Tumor necrosis factor‑α in systemic lupus erythematosus: Structure, function and therapeutic implications (Review)

  • Authors:
    • Farid Ghorbaninezhad
    • Patrizia Leone
    • Hajar Alemohammad
    • Basira Najafzadeh
    • Niloufar Sadat Nourbakhsh
    • Marcella Prete
    • Eleonora Malerba
    • Hossein Saeedi
    • Neda Jalili Tabrizi
    • Vito Racanelli
    • Behzad Baradaran
  • View Affiliations

  • Published online on: February 4, 2022     https://doi.org/10.3892/ijmm.2022.5098
  • Article Number: 43
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tumor necrosis factor‑α (TNF‑α) is a pleiotropic pro‑inflammatory cytokine that contributes to the pathophysiology of several autoimmune diseases, such as multiple sclerosis, inflammatory bowel disease, rheumatoid arthritis, psoriatic arthritis and systemic lupus erythematosus (SLE). The specific role of TNF‑α in autoimmunity is not yet fully understood however, partially, in a complex disease such as SLE. Through the engagement of the TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2), both the two variants, soluble and transmembrane TNF‑α, can exert multiple biological effects according to different settings. They can either function as immune regulators, impacting B‑, T‑ and dendritic cell activity, modulating the autoimmune response, or as pro‑inflammatory mediators, regulating the induction and maintenance of inflammatory processes in SLE. The present study reviews the dual role of TNF‑α, focusing on the different effects that TNF‑α may have on the pathogenesis of SLE. In addition, the efficacy and safety of anti‑TNF‑α therapies in preclinical and clinical trials SLE are discussed.

Introduction

Tumor necrosis factor-α (TNF-α) is a complex cytokine that impacts various physiological and pathological conditions. It can function as an immune regulator, contributing to the development and function regulation of B-cells, T-lymphocytes and dendritic cells, as a pro-inflammatory mediator, modulating the generation and preservation of inflammatory processes, or as an apoptotic inducer, promoting cell death (1,2). TNF-α is involved in the pathogenesis of numerous autoimmune disorders, such as rheumatoid arthritis (RA) (3), inflammatory bowel disease (4,5), psoriatic arthritis (6) and multiple sclerosis (7,8); however, its role in systemic lupus erythematosus (SLE) disease remains unclear. From the genetic point of view, several investigations have demonstrated a link between the TNF-α gene polymorphism and the susceptibility to SLE (911). Furthermore, there is a strong connection between TNF-α gene expression and clinical manifestations in patients with SLE (12).

Furthermore, TNF-α is a growth factor for B-lymphocytes, which can produce large quantities of TNF-α in an autocrine loop (1315). Serum levels of TNF-α have been discovered to be high in patients with SLE and have been linked to disease activity and several systemic manifestations, such as SLE-related cardiovascular disease and lupus nephritis (12,1620). The dysregulation of TNF-α is clearly linked to tissue destruction observed in lupus organ disease, to the death of lymphocytes and to the impaired clearance of apoptotic cells, resulting in the presentation of self-antigens and autoantibody formation (21). However, TNF-α has pro-apoptotic and anti-apoptotic properties, depending on the underlying contextual circumstances (2224). Treatment with anti-TNF drugs or recombinant TNF has been demonstrated to have conflicting results in murine models of SLE. As previously demonstrated, TNF-competent New Zealand Black (NZB) mice displayed an autoimmune phenotype, whereas TNF-deficient New Zealand Black (NZB) mice developed severe lupus-like disease (25,26).

Moreover, the disease was shown to be reversible by the administration of recombinant TNF; indeed, the early application of TNF in NZB/White (NZB/W) mice postponed the development of autoantibodies and lupus nephritis (25,27). In addition, TNF-blocking therapies have sometimes induced the production of antinuclear antibodies and IgM antibodies to double-stranded DNA in individuals with RA or Crohn's disease, suggesting the potential propensity of anti-TNF agents to stimulate pathogenic autoantibody production (28,29). These patients rarely exhibited a reversible drug-induced lupus-like syndrome (29). In this context, a critical view of the relevance of TNF in SLE is necessary. The present review article thus aimed to enhance the understanding of the functions of TNF-α in the pathogenesis of SLE and discuss the benefits associated with anti-TNF therapies in patients with SLE.

TNF-α: Structure and function

TNF-α, also known as cachectin, was first defined by Carswell et al (30) several decades ago as an endotoxin-inducing autoantibody molecule that leads to tumor necrosis. The human gene for TNF-α is located on the short arm of chromosome 6 between 6p21.1 and 6p21.3, which is within the human leukocyte antigen (HLA) class III region in humans (31). It includes exons, interrupted via 3 introns and is ~3 kb in length. Of note, >80% of the mature TNF-α sequence is encoded in exon 4. Exons 1 and 2 mainly include the sequence of leader peptides (32). In addition, multiple regulatory sites with sequences corresponding to the transcription factors, activator protein (AP-1/2), NF-κB and the cAMP-response element (CRE) have been identified on the 5′b-end of the TNF-α gene. TNF-α exists in two types, as a membrane-bound trimeric ligand (tmTNF) and as a soluble trimeric molecule (sTNF), each of which probably play a different physiological role. The sTNF-α is formed from tmTNF-α via the extracellular domain of the matrix metalloproteinase (MMP) TNF-α-converting enzyme (TACE). tmTNF-α can function as ligand-bound TNFR or as an intermediate receptor in transmitting external signals. The human TNF-α protein contains 233 amino acids with a predicted molecular weight of 25.6 kDa, which, following proteolytical cleavage by a specific protease, generates an active protein of 17 kDa. The hydrophobic transmembrane region includes 26–44 amino acids of the TNF-α pre-sequence, and the intracytoplasmic region comprises 50–76 amino acids (33). The soluble and membrane-bound forms function as biological homotrimers (similar to a triangular cone), each molecule interacting the other two substances. Each monomer includes two packed β-pleated sheets that are formed via eight antiparallel β-strands arranged in a β-jellyroll topology (34). It has also been reported that TNF-α undergoes post-translational alterations, including phosphorylation. The initial sequence of TNF-α indicates the existence of several phosphorylation consensus sites, providing a possible mechanism for regulating trimer formation and/or receptor binding. In addition, a previous study demonstrated that membrane-bound TNF-α was phosphorylated via creatine kinase (CKI) and dephosphorylated through phosphatase activation (35). TNF-α production has been detected in a wide range of cells, such as normal, malignant, hematopoietic and non-hematopoietic cells (36). Several factors can induce the production of TNF-α, including bacterial lipopolysaccharide (LPS, endotoxin), viral antigens, immune complexes, IL-1 and TNF-α itself via autocrine mechanisms. In addition, certain pathophysiological conditions, such as previous infection and inflammation, trauma, infarction and heart failure, can also induce the production of TNF-α (37,38). TNF-α performs its biological activities by interacting with two membrane-dependent receptors, TNF receptor (TNFR)1 and TNFR2, and via triggering a number of secondary proteins that elicit various responses in the cell, such as transcription factors, protein kinases and phospholipases (36,3941).

It has been demonstrated that TNF-α functions as a multifunctional cytokine which plays a vital role in controlling inflammation, secondary and tertiary lymphoid tissue development, and immune regulation (42). The functional mechanisms of TNF-α are highly diverse and somewhat complex. This protein plays conflicting roles: On the one hand, it combats certain types of infections and, on the other hand, induces pathological complications. This may be due to the stimulation of various signaling pathways involved in diverse cellular reactions, such as survival, differentiation, cell proliferation and cell death (43). A recent study indicated that TNF-α deregulation was directly associated with chronic inflammation, autoimmune diseases and other pathologies, such as neuroinflammation (44). Therefore, understanding the exact mechanisms of action of the TNF-α signaling pathways may lead to the development of effective therapies for the treatment of immune diseases.

TNF receptors: TNFR1 and TNFR2

The TNFR1 gene (also known as p55, p60, CD120a or TNFRSF1A), located on chromosome 12p13, has 10 exons and produces a 60-kDa protein (45). TNFR2 (also known as p75, p80, CD120b or TNFRSF1B) encoded via the gene located on chromosome 1p36.2, consists of 10 exons and gives rise to a protein of 80 kDa (46,47). These receptors are membrane glycoproteins and members of the TNF receptor superfamily (48). They are crucial to the development and homeostasis of the immune and neurological systems, and ectodermal organs (49,50). The extracellular domain is very similar between these two receptors and consists of multiple cysteine-rich domains involved in ligand binding; however, the intracellular domains clearly differ; thus, they can activate different signaling pathways by interacting with a variety of cytosolic proteins (51). Receptors are dependent on adapter proteins, including TNFR-associated death domain protein (TRADD), Fas-associated protein with death domain (FADD) and the TNFR-associated factor (TRAF)-1 to activate intracellular signaling pathways and induce a biological response. These proteins form a scaffold that allows other proteins to be absorbed to trigger the signaling pathway (52,53). TNFR1 expression has been observed in a number of cell types; however, TNFR2 expression has been observed in a small number of cells, such as T-cells and endothelial cells (54). TNF-α strongly binds to both receptors, and the differential engagement of the receptors is associated with distinct functions. sTNF interacts with both TNF receptors, while tmTNF mainly activates TNFR2 (51). TNFR2 has a lower affinity for TNF-α than for TNFR1, suggesting that TNFR2 can momentarily bind and can subsequently be release, playing a role in amplifying or synergizing TNFR1 signaling (55,56). The stimulation of TNFR1 is responsible for several biological effects of TNF-α, such as cytotoxicity and proliferation. The activation of TNFR1 stimulates various cellular responses, such as the induction of proliferation processes, apoptosis, or necroptosis, depending on the cell type and environmental conditions (56). TNFR1, in its cytoplasmic part, has a death domain (DD) related to TNF-α-mediated cytotoxicity, while TNFR2 lacks this domain (57). The engagement of TNF with TNFR1 leads to the successive formation of two different TNF receptor signaling complexes (complex I and complex II) that are separated both temporally and spatially. Complex I induces the expression of anti-apoptotic genes, which inhibit cell death processes mainly by activating transcription factors, such as NF-κB, whereas the second signaling pathway (complex II) leads to apoptosis or necroptosis (1). Compared to TNFR1, knowledge of TNFR2 signaling pathways is limited. Since TNFR2 lacks the DD, it cannot directly induce cell death. In contrast to the functions of TNFR1, which is able to induce inflammation or apoptotic responses, TNFR2 engagement significantly enhances cell stimulation, migration and propagation (58). The binding of TRAF2 to TNFR2 activates the canonical and non-canonical NF-κB signaling pathways (59). However, TNFR is able to activate NF-κB slowly, although with a longer activation time compared to TNFR (60). In addition, it has been shown that TNFR2 can induce cell survival (61). Other researchers have indicated that TNFR2 is required for antigen-associated differentiation and T-cell survival. TNFR2 regulates several adhesion molecules, including intercellular adhesion molecule-1 and selectin-E, which are central molecules in angiogenesis (62).

Downstream signaling of the TNF and TNFR axis

TNF signaling appears to be quite complex and can cover various downstream signaling pathways. TNFR1 is triggered through both membrane-bound and soluble TNF (51) (Fig. 1). The TNFR1 cytoplasmic DD allows interactions with other DD-containing proteins, including TRADD, E3 ubiquitin ligases, cellular inhibitor of apoptosis protein (cIAP)1/2, the receptor-interacting serine/threonine-protein kinase (RIPK)1 and TRAF2, resulting in complex I signaling (52,53). In turn, polyubiquitinated RIPK1 and cIAP1/2 proteins have a crucial function in the uptake of other proteins, such as the TGF-β-activated kinase 1 (TAK1) in the TAK1-binding protein (TAB)2/3 complex and the linear ubiquitin chain assembly complex (LUBAC), respectively (63,64). LUBAC can polyubiquitinate numerous molecules, such as LUBAC itself and NF-κB essential modulator (NEMO) in the IκB kinase (IKK) complex comprised of IKK1/IKKα, IKK2/IKKβ and NEMO/IKKγ (64,65). Furthermore, TAK1 phosphorylates IκB, a prerequisite for its ubiquitylation and proteasome degradation. NF-κB then translocates to the nucleus and prompts the transcription of target genes involved in inflammation and cell survival (64). In addition, TAK1 in complex with TAB2/TAB3 can also induce the triggering of AP-1 transcription factor through the phosphorylation of MAP kinases, such as cJun NH2-terminal kinase (JNK) and p38 (66,67). This signaling pathway activates the transcription of various pro-inflammatory genes. Moreover, TNF-TNFR1 interaction can activate other signaling pathways involved in programmed cell death, such as apoptosis and necroptosis through complex II and IIb signaling, respectively (68,69). In this case, the separation of RIPK1 and TRADD from complex I leads to the instability of complex I and in the formation of complex II, which includes FADD, cellular FLICE-inhibitory protein and pro-caspase-8 molecules, thus inducing apoptosis (70). In addition, when caspase is inhibited, the interaction of TNF with TNFR1 induces the formation of complex IIb, leading to the activation of the cell death pathway known as necroptosis. Complex IIb consists of the phosphorylated molecules, RIPK1 and RIPK3, and mixed lineage kinase domain-like pseudokinase (MLKL). Thereafter, MLKL oligomerization occurs, and phosphorylated MLKL is translocated to the plasma membrane, which is disrupted to stimulate necroptosis (71). In contrast to TNFR1, the interaction of TNFR2 with TNF (Fig. 2) causes the direct recruitment of TRAF1 or TRAF2 together with cIAP1/2 and LUBAC molecules (72,73). Subsequently, this signaling pathway may, similar to the TNFR1 signaling pathway, recruit the TAK1/TAB2/TAB3 and NEMO/IKKα/β complexes, resulting in the downstream stimulation of the canonical NF-κB pathway (60). Alternatively, the only membrane-bound TNF and non-soluble TNF trimers can activate, via TNFR2, the non-canonical NF-κB pathway via the TRAF2/cIAP1⁄2 complex interaction, resulting in the accumulation of NF-κB-inducing kinase (NIK). NIK phosphorylates the NF-κB precursor protein p100, thus eliciting its proteasomal proteolysis to p52, which results in the transcription of p52/RelB-containing NF-κB heterodimer (74,75). Under normal conditions, the basal level of NIK is maintained at a low level by TRAF3, which induces NIK ubiquitination and constitutive degradation by the proteasome. In response to tmTNF, NIK becomes stabilized due to TRAF3 degradation, and its accumulation activates non-canonical NF-κB signaling, resulting in autoimmune and inflammatory diseases (75).

In contrast to the common belief that TNFR1 signaling triggers apoptosis and TNFR2 signaling promotes pro-survival, there is increasing evidence to indicate that exclusive TNFR2 stimulation can induce apoptosis (despite the fact that TNFR2 does not contain a DD) through crosstalk among the two receptors and TRAFs, which are involved in initiating TNFR1 and TNFR2 signaling (60). Moreover, TNFR2 can enhance TNFR1-mediated apoptosis, despite the enhanced NF-κB activation (22,7680). The upregulation of TNFR2 induces proteasomal degradation and the consequent depletion of TRAF2 (77,78). Upon the activation of TNF, a member of the MAPK kinase family termed apoptosis signal-regulating kinase-1 is activated by TRAF2, and induces p38 and JNK activation (81). This pathway is triggered by stress stimuli and results in the regulation of apoptosis and inflammatory cytokine expression (66).

Role of TNF-α and TNFRs in autoimmune diseases

TNF-α is associated with the pathogenesis of various autoimmune diseases (Fig. 3), such as RA (3), inflammatory bowel disease, including ulcerative colitis and Crohn's disease (4,5), psoriatic arthritis (6) and multiple sclerosis (8). Along with IL-1β, TNF-α is involved in the onset and progression of RA (82). Elevated levels of TNF-α have been detected in the synovial fluid and synovium of patients with RA (82), causing local inflammation and ‘pannus’ structure formation, leading to tissue necrosis, cartilage erosion and bone destruction (82). In this context, synovial fibroblasts secrete IL-1β, monocyte chemoattractant protein-1, macrophage inflammatory protein-1α, MMP-1 and MMP-3, and receptor activator of nuclear factor-kB ligand, an osteoclastogenic cytokine, resulting in recruitment of immune cells (B-cells, T-cells, macrophages and neutrophils) and the perpetuation of the production of pro-inflammatory cytokines and mediators, such as IL-2, IL-1β and TNF-α (8284). Several studies using mouse models have demonstrated the interplay between TNF-α and IL-1β in vivo, and their crucial function in the onset and evolution of RA (8588). In addition, TNF-α reduces the frequency and function of regulatory T-cells in autoimmune-prone mice (89) and in patients with RA, and this effect has been shown to be reverted by TNF-α blockade (90).

A similar role is played by TNF-α in the gastrointestinal tract in patients with ulcerative colitis and Crohn's. TNF-α activates macrophages, enhances the T-cell response, induces the expression of adhesion molecules by the vascular endothelium, and the recruitment of neutrophils to local sites of inflammation, promotes tissue remodeling, edema and granuloma formation (4,5). TNF-α-dependent inflammation is extended through triggered NF-κβ-dependent pathways, which provide the release of MMPs with the consequent degradation of the mucosa and ulceration (91).

In patients with psoriatic arthritis, elevated levels of TNF-α stimulate DCs and macrophages to secrete high amounts of TNF-α and IL-23, promoting the differentiation of naive T-cells into Th17 cells, with the consequent overproduction of the pro-inflammatory cytokine, IL-17. IL-17 and TNF-α trigger the NF-κB signaling pathway, leading to keratinocyte activation and proliferation, the recruitment of inflammatory cells, epidermal hyperplasia and microabscess development (92).

There is also emerging evidence to support the involvement of TNF-α in the pathogenesis of SLE, which is discussed in the following section (Fig. 3).

Systemic lupus erythematosus and TNF-α

SLE is a systemic autoimmune disease featured by heterogeneous clinical manifestations and immunological abnormalities. Its pathogenesis remains poorly understood, and even though the etiology of SLE is undetermined, multiple elements are associated with disease development, including genetic (9395), epigenetic (96), immunoregulatory (97), ethnic (98), hormonal (99) and environmental factors (100103). The role TNF-α in the pathogenesis of SLE is controversial; some investigators have found that TNF-α confers SLE susceptibility (10,11,18,104), while others have described a protective role of TNF-α in patients with SLE (105,106). Multiple have studies indicated that TNF-α, along with other cytokines, such as IFN-α, IL-12, IL-4, IL-10, IL-6, A proliferation-inducing ligand (APRIL) and B cell-activating factor, IL-17 and IL-21 are the main SLE-related cytokines (107111). In particular, Svenungsson et al (19,20) emphasized the high triglyceride and low HDL levels as disease activity markers, and the elevated levels of TNF-α/TNFR in patients with SLE, as well as the link between inflammation, dyslipoproteinemia and cardiovascular disease in patients with SLE. Furthermore, an increased TNF-α concentration has been observed in the blood and in the inflamed kidneys of patients with SLE (112116). Further studies have also demonstrated a significant genetic relation between TNF-α promoter polymorphism and SLE susceptibility (911,117121). Increased levels of TNF-α have been found to be associated with disease severity in patients with SLE (18,107,122). Higher serum levels of TNF-α and its soluble receptors have been observed in patients with SLE with active disease compared with SLE patients with inactive disease (122). Moreover, patients with SLE with high TNF-α levels present T-lymphocytes which are more susceptible to apoptosis than T-cells from healthy controls (104). This enhanced TNF-α-induced apoptosis increases the autoantigen load, promoting autoimmune responses in patients with SLE (104). This enhanced TNF-α-induced apoptosis also increases the load of autoantigens, promoting autoimmune responses in patients with SLE (104).

Moreover, genetic variation at TNF alpha induced protein 3 (TNFAIP3) and TNF superfamily member 4 (TNFSF4) have been associated with lymphocyte dysregulation and different SLE ethnic groups (123126). Polymorphisms in TNFR2 may also play a role in the genetic susceptibility to SLE. A previous genotype analysis manifested that the existence of one 196R allele was sufficient for delivering SLE susceptibility in the Japanese population (127). Both TNFR1 and TNFR2 expression levels are highly enhanced in active the serum of patients with SLE (122,128,129), and sTNFRs are crucial modulators of the inflammatory responses in lupus nephritis (130,131). In Japanese patients, a mutation in exon 3 in position 61 of the tumor necrosis factor receptor superfamily 1A gene (TNFRSF1A) was shown to be associated with SLE. These patients were characterized by a high concentration of serum TNF, sTNFRSF1B and a low concentration of sTNFRSF1A (132).

On the contrary, some researchers have observed decreased levels of TNF-α in patients with SLE, particularly in patients with severe disease (105). Zhu et al (106) indicated that the expression levels of TNF-α adapter proteins TRADD, FADD, TRAF-2 and RIPK-1 in peripheral blood mononuclear cells were markedly diminished in patients with SLE and were negatively associated with the SLE activity index. Reduced levels of TNF-α adapter proteins have been shown to be related to advanced lymphocyte apoptosis and enormous autoantibody secretion, resulting in immune-pathogenic injury in patients with SLE (106). Moreover, several studies did not demonstrate any association between polymorphisms in the TNFR2 gene and SLE (133135). Sullivan et al (135) analyzed the frequency of genetic polymorphisms in the 3′ untranslated region of the TNFR2 gene in patients with SLE and did not find an association, although the study examined only Caucasian patients. Furthermore, Chadha et al (134) did not find any association between TNFRSF14, TNFRSF8, TNFRSF1B locus and SLE in European-Caucasian families. In line with this, Al-Ansari et al did not find any connection between the TNFRII 196R allele and SLE neither in Spanish or in UK populations (133).

Blocking of TNF: Therapeutic approaches in SLE; animal models and clinical trials

Murine disease models are genetically homogeneous populations used to research disease initiation and progression (136). There are different mouse models for lupus; some of them develop lupus spontaneously [e.g., NZB/W F1hybrid mice, medical research laboratory lymphoproliferation (MRL/lpr) mice, BXSB/Yaa mice], and in others, lupus is induced in the animals (e.g., pristane-induced lupus) (137). Due to the dual function of TNF-α (mediator of inflammation and regulator of autoimmunity), the efficacy of TNF-based therapies in SLE is controversial and can vary, depending on the subsets of patients (138). TNF-α is well observed in NZB/W F1 hybrid mice, MRL/lpr and C3H.SW lupus-prone mouse models. The NZB/W F1 lupus model denotes an F1 cross between the NZB and NZW strains (139). In 1988, Jacob and McDevitt (25) demonstrated that, unlike NZW mice (healthy mouse strains), NZB/W mice were defective in TNF-α production and developed severe lupus-like phenotypes. They also noted that the early application of recombinant TNF-α to NZB/W mice attenuated the progression of lupus nephritis (25). In 1989, Gordon et al (27) continued research on NZB/W mice, demonstrating that the use of TNF-α, even following the onset of renal symptoms, increased survival, reduced the progression of kidney damage and delayed the emergence of lupus in these mice. In 2000, the study by Kontoyiannis and Kollias (26) demonstrated that NZB mice with an engineered heterozygous TNF deficit developed lupus nephritis and autoimmunity due to a lower production of TNF. Contrary to these findings, Brennan et al (140) found high steady-state levels of TNF-α and IL-1 β in the renal cortices of NZB/W mice with lupus nephritis. They also noted that the administration of a lower dose of TNF-α increased kidney injury (140). Furthermore, in MRL/lpr mice, an elevation in TNF-α expression was previously detected, which was linked to the degree of inflammation and organ dysfunction (141143). The upregulation of TNF mRNA was discovered in the lungs of MRL/lpr mice in the study by Deguchi and Kishimoto (144). Overall these findings suggest that TNF-α may have both beneficial and harmful effects in experimental lupus models, based on its concentration and ability to play both immune-regulatory and pro-inflammatory functions (116). Thus, this cytokine can be considered as a therapeutic target in SLE. Rabbit anti-mouse TNF-α immunoglobulin (Ig)G antibody therapy has been shown to reduce autoimmune pulmonary inflammation in lupus-prone mice (144). It has been demonstrated that therapies directed at blocking TNF/TNFR interactions, such as soluble, dimericTNFR I (sTNFRI), which binds to TNF-α with high affinity, thus neutralizing it, reduce the infiltration of mononuclear cells into joints, lungs and skin in NZB/W mice, improving the symptoms of the disease and extending the lifespan (145). Bethunaickan et al (146) used a NZB/W murine model of IFN-induced lupus nephritis and treated mice with recombinant fusion proteins, such as TNFR2-Ig. They revealed that TNFR2-Ig treatment reduced the renal inflammatory response to immune complex deposition, stabilizing nephritis, thus prolonging survival (146).

Given the promising results of TNF blockade in SLE mouse models, the inhibition of this cytokine was previously investigated in patients with SLE. Clinically authorized TNF-α suppressors have been revealed to be effective in several autoimmune disorders, and novel TNF-α signaling blockers are currently being investigated in clinical trials. Infliximab (Remicade), adalimumab (Humira), certolizumab pegol (Cimzia), golimumab (Simponi) and etanercept (Enbrel) are the five anti-TNF drugs approved by the US Food and Drug Administration (FDA) for the treatment of rheumatic inflammatory diseases, such as RA, psoriasis, psoriatic arthritis and Crohn's disease (Table I) are currently being studied in patients with SLE (44,147).

Table I.

FDA-approved TNF-α inhibitors.

Table I.

FDA-approved TNF-α inhibitors.

DrugTrade nameType of agentFDA approval data (https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm)
Infliximab Remicade®Chimeric mouse/human mAbAugust, 1998
Etanercept Enbrel®A human soluble TNF-α receptorNovember, 1998
Adalimumab Humira®A fully human anti-TNF-α mAbDecember, 2002
Certolizumab pegol Cimzia®A PEGylated, Fab’-only, recombinant humanized mAbApril, 2008
Golimumab Simponi®A human IgG1κ anti-TNF-α mAbApril, 2009

[i] TNF-α, tumor necrosis factor-α; mAb, monoclonal antibody; PEG, polyethylene glycol.

However, these agents may induce autoimmunity, leading to the production of antinuclear antibodies and/or anti-double-stranded DNA antibodies, and may occasionally trigger the anti-TNF-α-induced lupus-like syndrome (ATIL) defined by clinical features suggestive of SLE (148). The majority of cases occur in patients with RA, inflammatory bowel disease and ankylosing spondylitis (29,149152). Previous studies have demonstrated that nephritis may occur following the administration of anti-TNF-α drugs (153,154).

Infliximab is a chimeric genetically modified monoclonal antibody that includes a murine variable region and a human IgG1 constant region. It is particular for all types of TNF in humans and effectively prevents TNF from attaching to both transmembrane and soluble receptors (147). Due to its chimeric structure, infliximab is the anti-TNF-α molecule with a larger degree of immunogenicity (152). Nevertheless, open-label studies and case reports have reported the effectiveness, acceptable safety and tolerability profile of infliximab in patients with SLE. Aringer et al (155157) observed that short-term induction therapy with infliximab along with azathioprine or methotrexate elicited long-term improvement in individuals with lupus nephritis. The majority of patients with SLE exhibited a transient elevation in autoantibodies against phospholipids and nuclear antigens, which was not associated with disease flares (NCT00368264) (155157). Other studies have confirmed the safety and efficiency of infliximab in patients with difficult-to-treat lupus nephritis (158,159). Hayat and Uppal (159) also demonstrated the efficacy of infliximab in a patient with difficult-to-treat active non-renal SLE. In a pilot study, Uppal et al (160) demonstrated that infliximab significantly decreased the SLE disease activity index (SLEDAI) without raising any safety concerns.

Etanercept is a full human monoclonal antibody with reduced immunogenicity. It is a fusion protein consisting of two equal extracellular regions of TNFR2 linked to the Fc fragment of human IgG1 and strongly binds to sTNF-α or tmTNF-α (161,162). The FDA has approved the therapeutic application of this drug for the treatment of RA, polyarticular juvenile idiopathic arthritis (JIA), psoriatic arthritis, ankylosing spondylitis and plaque psoriasis (44). Although the FDA has not yet approved etanercept for the treatment of SLE, it has been used in several clinical studies, including a randomized, double-blind, phase II, multi-center study for the treatment of lupus nephritis (NCT00447265), and in two phase II open-label trials for the treatment of discoid lupus erythematosus (NCT02656082 and NCT00797784). In an observational study, long-term treatment with etanercept was revealed to be relatively safe and efficacious in refractory lupus arthritis (163). In a previous case report study, an enhancement of clinical symptoms and the quality of life were described in subacute cutaneous lupus erythematosus individuals by etanercept treatment (164). The efficacy and the acceptable safety profile of etanercept were also shown to treat rhupus, a disease with characteristics of both RA and SLE (165,166). Micheloud et al (167) described a pregnant woman with SLE with a severe diffuse proliferative nephritis who was successfully treated with etanercept, plasmapheresis and high-dose intravenous gammaglobulin.

Using molecular docking approach, a recent study investigated the potential of selected anti-inflammatory peptides from plant and animal sources as novel inhibitors for the treatment of SLE. Protein-ligand and peptide-protein docking of twenty anti-inflammatory peptides targeting IFN-γ, IL-3 and TNF-α were developed to reduce inflammatory events which lead to autoantibody production. The study represents an initial step for employment of these peptides in the treatment of autoimmune disorders (168).

Blocking of TNFRs: Therapeutic approaches in SLE; animal models and clinical trials

The present review article has noted the paradoxical involvement of TNF-α in lupus and explained the advantages and disadvantages of blocking this cytokine in preclinical and clinical studies. In addition to inducing ATIL, and autoantibodies to dsDNA and phospholipids, TNF-α inhibitors can increase the risk of infections, malignancies (169), central nervous system demyelinating disorders, and other autoimmune diseases, such as type I diabetes, psoriasis and multiple sclerosis (53,170172). A probable cause of these side-effects is that prevailing TNF-α suppressors prevent the engagement between TNF-α and the receptors TNFR1 (with pro-inflammatory and pro-apoptotic role) and TNFR2 (with a regulatory function), leading to a loss of TNFR2 signaling regulatory function (173). Van Hauwermeiren et al (174) noted that TNFR1+/− mice, which express 50% of TNFR1 on cells, were highly resistant to lethal TNF-induced inflammation. Moreover, the decrease in p55TNFR mitigated TNF toxicity without compromising effectiveness (174), suggesting that TNFR can be considered as a therapeutic target (175). In SLE, the significance of TNF-α-TNFR1 interaction has been emphasized (176). Wu et al (177) demonstrated that the TNFR1 levels in the urine of mice and individuals with lupus nephritis increased; sTNFR1 and sTNFR2 levels have also been shown to be higher in patients with lupus nephritis (131). According to Deng et al (178) TNFR1 is abundantly expressed in skin lesions of MRL/lpr mice, unlike TNFR2, and the inhibition of TNFR1 signaling relieved skin lesions. On the other hand, thye acceleration of the disease course occurred in NZB/F1 mice defective in both TNFR1 and TNFR2 (179). However, the lack of the p55TNFR has been shown to lead to significantly increased lymphoproliferation and autoimmune disorder in the Fas deficient MRL-lpr/lpr mouse (180). Aderka et al (128) suggested that elevated serum sTNFR levels may be a valuable marker for assessing the progression of SLE. The effect of Brentuximab Vedotin targeting TNFR was investigated in adults with active SLE in a phase II, multi-center, randomized, double-blinded, multiple-ascending-dose study (NCT02533570) (Table II).

Table II.

Clinical trials of TNF-α/TNFR inhibition in SLE patients.

Table II.

Clinical trials of TNF-α/TNFR inhibition in SLE patients.

InterventionPhaseEnrollmentStatusStudy start dateNational clinical trial no.
Infliximab + azathioprineII/IIIOne participantTerminatedSeptember, 2006NCT00368264
Etanercept + lupus treatment-standard of care + placeboIIOne participantTerminatedFebruary, 2008NCT00447265
EtanerceptII25 participantsCompletedFebruary, 2016NCT02656082
EtanerceptII20 participantsUnknownOctober, 2008NCT00797784
Brentuximab vedotin + placeboII20 participantsTerminatedJuly, 2015NCT02533570

Conclusions and future perspectives

TNF-α is a potent pleiotropic cytokine with multiple cellular activities, also involved in developing autoimmune disorders. The impact of TNF-α on these diseases is not yet completely understood. On the one hand, TNF-α can play a pro-inflammatory and pro-apoptotic role, and on the other hand, it has a regulatory function. Currently, therapeutic strategies that target TNF-α are clinically utilized for the treatment of inflammatory and autoimmune diseases, such as RA, inflammatory bowel disease and psoriasis. However, notwithstanding their clinical achievement, the application of anti-TNF drugs is restricted due to severe side-effects and ATIL development. Alternative therapeutic strategies that selectively target TNFRs have exhibited immense therapeutic potential. Thus, the majority of available evidence suggests that the usability of anti-TNF drugs could be broadened. Understanding the dual role of TNF-α in autoimmunity is difficult, particularly in a complex disease, such as SLE. The use of drugs targeting TNF-α and TNFRs in SLE remains controversial. Further investigations are thus required to establish the favorable therapeutics benefits/risk ratio associated with the use of anti-TNF-α drugs, as well as to determine the treatment's effectiveness and side-effects in patients with SLE.

Acknowledgements

The authors would like to thank Dr Afshin Derakhshani, Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran, for critically reading and commenting on the manuscript.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

FG, PL, HA, BN, NSN, MP, EM, HS, NJT, VR and BB contributed to the conceptualization, methodology, data curation, investigation, visualization, and the drafting and editing of the manuscript. VR and BB critically reviewed the manuscript. All authors have read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Micheau O and Tschopp J: Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell. 114:181–190. 2003. View Article : Google Scholar : PubMed/NCBI

2 

Graninger WB, Steiner CW, Graninger MT, Aringer M and Smolen JS: Cytokine regulation of apoptosis and Bcl-2 expression in lymphocytes of patients with systemic lupus erythematosus. Cell Death Differ. 7:966–972. 2000. View Article : Google Scholar : PubMed/NCBI

3 

Choy EH and Panayi GS: Cytokine pathways and joint inflammation in rheumatoid arthritis. N Engl J Med. 344:907–916. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Sands BE and Kaplan GG: The role of TNFalpha in ulcerative colitis. J Clin Pharmacol. 47:930–941. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Adegbola SO, Sahnan K, Warusavitarne J, Hart A and Tozer P: Anti-TNF therapy in Crohn's disease. Int J Mol Sci. 19:22442018. View Article : Google Scholar : PubMed/NCBI

6 

Celis R, Cuervo A, Ramirez J and Cañete JD: Psoriatic synovitis: Singularity and potential clinical implications. Front Med (Lausanne). 6:142019. View Article : Google Scholar : PubMed/NCBI

7 

Derakhshani A, Asadzadeh Z, Safarpour H, Leone P, Shadbad MA, Heydari A, Baradaran B and Racanelli V: Regulation of CTLA-4 and PD-L1 expression in relapsing-remitting multiple sclerosis patients after treatment with fingolimod, IFNbeta-1α, glatiramer acetate, and dimethyl fumarate drugs. J Pers Med. 11:7212021. View Article : Google Scholar : PubMed/NCBI

8 

Pegoretti V, Baron W, Laman JD and Eisel ULM: Selective modulation of TNF-TNFRs signaling: Insights for multiple sclerosis treatment. Front Immunol. 9:9252018. View Article : Google Scholar : PubMed/NCBI

9 

Chen L, Huang Z, Liao Y, Yang B and Zhang J: Association between tumor necrosis factor polymorphisms and rheumatoid arthritis as well as systemic lupus erythematosus: A meta-analysis. Braz J Med Biol Res. 52:e79272019. View Article : Google Scholar : PubMed/NCBI

10 

Mahto H, Tripathy R, Meher BR, Prusty BK, Sharma M, Deogharia D, Saha AK, Panda AK and Das BK: TNF-α promoter polymorphisms (G-238A and G-308A) are associated with susceptibility to Systemic Lupus Erythematosus (SLE) and P. falciparum malaria: A study in malaria endemic area. Sci Rep. 9:117522019. View Article : Google Scholar : PubMed/NCBI

11 

Ramirez-Bello J, Cadena-Sandoval D, Mendoza-Rincon JF, Barbosa-Cobos RE, Sánchez-Muñoz F, Amezcua-Guerra LM, Sierra-Martínez M and Jiménez-Morales S: Tumor necrosis factor gene polymorphisms are associated with systemic lupus erythematosus susceptibility or lupus nephritis in Mexican patients. Immunol Res. 66:348–354. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Idborg H, Eketjall S, Pettersson S, Gustafsson JT, Zickert A, Kvarnström M, Oke V, Jakobsson PJ, Gunnarsson I and Svenungsson E: TNF-α and plasma albumin as biomarkers of disease activity in systemic lupus erythematosus. Lupus Sci Med. 5:e0002602018. View Article : Google Scholar : PubMed/NCBI

13 

Kehrl JH, Miller A and Fauci AS: Effect of tumor necrosis factor alpha on mitogen-activated human B cells. J Exp Med. 166:786–791. 1987. View Article : Google Scholar : PubMed/NCBI

14 

Boussiotis VA, Nadler LM, Strominger JL and Goldfeld AE: Tumor necrosis factor alpha is an autocrine growth factor for normal human B cells. Proc Natl Acad Sci USA. 91:7007–7011. 1994. View Article : Google Scholar : PubMed/NCBI

15 

Rieckmann P, Tuscano JM and Kehrl JH: Tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) in B-lymphocyte function. Methods. 11:128–132. 1997. View Article : Google Scholar : PubMed/NCBI

16 

Aringer M, Stummvoll GH, Steiner G, Köller M, Steiner CW, Höfler E, Hiesberger H, Smolen JS and Graninger WB: Serum interleukin-15 is elevated in systemic lupus erythematosus. Rheumatology (Oxford). 40:876–881. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Gabay C, Cakir N, Moral F, Roux-Lombard P, Meyer O, Dayer JM, Vischer T, Yazici H and Guerne PA: Circulating levels of tumor necrosis factor soluble receptors in systemic lupus erythematosus are significantly higher than in other rheumatic diseases and correlate with disease activity. J Rheumatol. 24:303–308. 1997.PubMed/NCBI

18 

Studnicka-Benke A, Steiner G, Petera P and Smolen JS: Tumour necrosis factor alpha and its soluble receptors parallel clinical disease and autoimmune activity in systemic lupus erythematosus. Br J Rheumatol. 35:1067–1074. 1996. View Article : Google Scholar : PubMed/NCBI

19 

Svenungsson E, Fei GZ, Jensen-Urstad K, de Faire U, Hamsten A and Frostegard J: TNF-alpha: A link between hypertriglyceridaemia and inflammation in SLE patients with cardiovascular disease. Lupus. 12:454–461. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Svenungsson E, Gunnarsson I, Fei GZ, Lundberg IE, Klareskog L and Frostegård J: Elevated triglycerides and low levels of high-density lipoprotein as markers of disease activity in association with up-regulation of the tumor necrosis factor alpha/tumor necrosis factor receptor system in systemic lupus erythematosus. Arthritis Rheum. 48:2533–2540. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Gordon C and Salmon M: Update on systemic lupus erythematosus: Autoantibodies and apoptosis. Clin Med (Lond). 1:10–14. 2001. View Article : Google Scholar : PubMed/NCBI

22 

Wang CY, Mayo MW, Korneluk RG, Goeddel DV and Baldwin AS Jr: NF-kappaB antiapoptosis: Induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation. Science. 281:1680–1683. 1998. View Article : Google Scholar : PubMed/NCBI

23 

Micheau O, Lens S, Gaide O, Alevizopoulos K and Tschopp J: NF-kappaB signals induce the expression of c-FLIP. Mol Cell Biol. 21:5299–5305. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Yan X, Xiao CW, Sun M, Tsang BK and Gibb W: Nuclear factor kappa B activation and regulation of cyclooxygenase type-2 expression in human amnion mesenchymal cells by interleukin-1beta. Biol Reprod. 66:1667–1671. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Jacob CO and McDevitt HO: Tumour necrosis factor-alpha in murine autoimmune ‘lupus’ nephritis. Nature. 331:356–358. 1988. View Article : Google Scholar : PubMed/NCBI

26 

Kontoyiannis D and Kollias G: Accelerated autoimmunity and lupus nephritis in NZB mice with an engineered heterozygous deficiency in tumor necrosis factor. Eur J Immunol. 30:2038–2047. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Gordon C, Ranges GE, Greenspan JS and Wofsy D: Chronic therapy with recombinant tumor necrosis factor-alpha in autoimmune NZB/NZW F1 mice. Clin Immunol Immunopathol. 52:421–434. 1989. View Article : Google Scholar : PubMed/NCBI

28 

Mohan AK, Edwards ET, Coté TR, Siegel JN and Braun MM: Drug-induced systemic lupus erythematosus and TNF-alpha blockers. Lancet. 360:6462002. View Article : Google Scholar : PubMed/NCBI

29 

Charles PJ, Smeenk RJ, Jong JD, Feldmann M and Maini RN: Assessment of antibodies to double-stranded DNA induced in rheumatoid arthritis patients following treatment with infliximab, a monoclonal antibody to tumor necrosis factor alpha: Findings in open-label and randomized placebo-controlled trials. Arthritis Rheum. 43:2383–2390. 2000. View Article : Google Scholar : PubMed/NCBI

30 

Carswell EA, Old LJ, Kassel RL, Green S, Fiore N and Williamson B: An endotoxin-induced serum factor that causes necrosis of tumors. Proc Natl Acad Sci USA. 72:3666–3670. 1975. View Article : Google Scholar : PubMed/NCBI

31 

Spriggs DR, Deutsch S and Kufe DW: Genomic structure, induction, and production of TNF-alpha. Immunol Ser. 56:3–34. 1992.PubMed/NCBI

32 

Shakhov AN, Collart MA, Vassalli P, Nedospasov SA and Jongeneel CV: Kappa B-type enhancers are involved in lipopolysaccharide-mediated transcriptional activation of the tumor necrosis factor alpha gene in primary macrophages. J Exp Med. 171:35–47. 1990. View Article : Google Scholar : PubMed/NCBI

33 

Smith RA and Baglioni C: The active form of tumor necrosis factor is a trimer. J Biol Chem. 262:6951–6954. 1987. View Article : Google Scholar : PubMed/NCBI

34 

Eck MJ and Sprang SR: The structure of tumor necrosis factor-alpha at 2.6 A resolution. Implications for receptor binding. J Biol Chem. 264:17595–17605. 1989. View Article : Google Scholar : PubMed/NCBI

35 

Watts AD, Hunt NH, Wanigasekara Y, Bloomfield G, Wallach D, Roufogalis BD and Chaudhri G: A casein kinase I motif present in the cytoplasmic domain of members of the tumour necrosis factor ligand family is implicated in ‘reverse signalling’. EMBO J. 18:2119–2126. 1999. View Article : Google Scholar : PubMed/NCBI

36 

Vilcek J and Lee TH: Tumor necrosis factor. New insights into the molecular mechanisms of its multiple actions. J Biol Chem. 266:7313–7316. 1991. View Article : Google Scholar : PubMed/NCBI

37 

Cairns CB, Panacek EA, Harken AH and Banerjee A: Bench to bedside: Tumor necrosis factor-alpha: From inflammation to resuscitation. Acad Emerg Med. 7:930–941. 2000. View Article : Google Scholar : PubMed/NCBI

38 

Camussi G, Albano E, Tetta C and Bussolino F: The molecular action of tumor necrosis factor-alpha. Eur J Biochem. 202:3–14. 1991. View Article : Google Scholar : PubMed/NCBI

39 

Yang L, Lindholm K, Konishi Y, Li R and Shen Y: Target depletion of distinct tumor necrosis factor receptor subtypes reveals hippocampal neuron death and survival through different signal transduction pathways. J Neurosci. 22:3025–3032. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Beyaert R and Fiers W: Molecular mechanisms of tumor necrosis factor-induced cytotoxicity. What we do understand and what we do not. FEBS Lett. 340:9–16. 1994. View Article : Google Scholar : PubMed/NCBI

41 

Darnay BG and Aggarwal BB: Signal transduction by tumour necrosis factor and tumour necrosis factor related ligands and their receptors. Ann Rheum Dis. 58 (Suppl 1):I2–I13. 1999. View Article : Google Scholar : PubMed/NCBI

42 

Kalliolias GD and Ivashkiv LB: TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat Rev Rheumatol. 12:49–62. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Fiers W: Tumor necrosis factor. Characterization at the molecular, cellular and in vivo level. FEBS Lett. 285:199–212. 1991. View Article : Google Scholar : PubMed/NCBI

44 

Jang DI, Lee AH, Shin HY, Song HR, Park JH, Kang TB, Lee SR and Yang SH: The role of tumor necrosis factor alpha (TNF-α) in autoimmune disease and current TNF-α inhibitors in therapeutics. Int J Mol Sci. 22:27192021. View Article : Google Scholar : PubMed/NCBI

45 

Fuchs P, Strehl S, Dworzak M, Himmler A and Ambros PF: Structure of the human TNF receptor 1 (p60) gene (TNFR1) and localization to chromosome 12p13 [corrected]. Genomics. 13:219–224. 1992. View Article : Google Scholar : PubMed/NCBI

46 

Wang XY, Kafka M, Dvilansky A and Nathan I: The roles of protein phosphorylation/dephosphorylation in tumor necrosis factor antitumor effects. J Interferon Cytokine Res. 16:1021–1025. 1996. View Article : Google Scholar : PubMed/NCBI

47 

Kemper O and Wallach D: Cloning and partial characterization of the promoter for the human p55 tumor necrosis factor (TNF) receptor. Gene. 134:209–216. 1993. View Article : Google Scholar : PubMed/NCBI

48 

Desplat-Jego S, Burkly L and Putterman C: Targeting TNF and its family members in autoimmune/inflammatory disease. Mediators Inflamm. 2014:6287482014. View Article : Google Scholar : PubMed/NCBI

49 

Bodmer JL, Schneider P and Tschopp J: The molecular architecture of the TNF superfamily. Trends Biochem Sci. 27:19–26. 2002. View Article : Google Scholar : PubMed/NCBI

50 

Locksley RM, Killeen N and Lenardo MJ: The TNF and TNF receptor superfamilies: Integrating mammalian biology. Cell. 104:487–501. 2001. View Article : Google Scholar : PubMed/NCBI

51 

Grell M: Tumor necrosis factor (TNF) receptors in cellular signaling of soluble and membrane-expressed TNF. J Inflamm. 47:8–17. 1995.PubMed/NCBI

52 

Hsu H, Xiong J and Goeddel DV: The TNF receptor 1-associated protein TRADD signals cell death and NF-kappa B activation. Cell. 81:495–504. 1995. View Article : Google Scholar : PubMed/NCBI

53 

Sedger LM and McDermott MF: TNF and TNF-receptors: From mediators of cell death and inflammation to therapeutic giants-past, present and future. Cytokine Growth Factor Rev. 25:453–472. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Faustman D and Davis M: TNF receptor 2 pathway: Drug target for autoimmune diseases. Nat Rev Drug Discov. 9:482–493. 2010. View Article : Google Scholar : PubMed/NCBI

55 

Grell M, Wajant H, Zimmermann G and Scheurich P: The type 1 receptor (CD120a) is the high-affinity receptor for soluble tumor necrosis factor. Proc Natl Acad Sci USA. 95:570–575. 1998. View Article : Google Scholar : PubMed/NCBI

56 

Wajant H, Pfizenmaier K and Scheurich P: Tumor necrosis factor signaling. Cell Death Differ. 10:45–65. 2003. View Article : Google Scholar : PubMed/NCBI

57 

Tartaglia LA, Ayres TM, Wong GH and Goeddel DV: A novel domain within the 55 kd TNF receptor signals cell death. Cell. 74:845–853. 1993. View Article : Google Scholar : PubMed/NCBI

58 

Bradley JR: TNF-mediated inflammatory disease. J Pathol. 214:149–160. 2008. View Article : Google Scholar : PubMed/NCBI

59 

Fischer R, Maier O, Naumer M, Krippner-Heidenreich A, Scheurich P and Pfizenmaier K: Ligand-induced internalization of TNF receptor 2 mediated by a di-leucin motif is dispensable for activation of the NFκB pathway. Cell Signal. 23:161–170. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Naude PJ, den Boer JA, Luiten PG and Eisel UL: Tumor necrosis factor receptor cross-talk. FEBS J. 278:888–898. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Marchetti L, Klein M, Schlett K, Pfizenmaier K and Eisel ULM: Tumor necrosis factor (TNF)-mediated neuroprotection against glutamate-induced excitotoxicity is enhanced by N-methyl-D-aspartate receptor activation. Essential role of a TNF receptor 2-mediated phosphatidylinositol 3-kinase-dependent NF-kappa B pathway. J Biol Chem. 279:32869–32881. 2004. View Article : Google Scholar : PubMed/NCBI

62 

Goto N, Tsurumi H, Takemura M, Hara T, Sawada M, Kasahara S, Kanemura N, Yamada T, Shimizu M, Takahashi T, et al: Serum-soluble tumor necrosis factor receptor 2 (sTNF-R2) level determines clinical outcome in patients with aggressive non-Hodgkin's lymphoma. Eur J Haematol. 77:217–225. 2006. View Article : Google Scholar : PubMed/NCBI

63 

Kanayama A, Seth RB, Sun L, Ea CK, Hong M, Shaito A, Chiu YH, Deng L and Chen ZJ: TAB2 and TAB3 activate the NF-kappaB pathway through binding to polyubiquitin chains. Mol Cell. 15:535–548. 2004. View Article : Google Scholar : PubMed/NCBI

64 

Haas TL, Emmerich CH, Gerlach B, Schmukle AC, Cordier SM, Rieser E, Feltham R, Vince J, Warnken U, Wenger T, et al: Recruitment of the linear ubiquitin chain assembly complex stabilizes the TNF-R1 signaling complex and is required for TNF-mediated gene induction. Mol Cell. 36:831–844. 2009. View Article : Google Scholar : PubMed/NCBI

65 

Tokunaga F, Sakata S, Saeki Y, Satomi Y, Kirisako T, Kamei K, Nakagawa T, Kato M, Murata S, Yamaoka S, et al: Involvement of linear polyubiquitylation of NEMO in NF-kappaB activation. Nat Cell Biol. 11:123–132. 2009. View Article : Google Scholar : PubMed/NCBI

66 

Sabio G and Davis RJ: TNF and MAP kinase signalling pathways. Semin Immunol. 26:237–245. 2014. View Article : Google Scholar : PubMed/NCBI

67 

Zeke A, Misheva M, Reményi A and Bogoyevitch MA: JNK signaling: Regulation and functions based on complex protein-protein partnerships. Microbiol Mol Biol Rev. 80:793–835. 2016. View Article : Google Scholar : PubMed/NCBI

68 

Itoh N and Nagata S: A novel protein domain required for apoptosis. Mutational analysis of human fas antigen. J Biol Chem. 268:10932–10937. 1993. View Article : Google Scholar : PubMed/NCBI

69 

Vercammen D, Beyaert R, Denecker G, Goossens V, Loo GV, Declercq W, Grooten J, Fiers W and Vandenabeele P: Inhibition of caspases increases the sensitivity of L929 cells to necrosis mediated by tumor necrosis factor. J Exp Med. 187:1477–1485. 1998. View Article : Google Scholar : PubMed/NCBI

70 

Shalini S, Dorstyn L, Dawar S and Kumar S: Old, new and emerging functions of caspases. Cell Death Differ. 22:526–539. 2015. View Article : Google Scholar : PubMed/NCBI

71 

Samson AL, Zhang Y, Geoghegan ND, Gavin XJ, Davies KA, Mlodzianoski MJ, Whitehead LW, Frank D, Garnish SE, Fitzgibbon C, et al: MLKL trafficking and accumulation at the plasma membrane control the kinetics and threshold for necroptosis. Nat Commun. 11:31512020. View Article : Google Scholar : PubMed/NCBI

72 

Rothe M, Wong SC, Henzel WJ and Goeddel DV: A novel family of putative signal transducers associated with the cytoplasmic domain of the 75 kDa tumor necrosis factor receptor. Cell. 78:681–692. 1994. View Article : Google Scholar : PubMed/NCBI

73 

Borghi A, Haegman M, Fischer R, Carpentier I, Bertrand MJM, Libert C, Afonina IS and Beyaert R: The E3 ubiquitin ligases HOIP and cIAP1 are recruited to the TNFR2 signaling complex and mediate TNFR2-induced canonical NF-kappaB signaling. Biochem Pharmacol. 153:292–298. 2018. View Article : Google Scholar : PubMed/NCBI

74 

Rauert H, Wicovsky A, Muller N, Siegmund D, Spindler V, Waschke J, Kneitz C and Wajant H: Membrane tumor necrosis factor (TNF) induces p100 processing via TNF receptor-2 (TNFR2). J Biol Chem. 285:7394–7404. 2010. View Article : Google Scholar : PubMed/NCBI

75 

Sun SC: The non-canonical NF-κB pathway in immunity and inflammation. Nat Rev Immunol. 17:545–558. 2017. View Article : Google Scholar : PubMed/NCBI

76 

Devin A, Cook A, Lin Y, Rodriguez Y, Kelliher M and Liu Z: The distinct roles of TRAF2 and RIP in IKK activation by TNF-R1: TRAF2 recruits IKK to TNF-R1 while RIP mediates IKK activation. Immunity. 12:419–429. 2000. View Article : Google Scholar : PubMed/NCBI

77 

Fotin-Mleczek M, Henkler F, Samel D, Reichwein M, Hausser A, Parmryd I, Scheurich P, Schmid JA and Wajant H: Apoptotic crosstalk of TNF receptors: TNF-R2-induces depletion of TRAF2 and IAP proteins and accelerates TNF-R1-dependent activation of caspase-8. J Cell Sci. 115:2757–2770. 2002. View Article : Google Scholar : PubMed/NCBI

78 

Li X, Yang Y and Ashwell JD: TNF-RII and c-IAP1 mediate ubiquitination and degradation of TRAF2. Nature. 416:345–347. 2002. View Article : Google Scholar : PubMed/NCBI

79 

Tada K, Okazaki T, Sakon S, Kobarai T, Kurosawa K, Yamaoka S, Hashimoto H, Mak TW, Yagita H, Okumura K, et al: Critical roles of TRAF2 and TRAF5 in tumor necrosis factor-induced NF-kappa B activation and protection from cell death. J Biol Chem. 276:36530–36534. 2001. View Article : Google Scholar : PubMed/NCBI

80 

Vince JE, Pantaki D, Feltham R, Mace PD, Cordier SM, Schmukle AC, Davidson AJ, Callus BA, Wong WWL, Gentle IE, et al: TRAF2 must bind to cellular inhibitors of apoptosis for tumor necrosis factor (tnf) to efficiently activate nf-{kappa}b and to prevent tnf-induced apoptosis. J Biol Chem. 284:35906–35915. 2009. View Article : Google Scholar : PubMed/NCBI

81 

Liu ZG, Hsu H, Goeddel DV and Karin M: Dissection of TNF receptor 1 effector functions: JNK activation is not linked to apoptosis while NF-kappaB activation prevents cell death. Cell. 87:565–576. 1996. View Article : Google Scholar : PubMed/NCBI

82 

Ridgley LA, Anderson AE and Pratt AG: What are the dominant cytokines in early rheumatoid arthritis? Curr Opin Rheumatol. 30:207–214. 2018. View Article : Google Scholar : PubMed/NCBI

83 

Kobayashi M, Squires GR, Mousa A, Tanzer M, Zukor DJ, Antoniou J, Feige U and Poole AR: Role of interleukin-1 and tumor necrosis factor alpha in matrix degradation of human osteoarthritic cartilage. Arthritis Rheum. 52:128–135. 2005. View Article : Google Scholar : PubMed/NCBI

84 

Mirza F, Lorenzo J, Drissi H, Lee FY and Soung DY: Dried plum alleviates symptoms of inflammatory arthritis in TNF transgenic mice. J Nutr Biochem. 52:54–61. 2018. View Article : Google Scholar : PubMed/NCBI

85 

Chen W, Li Z, Wang Z, Gao H, Ding J and He Z: Intraarticular injection of infliximab-loaded thermosensitive hydrogel alleviates pain and protects cartilage in rheumatoid arthritis. J Pain Res. 13:3315–3329. 2020. View Article : Google Scholar : PubMed/NCBI

86 

Rioja I, Bush KA, Buckton JB, Dickson MC and Life PF: Joint cytokine quantification in two rodent arthritis models: Kinetics of expression, correlation of mRNA and protein levels and response to prednisolone treatment. Clin Exp Immunol. 137:65–73. 2004. View Article : Google Scholar : PubMed/NCBI

87 

Williams RO, Marinova-Mutafchieva L, Feldmann M and Maini RN: Evaluation of TNF-alpha and IL-1 blockade in collagen-induced arthritis and comparison with combined anti-TNF-alpha/anti-CD4 therapy. J Immunol. 165:7240–7245. 2000. View Article : Google Scholar : PubMed/NCBI

88 

Yu D, Ye X, Che R, Wu Q, Qi J, Song L, Guo X, Zhang S, Wu H, Ren G and Li D: FGF21 exerts comparable pharmacological efficacy with Adalimumab in ameliorating collagen-induced rheumatoid arthritis by regulating systematic inflammatory response. Biomed Pharmacother. 89:751–760. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Wu AJ, Hua H, Munson SH and McDevitt HO: Tumor necrosis factor-alpha regulation of CD4+CD25+ T cell levels in NOD mice. Proc Natl Acad Sci USA. 99:12287–12292. 2002. View Article : Google Scholar : PubMed/NCBI

90 

Ehrenstein MR, Evans JG, Singh A, Moore S, Warnes G, Isenberg DA and Mauri C: Compromised function of regulatory T cells in rheumatoid arthritis and reversal by anti-TNFalpha therapy. J Exp Med. 200:277–285. 2004. View Article : Google Scholar : PubMed/NCBI

91 

Louis E: The immuno-inflammatory reaction in Crohn's disease and ulcerative colitis: Characterisation, genetics and clinical application. Focus on TNF alpha. Acta Gastroenterol Belg. 64:1–5. 2001.PubMed/NCBI

92 

Gottlieb AB, Chamian F, Masud S, Cardinale I, Abello MV, Lowes MA, Chen F, Magliocco M and Krueger JG: TNF inhibition rapidly down-regulates multiple proinflammatory pathways in psoriasis plaques. J Immunol. 175:2721–2729. 2005. View Article : Google Scholar : PubMed/NCBI

93 

Catalina MD, Owen KA, Labonte AC, Grammer AC and Lipsky PE: The pathogenesis of systemic lupus erythematosus: Harnessing big data to understand the molecular basis of lupus. J Autoimmun. 110:1023592020. View Article : Google Scholar : PubMed/NCBI

94 

Kwon YC, Chun S, Kim K and Mak A: Update on the genetics of systemic lupus erythematosus: Genome-wide association studies and beyond. Cells. 8:11802019. View Article : Google Scholar : PubMed/NCBI

95 

Marion TN and Postlethwaite AE: Chance, genetics, and the heterogeneity of disease and pathogenesis in systemic lupus erythematosus. Semin Immunopathol. 36:495–517. 2014. View Article : Google Scholar : PubMed/NCBI

96 

Hedrich CM: Epigenetics in SLE. Curr Rheumatol Rep. 19:582017. View Article : Google Scholar : PubMed/NCBI

97 

Pan L, Lu MP, Wang JH, Xu M and Yang SR: Immunological pathogenesis and treatment of systemic lupus erythematosus. World J Pediatr. 16:19–30. 2020. View Article : Google Scholar : PubMed/NCBI

98 

Maningding E, Dall'Era M, Trupin L, Murphy LB and Yazdany J: Racial and ethnic differences in the prevalence and time to onset of manifestations of systemic lupus erythematosus: The California lupus surveillance project. Arthritis Care Res (Hoboken). 72:622–629. 2020. View Article : Google Scholar : PubMed/NCBI

99 

Christou EAA, Banos A, Kosmara D, Bertsias GK and Boumpas DT: Sexual dimorphism in SLE: Above and beyond sex hormones. Lupus. 28:3–10. 2019. View Article : Google Scholar : PubMed/NCBI

100 

Barbhaiya M and Costenbader KH: Environmental exposures and the development of systemic lupus erythematosus. Curr Opin Rheumatol. 28:497–505. 2016. View Article : Google Scholar : PubMed/NCBI

101 

Constantin MM, Nita IE, Olteanu R, Constantin T, Bucur S, Matei C and Raducan A: Significance and impact of dietary factors on systemic lupus erythematosus pathogenesis. Exp Ther Med. 17:1085–1090. 2019.PubMed/NCBI

102 

Luo S, Long H and Lu Q: Recent advances in understanding pathogenesis and therapeutic strategies of systemic lupus erythematosus. Int Immunopharmacol. 89:1070282020. View Article : Google Scholar : PubMed/NCBI

103 

Parks CG, de Souza Espindola Santos A, Barbhaiya M and Costenbader KH: Understanding the role of environmental factors in the development of systemic lupus erythematosus. Best Pract Res Clin Rheumatol. 31:306–320. 2017. View Article : Google Scholar : PubMed/NCBI

104 

Habib HM, Taher TE, Isenberg DA and Mageed RA: Enhanced propensity of T lymphocytes in patients with systemic lupus erythematosus to apoptosis in the presence of tumour necrosis factor alpha. Scand J Rheumatol. 38:112–120. 2009. View Article : Google Scholar : PubMed/NCBI

105 

Gómez D, Correa PA, Gómez LM, Cadena J, Molina JF and Anaya JM: Th1/Th2 cytokines in patients with systemic lupus erythematosus: Is tumor necrosis factor alpha protective? Semin Arthritis Rheum. 33:404–413. 2004. View Article : Google Scholar : PubMed/NCBI

106 

Zhu L, Yang X, Chen W, Li X, Ji Y, Mao H, Nie J and Yu X: Decreased expressions of the TNF-alpha signaling adapters in peripheral blood mononuclear cells (PBMCs) are correlated with disease activity in patients with systemic lupus erythematosus. Clin Rheumatol. 26:1481–1489. 2007. View Article : Google Scholar : PubMed/NCBI

107 

McCarthy EM, Smith S, Lee RZ, Cunnane G, Doran MF, Donnelly S, Howard D, O'Connell P, Kearns G, Gabhann JN and Jefferies CA: The association of cytokines with disease activity and damage scores in systemic lupus erythematosus patients. Rheumatology (Oxford). 53:1586–1594. 2014. View Article : Google Scholar : PubMed/NCBI

108 

Prete M, Leone P, Frassanito MA, Desantis V, Marasco C, Cicco S, Dammacco F, Vacca A and Racanelli V: Belimumab restores Treg/Th17 balance in patients with refractory systemic lupus erythematosus. Lupus. 27:1926–1935. 2018. View Article : Google Scholar : PubMed/NCBI

109 

Su DL, Lu ZM, Shen MN, Li X and Sun LY: Roles of pro- and anti-inflammatory cytokines in the pathogenesis of SLE. J Biomed Biotechnol. 2012:3471412012. View Article : Google Scholar : PubMed/NCBI

110 

Tahernia L, Alimadadi H, Tahghighi F, Amini Z and Ziaee V: Frequency and type of hepatic and gastrointestinal involvement in juvenile systemic lupus erythematosus. Autoimmune Dis. 2017:80972732017.PubMed/NCBI

111 

Yap DY and Lai KN: The role of cytokines in the pathogenesis of systemic lupus erythematosus-from bench to bedside. Nephrology (Carlton). 18:243–255. 2013. View Article : Google Scholar : PubMed/NCBI

112 

Takemura T, Yoshioka K, Murakami K, Akano N, Okada M, Aya N and Maki S: Cellular localization of inflammatory cytokines in human glomerulonephritis. Virchows Arch. 424:459–464. 1994. View Article : Google Scholar : PubMed/NCBI

113 

Malide D, Russo P and Bendayan M: Presence of tumor necrosis factor alpha and interleukin-6 in renal mesangial cells of lupus nephritis patients. Hum Pathol. 26:558–564. 1995. View Article : Google Scholar : PubMed/NCBI

114 

Neale TJ, Ruger BM, Macaulay H, Dunbar PR, Hasan Q, Bourke A, Murray-McIntosh RP and Kitching AR: Tumor necrosis factor-alpha is expressed by glomerular visceral epithelial cells in human membranous nephropathy. Am J Pathol. 146:1444–1454. 1995.PubMed/NCBI

115 

Herrera-Esparza R, Barbosa-Cisneros O, Villalobos-Hurtado R and Avalos-Díaz E: Renal expression of IL-6 and TNFalpha genes in lupus nephritis. Lupus. 7:154–158. 1998. View Article : Google Scholar : PubMed/NCBI

116 

Aringer M and Smolen JS: The role of tumor necrosis factor-alpha in systemic lupus erythematosus. Arthritis Res Ther. 10:2022008. View Article : Google Scholar : PubMed/NCBI

117 

D'Alfonso S, Colombo G, Bella SD, Scorza R and Momigliano-Richiardi P: Association between polymorphisms in the TNF region and systemic lupus erythematosus in the Italian population. Tissue Antigens. 47:551–555. 1996. View Article : Google Scholar : PubMed/NCBI

118 

Dourmishev L, Kamenarska Z, Hristova M, Dodova R, Kaneva R and Mitev V: Association of TNF-α polymorphisms with adult dermatomyositis and systemic lupus erythematosus in Bulgarian patients. Int J Dermatol. 51:1467–1473. 2012. View Article : Google Scholar : PubMed/NCBI

119 

Lee YH, Harley JB and Nath SK: Meta-analysis of TNF-alpha promoter-308 A/G polymorphism and SLE susceptibility. Eur J Hum Genet. 14:364–371. 2006. View Article : Google Scholar : PubMed/NCBI

120 

Lin YJ, Chen RH, Wan L, Sheu JC, Huang CM, Lin CW, Chen SY, Lai CH, Lan YC, Hsueh KC, et al: Association of TNF-alpha gene polymorphisms with systemic lupus erythematosus in Taiwanese patients. Lupus. 18:974–979. 2009. View Article : Google Scholar : PubMed/NCBI

121 

Zúñiga J, Vargas-Alarcón G, Hernández-Pacheco G, Portal-Celhay C, Yamamoto-Furusho JK and Granados J: Tumor necrosis factor-alpha promoter polymorphisms in Mexican patients with systemic lupus erythematosus (SLE). Genes Immun. 2:363–366. 2001. View Article : Google Scholar : PubMed/NCBI

122 

Davas EM, Tsirogianni A, Kappou I, Karamitsos D, Economidou I and Dantis PC: Serum IL-6, TNFalpha, p55 srTNFalpha, p75srTNFalpha, srIL-2alpha levels and disease activity in systemic lupus erythematosus. Clin Rheumatol. 18:17–22. 1999. View Article : Google Scholar : PubMed/NCBI

123 

Adrianto I, Wen F, Templeton A, Wiley G, King JB, Lessard CJ, Bates JS, Hu Y, Kelly JA, Kaufman KM, et al: Association of a functional variant downstream of TNFAIP3 with systemic lupus erythematosus. Nat Genet. 43:253–258. 2011. View Article : Google Scholar : PubMed/NCBI

124 

Bates JS, Lessard CJ, Leon JM, Nguyen T, Battiest LJ, Rodgers J, Kaufman KM, James JA, Gilkeson GS, Kelly JA, et al: Meta-analysis and imputation identifies a 109 kb risk haplotype spanning TNFAIP3 associated with lupus nephritis and hematologic manifestations. Genes Immun. 10:470–477. 2009. View Article : Google Scholar : PubMed/NCBI

125 

Goulielmos GN, Zervou MI, Vazgiourakis VM, Ghodke-Puranik Y, Garyfallos A and Niewold TB: The genetics and molecular pathogenesis of systemic lupus erythematosus (SLE) in populations of different ancestry. Gene. 668:59–72. 2018. View Article : Google Scholar : PubMed/NCBI

126 

Manku H, Langefeld CD, Guerra SG, Malik TH, Alarcon-Riquelme M, Anaya JM, Bae SC, Boackle SA, Brown EE, Criswell LA, et al: Trans-ancestral studies fine map the SLE-susceptibility locus TNFSF4. PLoS Genet. 9:e10035542013. View Article : Google Scholar : PubMed/NCBI

127 

Komata T, Tsuchiya N, Matsushita M, Hagiwara K and Tokunaga K: Association of tumor necrosis factor receptor 2 (TNFR2) polymorphism with susceptibility to systemic lupus erythematosus. Tissue Antigens. 53:527–533. 1999. View Article : Google Scholar : PubMed/NCBI

128 

Aderka D, Wysenbeek A, Engelmann H, Cope AP, Brennan F, Molad Y, Hornik V, Levo Y, Maini RN and Feldmann M: Correlation between serum levels of soluble tumor necrosis factor receptor and disease activity in systemic lupus erythematosus. Arthritis Rheum. 36:1111–1120. 1993. View Article : Google Scholar : PubMed/NCBI

129 

Munroe ME, Vista ES, Guthridge JM, Thompson LF, Merrill JT and James JA: Proinflammatory adaptive cytokine and shed tumor necrosis factor receptor levels are elevated preceding systemic lupus erythematosus disease flare. Arthritis Rheumatol. 66:1888–1899. 2014. View Article : Google Scholar : PubMed/NCBI

130 

Horiuchi T, Mitoma H, Harashima S, Tsukamoto H and Shimoda T: Transmembrane TNF-alpha: Structure, function and interaction with anti-TNF agents. Rheumatology (Oxford). 49:1215–1228. 2010. View Article : Google Scholar : PubMed/NCBI

131 

Patel M, Oni L, Midgley A, Smith E, Tullus K, Marks SD, Jones CA, Pilkington C and Beresford MW: Increased concentration of plasma TNFR1 and TNFR2 in paediatric lupus nephritis. Lupus. 25:1040–1044. 2016. View Article : Google Scholar : PubMed/NCBI

132 

Ida H, Kawasaki E, Miyashita T, Tanaka F, Kamachi M, Izumi Y, Huang M, Tamai M, Origuchi T, Kawakami A, et al: A novel mutation (T61I) in the gene encoding tumour necrosis factor receptor superfamily 1A (TNFRSF1A) in a Japanese patient with tumour necrosis factor receptor-associated periodic syndrome (TRAPS) associated with systemic lupus erythematosus. Rheumatology (Oxford). 43:1292–1299. 2004. View Article : Google Scholar : PubMed/NCBI

133 

Al-Ansari AS, Ollier WE, Villarreal J, Ordi J, The LS and Hajeer AH: Tumor necrosis factor receptor II (TNFRII) exon 6 polymorphism in systemic lupus erythematosus. Tissue Antigens. 55:97–99. 2000. View Article : Google Scholar : PubMed/NCBI

134 

Chadha S, Miller K, Farwell L, Sacks S, Daly MJ, Rioux JD and Vyse TJ: Haplotype analysis of tumour necrosis factor receptor genes in 1p36: No evidence for association with systemic lupus erythematosus. Eur J Hum Genet. 14:69–78. 2006. View Article : Google Scholar : PubMed/NCBI

135 

Sullivan KE, Piliero LM, Goldman D and Petri MA: A TNFR2 3′ flanking region polymorphism in systemic lupus erythematosus. Genes Immun. 1:225–227. 2000. View Article : Google Scholar : PubMed/NCBI

136 

Fairhurst AM, Wandstrat AE and Wakeland EK: Systemic lupus erythematosus: Multiple immunological phenotypes in a complex genetic disease. Adv Immunol. 92:1–69. 2006. View Article : Google Scholar : PubMed/NCBI

137 

Li W, Titov AA and Morel L: An update on lupus animal models. Curr Opin Rheumatol. 29:434–441. 2017. View Article : Google Scholar : PubMed/NCBI

138 

Aringer M and Smolen JS: Therapeutic blockade of TNF in patients with SLE-promising or crazy? Autoimmun Rev. 11:321–325. 2012. View Article : Google Scholar : PubMed/NCBI

139 

Helyer BJ and Howie JB: Renal disease associated with positive lupus erythematosus tests in a cross-bred strain of mice. Nature. 197:1971963. View Article : Google Scholar : PubMed/NCBI

140 

Brennan DC, Yui MA, Wuthrich RP and Kelley VE: Tumor necrosis factor and IL-1 in New Zealand black/white mice. Enhanced gene expression and acceleration of renal injury. J Immunol. 143:3470–3475. 1989.PubMed/NCBI

141 

Boswell JM, Yui MA, Burt DW and Kelley VE: Increased tumor necrosis factor and IL-1 beta gene expression in the kidneys of mice with lupus nephritis. J Immunol. 141:3050–3054. 1988.PubMed/NCBI

142 

Yokoyama H, Kreft B and Kelley VR: Biphasic increase in circulating and renal TNF-alpha in MRL-lpr mice with differing regulatory mechanisms. Kidney Int. 47:122–130. 1995. View Article : Google Scholar : PubMed/NCBI

143 

Tsai CY, Wu TH, Huang SF, Sun KH, Hsieh SC, Han SH, Yu HS and Yu CL: Abnormal splenic and thymic IL-4 and TNF-alpha expression in MRL-lpr/lpr mice. Scand J Immunol. 41:157–163. 1995. View Article : Google Scholar : PubMed/NCBI

144 

Deguchi Y and Kishimoto S: Tumour necrosis factor/cachectin plays a key role in autoimmune pulmonary inflammation in lupus-prone mice. Clin Exp Immunol. 85:392–395. 1991. View Article : Google Scholar : PubMed/NCBI

145 

Su X, Zhou T, Yang P, Edwards CK III and Mountz JD: Reduction of arthritis and pneumonitis in motheaten mice by soluble tumor necrosis factor receptor. Arthritis Rheum. 41:139–149. 1998. View Article : Google Scholar : PubMed/NCBI

146 

Bethunaickan R, Sahu R, Liu Z, Tang YT, Huang W, Edegbe O, Tao H, Ramanujam M, Madaio MP and Davidson A: Anti-tumor necrosis factor alpha treatment of interferon-α-induced murine lupus nephritis reduces the renal macrophage response but does not alter glomerular immune complex formation. Arthritis Rheum. 64:3399–3408. 2012. View Article : Google Scholar : PubMed/NCBI

147 

Monaco C, Nanchahal J, Taylor P and Feldmann M: Anti-TNF therapy: Past, present and future. Int Immunol. 27:55–62. 2015. View Article : Google Scholar : PubMed/NCBI

148 

Williams EL, Gadola S and Edwards CJ: Anti-TNF-induced lupus. Rheumatology (Oxford). 48:716–720. 2009. View Article : Google Scholar : PubMed/NCBI

149 

Aghdashi MA, Khadir M and Dinparasti-Saleh R: Antinuclear antibodies and lupus-like manifestations in rheumatoid arthritis and ankylosing spondylitis patients at 4 months' follow-up after treatment with infliximab and etanercept. Curr Rheumatol Rev. 16:61–66. 2020. View Article : Google Scholar : PubMed/NCBI

150 

Gonnet-Gracia C, Barnetche T, Richez C, Blanco P, Dehais J and Schaeverbeke T: Anti-nuclear antibodies, anti-DNA and C4 complement evolution in rheumatoid arthritis and ankylosing spondylitis treated with TNF-alpha blockers. Clin Exp Rheumatol. 26:401–407. 2008.PubMed/NCBI

151 

Ramos-Casals M, Brito-Zeron P, Munoz S, Soria N, Galiana D, Bertolaccini L, Cuadrado MJ and Khamashta MA: Autoimmune diseases induced by TNF-targeted therapies: Analysis of 233 cases. Medicine (Baltimore). 86:242–251. 2007. View Article : Google Scholar : PubMed/NCBI

152 

Santos CS, Castro CA, Morales CM and Álvarez ED: Anti-TNF-α-induced lupus syndrome: Two case reports and review of current literature. Z Rheumatol. 80:481–486. 2021. View Article : Google Scholar : PubMed/NCBI

153 

Stokes MB, Foster K, Markowitz GS, Ebrahimi F, Hines W, Kaufman D, Moore B, Wolde D and D'Agati VD: Development of glomerulonephritis during anti-TNF-alpha therapy for rheumatoid arthritis. Nephrol Dial Transplant. 20:1400–1406. 2005. View Article : Google Scholar : PubMed/NCBI

154 

Mor A, Bingham CO III, Barisoni L, Lydon E and Belmont HM: Proliferative lupus nephritis and leukocytoclastic vasculitis during treatment with etanercept. J Rheumatol. 32:740–743. 2005.PubMed/NCBI

155 

Aringer M, Graninger WB, Steiner G and Smolen JS: Safety and efficacy of tumor necrosis factor alpha blockade in systemic lupus erythematosus: An open-label study. Arthritis Rheum. 50:3161–3169. 2004. View Article : Google Scholar : PubMed/NCBI

156 

Aringer M, Steiner G, Graninger WB, Höfler E, Steiner CW and Smolen JS: Effects of short-term infliximab therapy on autoantibodies in systemic lupus erythematosus. Arthritis Rheum. 56:274–279. 2007. View Article : Google Scholar : PubMed/NCBI

157 

Aringer M, Houssiau F, Gordon C, Graninger WB, Voll RE, Rath E, Steiner G and Smolen JS: Adverse events and efficacy of TNF-alpha blockade with infliximab in patients with systemic lupus erythematosus: long-term follow-up of 13 patients. Rheumatology (Oxford). 48:1451–1454. 2009. View Article : Google Scholar : PubMed/NCBI

158 

Matsumura R, Umemiya K, Sugiyama T, Sueishi M, Umibe T, Ichikawa K and Yoshimura M; Study Group on Nephrology at the National Hospital Organization of Japan, : Anti-tumor necrosis factor therapy in patients with difficult-to-treat lupus nephritis: A prospective series of nine patients. Clin Exp Rheumatol. 27:416–421. 2009.PubMed/NCBI

159 

Hayat SJ and Uppal SS: Therapeutic efficacy and safety profile of infliximab in active systemic lupus erythematosus. Mod Rheumatol. 17:174–177. 2007. View Article : Google Scholar : PubMed/NCBI

160 

Uppal SS, Hayat SJ and Raghupathy R: Efficacy and safety of infliximab in active SLE: A pilot study. Lupus. 18:690–697. 2009. View Article : Google Scholar : PubMed/NCBI

161 

Moreland LW, Baumgartner SW, Schiff MH, Tindall EA, Fleischmann RM, Weaver AL, Ettlinger RE, Cohen S, Koopman WJ, Mohler K, et al: Treatment of rheumatoid arthritis with a recombinant human tumor necrosis factor receptor (p75)-Fc fusion protein. N Engl J Med. 337:141–147. 1997. View Article : Google Scholar : PubMed/NCBI

162 

Anderson PJ: Tumor necrosis factor inhibitors: Clinical implications of their different immunogenicity profiles. Semin Arthritis Rheum. 34 5 Suppl 1:S19–S22. 2005. View Article : Google Scholar : PubMed/NCBI

163 

Cortes-Hernandez J, Egri N, Vilardell-Tarres M and Ordi-Ros J: Etanercept in refractory lupus arthritis: An observational study. Semin Arthritis Rheum. 44:672–679. 2015. View Article : Google Scholar : PubMed/NCBI

164 

Norman R, Greenberg RG and Jackson JM: Case reports of etanercept in inflammatory dermatoses. J Am Acad Dermatol. 54 3 Suppl 2:S139–S142. 2006. View Article : Google Scholar : PubMed/NCBI

165 

Yang BB, Xiao H, Li XJ and Zheng M: Safety and efficacy of etanercept-methotrexate combination therapy in patients with rhupus: An observational study of non-glucocorticoid treatment for rheumatic diseases. Discov Med. 25:14–20. 2018.PubMed/NCBI

166 

Danion F, Sparsa L, Arnaud L, Alsaleh G, Lefebvre F, Gies V, Martin T, Lukas C, Durckel J, Ardizzone M, et al: Long-term efficacy and safety of antitumour necrosis factor alpha treatment in rhupus: An open-label study of 15 patients. RMD Open. 3:e0005552017. View Article : Google Scholar : PubMed/NCBI

167 

Micheloud D, Nuno L, Rodriguez-Mahou M, Sánchez-Ramón S, Ortega MC, Aguarón A, Junco E, Carbone J, Fernández-Cruzl E, Carreño L and López-Longo FJ: Efficacy and safety of Etanercept, high-dose intravenous gammaglobulin and plasmapheresis combined therapy for lupus diffuse proliferative nephritis complicating pregnancy. Lupus. 15:881–885. 2006. View Article : Google Scholar : PubMed/NCBI

168 

Mustafa G, Mahrosh HS, Salman M, Sharif S, Jabeen R, Majeed T and Tahir H: Identification of peptides as novel inhibitors to target IFN-γ, IL-3, and TNF-α in systemic lupus erythematosus. Biomed Res Int. 2021:11240552021. View Article : Google Scholar : PubMed/NCBI

169 

Bongartz T, Sutton AJ, Sweeting MJ, Buchan I, Matteson EL and Montori V: Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies: Systematic review and meta-analysis of rare harmful effects in randomized controlled trials. JAMA. 295:2275–2285. 2006. View Article : Google Scholar : PubMed/NCBI

170 

Tack CJ, Kleijwegt FS, Van Riel PL and Roep BO: Development of type 1 diabetes in a patient treated with anti-TNF-alpha therapy for active rheumatoid arthritis. Diabetologia. 52:1442–1444. 2009. View Article : Google Scholar : PubMed/NCBI

171 

Bloom BJ: Development of diabetes mellitus during etanercept therapy in a child with systemic-onset juvenile rheumatoid arthritis. Arthritis Rheum. 43:2606–2608. 2000. View Article : Google Scholar : PubMed/NCBI

172 

Kollias G and Kontoyiannis D: Role of TNF/TNFR in autoimmunity: Specific TNF receptor blockade may be advantageous to anti-TNF treatments. Cytokine Growth Factor Rev. 13:315–321. 2002. View Article : Google Scholar : PubMed/NCBI

173 

Zhang N, Wang Z and Zhao Y: Selective inhibition of Tumor necrosis factor receptor-1 (TNFR1) for the treatment of autoimmune diseases. Cytokine Growth Factor Rev. 55:80–85. 2020. View Article : Google Scholar : PubMed/NCBI

174 

Van Hauwermeiren F, Armaka M, Karagianni N, Kranidioti K, Vandenbroucke RE, Loges S, Roy MV, Staelens J, Puimège L, Palagani A, et al: Safe TNF-based antitumor therapy following p55TNFR reduction in intestinal epithelium. J Clin Invest. 123:2590–2603. 2013. View Article : Google Scholar : PubMed/NCBI

175 

Puimege L, Libert C and Van Hauwermeiren F: Regulation and dysregulation of tumor necrosis factor receptor-1. Cytokine Growth Factor Rev. 25:285–300. 2014. View Article : Google Scholar : PubMed/NCBI

176 

Vinay DS and Kwon BS: The tumour necrosis factor/TNF receptor superfamily: Therapeutic targets in autoimmune diseases. Clin Exp Immunol. 164:145–157. 2011. View Article : Google Scholar : PubMed/NCBI

177 

Wu T, Xie C, Wang HW, Zhou XJ, Schwartz N, Calixto S, Mackay M, Aranow C, Putterman C and Mohan C: 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. 179:7166–7175. 2007. View Article : Google Scholar : PubMed/NCBI

178 

Deng GM, Liu L and Tsokos GC: Targeted tumor necrosis factor receptor I preligand assembly domain improves skin lesions in MRL/lpr mice. Arthritis Rheum. 62:2424–2431. 2010. View Article : Google Scholar : PubMed/NCBI

179 

Jacob N, Yang H, Pricop L, Liu Y, Gao X, Zheng SG, Wang J, Gao HX, Putterman C, Koss MN, et al: Accelerated pathological and clinical nephritis in systemic lupus erythematosus-prone New Zealand mixed 2328 mice doubly deficient in TNF receptor 1 and TNF receptor 2 via a Th17-associated pathway. J Immunol. 182:2532–2541. 2009. View Article : Google Scholar : PubMed/NCBI

180 

Zhou T, Edwards CK III, Yang P, Wang Z, Bluethmann H and Mountz JD: Greatly accelerated lymphadenopathy and autoimmune disease in lpr mice lacking tumor necrosis factor receptor I. J Immunol. 156:2661–2665. 1996.PubMed/NCBI

Related Articles

Journal Cover

April-2022
Volume 49 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ghorbaninezhad F, Leone P, Alemohammad H, Najafzadeh B, Nourbakhsh NS, Prete M, Malerba E, Saeedi H, Tabrizi NJ, Racanelli V, Racanelli V, et al: Tumor necrosis factor‑α in systemic lupus erythematosus: Structure, function and therapeutic implications (Review). Int J Mol Med 49: 43, 2022
APA
Ghorbaninezhad, F., Leone, P., Alemohammad, H., Najafzadeh, B., Nourbakhsh, N.S., Prete, M. ... Baradaran, B. (2022). Tumor necrosis factor‑α in systemic lupus erythematosus: Structure, function and therapeutic implications (Review). International Journal of Molecular Medicine, 49, 43. https://doi.org/10.3892/ijmm.2022.5098
MLA
Ghorbaninezhad, F., Leone, P., Alemohammad, H., Najafzadeh, B., Nourbakhsh, N. S., Prete, M., Malerba, E., Saeedi, H., Tabrizi, N. J., Racanelli, V., Baradaran, B."Tumor necrosis factor‑α in systemic lupus erythematosus: Structure, function and therapeutic implications (Review)". International Journal of Molecular Medicine 49.4 (2022): 43.
Chicago
Ghorbaninezhad, F., Leone, P., Alemohammad, H., Najafzadeh, B., Nourbakhsh, N. S., Prete, M., Malerba, E., Saeedi, H., Tabrizi, N. J., Racanelli, V., Baradaran, B."Tumor necrosis factor‑α in systemic lupus erythematosus: Structure, function and therapeutic implications (Review)". International Journal of Molecular Medicine 49, no. 4 (2022): 43. https://doi.org/10.3892/ijmm.2022.5098