Neutrophils (Polymorphonuclear Leukocytes) in Systemic Lupus Erythematosus

Chapter 23 Neutrophils (Polymorphonuclear Leukocytes) in Systemic Lupus Erythematosus





LE CELLS: A NEUTROPHIL-MEDIATED PHENOMENON


The discovery of the lupus erythematosus (LE) cell phenomenon by Hangraves and colleagues in 19481 was a pivotal milestone not only from a diagnostic aspect but from the standpoint of its pathophysiologic significance. As recent studies show, the LE cell represents a hallmark stage in the initiation and perpetuation of the autoimmune process that underlies the role of apoptosis (programmed cell death) in the pathogenesis of systemic lupus erythematosus (SLE). It has recently been shown that the LE cell results from phagocytosis of apoptotic bodies by neutrophils [polymorphonuclear cells (PMN)] induced by anti-double-stranded (ds) DNA autoantibodies.1 Studies using flow cytometric analysis (FACS) of LE cells demonstrated the neutrophil as the phagocyte that engulfs apoptotic cells.1 Anti-dsDNA antibodies are a serologic hallmark as well as a crucial factor in the pathogenesis of immune complex–mediated tissue damage in SLE.1


The mechanism suggested to be involved in the phagocytic uptake of nuclei from apoptotic cells by neutrophils is induced by anti-dsDNA antibody-mediated inhibition of enzymatic cleavage of the nuclei.1 The elucidation of the mechanism of production of the LE cells sheds light on the relevance and possible active role neutrophils play in the relationship between innate immune reaction presented by neutrophils and the specific immune response presented by antinuclear antibodies that underlie the pathogenesis of SLE.



NEUTROPHILS: THE ROLE OF INNATE IMMUNITY IN THE PATHOGENESIS OF SLE


Neutrophils (PMNs) are a key component of the primary host defense line against noxious pathogens (together with macrophages and dendritic cells). This host defense line comprises the cellular arm of the innate immunity. Impaired clearance of apoptotic cells has been suggested as a crucial factor in the pathogenesis of SLE.1 To succeed with their physiologic reactions (such as chemotaxis, degranulation and phagocytosis) neutrophils become activated by various stimuli, including bacterial products, complement split products (such as C5a), immune complexes (ICs), chemokines, and cytokines [such as interleukin (IL)-1 and −8]. PMN activation is initiated upon recognition of antibody- or complement-opsonized particles as well as directly by microbial products via the toll-like receptors.7,8 Table 23.1 summarizes the main neutrophil surface receptors and their expression regulation in SLE. Neutrophils constitutively express surface complement receptors (CR), CR1 (CD35), and CR3 (β2-integrin, CD11b/CD18, Mac-1), which recognize C3b/iC3b split products of the complement system.


TABLE 23.1 NEUTROPHIL SURFACE RECEPTORS AND THEIR EXPRESSION IN SLE















































































Surface Receptor Counter Ligand Surface Expression in SLE
Complement receptor-1 (CD35) C1q, C3b, iC3b, mannose binding lectin Normal
Complement receptor-3 (CD11b/CD18) ICb3, intercellular adhesion molecule-1 (ICAM-1), E-selectin, junctional adhesion molecule C (JAM-C), fibrinogen, heparin, elastase, neutrophil inhibitory factor, complement factor-H, glycoprotein Iba, urokinase receptor (uPAR), laminin, collagen, vitronectin, connective tissue growth factor Increased
L-selectin (CD62L) Sialomucins (CD34 family) Decreased
Fc gamma receptor I (CD64) Immunoglobulins, immunocomplexes NPD
Fc gamma receptor II (CD16) Immunoglobulins, immunocomplexes NPD
Fc gamma receptor III (CD32) Immunoglobulins, immunocomplexes NPD
C5a receptor C5a Normal
Interleukin-8 receptor Interleukin-8 NPD
Interleukin-1 receptor Interleukin-1 NPD
Interferon I receptor Interferons NPD
Tumor necrosis factor-a-receptor (TNFa) Tumor necrosis factor-α NPD
TNFa-related apoptosis—inducing ligand (TRAIL) receptor-3 TNF α-related apoptosis-inducing ligand (TRAIL) Normal/decreased in patients with neutropenia
Glycoprotein Fas (CD95) Fas-ligand Increased
Lipopolysaccharide receptor (CD14, toll-like receptor-4) Lipopolysaccharide NPD
Toll-like receptor-9 CpG motifs of bacterilal DNA NPD
Granulocyte-colony stimulating factor (G-CSF) G-CSF NPD
Granulocyte and macrophage-CSF GM-CSF NPD
Glycoprotein receptor (CD44) Hyaluronan, fibronectin, collagen, fibrin Decreased

NPD, no published data


IC are abundantly present in the serum as well as deposited in target organs affected in SLE. Neutrophil interaction with IC is mediated through binding to CR and Fcγ receptors (FcγR) present on these cells. Neutrophil activation in patients with SLE is mediated by IC binding to FcγR, as reflected by a unique cell adhesion expression. Neutrophils constitutively express β2 integrins (CR3, CD11b/CD18) and L-selectin (LS, CD62L) on their surface, which play a role in the adhesion of neutrophils to endothelium and their egress to an extravascular site of inflammation. Upon neutrophil activation by chemoattractants [such as C5a, IL-8, and formylmethionyl-leucyl phenylalanine (FMLP)] there is an up-regulation of β2-integrin with a concomitant shedding (down-regulation) of LS.


By contrast, in patients with active SLE there is a marked up-regulation of β2 integrin surface molecules with no change in the expression of LS.1 This discrepancy between the lack of LS shedding despite an up-regulation of β2-integrin was shown to be related to FcγR activation by IC.9,10 Other studies confirmed that neutrophils are activated in the circulation of patients with active SLE, as reflected by increased expression of CD11b/CD18 on their surface with no decrease in LS expression.1 Moreover, the expression of β2-integrin on the surface of circulating neutrophils correlated positively with activity scores in patients with SLE.11,12 In contrast, there was no correlation between LS expression and disease activity.1


Interaction between FcR expressed on phagocytic cells and antibodies plays a critical role in innate immune response. Neutrophils (the major type of phagocytic cell in the blood) express two types of FcR [FcγR-IIA (CD32) and FcγR-IIIB (CD16)], both of which bind effectively to IC. IC binding to FcR results in neutrophil activation.1 In addition, FcR expressed on phagocytic cells are important in the process of IC clearance from the circulation. There is evidence suggesting that Fc-mediated clearance of IC is defective in patients with SLE.1 Allelic variants of FcγR are common within the general population. However, certain polymorphism patterns have been linked to increased susceptibility to SLE.1 Several studies addressing the association of FcγR-IIA and IIIA polymorphism and susceptibility for SLE in various ethnic groups yielded inconclusive results.


In a meta-analysis comprising 17 studies, the homozygosity for the R131 allele of the FcγR-IIA was associated with a 1.3-fold greater risk for developing SLE but not for lupus nephritis.1 The evidence for an SLE susceptibility was not conclusive for patients of European descent compared to those of Asian and African descent. Another meta-analysis looking for the association of V/F158 genotype of FcγR IIIA demonstrated that the F158 allele poses a 20% greater risk for the development of lupus nephritis.1 FF homozygotes were more prone to renal damage as compared to VV homozygotes (odds ratio 1.47).


These polymorphisms are associated with low-affinity FcγR, which have a lower capacity to clear ICs. The half-life of IgG-coated erythrocytes in the blood was prolonged in lupus patients expressing the FcγR-IIA-R/R131 genotype, whereas the homozygous genotype FcγR-IIIA-F (F158) was associated with arthritis and/or serositis in these patients.1 This association of FcγR polymorphism with an increased risk of SLE and/or certain manifestations of the disease emphasizes the importance of neutrophils in the process of IC clearance, in that a delay in clearance results in greater IC deposition in tissues and organs such as the kidneys and blood vessels.


FcγRs play another role in the pathogenesis of SLE; namely, clearance of apoptotic cells. The half-life of circulating neutrophils is about 7 hours, and following egress to an extravascular tissue they die within 1 to 2 days. Under normal conditions, neutrophils are removed from the circulation as well as from inflamed tissue by apoptosis, genetically programmed cell death.1 Delayed neutrophil apoptosis has been associated with autoimmune disorders, especially so in SLE where nuclear materials serve as autoantigens for the production of an array of antinuclear antibodies.1 Nucleosomes are known as major autoantigens in SLE and are exposed at the cell surface in apoptosis. Thus, increased apoptosis and/or delayed clearance of apoptotic cells could contribute to an overload of autoantigens in the circulation that will eventually lead to an increased nucleosome-containing IC deposition in target tissues. Both increased neutrophil apoptosis and delayed removal of apoptotic neutrophils from the circulation have been described in lupus.21,22


Increased neutrophil apoptosis was correlated with disease activity and could serve as a source for dsDNA, as suggested by a positive correlation between antibodies to dsDNA and increased neutrophil apoptosis found in lupus patients.1 Moreover, delayed clearance of apoptotic neutrophils was observed in lupus and suggested as a major step in the pathogenesis of SLE.1 A decreased ability of macrophages to ingest apoptotic neutrophils was found in SLE and inversely correlated with activity scores.1 Taken together, these data indicate that increased neutrophil apoptosis together with decreased macrophage clearance of apoptotic cells yield a large nuclear-derived autoantigen burden that may induce or facilitate the formation of nucleosome-immunoglobulin complexes.


Various mechanisms have been suggested to explain the delayed clearance of apoptotic cells in lupus, such as complement deficiency and low pentraxin levels. With regard to delayed clearance of apoptotic neutrophils in SLE, reduced expression of CD44 and resistance to the apoptosis-inhibiting effects of granulocyte-colony stimulation factors (G-CSFs) were suggested as possible underlying mechanisms.23,24 In conclusion, cumulative data suggest that neutrophil apoptosis plays a major role in the pathogenesis of SLE by serving as the largest source of nucleosomes and nuclear debris for autoantigens resulting in production of anti-dsDNA and other antinuclear antibodies in lupus.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jul 24, 2018 | Posted by in MUSCULOSKELETAL MEDICINE | Comments Off on Neutrophils (Polymorphonuclear Leukocytes) in Systemic Lupus Erythematosus

Full access? Get Clinical Tree

Get Clinical Tree app for offline access