Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Jun 8.
Published in final edited form as: J Immunol. 2008 Jun 1;180(11):7102–7106. doi: 10.4049/jimmunol.180.11.7102

Soluble IL-6R is produced by IL-6R ectodomain shedding in activated CD4 T cells

Eva M Briso *, Oliver Dienz *, Mercedes Rincon *
PMCID: PMC2692633  NIHMSID: NIHMS105489  PMID: 18490707

Abstract

IL-6 trans-signaling via the soluble IL-6R (sIL-6R) plays an important role in the progression of several autoimmune diseases and cancer by providing IL-6-responsiveness to cells lacking IL-6R. However, the potential sources of sIL-6R are less understood. In this study we show that sIL-6R is produced by both naïve and memory CD4 T cells upon TCR activation. The production of sIL-6R by activated CD4 T cells is mediated by shedding of the membrane-bound IL-6R and this process correlates with the expression of the metalloproteinase ADAM17 in these cells. In contrast to CD4 T cells, CD8 T cells do not express ADAM17 and their production of sIL-6R is negligible. Thus, during an immune response CD4 T cells are an important source of sIL-6R. Production of sIL-6R by autoreactive CD4 T cells may contribute to their role in the development of autoimmune disease by conferring IL-6-responsiveness to cells lacking IL-6R such as synoviocytes.

Keywords: T cells, cytokine receptors, cell activation, rheumatoid arthritis, cytokines

INTRODUCTION

IL-6 is a cytokine produced by professional antigen presenting cells, but also by cells from non-hematopoietic origin (1). IL-6 binds to the IL-6R present on the cell surface in association with gp130, the transmembrane signal transducer receptor that is shared by other cytokines of the same family (1). Neither IL-6R nor IL-6 binds to gp130 alone. A complex of IL-6R-IL6 is necessary for binding to gp130 and forms a hexamer consisting of two IL-6R, two IL-6 and two gp130 molecules (2). The gp130 receptor is broadly expressed in most cell types, but the expression of IL-6R is more restricted to liver cells and leukocytes (3). However, IL-6 has also been shown to have an effect on cells that normally lack IL-6R including cancer cells, neurons, osteoclasts (1). This effect is mediated by a soluble form of IL-6R (sIL-6R) containing the extracellular domain sufficient for binding to IL-6 and gp130. Once sIL-6R is secreted, it can bind to IL-6 and the complex interacts with the ubiquitously expressed gp130 in a process that has been termed trans-signaling of IL-6 (4). This process makes IL-6R-lacking cells responsive to IL-6 when there is a source that produces sIL-6R. sIL-6R and IL-6 trans-signaling have now been implicated in a number of pathological diseases, including rheumatoid arthritis and Crohn’s disease (57), as well as colon cancer (5, 8).

Two major mechanisms for the production of sIL-6R have been proposed (9). In addition to the transmembrane form of the IL-6R, a mRNA transcript lacking the transmembrane domain can be generated by alternative splicing using splicing donor and acceptor sites that flank the region coding for the transmembrane domain. The alternative splicing also introduces a frameshift leading to the incorporation of 10 additional amino acids at the C-terminus of the sIL-6R (10, 11). A second mechanism for the generation of sIL-6R is the proteolytic cleavage (“shedding”) of the membrane-bound IL-6R at the cell surface interface through a putative proteolytic site immediately prior to the transmembrane domain (12). Initial studies using pharmacological inhibitors suggested that the cleavage of the membrane-bound IL-6R is mediated by metalloproteinases (13, 14). Recent studies in monocytes, fibroblasts and cancer cell lines have shown that two specific members of the ADAM (a disintegrin and metalloproteinase domain) family of zinc-dependent metalloproteinases, ADAM17 (also named TACE for TNFα-converting enzyme) and ADAM10, contribute to the IL-6R shedding (15, 16).

Less is known about the potential cellular sources that produce this receptor. Secretion of sIL-6R has been reported in neutrophils, monocytic and hepatocytic cell lines and human T lymphotropic virus type I (HTLV1)-transformed, but not nontransformed, T cell lines (6, 11, 17, 18). In this study we show for the first time that human naïve and memory CD4 T cells, but not CD8 T cells, produce sIL-6R upon TCR activation. The secretion of sIL-6R is primarily mediated by shedding of the membrane-bound receptor and correlates with the expression of ADAM17 in CD4 T cells.

MATERIAL AND METHODS

Isolation and activation of human CD4 T cells

PBMCs were obtained from healthy volunteers according to institutional research protection guidelines. CD4 and CD8 T cells were purified by positive selection using MACS kits as recommended by the manufacturer (Miltenyi Biotech). Isolation of naïve and memory CD4 T cells was performed by FACS sorting. Cells were activated with plate-bound anti-CD3 mAb (3 μg/ml) and soluble anti-CD28 mAb (2 μg/ml) (BD Pharmingen). 50 μM TNFα protease inhibitor (TAPI)-0 (Peptides International) was used.

RT-PCR

Total RNA was isolated from cells using the RNeasy Kit (Qiagen). RT-PCR was performed to examine the expression of membrane-bound IL-6R and alternative spliced sIL-6R using the IL-6R-sense (5′-cattgccattgttctgaggttc-3′) or sIL-6R-sense: (5′-gcgacaagcctcccaggttc-3′) and the shared IL-6R-anti-sense (5′-gtgccacccagccagctatc-3′) oligonucleotides as previously described (19). ADAM17 and ADAM10 expression was examined by RT-PCR using the previously described primers (20).

ELISA

Human soluble IL-6R production was determined by ELISA using the DuoSet kit as recommended by the manufacturer (R&D). IL-2 production was determined by ELISA using the capture (2 μg/ml) and biotinylated (1 μg/ml) (BD Pharmingen) antibodies.

Flow cytometry analysis

The following mAbs were used for staining: anti-CD14, CD8, CD4, CD62L (Caltag), and IL-6R (BD Pharmingen). Cells were analyzed using the LSR II flow cytometer (BD Bioscience). For isolation of naïve and memory CD4 T cells, PBMC were stained with anti-CD4, CD45RO (Caltag) and CD45RA (Biolegend) mAbs, and CD4 CD45RA and CD4 CD45RO populations were isolated by FACS sorting using the FACSARIA instrument (BD Bioscience).

RESULTS AND DISCUSSION

TCR signals induce the secretion of soluble IL-6R in CD4 T cells, but not CD8 T cells

Soluble IL-6R and IL-6 trans-signaling have been involved in a number of inflammatory diseases (4, 5, 9). IL-6 is known to be produced by a variety of cells. However, it remains unclear which cells produce sIL-6R. It is believed that neutrophils and macrophages are most likely the major source of sIL-6R. Because CD4 T cells play an important role in autoimmune diseases, we examined the production of sIL-6R in this population. CD4 T cells were purified from peripheral blood of healthy volunteers and activated with anti-CD3 and anti-CD28 mAbs for different periods of time. Secretion of sIL-6R into culture supernatant was determined by ELISA. Significant levels of sIL-6R were detected already one day upon activation, and these increased over the activation period (Fig. 1A). No sIL-6R could be detected in CD4 T cells incubated in medium alone in the absence of TCR signals (data not shown). Unlike CD4 T cells, CD8 T cells do not seem to play a major role in autoimmune diseases. To determine whether CD8 T cells are also capable of secreting sIL-6R we examined its production by purified CD8 T cells upon activation with anti-CD3 and anti-CD28 mAbs. In contrast to CD4 T cells, CD8 T cells produced minimal levels of sIL-6R which did not increase over the time of activation (Fig. 1B). Analysis of IL-2 production by CD8 T cells showed that these cells were activated since they produced high levels of IL-2 despite their inability to secrete sIL-6R (Fig. 1C). The purity (>95%) in both populations was comparable, and monocytes could not be detected in either purified CD4 or CD8 T cells (Fig. 1D), indicating that the differential production of sIL-6R was not due to a difference in the purity of these cells. CD4 T cells, but not CD8 T cells, are therefore a major source of sIL-6R upon TCR activation. Analysis of membrane-bound IL-6R in CD4 and CD8 T cells by flow cytometry showed increased levels of this receptor on the cell surface of CD4 T cells compared with the levels on CD8 T cells (Fig. 1E), suggesting that CD4 T cells could also help CD8 T cells by providing sIL-6R and increasing their responsiveness to IL-6.

Figure 1.

Figure 1

Production of sIL-6R by human CD4 T cells upon TCR stimulation. A, Human CD4 T cells were activated with anti-CD3 and anti-CD28 mAbs for the indicated periods of time, and sIL-6R production was examined by ELISA. The mean ± SD of three independent experiments with cells from different donors is shown. B, Purified CD4 and CD8 T cells were activated with anti-CD3 and anti-CD28 mAbs, and sIL-6R production was examined by ELISA (data represent the mean ± SD of triplicate determinations). A representative experiment of two performed is shown. C, CD8 T cells were activated as in (B), and IL-2 production was examined. D, CD4 and CD8 T cells were stained for CD4 or CD8 and CD14, and analyzed by flow cytometry. Numbers indicate the percentage of the corresponding population. E, CD4 (thin line) and CD8 (thick line) T cells were stained for CD4 or CD8 and IL-6R and analyzed by flow cytometry analysis. Histograms show the fluorescence intensity for IL-6R in each population. Filled histogram represents unstained cells.

Secretion of sIL-6R by CD4 T cells is mediated by the shedding of membrane-bound IL-6R

sIL-6R can be produced by alternative splicing of the mRNA that normally encodes for the membrane-bound form of IL-6R, or by proteolytic cleavage of the membrane-bound form. To determine whether the sIL-6R secreted by CD4 T cells was produced by alternative splicing of the IL-6R mRNA induced upon activation we examined the expression of the two isoforms (i.e. membrane-bound and secreted) by RT-PCR using RNA from CD4 T cells prior to and after activation. Both the membrane-bound and the soluble isoforms of IL-6R were already present in CD4 T cells prior to stimulation (Fig. 2A) and their expression was slightly decreased with activation (Fig. 2A), further demonstrated by semiquantitative analysis (data not shown). Thus, the production of sIL-6R by activated CD4 T cells does not seem to be caused by switching from the membrane-bound to the soluble alternatively spliced isoform. In addition, since no sIL-6R was detected if CD4 T cells were incubated in medium alone in the absence of anti-CD3 and anti-CD28 mAbs (data not shown), these results also show that the presence of the alternatively spliced IL-6R mRNA is not indicative of protein expression and secretion of the sIL-6R.

Figure 2.

Figure 2

Production of sIL-6R by CD4 T cells is due to shedding of the membrane-bound IL-6R. A, Total RNA was isolated from unstimulated CD4 T cells (u.s.) and from cells activated with anti-CD3 and anti-CD28 mAbs. Expression of membrane-bound IL-6R (mb IL6R) and sIL-6R generated by alternative splicing (as sIL-6R) was examined by RT-PCR. HPRT was also examined as an endogenous control. A representative experiment of four performed is shown. B, Cell surface expression of membrane-bound IL-6R was examined in unstimulated CD4 T cells (filled histogram, u.s.), and in CD4 T cells activated with anti-CD3 and anti-CD28 mAbs for 24 h (thin line) or 48 h (thick line) by flow cytometry analysis. Horizontal line denotes the gate relative to unstained cells. C, CD4 T cells were activated with anti-CD3 and anti-CD28 mAbs in the absence or presence of TAPI. sIL-6R production was determined by ELISA. A representative experiment of four performed is shown. D, CD4 T cells were activated as described in (C). IL-2 production was examined by ELISA. The mean ± SD of three independent experiments is shown.

Analysis of the IL-6R expression on the cell surface of CD4 T cells during activation by flow cytometry showed that the levels of IL-6R decreased significantly upon activation (Fig. 2B). This downmodulation of IL-6R on the surface during activation could be associated with the production of sIL-6R by shedding of the membrane-bound form. Cleavage of the membrane-bound sIL-6R is mediated primarily by specific members of the ADAM family of metalloproteinases. To determine whether production of sIL-6R by CD4 T cells could be due to the shedding of membrane-bound IL-6R, we examined the effect of TAPI, an inhibitor of metalloproteinases that has been shown to prevent IL-6R shedding in other cell types (13, 14). CD4 T cells were activated in the presence or absence of TAPI and the production of sIL-6R in the supernatant was measured by ELISA. The presence of TAPI during activation practically abolished the production of sIL-6R (Fig. 2C). No effect of TAPI on survival and/or cell expansion was observed (data not shown), indicating that the effect was not due to toxicity of TAPI in these cells. Furthermore, analysis of IL-2 production showed that the levels of IL-2 in the supernatant were not decreased by the presence of TAPI (Fig. 2D). Together, these results indicate that CD4 T cells produce high levels sIL-6R primarily by metalloproteinase-mediated shedding of the membrane-bound IL-6R form. The progressive increase in the levels of sIL-6R during the activation is probably due to the continuous expression and shedding of membrane-bound IL-6R. Since low levels of sIL-6R could still be detected in the presence of TAPI, we cannot rule out that a minimal fraction of the secreted sIL-6R may also be derived from the expression of the alternatively spliced form.

Differential expression of ADAM17 and ADAM10 in CD4 and CD8 T cells

ADAM10 and ADAM17 have been shown to contribute to the shedding of IL-6R and production of sIL-6R in monocytes, fibroblasts or osteogenic cell lines. The expression of ADAMs is not well characterized in CD4 T cells. Thus, to determine whether these two metalloproteinases could also be the target of TAPI in CD4 T cells, we examined their expression in these cells by RT-PCR. ADAM17 mRNA was readily detected in freshly isolated CD4 T cells and its expression was slightly upregulated during T cell activation (Fig. 3A), further demonstrated by semiquantitative analysis (data not shown). The levels of ADAM10 mRNA in CD4 T cells prior to stimulation were almost undetectable, but appeared to increase later during activation (Fig. 3A). Thus, secretion of sIL-6R by shedding in CD4 T cells is most likely mediated by ADAM17, but ADAM10 may also contribute.

Figure 3.

Figure 3

Expression of ADAM17 and ADAM10 in CD4 and CD8 T cells. A, ADAM17 and ADAM10 mRNA levels were examined in unstimulated CD4 T cells (u.s.) and CD4 T cells stimulated with anti-CD3 and anti-CD28 mAbs by RT-PCR. A representative experiment of three performed is shown. B, CD4 T cells were treated (thick line) or non-treated (thin line) with anti-CD3 and anti-CD28 mAbs for 60 min. IL-6R and CD62L were examined by flow cytometry analysis. Unstained cells are shown by the filled histograms. C, CD4 T cells were treated with medium alone (thin line), PMA (thick line) or PMA and TAPI (dashed line) for 60 min. IL-6R and CD62L were examined by flow cytometry. D, Expression of ADAM17 in unstimulated CD4 and CD8 T cells, as well as in CD8 T cells activated as in (A) was examined by RT-PCR.

It is believed that shedding mediated by ADAM17 in T cells occurs very rapidly upon activation since the cell surface expression of CD62L (another ADAM17 target) can be downregulated within 30 to 60 min of activation (2123). Analysis of IL-6R expression in CD4 T cells upon 60 min of activation with anti-CD3 and anti-CD28 mAbs showed no significant reduction in the cell surface levels (Fig. 3B). However, no reduction in CD62L cell surface levels was observed either (Fig. 3B). Similarly, minimal changes in the cell surface levels for either molecule could be found after 4 h of activation (data not shown). Since most studies showing a rapid shedding of CD62L were performed using PMA as stimulus, we also examined the expression of IL-6R and CD62L after 60 min of treatment with PMA. Interestingly, PMA almost abrogated cell surface expression of both CD62L and IL-6R, and this effect was blocked by TAPI (Fig. 3C). Thus, shedding of IL-6R in CD4 T cells is rapidly induced by PMA, but it seems to be delayed and progressive in response to TCR-mediated signals.

Because CD8 and CD4 T cells differ in their ability to produce sIL-6R, we examined the expression of ADAM10 and ADAM17 in CD8 T cells. The levels of ADAM17 in CD8 T cells prior to stimulation were almost undetectable compared with the levels in CD4 T cells (Fig. 3D). ADAM17 expression however seems to be induced later during the activation of CD8 T cells (Fig. 3D). The expression of ADAM10 in CD8 T cells was also practically undetectable, similarly to the levels in CD4 T cells (data not shown). The lack of ADAM10 and ADAM17 in CD8 T cells could explain the inability of these cells to cleave the membrane-bound IL-6R and produce sIL-6R upon activation.

Naïve and memory CD4 T cells can produce sIL-6R upon activation

While naïve CD4 T cells produce very limited amounts of effector cytokines such as IL-4 or IFNγ, memory cells have the ability to produce large amounts of a variety of cytokines very rapidly upon activation. We determined whether only memory or both naïve and memory populations contribute to the production of sIL-6R observed in total CD4 T cells. We first examined the cell surface expression of membrane-bound IL-6R in naïve (CD45RA+) and memory (CD45RO+) CD4 T cells by flow cytometry analysis. The levels of membrane-bound IL-6R were comparable in both populations (Fig. 4A). To examine the presence of the alternatively spliced sIL-6R in each population, naïve and memory CD4 cells were isolated from PBMCs by FACS sorting. Similar to the expression on the surface, no difference in mRNA levels for membrane-bound IL-6R was observed between naïve and memory CD4 cells (Fig. 4B). In contrast, increased levels of the alternatively spliced form of sIL-6R were detected in memory relative to naïve CD4 T cells (Fig. 4B).

Figure 4.

Figure 4

Naïve and memory CD4 T cells produce sIL-6R upon activation. A, PBMCs were stained for CD4, CD45RO, CD45RA and IL-6R, and analyzed by flow cytometry. Profiles show cell surface expression of IL-6R in gated CD4 CD45RA (naïve) (thin line) and CD4 CD45RO (memory) (thick line) cells. Filled histogram represents unstained cells. B, Naïve and memory cells were purified by FACS sorting. The expression of sIL-6R generated by alternative splicing (as sIL-6R) and membrane-bound IL-6R (mb IL-6R) was examined by RT-PCR. A representative experiment of three performed is shown. C, FACS-sorted naïve and memory cells were activated with anti-CD3 and anti-CD28 mAbs, and sIL-6R production was determined by ELISA. A representative experiment of three performed is shown. D, FACS-sorted naïve cells were activated with anti-CD3 and anti-CD28 mAbs in the presence or absence of TAPI, and sIL-6R production was determined by ELISA. E, Expression of ADAM17 in FACS-sorted naïve and memory CD4 T cells was examined by RT-PCR.

We therefore examined the production of sIL-6R in both CD4 T cell populations upon activation. FACS sorted naïve and memory CD4 cells were activated and sIL-6R production was examined. The levels of sIL-6R produced by naïve CD4 cells were comparable to those produced by memory CD4 T cells (Fig. 4C). These results indicate that both populations were capable of producing sIL-6R despite the differential expression of the alternatively spliced form, and suggested that shedding of the membrane-bound IL-6R is the major mechanism that contributes to the production of sIL-6R in naïve CD4 T cells. Analysis of sIL-6R production in naïve cells activated in the presence of TAPI further showed that sIL-6R production in these cells is primarily due to the shedding of membrane-bound IL-6R (Fig. 4D). We also examined the expression of ADAM17 in naïve and memory CD4 T cells by RT-PCR. ADAM17 was similarly expressed in both cell populations (Fig. 4E). No expression of ADAM10 was detected in either population (data not shown), further supporting that shedding of sIL-6R is primarily mediated by ADAM17.

In summary, our data show for the first time that both naïve and memory human CD4 T cells secrete sIL-6R upon TCR activation. In addition, we also demonstrate that despite the constitutive expression of the alternatively spliced form of the sIL-6R in CD4 T cells, shedding of the membrane-bound IL-6R is the major mechanism that contributes to the secretion of sIL-6R by these cells. Secretion of sIL-6R by shedding of the membrane-bound IL-6R correlates with the presence of ADAM17 in CD4 T cells prior to stimulation.

Although macrophages and neutrophils are believed to be the major source of sIL-6R, our data indicate that CD4 T cells are also capable of producing sIL-6R upon TCR activation. While several studies have examined the effect of sIL-6R and IL-6 trans-signaling in CD4 T cells (24, 25), this is the first report showing that CD4 T cells indeed represent an additional and likely critical source of sIL-6R during an immune response. The ability of producing sIL-6R however is restricted to CD4 T cells since CD8 T cells did not secrete significant levels of sIL-6R. Although naïve CD4 T cells display limited ability as effector cells since they cannot produce significant amounts of cytokines, we show here that they can produce substantial amounts of sIL-6R. Thus, naïve CD4 T cells may also act as effector cells by providing responsiveness to IL-6. Since IL-6 plays a critical role in generating IL-17 expressing Th17 cells (26), the sIL-6R produced by activated CD4 T cells may enhance IL-6 responsiveness and therefore Th17 differentiation of naïve CD4 T cells.

The role of sIL-6R in arthritis has been well described (7), but it remains unclear which cells are responsible for the generation of sIL-6R. CD4 T cells are clearly involved in the development and progression of rheumatoid arthritis. It is therefore possible that autoreactive CD4 T cells present in the joints, instead of macrophages, are the main source of sIL-6R, while macrophages and/or fibroblasts could be the major source of IL-6.

Acknowledgments

We thank Colette Charland (Flow Cytometry Facility) for technical assistance with flow cytometry analysis and cell sorting.

Footnotes

This work was supported by the National Institute of Health grant PO1 AI045666 (M.R.).

References

  • 1.Kamimura D, Ishihara K, Hirano T. IL-6 signal transduction and its physiological roles: the signal orchestration model. Rev Physiol Biochem Pharmacol. 2003;149:1–38. doi: 10.1007/s10254-003-0012-2. [DOI] [PubMed] [Google Scholar]
  • 2.Boulanger MJ, Chow DC, Brevnova EE, Garcia KC. Hexameric structure and assembly of the interleukin-6/IL-6 alpha-receptor/gp130 complex. Science. 2003;300:2101–2104. doi: 10.1126/science.1083901. [DOI] [PubMed] [Google Scholar]
  • 3.Saito M, Yoshida K, Hibi M, Taga T, Kishimoto T. Molecular cloning of a murine IL-6 receptor-associated signal transducer, gp130, and its regulated expression in vivo. J Immunol. 1992;148:4066–4071. [PubMed] [Google Scholar]
  • 4.Scheller J, Rose-John S. Interleukin-6 and its receptor: from bench to bedside. Med Microbiol Immunol. 2006;195:173–183. doi: 10.1007/s00430-006-0019-9. [DOI] [PubMed] [Google Scholar]
  • 5.Rose-John S, Scheller J, Elson G, Jones SA. Interleukin-6 biology is coordinated by membrane-bound and soluble receptors: role in inflammation and cancer. J Leukoc Biol. 2006;80:227–236. doi: 10.1189/jlb.1105674. [DOI] [PubMed] [Google Scholar]
  • 6.Atreya R, Mudter J, Finotto S, Mullberg J, Jostock T, Wirtz S, Schutz M, Bartsch B, Holtmann M, Becker C, Strand D, Czaja J, Schlaak JF, Lehr HA, Autschbach F, Schurmann G, Nishimoto N, Yoshizaki K, Ito H, Kishimoto T, Galle PR, Rose-John S, Neurath MF. Blockade of interleukin 6 trans signaling suppresses T-cell resistance against apoptosis in chronic intestinal inflammation: evidence in crohn disease and experimental colitis in vivo. Nat Med. 2000;6:583–588. doi: 10.1038/75068. [DOI] [PubMed] [Google Scholar]
  • 7.Nowell MA, Richards PJ, Horiuchi S, Yamamoto N, Rose-John S, Topley N, Williams AS, Jones SA. Soluble IL-6 receptor governs IL-6 activity in experimental arthritis: blockade of arthritis severity by soluble glycoprotein 130. J Immunol. 2003;171:3202–3209. doi: 10.4049/jimmunol.171.6.3202. [DOI] [PubMed] [Google Scholar]
  • 8.Becker C, Fantini MC, Schramm C, Lehr HA, Wirtz S, Nikolaev A, Burg J, Strand S, Kiesslich R, Huber S, Ito H, Nishimoto N, Yoshizaki K, Kishimoto T, Galle PR, Blessing M, Rose-John S, Neurath MF. TGF-beta suppresses tumor progression in colon cancer by inhibition of IL-6 trans-signaling. Immunity. 2004;21:491–501. doi: 10.1016/j.immuni.2004.07.020. [DOI] [PubMed] [Google Scholar]
  • 9.Jones SA, Horiuchi S, Topley N, Yamamoto N, Fuller GM. The soluble interleukin 6 receptor: mechanisms of production and implications in disease. Faseb J. 2001;15:43–58. doi: 10.1096/fj.99-1003rev. [DOI] [PubMed] [Google Scholar]
  • 10.Lust JA, Donovan KA, Kline MP, Greipp PR, Kyle RA, Maihle NJ. Isolation of an mRNA encoding a soluble form of the human interleukin-6 receptor. Cytokine. 1992;4:96–100. doi: 10.1016/1043-4666(92)90043-q. [DOI] [PubMed] [Google Scholar]
  • 11.Horiuchi S, Koyanagi Y, Zhou Y, Miyamoto H, Tanaka Y, Waki M, Matsumoto A, Yamamoto M, Yamamoto N. Soluble interleukin-6 receptors released from T cell or granulocyte/macrophage cell lines and human peripheral blood mononuclear cells are generated through an alternative splicing mechanism. Eur J Immunol. 1994;24:1945–1948. doi: 10.1002/eji.1830240837. [DOI] [PubMed] [Google Scholar]
  • 12.Mullberg J, Oberthur W, Lottspeich F, Mehl E, Dittrich E, Graeve L, Heinrich PC, Rose-John S. The soluble human IL-6 receptor. Mutational characterization of the proteolytic cleavage site. J Immunol. 1994;152:4958–4968. [PubMed] [Google Scholar]
  • 13.Mullberg J, Durie FH, Otten-Evans C, Alderson MR, Rose-John S, Cosman D, Black RA, Mohler KM. A metalloprotease inhibitor blocks shedding of the IL-6 receptor and the p60 TNF receptor. J Immunol. 1995;155:5198–5205. [PubMed] [Google Scholar]
  • 14.Jones SA, Horiuchi S, Novick D, Yamamoto N, Fuller GM. Shedding of the soluble IL-6 receptor is triggered by Ca2+ mobilization, while basal release is predominantly the product of differential mRNA splicing in THP-1 cells. Eur J Immunol. 1998;28:3514–3522. doi: 10.1002/(SICI)1521-4141(199811)28:11<3514::AID-IMMU3514>3.0.CO;2-T. [DOI] [PubMed] [Google Scholar]
  • 15.Matthews V, Schuster B, Schutze S, Bussmeyer I, Ludwig A, Hundhausen C, Sadowski T, Saftig P, Hartmann D, Kallen KJ, Rose-John S. Cellular cholesterol depletion triggers shedding of the human interleukin-6 receptor by ADAM10 and ADAM17 (TACE) J Biol Chem. 2003;278:38829–38839. doi: 10.1074/jbc.M210584200. [DOI] [PubMed] [Google Scholar]
  • 16.Althoff K, Reddy P, Voltz N, Rose-John S, Mullberg J. Shedding of interleukin-6 receptor and tumor necrosis factor alpha. Contribution of the stalk sequence to the cleavage pattern of transmembrane proteins. Eur J Biochem. 2000;267:2624–2631. doi: 10.1046/j.1432-1327.2000.01278.x. [DOI] [PubMed] [Google Scholar]
  • 17.Jostock T, Mullberg J, Ozbek S, Atreya R, Blinn G, Voltz N, Fischer M, Neurath MF, Rose-John S. Soluble gp130 is the natural inhibitor of soluble interleukin-6 receptor transsignaling responses. Eur J Biochem. 2001;268:160–167. doi: 10.1046/j.1432-1327.2001.01867.x. [DOI] [PubMed] [Google Scholar]
  • 18.Horiuchi S, Yamamoto N, Dewan MZ, Takahashi Y, Yamashita A, Yoshida T, Nowell MA, Richards PJ, Jones SA, Yamamoto N. Human T-cell leukemia virus type-I Tax induces expression of interleukin-6 receptor (IL-6R): Shedding of soluble IL-6R and activation of STAT3 signaling. Int J Cancer. 2006;119:823–830. doi: 10.1002/ijc.21918. [DOI] [PubMed] [Google Scholar]
  • 19.Nakanishi H, Yoshioka K, Joyama S, Araki N, Myoui A, Ishiguro S, Ueda T, Yoshikawa H, Itoh K. Interleukin-6/soluble interleukin-6 receptor signaling attenuates proliferation and invasion, and induces morphological changes of a newly established pleomorphic malignant fibrous histiocytoma cell line. Am J Pathol. 2004;165:471–480. doi: 10.1016/S0002-9440(10)63312-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Allinson TM, Parkin ET, Condon TP, Schwager SL, Sturrock ED, Turner AJ, Hooper NM. The role of ADAM10 and ADAM17 in the ectodomain shedding of angiotensin converting enzyme and the amyloid precursor protein. Eur J Biochem. 2004;271:2539–2547. doi: 10.1111/j.1432-1033.2004.04184.x. [DOI] [PubMed] [Google Scholar]
  • 21.Borland G, Murphy G, Ager A. Tissue inhibitor of metalloproteinases-3 inhibits shedding of L-selectin from leukocytes. J Biol Chem. 1999;274:2810–2815. doi: 10.1074/jbc.274.5.2810. [DOI] [PubMed] [Google Scholar]
  • 22.Galkina E, Tanousis K, Preece G, Tolaini M, Kioussis D, Florey O, Haskard DO, Tedder TF, Ager A. L-selectin shedding does not regulate constitutive T cell trafficking but controls the migration pathways of antigen-activated T lymphocytes. J Exp Med. 2003;198:1323–1335. doi: 10.1084/jem.20030485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Li N, Boyd K, Dempsey PJ, Vignali DA. Non-cell autonomous expression of TNF-alpha-converting enzyme ADAM17 is required for normal lymphocyte development. J Immunol. 2007;178:4214–4221. doi: 10.4049/jimmunol.178.7.4214. [DOI] [PubMed] [Google Scholar]
  • 24.Dominitzki S, Fantini MC, Neufert C, Nikolaev A, Galle PR, Scheller J, Monteleone G, Rose-John S, Neurath MF, Becker C. Cutting edge: trans-signaling via the soluble IL-6R abrogates the induction of FoxP3 in naive CD4+CD25 T cells. J Immunol. 2007;179:2041–2045. doi: 10.4049/jimmunol.179.4.2041. [DOI] [PubMed] [Google Scholar]
  • 25.Doganci A, Eigenbrod T, Krug N, De Sanctis GT, Hausding M, Erpenbeck VJ, Haddad el B, Lehr HA, Schmitt E, Bopp T, Kallen KJ, Herz U, Schmitt S, Luft C, Hecht O, Hohlfeld JM, Ito H, Nishimoto N, Yoshizaki K, Kishimoto T, Rose-John S, Renz H, Neurath MF, Galle PR, Finotto S. The IL-6R alpha chain controls lung CD4+CD25+ Treg development and function during allergic airway inflammation in vivo. J Clin Invest. 2005;115:313–325. doi: 10.1172/JCI22433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F. Interleukins 1β and 6 but not transforming growth factor-β are essential for the differentiation of interleukin 17-producing human T helper cells. Nat Immunol. 2007;8:942–949. doi: 10.1038/ni1496. [DOI] [PubMed] [Google Scholar]

RESOURCES