 156(3):766–777PubMedCrossRef
139.
go back to reference Agodoa LY, Gauthier VJ, Mannik M (1985) Antibody localization in the glomerular basement membrane may precede in situ immune deposit formation in rat glomeruli. J Immunol 134(2):880–884PubMed Agodoa LY, Gauthier VJ, Mannik M (1985) Antibody localization in the glomerular basement membrane may precede in situ immune deposit formation in rat glomeruli. J Immunol 134(2):880–884PubMed
140.
141.
go back to reference Bergtold A, Gavhane A, D’Agati V, Madaio M, Clynes R (2006) FcR-bearing myeloid cells are responsible for triggering murine lupus nephritis. J Immunol 177(10):7287–7295PubMed Bergtold A, Gavhane A, D’Agati V, Madaio M, Clynes R (2006) FcR-bearing myeloid cells are responsible for triggering murine lupus nephritis. J Immunol 177(10):7287–7295PubMed
143.
go back to reference Qing X, Pitashny M, Thomas DB, Barrat FJ, Hogarth MP, Putterman C (2008) Pathogenic anti-DNA antibodies modulate gene expression in mesangial cells: involvement of HMGB1 in anti-DNA antibody-induced renal injury. Immunol Lett 121(1):61–73. doi:10.1016/j.imlet.2008.08.007 PubMedCrossRef Qing X, Pitashny M, Thomas DB, Barrat FJ, Hogarth MP, Putterman C (2008) Pathogenic anti-DNA antibodies modulate gene expression in mesangial cells: involvement of HMGB1 in anti-DNA antibody-induced renal injury. Immunol Lett 121(1):61–73. doi:10.​1016/​j.​imlet.​2008.​08.​007 PubMedCrossRef
144.
go back to reference Uciechowski P, Schwarz M, Gessner JE, Schmidt RE, Resch K, Radeke HH (1998) IFN-gamma induces the high-affinity Fc receptor I for IgG (CD64) on human glomerular mesangial cells. Eur J Immunol 28(9):2928–2935PubMedCrossRef Uciechowski P, Schwarz M, Gessner JE, Schmidt RE, Resch K, Radeke HH (1998) IFN-gamma induces the high-affinity Fc receptor I for IgG (CD64) on human glomerular mesangial cells. Eur J Immunol 28(9):2928–2935PubMedCrossRef
145.
go back to reference Lopez-Armada MJ, Gomez-Guerrero C, Egido J (1996) Receptors for immune complexes activate gene expression and synthesis of matrix proteins in cultured rat and human mesangial cells: role of TGF-beta. J Immunol 157(5):2136–2142PubMed Lopez-Armada MJ, Gomez-Guerrero C, Egido J (1996) Receptors for immune complexes activate gene expression and synthesis of matrix proteins in cultured rat and human mesangial cells: role of TGF-beta. J Immunol 157(5):2136–2142PubMed
146.
go back to reference Morcos M, Hansch GM, Schonermark M, Ellwanger S, Harle M, Heckl-Ostreicher B (1994) Human glomerular mesangial cells express CD16 and may be stimulated via this receptor. Kidney Int 46(6):1627–1634PubMedCrossRef Morcos M, Hansch GM, Schonermark M, Ellwanger S, Harle M, Heckl-Ostreicher B (1994) Human glomerular mesangial cells express CD16 and may be stimulated via this receptor. Kidney Int 46(6):1627–1634PubMedCrossRef
147.
go back to reference Radeke HH, Janssen-Graalfs I, Sowa EN, Chouchakova N, Skokowa J, Loscher F, Schmidt RE, Heeringa P, Gessner JE (2002) Opposite regulation of type II and III receptors for immunoglobulin G in mouse glomerular mesangial cells and in the induction of anti-glomerular basement membrane (GBM) nephritis. J Biol Chem 277(30):27535–27544. doi:10.1074/jbc.M200419200 PubMedCrossRef Radeke HH, Janssen-Graalfs I, Sowa EN, Chouchakova N, Skokowa J, Loscher F, Schmidt RE, Heeringa P, Gessner JE (2002) Opposite regulation of type II and III receptors for immunoglobulin G in mouse glomerular mesangial cells and in the induction of anti-glomerular basement membrane (GBM) nephritis. J Biol Chem 277(30):27535–27544. doi:10.​1074/​jbc.​M200419200 PubMedCrossRef
148.
go back to reference Kovalenko P, Fujinaka H, Yoshida Y, Kawamura H, Qu Z, El-Shemi AG, Li H, Matsuki A, Bilim V, Yaoita E, Abo T, Uchiyama M, Yamamoto T (2004) Fc receptor-mediated accumulation of macrophages in crescentic glomerulonephritis induced by anti-glomerular basement membrane antibody administration in WKY rats. Int Immunol 16(5):625–634. doi:10.1093/intimm/dxh058 PubMedCrossRef Kovalenko P, Fujinaka H, Yoshida Y, Kawamura H, Qu Z, El-Shemi AG, Li H, Matsuki A, Bilim V, Yaoita E, Abo T, Uchiyama M, Yamamoto T (2004) Fc receptor-mediated accumulation of macrophages in crescentic glomerulonephritis induced by anti-glomerular basement membrane antibody administration in WKY rats. Int Immunol 16(5):625–634. doi:10.​1093/​intimm/​dxh058 PubMedCrossRef
150.
go back to reference Taylor KE, Chung SA, Graham RR, Ortmann WA, Lee AT, Langefeld CD, Jacob CO, Kamboh MI, Alarcon-Riquelme ME, Tsao BP, Moser KL, Gaffney PM, Harley JB, Petri M, Manzi S, Gregersen PK, Behrens TW, Criswell LA (2011) Risk alleles for systemic lupus erythematosus in a large case–control collection and associations with clinical subphenotypes. PLoS Genet 7(2):e1001311. doi:10.1371/journal.pgen.1001311 PubMedCrossRef Taylor KE, Chung SA, Graham RR, Ortmann WA, Lee AT, Langefeld CD, Jacob CO, Kamboh MI, Alarcon-Riquelme ME, Tsao BP, Moser KL, Gaffney PM, Harley JB, Petri M, Manzi S, Gregersen PK, Behrens TW, Criswell LA (2011) Risk alleles for systemic lupus erythematosus in a large case–control collection and associations with clinical subphenotypes. PLoS Genet 7(2):e1001311. doi:10.​1371/​journal.​pgen.​1001311 PubMedCrossRef
151.
go back to reference Moser KL, Neas BR, Salmon JE, Yu H, Gray-McGuire C, Asundi N, Bruner GR, Fox J, Kelly J, Henshall S, Bacino D, Dietz M, Hogue R, Koelsch G, Nightingale L, Shaver T, Abdou NI, Albert DA, Carson C, Petri M, Treadwell EL, James JA, Harley JB (1998) Genome scan of human systemic lupus erythematosus: evidence for linkage on chromosome 1q in african-american pedigrees. Proc Natl Acad Sci U S A 95(25):14869–14874PubMedCrossRef Moser KL, Neas BR, Salmon JE, Yu H, Gray-McGuire C, Asundi N, Bruner GR, Fox J, Kelly J, Henshall S, Bacino D, Dietz M, Hogue R, Koelsch G, Nightingale L, Shaver T, Abdou NI, Albert DA, Carson C, Petri M, Treadwell EL, James JA, Harley JB (1998) Genome scan of human systemic lupus erythematosus: evidence for linkage on chromosome 1q in african-american pedigrees. Proc Natl Acad Sci U S A 95(25):14869–14874PubMedCrossRef
152.
go back to reference Agnello V, Koffler D, Kunkel HG (1973) Immune complex systems in the nephritis of systemic lupus erythematosus. Kidney Int 3(2):90–99PubMedCrossRef Agnello V, Koffler D, Kunkel HG (1973) Immune complex systems in the nephritis of systemic lupus erythematosus. Kidney Int 3(2):90–99PubMedCrossRef
153.
go back to reference Godau J, Heller T, Hawlisch H, Trappe M, Howells E, Best J, Zwirner J, Verbeek JS, Hogarth PM, Gerard C, Van Rooijen N, Klos A, Gessner JE, Kohl J (2004) C5a initiates the inflammatory cascade in immune complex peritonitis. J Immunol 173(5):3437–3445PubMed Godau J, Heller T, Hawlisch H, Trappe M, Howells E, Best J, Zwirner J, Verbeek JS, Hogarth PM, Gerard C, Van Rooijen N, Klos A, Gessner JE, Kohl J (2004) C5a initiates the inflammatory cascade in immune complex peritonitis. J Immunol 173(5):3437–3445PubMed
154.
go back to reference Wang Y, Hu Q, Madri JA, Rollins SA, Chodera A, Matis LA (1996) Amelioration of lupus-like autoimmune disease in NZB/WF1 mice after treatment with a blocking monoclonal antibody specific for complement component C5. Proc Natl Acad Sci U S A 93(16):8563–8568PubMedCrossRef Wang Y, Hu Q, Madri JA, Rollins SA, Chodera A, Matis LA (1996) Amelioration of lupus-like autoimmune disease in NZB/WF1 mice after treatment with a blocking monoclonal antibody specific for complement component C5. Proc Natl Acad Sci U S A 93(16):8563–8568PubMedCrossRef
155.
go back to reference Shushakova N, Skokowa J, Schulman J, Baumann U, Zwirner J, Schmidt RE, Gessner JE (2002) C5a Anaphylatoxin is a major regulator of activating versus inhibitory FcgammaRs in immune complex-induced lung disease. J Clin Invest 110(12):1823–1830. doi:10.1172/JCI16577 PubMed Shushakova N, Skokowa J, Schulman J, Baumann U, Zwirner J, Schmidt RE, Gessner JE (2002) C5a Anaphylatoxin is a major regulator of activating versus inhibitory FcgammaRs in immune complex-induced lung disease. J Clin Invest 110(12):1823–1830. doi:10.​1172/​JCI16577 PubMed
156.
go back to reference Skokowa J, Ali SR, Felda O, Kumar V, Konrad S, Shushakova N, Schmidt RE, Piekorz RP, Nurnberg B, Spicher K, Birnbaumer L, Zwirner J, Claassens JW, Verbeek JS, van Rooijen N, Kohl J, Gessner JE (2005) Macrophages induce the inflammatory response in the pulmonary arthus reaction through G alpha i2 activation that controls C5aR and Fc receptor cooperation. J Immunol 174(5):3041–3050PubMed Skokowa J, Ali SR, Felda O, Kumar V, Konrad S, Shushakova N, Schmidt RE, Piekorz RP, Nurnberg B, Spicher K, Birnbaumer L, Zwirner J, Claassens JW, Verbeek JS, van Rooijen N, Kohl J, Gessner JE (2005) Macrophages induce the inflammatory response in the pulmonary arthus reaction through G alpha i2 activation that controls C5aR and Fc receptor cooperation. J Immunol 174(5):3041–3050PubMed
158.
go back to reference Korb LC, Ahearn JM (1997) C1q Binds directly and specifically to surface blebs of apoptotic human keratinocytes: complement deficiency and systemic lupus erythematosus revisited. J Immunol 158(10):4525–4528PubMed Korb LC, Ahearn JM (1997) C1q Binds directly and specifically to surface blebs of apoptotic human keratinocytes: complement deficiency and systemic lupus erythematosus revisited. J Immunol 158(10):4525–4528PubMed
159.
go back to reference Mevorach D, Mascarenhas JO, Gershov D, Elkon KB (1998) Complement-dependent clearance of apoptotic cells by human macrophages. J Exp Med 188(12):2313–2320PubMedCrossRef Mevorach D, Mascarenhas JO, Gershov D, Elkon KB (1998) Complement-dependent clearance of apoptotic cells by human macrophages. J Exp Med 188(12):2313–2320PubMedCrossRef
162.
go back to reference von Bruhl ML, Stark K, Steinhart A, Chandraratne S, Konrad I, Lorenz M, Khandoga A, Tirniceriu A, Coletti R, Kollnberger M, Byrne RA, Laitinen I, Walch A, Brill A, Pfeiler S, Manukyan D, Braun S, Lange P, Riegger J, Ware J, Eckart A, Haidari S, Rudelius M, Schulz C, Echtler K, Brinkmann V, Schwaiger M, Preissner KT, Wagner DD, Mackman N, Engelmann B, Massberg S (2012) Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med 209(4):819–835. doi:10.1084/jem.20112322 CrossRef von Bruhl ML, Stark K, Steinhart A, Chandraratne S, Konrad I, Lorenz M, Khandoga A, Tirniceriu A, Coletti R, Kollnberger M, Byrne RA, Laitinen I, Walch A, Brill A, Pfeiler S, Manukyan D, Braun S, Lange P, Riegger J, Ware J, Eckart A, Haidari S, Rudelius M, Schulz C, Echtler K, Brinkmann V, Schwaiger M, Preissner KT, Wagner DD, Mackman N, Engelmann B, Massberg S (2012) Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med 209(4):819–835. doi:10.​1084/​jem.​20112322 CrossRef
163.
go back to reference Megens RT, Vijayan S, Lievens D, Doring Y, van Zandvoort MA, Grommes J, Weber C, Soehnlein O (2012) Presence of luminal neutrophil extracellular traps in atherosclerosis. Thromb Haemost 107(3):597–598. doi:10.1160/TH11-09-0650 PubMedCrossRef Megens RT, Vijayan S, Lievens D, Doring Y, van Zandvoort MA, Grommes J, Weber C, Soehnlein O (2012) Presence of luminal neutrophil extracellular traps in atherosclerosis. Thromb Haemost 107(3):597–598. doi:10.​1160/​TH11-09-0650 PubMedCrossRef
165.
go back to reference Daigo K, Yamaguchi N, Kawamura T, Matsubara K, Jiang S, Ohashi R, Sudou Y, Kodama T, Naito M, Inoue K, Hamakubo T (2012) The proteomic profile of circulating pentraxin 3 (PTX3) complex in sepsis demonstrates the interaction with azurocidin 1 and other components of neutrophil extracellular traps. Mol Cell Proteomics 11(6):M111 015073. doi:10.1074/mcp.M111.015073 PubMed Daigo K, Yamaguchi N, Kawamura T, Matsubara K, Jiang S, Ohashi R, Sudou Y, Kodama T, Naito M, Inoue K, Hamakubo T (2012) The proteomic profile of circulating pentraxin 3 (PTX3) complex in sepsis demonstrates the interaction with azurocidin 1 and other components of neutrophil extracellular traps. Mol Cell Proteomics 11(6):M111 015073. doi:10.​1074/​mcp.​M111.​015073 PubMed
166.
go back to reference Schorn C, Janko C, Krenn V, Zhao Y, Munoz LE, Schett G, Herrmann M (2012) Bonding the foe—NETting neutrophils immobilize the pro-inflammatory monosodium urate crystals. Front Immunol 3:376. doi:10.3389/fimmu.2012.00376 PubMed Schorn C, Janko C, Krenn V, Zhao Y, Munoz LE, Schett G, Herrmann M (2012) Bonding the foe—NETting neutrophils immobilize the pro-inflammatory monosodium urate crystals. Front Immunol 3:376. doi:10.​3389/​fimmu.​2012.​00376 PubMed
167.
go back to reference Wermeling F, Chen Y, Pikkarainen T, Scheynius A, Winqvist O, Izui S, Ravetch JV, Tryggvason K, Karlsson MC (2007) Class a scavenger receptors regulate tolerance against apoptotic cells, and autoantibodies against these receptors are predictive of systemic lupus. J Exp Med 204(10):2259–2265. doi:10.1084/jem.20070600 PubMedCrossRef Wermeling F, Chen Y, Pikkarainen T, Scheynius A, Winqvist O, Izui S, Ravetch JV, Tryggvason K, Karlsson MC (2007) Class a scavenger receptors regulate tolerance against apoptotic cells, and autoantibodies against these receptors are predictive of systemic lupus. J Exp Med 204(10):2259–2265. doi:10.​1084/​jem.​20070600 PubMedCrossRef
168.
go back to reference Parvatiyar K, Zhang Z, Teles RM, Ouyang S, Jiang Y, Iyer SS, Zaver SA, Schenk M, Zeng S, Zhong W, Liu ZJ, Modlin RL, Liu YJ, Cheng G (2012) The helicase DDX41 recognizes the bacterial secondary messengers cyclic di-GMP and cyclic di-AMP to activate a type I interferon immune response. Nat Immunol 13(12):1155–1161. doi:10.1038/ni.2460 PubMedCrossRef Parvatiyar K, Zhang Z, Teles RM, Ouyang S, Jiang Y, Iyer SS, Zaver SA, Schenk M, Zeng S, Zhong W, Liu ZJ, Modlin RL, Liu YJ, Cheng G (2012) The helicase DDX41 recognizes the bacterial secondary messengers cyclic di-GMP and cyclic di-AMP to activate a type I interferon immune response. Nat Immunol 13(12):1155–1161. doi:10.​1038/​ni.​2460 PubMedCrossRef
171.
go back to reference Guimaraes-Costa AB, Nascimento MT, Froment GS, Soares RP, Morgado FN, Conceicao-Silva F, Saraiva EM (2009) Leishmania amazonensis promastigotes induce and are killed by neutrophil extracellular traps. Proc Natl Acad Sci U S A 106(16):6748–6753. doi:10.1073/pnas.0900226106 PubMedCrossRef Guimaraes-Costa AB, Nascimento MT, Froment GS, Soares RP, Morgado FN, Conceicao-Silva F, Saraiva EM (2009) Leishmania amazonensis promastigotes induce and are killed by neutrophil extracellular traps. Proc Natl Acad Sci U S A 106(16):6748–6753. doi:10.​1073/​pnas.​0900226106 PubMedCrossRef
173.
go back to reference Kirchner T, Moller S, Klinger M, Solbach W, Laskay T, Behnen M (2012) The impact of various reactive oxygen species on the formation of neutrophil extracellular traps. Mediators Inflamm 2012:849136. doi:10.1155/2012/849136 PubMedCrossRef Kirchner T, Moller S, Klinger M, Solbach W, Laskay T, Behnen M (2012) The impact of various reactive oxygen species on the formation of neutrophil extracellular traps. Mediators Inflamm 2012:849136. doi:10.​1155/​2012/​849136 PubMedCrossRef
174.
go back to reference Willis VC, Gizinski AM, Banda NK, Causey CP, Knuckley B, Cordova KN, Luo Y, Levitt B, Glogowska M, Chandra P, Kulik L, Robinson WH, Arend WP, Thompson PR, Holers VM (2011) N-alpha-benzoyl-N5-(2-chloro-1-iminoethyl)-l-ornithine amide, a protein arginine deiminase inhibitor, reduces the severity of murine collagen-induced arthritis. J Immunol 186(7):4396–4404. doi:10.4049/jimmunol.1001620 PubMedCrossRef Willis VC, Gizinski AM, Banda NK, Causey CP, Knuckley B, Cordova KN, Luo Y, Levitt B, Glogowska M, Chandra P, Kulik L, Robinson WH, Arend WP, Thompson PR, Holers VM (2011) N-alpha-benzoyl-N5-(2-chloro-1-iminoethyl)-l-ornithine amide, a protein arginine deiminase inhibitor, reduces the severity of murine collagen-induced arthritis. J Immunol 186(7):4396–4404. doi:10.​4049/​jimmunol.​1001620 PubMedCrossRef
175.
go back to reference van Montfoort ML, Stephan F, Lauw MN, Hutten BA, Van Mierlo GJ, Solati S, Middeldorp S, Meijers JC, Zeerleder S (2013) Circulating nucleosomes and neutrophil activation as risk factors for deep vein thrombosis. Arterioscler Thromb Vasc Biol 33(1):147–151. doi:10.1161/ATVBAHA.112.300498 PubMedCrossRef van Montfoort ML, Stephan F, Lauw MN, Hutten BA, Van Mierlo GJ, Solati S, Middeldorp S, Meijers JC, Zeerleder S (2013) Circulating nucleosomes and neutrophil activation as risk factors for deep vein thrombosis. Arterioscler Thromb Vasc Biol 33(1):147–151. doi:10.​1161/​ATVBAHA.​112.​300498 PubMedCrossRef
Metadata
Title
Neutrophil extracellular chromatin traps connect innate immune response to autoimmunity
Authors
Marko Radic
Tony N. Marion
Publication date
01-07-2013
Publisher
Springer-Verlag
Published in
Seminars in Immunopathology / Issue 4/2013
Print ISSN: 1863-2297
Electronic ISSN: 1863-2300
DOI
https://doi.org/10.1007/s00281-013-0376-6

Other articles of this Issue 4/2013

Seminars in Immunopathology 4/2013 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

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.