GET THE APP

Pro- and Anti-inflammatory Neutrophils in Lupus
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Review Article - (2014) Volume 0, Issue 0

Pro- and Anti-inflammatory Neutrophils in Lupus

Evan Der, Abhishek Trigunaite, Ayesha Khan and Trine N. Jørgensen*
Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Ohio, USA
*Corresponding Author: Dr. Trine N. Jørgensen, Department of Immunology, NE40, Lerner Research Institute, Cleveland Clinic Foundation, Ohio, USA, Tel: +1 216-444-7454, Fax: +1 216-444-9329 Email:

Abstract

A hallmark of SLE is the presence of elevated levels of circulating anti-nuclear autoantibodies specific towards chromatin, histones or dsDNA. Understanding the regulation of antibody production is therefore of utmost importance in understanding lupus pathogenesis. Spearheaded by the identification of accumulating immunosuppressive neutrophils in cancer patients, the nature and function of neutrophils have expanded from a uniform pro-inflammatory cell population to a heterogeneous population of cells with pro-inflammatory or immunosuppressive capacities. While much is known about pro-inflammatory neutrophils and the likely pathogenic function of such cells in lupus, a potential role for immunosuppressive neutrophils in protecting genetically predisposed individuals has only recently emerged. For example, SLE-derived neutrophils spontaneously produce type I interferons (IFNα), strongly associated with disease development, release chromatin-containing neutrophil extracellular traps (NETs), potentially functioning as a source of nuclear auto-antigen, and may activate B cells in a T cell independent fashion. In contrast, levels and functions of regulatory neutrophils (Nregs) involved in T celldependent B cell differentiation and germinal center reactions, are dysregulated in female lupus-prone mice during disease development. Here we review data supporting a role for both pro- and anti-inflammatory neutrophils in lupus.

Keywords: Systemic lupus erythematosus; Autoantibody; Neutrophils; Pro-inflammatory; Immunosuppression

Abbreviations

ANA: Anti-Nuclear Antibodies; APRIL: A Proliferation Inducing Ligand; BAFF: B cell Activation Factor; BM: Bone Marrow; CNS: Central Nervous System; GC: Germinal Center; ds: double stranded; IC: Immune Complex; IL: Interleukin; IFN: Interferon; LDG: Low Density Granulocyte; NET: Neutrophil Extracellular Trap; Nreg: Regulatory Neutrophil; SLE: Systemic Lupus Erythematosus; TFH: T Follicular Helper Cell

Introduction

Systemic Lupus Erythematosus (SLE) is characterized by elevated levels of antinuclear antibodies (ANA) and circulating immune complexes (IC) known to deposit in organs such as the skin, kidney, heart, and lung, promoting mononuclear cell infiltration and tissue damage. The development of SLE is attributed both environmental triggers and genetic predisposition, and presents with an overwhelming female bias [1]. Since pathogenic autoantibodies characterize the disorder, decades of research have focused on the function and dysregulation of autoreactive B cells. However, since the discovery of elevated levels of neutrophil-associated gene transcripts in peripheral blood mononuclear cell samples from SLE patients as compared with healthy controls [2], an increasing number of studies have investigated the role of neutrophils in the pathogenesis of SLE.

Neutrophils are the most abundant circulating leukocyte. The cells are short-lived, granule-rich and constitute the primary defense against microbial and fungal infections [3]. During infections, circulating neutrophils are recruited to the site of infection by chemotactic cytokines. The classic effector functions of neutrophils include phagocytosis and the release of antimicrobial proteins, reactive oxygen species (ROS), metalloproteinases (MMPs) and other endopeptidases such as trypsin and neutrophil elastase [4]. While the functions of these effector mechanisms are detrimental to pathogens, these enzymes may also harm the host resulting in tissue damage and necrosis. More recent evidence suggests that neutrophils can also function as antigen presenting cells [5,6], as a source of autoantigen and type I interferon (IFNα) in autoimmunity [7-10], or even as negative regulators of T and B cell responses during systemic inflammation [11-14]. In this review we discuss evidence from human and mouse studies supporting a key role for pro- and anti-inflammatory neutrophils in lupus pathogenesis.

Pro-inflammatory Neutrophils in Accumulate in SLE Patients and Mice with Lupus-like Disease

Despite chronic neutropenia and hyper-susceptibility to bacterial infections [15-17], a significant proportion of SLE patients display elevated levels of immature neutrophils and a pronounced granulopoiesis gene expression signature in cells from both peripheral blood and bone marrow exudates [2,18,19]. In addition, neutrophils are known to infiltrate target organs such as the skin, kidney or vasculature in SLE patients [20-22], although the pathogenic nature of such infiltration remains unknown. The population of neutrophil-like cells accumulating in SLE patients can be divided into at least two subpopulations based on their granularity: high density (mature granulocytic) neutrophils and low density (immature monocytic) granulocytes (LDGs) [2,22]. The functional capacities of both cell subsets are dysregulated in SLE patients however it is not yet clear whether such defects are in themselves causative in disease development [7,8].

Likewise, in virtually all murine models of SLE, myeloid lineage cells accumulate in the circulation, spleen and/or target organs (CNS, kidneys, lungs and vasculature) as disease progresses [23-26]. The appearance of elevated numbers of CD11b+ cells most often correlates with the development of other disease markers such as ANA production and/or kidney infiltration, and thus, these cells are generally believed to exert pro-inflammatory functions. CD11b expression is shared among many cell subsets and additional surface markers along with nuclear morphology analyses are commonly used to define neutrophil populations. Thus, accumulating neutrophil-like cells co-express the surface marker Ly6C and show a monocytic nuclear morphology in female autoimmune (NZB x NZW)F1 mice [27,28], while the dominant population of neutrophil-like cells in protected male (NZB x NZW)F1 mice express Ly6G and a multilobe nuclear morphology [28] (please see below for more details). In MRL/lpr lupus-prone mice and pristane-induced lupus models both Ly6C+ and Ly6G+ cell populations accumulate [23,29,30], while neither of these two markers are present on accumulating myeloid cells in BXSB mice [31-33].

Neutrophil-associated Effector Functions are Abnormal in SLE Patients and Mouse Models of Lupus

Phagocytosis

Both adult and juvenile-onset SLE patients and patients with chronic granulomatous disease are known to have an increased risk of developing lupus, present with elevated levels of apoptotic neutrophils [34-38]. Under non-inflammatory conditions, neutrophils have a short life span ending with spontaneous apoptosis and subsequent removal by phagocytes [39]. During inflammation, however, neutrophils are kept alive to help fight off the infection. It is therefore conceivable that apoptotic neutrophils accumulate in SLE patients, not as a result of increased apoptosis, but rather, due to impaired clearance of apoptotic bodies by professional phagocytes including macrophages and neutrophils themselves. Defective phagocytosis may stem from SLE-associated gene polymorphisms affecting the expression and/or function of Fc-receptors (FcgRIIa, FcgRIIb and FcgRIII) or complement receptor 3 (CR3; CD11b/CD18) [40-42], all of which are involved in macrophage and neutrophil-dependent phagocytosis.

Production of reactive oxygen and nitrogen species

A major effector mechanism of neutrophils is their ability to produce and release antimicrobial products and reactive oxygen species (ROS) upon encounter of pathogens. Correlating with the accumulation of neutrophils, patients with active SLE display elevated production of ROS by circulating neutrophils and increased levels of oxidative damage in kidney biopsies [43-46]. Abnormal oxidative protein modifications have also been associated with disease development in MRL/lpr lupus-prone mice and increased immunity in rabbits [47,48]. Increased ROS production could be a result of accumulating neutrophils as described above, or a consequence of decreased expression or inhibited function of superoxide dismutase (SOD1). SOD1 is a critical antioxidant involved in the consumption of free oxygen radicals that has been shown to be significantly reduced in SLE patients with active disease [49,50]. Alternatively, a polymorphism in the gene neutrophil cytosolic factor 2 (NCF2) encoding for p67phox, a critical component of the NADPH oxidase and thus involved in the production of ROS, has been found in several cohorts of SLE patients [51,52].

Cytokine production

While the mechanisms of tissue damage are not completely characterized, cytokine and chemokine production by inflammatory myeloid cells have been observed in both SLE patients and several of the murine models of lupus. As such, many cytokines have been associated with the initiation and progression of lupus including interleukin-6 (IL-6), IL-10, IL-17, IL-18, IL-21, tumor necrosis factor-alpha (TNFα) and interferon-alpha (IFNα) [53,54].

IFNα has received significant attention as PBMCs from SLE patients often express elevated levels of IFNα-induced gene transcripts [2,55,56]. IFNα is induced in response to a broad range of signals, but most efficiently in response to intracellular toll-like receptors: TLR3, TLR7, TLR8 and TLR9 [57]. Interestingly, chronic TLR7-stimulation was recently shown to induce myelopoiesis leading to monocyte and neutrophil accumulation in an IFNα-dependent manner [58]. The most potent IFNα-producing cell type is the plasmacytoid dendritic cell [59]; however neutrophils can produce IFNα under chronic inflammatory conditions as well [22,60]. This is true in humans, as well as in mice, and has recently been reviewed elsewhere [61].

B cell manipulating factors such as IL-6, IL-21, BAFF and APRIL, can all be produced by neutrophils and have been associated with lupus pathogenesis. IL-6 is a powerful regulator of B cell differentiation and antibody production and manipulation of IL-6 levels affect lupus pathogenesis in both female (NZB x NZW)F1 mice and pristane-induced lupus [62-64]. IL-21 is required for functional T follicular helper (TFH) cell differentiation and the formation of germinal centers (GC), and can be produced by neutrophils during chronic inflammation. In SLE, such IL-21 may also lead to T-cell independent B cell activation and differentiation [11]. Interestingly, these cells also produced A Proliferation Inducing Ligand (APRIL), crucial for plasma cell survival. Further evidence for communications between neutrophils and B cells comes from studies showing that tissue infiltrating, G-CSF-triggered neutrophils produce and secrete BAFF under inflammatory conditions [65], while BM neutrophils from SLE patients produce both APRIL and BAFF supporting early B cell development and plasma cell survival [66].

Finally, in two recent studies of pristane-induced lupus, bone-marrow derived neutrophils (Ly6GhiLy6Clow) were reported to produce significant levels of TNFα and IL-17A, resulting in myelopoiesis and the accumulation of neutrophil-like cells [67,68]. Interestingly, TNFα have been associated with many neutrophil effector functions including ROS production, while IL-17A is emerging as a key cytokine controlling both T and B cell dependent immune responses [69,70].

NETosis

Neutrophils are known to release extracellular traps, known as neutrophil extracellular traps or NETs, as a mechanism to immobilize pathogens [4]. This process leads to the subsequent death of the neutrophil; a process known as NETosis [71]. From a lupus perspective, NETs offer an enticing source of nuclear antigen, as these structures consist of strands of elastase covered with antimicrobial proteins, such as MPO and LL37, alongside histones and free DNA [4,21] and excellently reviewed in [61,72]. In support hereof, recent data show that LL37/DNA-containing NETs effectively activate plasmacytoid DCs via TLR9 cross linking resulting in IFNα production [7]. Interestingly, the production of NETs by neutrophils can also be induced by many inflammatory signals including IFNα and IC [7,8]. Finally, it has been shown that low density granulocytes (LDGs) from SLE patients spontaneously undergo NETosis in vitro [21,60], while NETs have been observed in cultures of Ly6ChighCD11b+ neutrophils from pristane-treated lupus mice [73], further supporting a role for NETs in lupus pathogenesis.

In addition to the spontaneous and chronic generation of NETs, dysregulated removal of NETs may play a role in lupus pathogenesis [74]. Removal of NETs relies on DNaseI activity, and impaired DNaseI activity has been associated with kidney damage in lupus nephritis patients and mouse models of SLE [74-76]. Moreover, the presence of DNaseI inhibitors and anti-NET antibodies preventing DNaseI from binding and degrading the NETs, have been identified in serum samples from SLE patients [74], further adding to the pathogenicity of NETs.

Anti-inflammatory Properties of Neutrophils in Lupus

The biological importance of immunosuppressive tumor-infiltrating Ly6G+ and Ly6C+ myeloid cells was spearheaded by the identification of ROS-dependent T cell suppression in cancer patients [77] and numerous studies have since then explored the tumor-promoting function of these cells in both mouse models and cancer patients [78]. Recently, immunosuppressive functions of neutrophils have been described in several studies of systemic inflammation and autoimmunity including sepsis and SLE [11,12]. Still, the mechanism(s) of suppression utilized by these cells remain to be identified.

We recently reported the presence and function of populations of regulatory Ly6G+Gr1highCD11b+ and Ly6ChighGr1lowCD11b+ neutrophils in the (NZB x NZW)F1 mouse model of SLE [13,28]. Since, lupus presents with an overwhelming female bias, we hypothesized that such regulatory neutrophils could be a mechanism preventing disease in otherwise genetically predisposed males. Consistent with a sex-dependent immunosuppressive function, we found that male (NZB x NZW)F1 mice express increased numbers of Gr1+ cells throughout life [28] (Table 1). In young <9 week old lupus-prone male, as well as female, (NZB x NZW)F1 mice, Ly6G+Gr1highCD11b+ and Ly6ChighGr1lowCD11b+ neutrophils were found to be immunosuppressive towards B and T cells, respectively. As female mice aged, however, the cells lost their immunosuppressive capability both in vitro and in vivo [13,28]. A similar loss of function was also observed in males, albeit not until much later in life. Interestingly, over time Ly6ChighGr1lowCD11b+ neutrophils became immunostimulatory to both T and B cells, resembling the function of pro-inflammatory LDGs as described above.

  Gr1highCD11b+ cells Gr1lowCD11b+ cells
Expression levels (spleen) Elevated in males 4-26 weeks of age Elevated in females (>9 wks)
Immunosuppressive in vitro Yes (females only <16 wk) Yes (females only <9 wk)
Cellular target B cell differentiation T cell differentiation and proliferation
Cell-Cell contact No (males); Yes (females) ns*
Effector mechanism Unknown (males); ROS/NO (females) ns*
Immunostimulatory in vitro No Yes (female ≥ 9wk old; males >16 wk old)
Cellular target n/a B cell differentiation and T cell proliferation
  All Gr1+ cells
Immunosuppressive in vivo Yes (males) No (females ≥ 9 wks of age)
Cellular target GC reaction via control of TFH cell differentiation (males) n/a
*ns: not studied; n/a: not applicable [13,28]

Table 1: Neutrophil-like cells differ in numbers and functions between protected males and disease-prone females in (NZB x NZW)F1 lupus mice.

In addition to the temporal difference in immunosuppression between male and female-derived Nregs, our studies also showed that Ly6G+Gr1highCD11b+ and Ly6ChighGr1lowCD11b+ neutrophils not only targeted different lymphoid cell populations in vitro, but also utilized different immunosuppressive mechanisms [13,28]. For example, female Gr1highCD11b+ cells depended on cell-cell contact and utilized ROS/NO as their mechanism of inhibition in B cell differentiation assays, while male cells exerted their suppressive capacity via an unknown secreted, soluble factor ([28] and unpublished results). These data are consistent with a study showing that immunosuppressive Gr1-CD11b+ myeloid cells from MRL/lpr mice suppress T cell proliferation in an arginase-1-dependent manner [79], and the observation that MRL/lpr mice defective in ROS-production display an accelerated disease profile [80].

Several lines of evidence suggest that the observed immunosuppressive function in male lupus-prone mice is reminiscent with the normal behavior of such cells. For example, studies in non-autoimmune mice have shown that neutrophils can limit immune reactions in response to inflammation or immunization ([81,82] and our unpublished observation). In addition, complement factor 4 produced by myeloid cells may regulate spontaneous GC reactions in self-reactive B cell receptor transgenic mice [14]. We therefore suggest that the chronic inflammatory milieu developing in female (NZB x NZW)F1 mice affect a population of regulatory neutrophils (Nregs) either via the induction of apoptosis (or maybe even NETosis) [4,83] or via the induction of a differentiation program driving the development of non-immunosuppressive cells such as dendritic cells, macrophages or mature neutrophils, as previously suggested [83-85].

Summary

There is still much to learn about pro- and anti-inflammatory neutrophils, their effector functions and role in the pathogenesis of SLE. We here suggest a model in which immunosuppressive neutrophil-like cells (Nregs) constitute a normal regulatory component involved in the control of adaptive immune responses (Figure 1). We suggest that the normal function of Nregs is to control TFH cell differentiation and GC reactions either directly via the production of immunomodulatory cytokines affecting differentiation of T and B cells or via the production of chemokines affecting the assembly of GCs during T-dependent antibody responses. During systemic inflammation (including infections, cancer, and SLE) Nregs are targeted to either die, hereby promoting access to nuclear autoantigens, or differentiate into pro-inflammatory dendritic cells, macrophages and mature neutrophils driving T and B cell activation and eventually autoantibody production. Future studies identifying factors targeting and changing the function of Nregs during chronic inflammation (including SLE) may provide additional targets for early intervention therapy in lupus-susceptible individuals.

clinical-cellular-immunology-Neutrophils-Lupus

Figure 1: Neutrophils in Lupus. Model suggesting that a subset of neutrophils (Nregs) possess regulatory abilities involved in the control of humoral immunity. Under non-inflammatory conditions, such Nregs are located to the secondary lymphoid organs where they regulate T and B cell responses to foreign antigen. During chronic inflammation (Lupus), neutrophils display pro-inflammatory abilities including the production of IFNα and secretion of NETs by LDGs, and the production of reactive oxygen and nitrogen species involved in tissue damage by mature neutrophils. Signals driving the generation of pro-inflammatory neutrophils may include estrogen, IC and cytokines, all of which are associated with the development of lupus in genetically predisposed females.

References

  1. Kotzin BL, West SG (2001) Systemic Lupus Erythrematosus. In: Rich R, Shearer WT, Kotzin BL, Schroeder Jr HW (Eds), Clinical Immunology: Principles and Practice. Mosby International Limited.
  2. Bennett L, Palucka AK, Arce E, Cantrell V, Borvak J, et al. (2003) Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med 197: 711-723.
  3. Nathan C (2006) Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol 6: 173-182.
  4. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, et al. (2004) Neutrophil extracellular traps kill bacteria. Science 303: 1532-1535.
  5. Beauvillain C, Delneste Y, Scotet M, Peres A, Gascan H, et al. (2007) Neutrophils efficiently cross-prime naive T cells in vivo. Blood 110: 2965-2973.
  6. Abi Abdallah DS, Egan CE, Butcher BA, Denkers EY (2011) Mouse neutrophils are professional antigen-presenting cells programmed to instruct Th1 and Th17 T-cell differentiation. Int Immunol 23: 317-326.
  7. Lande R, Ganguly D, Facchinetti V, Frasca L, Conrad C, et al. (2011) Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus. Sci Transl Med 3: 73ra19.
  8. Garcia-Romo GS1, Caielli S, Vega B, Connolly J, Allantaz F, et al. (2011) Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci Transl Med 3: 73ra20.
  9. Diana J, Simoni Y, Furio L, Beaudoin L, Agerberth B, et al. (2013) Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes. Nat Med 19: 65-73.
  10. Khandpur R, Carmona-Rivera C, Vivekanandan-Giri A, Gizinski A, Yalavarthi S, et al. (2013) NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci Transl Med 5: 178ra40.
  11. Puga I, Cols M, Barra CM, He B, Cassis L, et al. (2011) B cell-helper neutrophils stimulate the diversification and production of immunoglobulin in the marginal zone of the spleen. Nat Immunol 13: 170-180.
  12. Pillay J1, Tak T, Kamp VM, Koenderman L (2013) Immune suppression by neutrophils and granulocytic myeloid-derived suppressor cells: similarities and differences. Cell Mol Life Sci 70: 3813-3827.
  13. Der E, Dimo J, Trigunaite A, Jones J, Jørgensen TN (2014) Gr1+ cells suppress T-dependent antibody responses in (NZB x NZW)F1 male mice through inhibition of T follicular helper cells and germinal center formation. J Immunol 192: 1570-1576.
  14. Chatterjee P, Agyemang AF, Alimzhanov MB, Degn S, Tsiftsoglou SA, et al. (2013) Complement C4 maintains peripheral B-cell tolerance in a myeloid cell dependent manner. Eur J Immunol 43: 2441-2450.
  15. Yamasaki K, Niho Y, Yanase T (1983) Granulopoiesis in systemic lupus erythematosus. Arthritis Rheum 26: 516-521.
  16. López-Karpovitch X, Cardiel M, Cardenas R, Piedras J, Alarcón-Segovia D (1989) Circulating colony-forming units of granulocytes and monocytes/macrophages in systemic lupus erythematosus. Clin Exp Immunol 77: 43-46.
  17. Martínez-Baños D, Crispín JC, Lazo-Langner A, Sánchez-Guerrero J (2006) Moderate and severe neutropenia in patients with systemic lupus erythematosus. Rheumatology (Oxford) 45: 994-998.
  18. Nakou M, Knowlton N, Frank MB, Bertsias G, Osban J, et al. (2008) Gene expression in systemic lupus erythematosus: bone marrow analysis differentiates active from inactive disease and reveals apoptosis and granulopoiesis signatures. Arthritis Rheum 58: 3541-3549.
  19. Umiker BR, Andersson S, Fernandez L, Korgaokar P, Larbi A, et al. (2014) Dosage of X-linked Toll-like receptor 8 determines gender differences in the development of systemic lupus erythematosus. Eur J Immunol 44: 1503-1516.
  20. Peterson KS, Huang JF, Zhu J, D'Agati V, Liu X, et al. (2004) Characterization of heterogeneity in the molecular pathogenesis of lupus nephritis from transcriptional profiles of laser-captured glomeruli. J Clin Invest 113: 1722-1733.
  21. Villanueva E, Yalavarthi S, Berthier CC, Hodgin JB, Khandpur R, et al. (2011) Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immunostimulatory molecules in systemic lupus erythematosus. J Immunol 187: 538-552.
  22. Denny MF, Yalavarthi S, Zhao W, Thacker SG, Anderson M, et al. (2010) A distinct subset of proinflammatory neutrophils isolated from patients with systemic lupus erythematosus induces vascular damage and synthesizes type I IFNs. J Immunol 184: 3284-3297.
  23. James WG, Hutchinson P, Bullard DC, Hickey MJ (2006) Cerebral leucocyte infiltration in lupus-prone MRL/MpJ-fas lpr mice--roles of intercellular adhesion molecule-1 and P-selectin. Clin Exp Immunol 144: 299-308.
  24. Menke J, Rabacal WA, Byrne KT, Iwata Y, Schwartz MM, et al. (2009) Circulating CSF-1 promotes monocyte and macrophage phenotypes that enhance lupus nephritis. J Am Soc Nephrol 20: 2581-2592.
  25. Chowdhary VR, Grande JP, Luthra HS, David CS (2007) Characterization of haemorrhagic pulmonary capillaritis: another manifestation of Pristane-induced lupus. Rheumatology (Oxford) 46: 1405-1410.
  26. Hayashi T, Hasegawa K, Ichinohe N (2005) ICAM-1 expression on endothelium and systemic cytokine production in cutaneous neutrophilic leukocytoclastic vasculitis in NZBxNZWF1 mice. Histol Histopathol 20: 45-52.
  27. Bethunaickan R, Berthier CC, Ramanujam M, Sahu R, Zhang W, et al. (2011) A unique hybrid renal mononuclear phagocyte activation phenotype in murine systemic lupus erythematosus nephritis. J Immunol 186: 4994-5003.
  28. Trigunaite A, Khan A, Der E, Song A, Varikuti S, et al. (2013) Gr-1(high) CD11b+ cells suppress B cell differentiation and lupus-like disease in lupus-prone male mice. Arthritis Rheum 65: 2392-2402.
  29. Brown NJ, Hutcheson J, Bickel E, Scatizzi JC, Albee LD, et al. (2004) Fas death receptor signaling represses monocyte numbers and macrophage activation in vivo. J Immunol 173: 7584-7593.
  30. Xu Y, Lee PY, Li Y, Liu C, Zhuang H, et al. (2012) Pleiotropic IFN-dependent and -independent effects of IRF5 on the pathogenesis of experimental lupus. J Immunol 188: 4113-4121.
  31. Wofsy D, Kerger CE, Seaman WE (1984) Monocytosis in the BXSB model for systemic lupus erythematosus. J Exp Med 159: 629-634.
  32. Vieten G, Hadam MR, De Boer H, Olp A, Fricke M, et al. (1992) Expanded macrophage precursor populations in BXSB mice: possible reason for the increasing monocytosis in male mice. Clin Immunol Immunopathol 65: 212-218.
  33. Santiago-Raber ML, Amano H, Amano E, Baudino L, Otani M, et al. (2009) Fcgamma receptor-dependent expansion of a hyperactive monocyte subset in lupus-prone mice. Arthritis Rheum 60: 2408-2417.
  34. Courtney PA, Crockard AD, Williamson K, Irvine AE, Kennedy RJ, et 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: 309-314.
  35. Su YJ, Cheng TT, Chen CJ, Chiu WC, Hsu CY, et al. (2013) The association among leukocyte apoptosis, autoantibodies and disease severity in systemic lupus erythematosus. J Transl Med 11: 261.
  36. Ren Y, Tang J, Mok MY, Chan AW, Wu A, et al. (2003) Increased apoptotic neutrophils and macrophages and impaired macrophage phagocytic clearance of apoptotic neutrophils in systemic lupus erythematosus. Arthritis Rheum 48: 2888-2897.
  37. Midgley A, McLaren Z, Moots RJ, Edwards SW, Beresford MW (2009) The role of neutrophil apoptosis in juvenile-onset systemic lupus erythematosus. Arthritis Rheum 60: 2390-2401.
  38. Sanford AN, Suriano AR, Herche D, Dietzmann K, Sullivan KE (2006) Abnormal apoptosis in chronic granulomatous disease and autoantibody production characteristic of lupus. Rheumatology (Oxford) 45: 178-181.
  39. Geering B, Simon HU (2011) Peculiarities of cell death mechanisms in neutrophils. Cell Death Differ 18: 1457-1469.
  40. Rhodes B, Fürnrohr BG, Roberts AL, Tzircotis G, Schett G, et al. (2012) The rs1143679 (R77H) lupus associated variant of ITGAM (CD11b) impairs complement receptor 3 mediated functions in human monocytes. Ann Rheum Dis 71: 2028-2034.
  41. MacPherson M, Lek HS, Prescott A, Fagerholm SC (2011) A systemic lupus erythematosus-associated R77H substitution in the CD11b chain of the Mac-1 integrin compromises leukocyte adhesion and phagocytosis. J Biol Chem 286: 17303-17310.
  42. Fossati-Jimack L, Ling GS, Cortini A, Szajna M, Malik TH, et al. (2013) Phagocytosis is the main CR3-mediated function affected by the lupus-associated variant of CD11b in human myeloid cells. PLoS One 8: e57082.
  43. Perazzio SF, Salomão R, Silva NP, Andrade LE (2012) Increased neutrophil oxidative burst metabolism in systemic lupus erythematosus. Lupus 21: 1543-1551.
  44. Moroni G, Novembrino C, Quaglini S, De Giuseppe R, Gallelli B, et al. (2010) Oxidative stress and homocysteine metabolism in patients with lupus nephritis. Lupus 19: 65-72.
  45. Alves CM, Marzocchi-Machado CM, Louzada-Junior P, Azzolini AE, Polizello AC, et al. (2008) Superoxide anion production by neutrophils is associated with prevalent clinical manifestations in systemic lupus erythematosus. Clin Rheumatol 27: 701-708.
  46. Jiang T, Tian F, Zheng H, Whitman SA, Lin Y, et al. (2014) Nrf2 suppresses lupus nephritis through inhibition of oxidative injury and the NF-κB-mediated inflammatory response. Kidney Int 85: 333-343.
  47. Wang G, Li H, Firoze KM (2012) Differential oxidative modification of proteins in MRL+/+ and MRL/lpr mice: Increased formation of lipid peroxidation-derived aldehyde-protein adducts may contribute to accelerated onset of autoimmune response. Free Radic Res 46: 1472-1481.
  48. Scofield RH, Kurien BT, Ganick S, McClain MT, Pye Q, et al. (2005) Modification of lupus-associated 60-kDa Ro protein with the lipid oxidation product 4-hydroxy-2-nonenal increases antigenicity and facilitates epitope spreading. Free Radic Biol Med 38: 719-728.
  49. Kurien BT, Scofield RH (2003) Free radical mediated peroxidative damage in systemic lupus erythematosus. Life Sci 73: 1655-1666.
  50. Wang G, Pierangeli SS, Papalardo E, Ansari GA, Khan MF (2010) Markers of oxidative and nitrosative stress in systemic lupus erythematosus: correlation with disease activity. Arthritis Rheum 62: 2064-2072.
  51. Kim-Howard X, Sun C, Molineros JE, Maiti AK, Chandru H, et al. (2014) Allelic heterogeneity in NCF2 associated with systemic lupus erythematosus (SLE) susceptibility across four ethnic populations. Hum Mol Genet 23: 1656-1668.
  52. Yu B, Chen Y, Wu Q, Li P, Shao Y, et al. (2011) The association between single-nucleotide polymorphisms of NCF2 and systemic lupus erythematosus in Chinese mainland population. Clin Rheumatol 30: 521-527.
  53. Clark DN, Markham JL, Sloan CS, Poole BD (2013) Cytokine inhibition as a strategy for treating systemic lupus erythematosus. Clin Immunol 148: 335-343.
  54. Yap DY, Lai KN (2013) The role of cytokines in the pathogenesis of systemic lupus erythematosus - from bench to bedside. Nephrology (Carlton) 18: 243-255.
  55. Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, et al. (2003) Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc Natl Acad Sci U S A 100: 2610-2615.
  56. Kirou KA, Lee C, George S, Louca K, Peterson MG, et al. (2005) Activation of the interferon-alpha pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease. Arthritis Rheum 52: 1491-1503.
  57. Takeda K, Kaisho T, Akira S (2003) Toll-like receptors. Annu Rev Immunol 21: 335-376.
  58. Buechler MB, Teal TH, Elkon KB, Hamerman JA (2013) Cutting edge: Type I IFN drives emergency myelopoiesis and peripheral myeloid expansion during chronic TLR7 signaling. J Immunol 190: 886-891.
  59. Siegal FP, Kadowaki N, Shodell M, Fitzgerald-Bocarsly PA, Shah K, et al. (1999) The nature of the principal type 1 interferon-producing cells in human blood. Science 284: 1835-1837.
  60. Lindau D, Mussard J, Rabsteyn A, Ribon M, Kötter I, et al. (2013) TLR9 independent interferon a production by neutrophils on NETosis in response to circulating chromatin, a key lupus autoantigen. Ann Rheum Dis .
  61. Carmona-Rivera C1, Kaplan MJ (2013) Low-density granulocytes: a distinct class of neutrophils in systemic autoimmunity. Semin Immunopathol 35: 455-463.
  62. Finck BK, Chan B, Wofsy D (1994) Interleukin 6 promotes murine lupus in NZB/NZW F1 mice. J Clin Invest 94: 585-591.
  63. Richards HB, Satoh M, Shaw M, Libert C, Poli V, et al. (1998) Interleukin 6 dependence of anti-DNA antibody production: evidence for two pathways of autoantibody formation in pristane-induced lupus. J Exp Med 188: 985-990.
  64. Ryffel B, Car BD, Gunn H, Roman D, Hiestand P, et al. (1994) Interleukin-6 exacerbates glomerulonephritis in (NZB x NZW)F1 mice. Am J Pathol 144: 927-937.
  65. Scapini P, Carletto A, Nardelli B, Calzetti F, Roschke V, et al. (2005) Proinflammatory mediators elicit secretion of the intracellular B-lymphocyte stimulator pool (BLyS) that is stored in activated neutrophils: implications for inflammatory diseases. Blood 105: 830-837.
  66. Palanichamy A, Bauer JW, Yalavarthi S, Meednu N, Barnard J, et al. (2014) Neutrophil-mediated IFN activation in the bone marrow alters B cell development in human and murine systemic lupus erythematosus. J Immunol 192: 906-918.
  67. Zhuang H, Han S, Xu Y, Li Y, Wang H, et al. (2014) Toll-like receptor 7-stimulated tumor necrosis factor a causes bone marrow damage in systemic lupus erythematosus. Arthritis Rheumatol 66: 140-151.
  68. Summers SA, Odobasic D, Khouri MB, Steinmetz OM, Yang Y, et al. (2014) Endogenous interleukin (IL)-17A promotes pristane-induced systemic autoimmunity and lupus nephritis induced by pristane. Clin Exp Immunol 176: 341-350.
  69. Tanaka H, Ogura H, Yokota J, Sugimoto H, Yoshioka T, et al. (1991) Acceleration of superoxide production from leukocytes in trauma patients. Ann Surg 214: 187-192.
  70. Ding Y, Li J, Wu Q, Yang P, Luo B, et al. (2013) IL-17RA is essential for optimal localization of follicular Th cells in the germinal center light zone to promote autoantibody-producing B cells. J Immunol 191: 1614-1624.
  71. Kaplan MJ, Radic M (2012) Neutrophil extracellular traps: double-edged swords of innate immunity. J Immunol 189: 2689-2695.
  72. Yu Y, Su K (2013) Neutrophil Extracellular Traps and Systemic Lupus Erythematosus. J Clin Cell Immunol 4.
  73. Herman S, Kny A, Schorn C, Pfatschbacher J, Niederreiter B, et al. (2012) Cell death and cytokine production induced by autoimmunogenic hydrocarbon oils. Autoimmunity 45: 602-611.
  74. Hakkim A, Fürnrohr BG, Amann K, Laube B, Abed UA, et al. (2010) Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc Natl Acad Sci U S A 107: 9813-9818.
  75. Leffler J, Martin M, Gullstrand B, Tydén H, Lood C, et al. (2012) Neutrophil extracellular traps that are not degraded in systemic lupus erythematosus activate complement exacerbating the disease. J Immunol 188: 3522-3531.
  76. 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.
  77. Schmielau J, Finn OJ (2001) Activated granulocytes and granulocyte-derived hydrogen peroxide are the underlying mechanism of suppression of t-cell function in advanced cancer patients. Cancer Res 61: 4756-4760.
  78. Gabrilovich DI, Nagaraj S (2009) Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 9: 162-174.
  79. Iwata Y, Furuichi K, Kitagawa K, Hara A, Okumura T, et al. (2010) Involvement of CD11b+ GR-1 low cells in autoimmune disorder in MRL-Fas lpr mouse. Clin Exp Nephrol 14: 411-417.
  80. Campbell AM, Kashgarian M, Shlomchik MJ (2012) NADPH oxidase inhibits the pathogenesis of systemic lupus erythematosus. Sci Transl Med 4: 157ra141.
  81. Delano MJ, Scumpia PO, Weinstein JS, Coco D, Nagaraj S, et al. (2007) MyD88-dependent expansion of an immature GR-1(+)CD11b(+) population induces T cell suppression and Th2 polarization in sepsis. J Exp Med 204: 1463-1474.
  82. Yang CW, Strong BS, Miller MJ, Unanue ER (2010) Neutrophils influence the level of antigen presentation during the immune response to protein antigens in adjuvants. J Immunol 185: 2927-2934.
  83. Sander LE, Sackett SD, Dierssen U, Beraza N, Linke RP, et al. (2010) Hepatic acute-phase proteins control innate immune responses during infection by promoting myeloid-derived suppressor cell function. J Exp Med 207: 1453-1464.
  84. Sade-Feldman M, Kanterman J, Ish-Shalom E, Elnekave M, Horwitz E, et al. (2013) Tumor necrosis factor-a blocks differentiation and enhances suppressive activity of immature myeloid cells during chronic inflammation. Immunity 38: 541-554.
  85. Shirota Y, Shirota H, Klinman DM (2012) Intratumoral injection of CpG oligonucleotides induces the differentiation and reduces the immunosuppressive activity of myeloid-derived suppressor cells. J Immunol 188: 1592-1599.
Citation: Der E, Trigunaite A, Khan A, Jørgensen TN (2014) Pro- and Anti-inflammatory Neutrophils in Lupus. J Clin Cell Immunol 5:239.

Copyright: © 2014 Der E, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top