Elsevier

Pharmacological Research

Volume 115, January 2017, Pages 124-132
Pharmacological Research

Invited review
Transmembrane TNF-alpha reverse signaling leading to TGF-beta production is selectively activated by TNF targeting molecules: Therapeutic implications

https://doi.org/10.1016/j.phrs.2016.11.025Get rights and content

Abstract

Tumor necrosis factor (TNF)-α is a potent pro-inflammatory cytokine exerting pleiotropic effects on various cell types. It is synthesized in a precursor form called transmembrane TNF-α (mTNF-α) which, after being processed by metalloproteinases, is released in a soluble form to mediate its biological activities through Type 1 and 2 TNF receptors in TNF receptor expressing cells. In addition to acting in soluble form, TNF-α also acts in the transmembrane form both as a ligand by activating TNF receptors, as well as a receptor that transmits outside-to-inside (reverse) signals back into mTNF-α bearing cells. Since the discovery that TNF-α plays a determining role in the pathogenesis of several chronic inflammatory diseases, anti-TNF agents are increasingly being used in the treatment of a rapidly expanding number of rheumatic and systemic autoimmune diseases, such as rheumatoid arthritis, Crohn’s disease, psoriasis, psoriatic arthritis, ankyloting spondylitis, Wegener granulomatosis and sarcoidosis. There are 5 TNF antagonists currently available: etanercept, a soluble TNF receptor construct; infliximab, a chimeric monoclonal antibody; adalimumab and golimumab, fully human antibodies; and certolizumab pegol, an Fab’ fragment of a humanized anti-TNF-α antibody. Though each compound can efficiently neutralize TNF-α, increasing evidence suggests that they show different efficacy in the treatment of these diseases. These observations indicate that in addition to neutralizing TNF-α, other biological effects induced by TNF-α targeting molecules dictate the success of the therapy. Recently, we found that mTNF-α reverse signaling leads to transforming growth factor (TGF)-β production in macrophages and anti-TNF agents selectively trigger this pathway. In this review we will focus on the potential contribution of the activation of the mTNF-α signaling pathway to the success of the anti-TNF therapy.

Introduction

Tumor necrosis factor (TNF)-α is a powerful pro-inflammatory cytokine which plays a critical role in the pathogenesis of various chronic inflammatory diseases, such as rheumatoid arthritis [1]. Though TNF-α is a pleiotropic cytokine, it is produced primarily by cells of the monocytic lineage − such as macrophages, Langerhans cells, microglia, astroglia, Kupffer cells, and alveolar macrophages [2], [3]. In addition, other cells and tissues including lymphoid cells, endothelial cells, mast cells, fibroblasts, and neuronal tissue were also reported to express TNF-α [4]⋅ TNF-α is barely expressed by quiescent cells. Its production in macrophages can be induced by a wide variety of stimuli, including bacteria, tumor cells, immune complexes, viruses, complement factors, cytokines, irradiation, ischemia/hypoxia and trauma. The biosynthesis of TNF-α is strictly controlled. Its gene expression is regulated at the transcriptional level by several transcription factors, such as nuclear factor kappa B (NFκB) and nuclear factor activated T cells (NF-AT). The TNF-α mRNA appears within 30 min in response to most of the stimuli, but the TNF protein expression is controlled mostly post-transcriptionally. Thus, TNF-α production is also regulated at the translational level via the UA-rich sequence in the 3′ untranslated region of human TNF-α mRNA [5]. Translation of TNF mRNA leads to the intracellular production of trimeric pro-TNF protein, which matures and is inserted into the plasma membrane as transmembrane TNF-α (mTNF-α).

Transmembrane TNF-α is expressed as a cell surface II polypeptide consisting of 233 amino acid residues [6], [7], [8]. After being processed by metalloproteinases such as TNF-α-converting enzyme [9], [10], the soluble form of TNF-α (s TNF-α) of 157 amino acid residues is released and mediates its biological activities by triggering Type 1 and 2 TNF receptors (TNF-R), which also function as receptor trimers [11]. However, increasing evidences suggest that not only soluble TNF-α, but also its precursor form, mTNF-α, is involved in the regulation of the inflammatory response acting primarily locally.

TNF-R1 is constitutively expressed in most tissues, whereas protein levels of TNF-R2 are highly regulated, and the receptor typically appears in cells of the immune system. Binding of TNF-α onto TNF-R1 is considered to be irreversible, whereas binding of TNF-α onto TNF-R2 has both rapid on and off kinetics. Therefore, it is believed that TNF-R2 might act as a “ligand passer” to TNF-R1 in some cells by elevating the local concentration of TNF-α at the cell surface [12]. In the vast majority of cells, TNF-R1 is the key component in mediating TNF signaling, whereas in the lymphoid system TNF-R2 plays the determining role [11]. Because TNF-R2 can only be fully activated by the transmembrane, but not by the soluble form of TNF, it is very likely that its real contribution to the immune responses is generally underestimated [13]. The reason for this difference in binding is not fully explained yet, but the different stabilities of the individual ligand/receptor complexes very likely contribute to the phenomenon [13], [14]. Though TNF-R1 activation generally triggers pro-inflammatory responses, apoptosis-related signaling events can also be induced, whereas the TNF-R2 signal, especially in activated T cells, induces cell survival pathways that can contribute to the initiation of cell proliferation [15].

In response to an inflammatory signal, similar to mTNF-α, the extracellular domains of both TNF-Rs can also be proteolytically cleaved by matrix metalloproteinase family of enzymes, yielding soluble receptor fragments (sTNF-Rs) [16]. sTNF-Rs retain the ability to bind TNF-α, and thus function as endogenous inhibitors of it [17]. Due to the lack of cooperativity in ligand binding, however, the affinities of sTNF-Rs are low compared to their membrane-bound forms (mTNF-Rs). TACE was suggested to act as a processing enzyme for TNF-R1 cleavage as well [18], and during inflammation this cleavage plays a determining role in controlling cellular TNF responsiveness, as cleavage-resistant TNF-R1 mutations are linked with dominantly inherited autoinflammatory syndromes named TNF-R1-associated periodic syndromes [19] (Fig. 1).

Even though TNF-α participates in various normal homeostatic mechanisms including host defense or wound healing [20], dysregulated TNF-α production has been detected in a wide variety of inflammatory diseases. In addition, TNF-α-activated macrophages are well known to contribute to the immunopathology of many autoimmune diseases [21]. Thus, macrophage-derived TNF-α has been implicated in the pathomechanism of rheumatoid arthritis (RA), inflammatory bowel disease (IBD), Wegener granulomatosis (WG), sarcoidosis (S), psoriasis (P), psoriatic arthritis, ankylosing spondylitis (AS), juvenile chronic arthritis, atherosclerosis, and sepsis [22], [23], [24], [25], [26], [27].

Nowadays five TNF-α blocking agents are currently licensed for treating TNF-α-driven diseases. These five drugs are: 1) etanercept, a recombinant human soluble fusion protein of TNF-R2 coupled to the Fc portion of IgG [28]; 2) infliximab, an anti-TNF human-murine chimeric IgG1 monoclonal antibody [29]; 3) adalimumab, a human anti-human TNF-α antibody that was produced by phage display [30]; (4); golimumab, a human anti-TNF-α IgG1κ monoclonal antibody that can be administered by the patient [31]; and 5) certolizumab pegol, an Fab’ fragment of a humanized anti-TNF-α antibody [32]. Though the pharmacokinetic properties of Fab’ in vitro are usually poor, attachment of a 40 kDa polyethylene glycol (PEG) moiety markedly increased the half-life time of certolizumab to a value comparable with that of a whole antibody. These drugs are almost equal in neutralizing of both soluble and membrane-bound TNF-α [33], however, significant differences can be found as to the disease they are effective. Thus, infliximab, golimumab, adalimumab and certolizumab pegol are effective, while etanercep failed in the trials of Crohn’s disease, Wegener granulomatosis and sarcoidosis and is less effective in the treatment of skin psoriasis [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45], [46], [47], [48], [49] (Table 1). These observations suggest that in addition to the neutralization of TNF-α, other effects induced by anti-TNF targeting molecules dictate the therapeutic results.

Section snippets

Binding properties of anti-TNF-α targeting molecules to sTNF-α and to mTNF-α

A number of studies have addressed the binding of anti-TNF-α targeting molecules to soluble and membrane-bound TNF-α [33], [50], [51]. All the compounds effectively neutralize both the soluble and the membrane bound forms of TNF-α [33]. However, etanercept binding is restricted to the trimer form of TNF-α molecule and forms a 1:1 complex with the TNF trimer in which only two of the three receptor binding sites on TNF are occupied by etanercept [50]. In addition, since the p75 TNF receptor, from

Neutralizing of mTNF-α acting as a ligand

Membrane bound TNF-α, similar to soluble TNF-α, can act as a ligand via interacting with both type 1 and type 2 TNF-Rs on the surrounding cells regulating immune responses primarily locally in a strong cellular context. Thus, mTNF-α expressed by activated macrophages, monocytes, lymphocytes or freshly isolated NK cells was shown to induce apoptosis in various cell types including tumor cells, hepatocytes and HIV-infected lymphocytes [53], [54], [55], [56], [57]. Cell surface TNF-α induces

Cell-dependent cytotoxicity and antibody-dependent cell-mediated cytotoxicity

Some of the cells participating in the pro-inflammatory response express Fc receptors. Antibodies that can bind to the Fc receptors of cells can induce both cell-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC) of Fc receptor bearing cells. In line with the expectations, those TNF-α targeting molecules (such as etanercept, infliximab, adalimumab and golimumab), which are based on human IgG1 Fc (which has the capability of fixing complement and binding to Fc

Cell death of activated human peripheral blood lymphocytes and monocytes in vivo

In addition to neutralizing TNF-α, the amount of pro-inflammatory TNF-α can also be decreased by TNF-targeting molecules, if TNF-α expressing cells are efficiently eliminated by them from the body. Resistance of cells, such as T cells, to apoptosis has been proposed to play an important role in Crohn’s disease [68]. Therefore, a number of studies have examined the effect of anti-TNF-α targeting molecules to mediate apoptosis. According to these data infliximab, adalimumab and golimumab are able

Infliximab, adalimumab, golimumab and certolizumab pegol trigger TGF-β production in human macrophages, while etanercept does not

A number of studies, which tried to explain the difference in the therapeutic efficacy of TNF-α targeting molecules, found that etanercept was unique in inducing cell responses that are attributed to mTNF-α reverse signaling. Thus, infliximab, adalimumab and golimumab were able to induce apoptosis in monocyte or T cell cultures, and the apoptosis-inducing effect was dependent on serine phosphorylation of mTNF-α. Etanercept was ineffective [33], [66], [67]. Certolizumab pegol was also unable to

Possible contribution of TGF-β production to the therapeutic efficacy of TNF-targeting molecules

Etanercept is the only TNF targeting molecule, which was found to be ineffective in the treatment of three granulomatous diseases: Crohn’s disease, Wegener granulomatosis and sarcoidosis, and less effective in the treatment of psoriasis. The data available indicate that though etanercept can neutralize mTNF-α efficiently and thus it can interfere with sTNF- α and with mTNF-α as a ligand, it is the only TNF targeting molecule, which cannot trigger full mTNF-α reverse signaling, and thus it does

Conclusion

TNF targeting molecules are increasingly applied in the treatment of various chronic inflammatory diseases. All the five TNF targeting molecules used in the clinical practice efficiently neutralize both the soluble and the transmembrane form of TNF-α, thus efficiently interfere with the signaling pathways that are triggered by TNF-Rs. However, because their structure is different, they behave differently in whether they can induce CDC or ADCC, and more importantly in whether they can or cannot

Conflict of interest

The authors acknowledge no conflict of interest.

Acknowledgements

This work was supported by the National Research Fund (OTKA T104228), by TÉT_10-1-2011-0028 and by the GINOP-2.3.2-15-2016-00006 project (co-financed by the European Union and the European Regional Development Fund).

References (119)

  • J.R. Maneiro et al.

    BIOBADASER Study Group, efficacy and safety of TNF antagonists in sarcoidosis: data from the Spanish registry of biologics BIOBADASER and a systematic review

    Semin. Arthritis Rheum.

    (2012)
  • P.J. Mease et al.

    Etanercept in the treatment of psoriatic arthritis and psoriasis: a randomised trial

    Lancet

    (2000)
  • J.F. Curtis et al.

    The use of biologics in rheumatoid arthritis: current and emerging paradigms of care

    Clin. Ther.

    (2011)
  • R. Peck et al.

    Cell surface tumor necrosis factor (TNF) accounts for monocyte- and lymphocyte-mediated killing of TNF-resistant target cells

    Cell. Immunol.

    (1989)
  • E.F. Schmid et al.

    Both tumor necrosis factor receptors TNFR60 and TNFR80, are involved in signaling endothelial tissue factor expression by juxtacrine tumor necrosis factor alpha

    Blood

    (1995)
  • M. Grell et al.

    The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor

    Cell

    (1995)
  • G. Eissner et al.

    Ligands working as receptors: reverse signaling by members of the TNF superfamily enhance the plasticity of the immune system

    Cytokine Growth Factor Rev.

    (2004)
  • M. Yu et al.

    Influence of reverse signaling via membrane TNF-alpha on cytotoxicity of NK92 cells

    Eur. J. Cell Biol.

    (2009)
  • H. Mitoma et al.

    Infliximab induces potent anti-inflammatory responses by outside-to-inside signals through transmembrane TNF-α

    Gastroenterology

    (2005)
  • S. Kirchner et al.

    Effect of different tumor necrosis factor (TNF) reactive agents on reverse signaling of membrane integrated TNF in monocytes

    Cytokine

    (2004)
  • G.H. Waetzig et al.

    Differential p38 mitogen-activated protein kinase target phosphorylation in responders and nonresponders to infliximab

    Gastroenterology

    (2003)
  • J.R. Bradley

    TNF-mediated inflammatory disease

    J. Pathol.

    (2008)
  • K.D. Pfeffer et al.

    Expression and regulation of tumor necrosis factor in macrophages from cystic fibrosis patients

    Am. J. Respir. Cell. Mol. Biol.

    (1993)
  • E.A. Moelants et al.

    Regulation of TNF-α with a focus on rheumatoid arthritis

    Immunol. Cell Biol.

    (2013)
  • D. Pennica et al.

    Human tumour necrosis factor: precursor structure, expression and homology to lymphotoxin

    Nature

    (1984)
  • B. Luettiq et al.

    Evidence for the existence of two forms of membrane tumor necrosis factor: an integral protein and a molecule attached to its receptor

    J. Immunol.

    (1989)
  • M.L. Moss et al.

    Cloning of a disintegrin metalloproteinase that processes precursor tumour-necrosis factor-alpha

    Nature

    (1997)
  • R.A. Black et al.

    A metalloproteinase disintegrin that releases tumor-necrosis factor-alpha from cells

    Nature

    (1997)
  • M. Grell et al.

    The type 1 receptor (CD120a) is the high-affinity receptor for soluble tumor necrosis factor

    Proc. Natl. Acad. Sci. U. S. A.

    (1998)
  • D. Faustman et al.

    TNF receptor 2 pathway: drug target for autoimmune diseases

    Nat. Rev. Drug Discov.

    (2010)
  • D. Wallach et al.

    Soluble and cell surface receptors for tumor necrosis factor

    Agents Actions Suppl.

    (1991)
  • K.J. Van Zee et al.

    Tumor necrosis factor soluble receptors circulate during experimental and clinical inflammation and can protect against excessive tumor necrosis factor alpha in vitro and in vivo

    Proc. Natl. Acad. Sci. U. S. A.

    (1992)
  • B. Beutler et al.

    The biology of cachectin/TNF–a primary mediator of the host response

    Annu. Rev. Immunol.

    (1989)
  • R.A. Flavell

    The relationship of inflammation and initiation of autoimmune disease: role of TNF super family members

    Curr. Top. Microbiol. Immunol.

    (2002)
  • M. Feldmann et al.

    Cytokine expression and networks in rheumatoid arthritis: rationale for anti-TNF alpha antibody therapy and its mechanism of action

    J. Inflamm.

    (1995)
  • G. Kollias et al.

    The function of tumour necrosis factor and receptors in models of multi-organ inflammation rheumatoid arthritis, multiple sclerosis and inflammatory bowel disease

    Ann. Rheum. Dis.

    (1999)
  • F.C. Eberle et al.

    Recent advances in understanding psoriasis

    F1000Res.

    (2016)
  • R. Kleemann et al.

    Cytokines and atherosclerosis: a comprehensive review of studies in mice

    Cardiovasc. Res.

    (2008)
  • F. Moosig et al.

    Wegener’s granulomatosis: the current view

    Clin. Rev. Allergy Immunol.

    (2008)
  • M.E. Weinblatt et al.

    A trial of etanercept a recombinant tumor necrosis factor receptor:Fc fusion protein, in patients with rheumatoid arthritis receiving methotrexate

    N. Engl. J. Med.

    (1999)
  • R.N. Maini et al.

    Monoclonal anti-TNF alpha antibody as a probe of pathogenesis and therapy of rheumatoid disease

    Immunol. Rev.

    (1995)
  • A. den Broeder et al.

    A single dose, placebo controlled study of the fully human anti-tumor necrosis factor-alpha antibody adalimumab (D2E7) in patients with rheumatoid arthritis

    J. Rheumatol.

    (2002)
  • H. Zhou et al.

    Pharmacokinetics and safety of golimumab a fully human anti-TNF-alpha monoclonal antibody, in subjects with rheumatoid arthritis

    J. Clin. Pharmacol.

    (2007)
  • E.H. Choy et al.

    Efficacy of a novel PEGylated humanized anti-TNF fragment (CDP870) in patients with rheumatoid arthritis: a phase II double-blinded randomized, dose-escalating trial

    Rheumatology (Oxford)

    (2002)
  • A. Nesbitt et al.

    Mechanism of action of certolizumab pegol (CDP870): in vitro comparison with other anti-tumor necrosis factor alpha agents

    Inflamm. Bowel Dis.

    (2007)
  • M.E. Weinblatt et al.

    Safety and efficacy of etanercept beyond 10 years of therapy in North American patients with early and longstanding rheumatoid arthritis

    Arthritis Care Res.

    (2011)
  • C. Mukhtyar et al.

    Current state of tumour necrosis factor α blockade in Wegener’s granulomatosis

    Ann. Rheum. Dis.

    (2005)
  • T. Frech

    Treatment of ankylosing spondylitis: focus on etanercept

    Biologics

    (2007)
  • A.C. Moss

    Optimizing the use of biological therapy in patients with inflammatory bowel disease Optimizing the use of biological therapy in patients with inflammatory bowel disease

    Gastroenterol. Rep.

    (2015)
  • P. Lamprecht et al.

    Effectiveness of TNF-a blockade with infliximab in refractory Wegener’s granulomatosis

    Rheumatology

    (2002)
  • Cited by (72)

    • Lymphocyte activation gene-3 (LAG-3) regulatory T cells: An evolving biomarker for treatment response in autoimmune diseases

      2022, Autoimmunity Reviews
      Citation Excerpt :

      In RA, the combination of anti-TNFs and MTX as opposed to monotherapy has the advantage of better efficacy and reduced immunogenicity against anti-TNF agents [111,112]. Anti-TNF agents neutralize TNF cytokine, but their therapeutic efficacy results from the downstream signaling through binding to membrane TNF on monocytes (a process known as “reverse signaling”) which subsequently modulates T cell populations [113,114]. MTX specifically inhibits lymphocytes [97].

    • APOLLO: An accurate and independently validated prediction model of lower-grade gliomas overall survival and a comparative study of model performance

      2022, eBioMedicine
      Citation Excerpt :

      HMGA1 and TFG are regulated by NRF1, and can affect the prognosis of gliomas.45-47 FAS48 and SMAD449 are important members of the TNF-receptor superfamily and TGF-β signaling pathway, respectively, play a major role in tumor microenvironment and have antagonistic interactions.50 Though the biological function of the interaction between CTNND2 and GOLGA5 remains unclear, overexpressed CTNND2 is likely to increase tumor invasion of gliomas.51

    View all citing articles on Scopus
    View full text