Next Article in Journal
Techniques and Strategies in Drug Design and Discovery
Previous Article in Journal
Activated Inositol Phosphate, Substrate for Synthesis of Prostaglandylinositol Cyclic Phosphate (Cyclic PIP)—The Key for the Effectiveness of Inositol-Feeding
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Nonspecific Inhibition of IL6 Family Cytokine Signalling by Soluble gp130

by
Anissa A. Widjaja
1,* and
Stuart A. Cook
1,2,3,*
1
Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857, Singapore
2
National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore 169609, Singapore
3
MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London W6 8RF, UK
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(3), 1363; https://doi.org/10.3390/ijms25031363
Submission received: 21 December 2023 / Revised: 17 January 2024 / Accepted: 21 January 2024 / Published: 23 January 2024
(This article belongs to the Section Molecular Biology)

Abstract

:
IL6 is a proinflammatory cytokine that binds to membrane-bound IL6 receptor (IL6R) or soluble IL6R to signal via gp130 in cis or trans, respectively. We tested the hypothesis that sgp130Fc, which is believed to be a selective IL6 trans-signalling inhibitor, is in fact a non-specific inhibitor of gp130 signalling. In human cancer and primary cells, sgp130Fc inhibited IL6, IL11, OSM and CT1 cis-signalling. The IC50 values of sgp130Fc for IL6 and OSM cis-signalling were markedly (20- to 200-fold) lower than the concentrations of sgp130Fc used in mouse studies and clinical trials. sgp130 inhibited IL6 and OSM signalling in the presence of an ADAM10/17 inhibitor and the absence of soluble IL6R or OSMR, with effects that were indistinguishable from those of a gp130 neutralising antibody. These data show that sgp130Fc does not exclusively block IL6 trans-signalling and reveal instead that broad inhibition of gp130 signalling likely underlies its therapeutic effects. This proposes global or modular inhibition of gp130 as a therapeutic approach for treating human disease.

1. Introduction

Interleukin 6 (IL6) is a proinflammatory cytokine that binds membrane-bound IL6R to signal in cis via the shared glycoprotein 130 (gp130) receptor. Soluble IL6 receptor (sIL6R), which is released from cell membranes by ADAM proteases, can also bind to IL6 to form IL6:sIL6R complexes that amplify IL6 signalling in autocrine and activate gp130 in cells lacking IL6R, in paracrine [1,2,3,4]. This secondary signalling mechanism is termed trans-signalling and is thought to be strongly pro-inflammatory and disease causing, as opposed to IL6 cis-signalling that is believed homeostatic [2]. gp130 itself can be cleaved from the plasma membrane to form soluble gp130 (sgp130) that acts as an inhibitor of IL6 trans-signalling by decoying/trapping IL6:sIL6R complexes [5].
The field of IL6 trans-signalling relies heavily on experiments using a synthetic protein i.e., sgp130Fc [6] that comprises two sgp130 molecules linked to an IgG1 antibody Fc domain. sgp130Fc is taught as a specific inhibitor of IL6 trans-signalling that leaves IL6 cis-signalling intact. sgp130 administered to rodents is protective against diseases in multiple mouse and rat models, with superiority over anti-IL6/IL6R monotherapy in some instances [7,8,9]. Following proof-of-concept (POC) therapeutic studies in rodents, sgp130Fc was developed further as an anti-inflammatory drug (olamkicept), which is now in clinical trials [2,6,10,11]. sgp130Fc has been described as “the first selective inhibitor of IL6 trans-signalling” [2] and “a decoy protein that exclusively blocks IL6 proinflammatory trans-signalling” [11].
While sgp130 is widely recognized and accepted as a specific inhibitor of IL6 trans-signalling, it was suggested to inhibit Oncostatin M (OSM) and Leukaemia Inhibitory Factor (LIF) signalling when used at high concentrations in early publications [1,5,6,12]. We postulated that sgp130Fc might—at high concentration—act as a decoy for membrane bound IL6:IL6R and/or Interleukin 11 (IL11):IL11RA cis-signalling complexes. Additionally, sgp130 may also directly serve as a decoy for OSM, which is unable to signal in trans as it does not bind to its alpha receptors until after it has formed a low affinity complex with gp130 [13,14,15,16].
Here, we tested the hypothesis that sgp130Fc, at concentrations relevant to those used in mouse experiments and in clinical trials [7,10,11,17], is a non-specific inhibitor of IL6 family cytokine signalling, rather than being a selective inhibitor of IL6 trans-signalling.

2. Results

2.1. sgp130Fc Inhibits IL6, IL11, OSM, and CT1 Signalling

We tested our hypothesis by conducting experiments in a lung epithelial cancer cell line (A549), primary human hepatocytes and primary human hepatic stellate cells (HSCs, a stromal cell type), which express the diversity of IL6 family cytokine receptors, including IL11RA. This approach helps to avoid potential bias in IL6 family receptor expression that might occur in a single cell type and tests whether detected effects are cell-type specific or more generalisable.
For IL6 or IL11 to signal in trans, IL6R or IL11RA must first be cleaved from the plasma membrane, which is primarily achieved by ADAM10/17 [3,4]. We screened the supernatants of cells incubated (24 h) in media for the presence of sIL6R and sIL11RA and extended analyses to sOSMRβ (sOSMR) and sLIFR (Figure 1A). sIL6R was only detected in A549 supernatants with levels below the lower limit of detection (LLOD: 15.2 pg/mL) in hepatocytes and HSCs. sIL11RA was abundant in all supernatants with highest levels seen for hepatocytes, sOSMR was undetectable in all supernatants (LLOD: 0.094 ng/mL), and sLIFR was equally detected in A549 and hepatocyte conditioned media but not in HSC media (LLOD: 0.188 ng/mL) (Figure 1A).
In the presence or absence of sgp130Fc (5 µg/mL), we assessed the phosphorylation of STAT3 (p-STAT3) in response to IL6 family cytokines that have been reported to signal in trans (IL6 and IL11), cytokines not known to signal in trans (Cardiotrophin 1 (CT1), ciliary neurotrophic factor (CNTF), and LIF), and an IL6 family cytokine previously reported as incapable of trans-signalling (OSM).
While there was some variation in the p-STAT3/STAT3 response to cytokine stimulation, results were similar across cell types (Figure 1B–D and Supplementary Figure S1A–C). For IL6, IL11, CT1 and OSM, p-STAT3/STAT3 ratios were significantly inhibited by sgp130Fc. CNTF and LIF-stimulated STAT3 activation were unaffected by the presence of sgp130Fc.
It was notable that following OSM stimulation (15 min), total STAT3 levels were diminished and p-STAT3/STAT3 ratios notably increased, which would be consistent with phosphorylation-induced protein degradation secondary to strong phosphorylation of STAT3 by OSM, as compared to other IL6 family cytokines [18,19]. The specificity of this effect was confirmed by probing samples stimulated with OSM for GAPDH, which was unchanged.
These data showed that sgp130Fc at a concentration of 5 µg/mL inhibits gp130-mediated STAT3 activation by varied IL6 family cytokines, including OSM that does not signal in trans [13,14,15,16]. Furthermore, effects of sgp130Fc on STAT3 activation were unrelated to the presence, abundance, or absence of soluble receptors for IL6 family cytokines, suggesting trans effects are not a major component of the signalling we observed.

2.2. sgp130FC Inhibits IL6 and OSM Cis-Signalling

To further determine if sgp130Fc inhibits IL6 cis-signalling we pre-incubated A549 cells with either vehicle control (0.1% DMSO) or GW280264X (1 μM in 0.1% DMSO, 16 h), which potently inhibits ADAM10 and 17 (IC50, 8.0 nM and 11.5 nM, respectively), to prevent IL6R cleavage and sIL6R generation. On the day of the experiment, cells were washed with PBS, supplemented with fresh media, again with GW280264X present or not, and immediately stimulated (15 min) cells with IL6 with or without the addition of sgp130Fc (5 µg/mL).
Immunoblotting for p-STAT3/STAT3 revealed robust STAT3 activation in cells incubated with or without GW280264X and equally potent inhibition of this effect by sgp130Fc. At the end of the experiment, supernatants were assessed for sIL6R that was detectable in the absence of GW280264X but not detected in media from cells incubated with GW280264X. (Figure 2A,B and Supplementary Figure S2A,B). These data reinforce the fact that sgp130Fc inhibits IL6 cis-signalling.
Experiments using GW280264X were repeated in primary human hepatocytes to exclude the possibility of an undetected presence [and effect] of IL6R in hepatocyte media, despite the negative ELISA results (Figure 1A). Comparable levels of STAT3 activation were observed following IL6 stimulation of DMSO (control) and GW280264X-treated hepatocytes, which reconfirmed the absence of an IL6:sIL6R trans-signalling effect in hepatocyte media (Figure 2A,B). There was similar inhibition of STAT3 activation by sgp130Fc in the presence or absence of ADAM10/17 inhibitor, excluding a trans-signalling component (Figure 2A,B).
We then explored the effects of sgp130Fc on IL6 and OSM cis-signalling in A549 cells across a dose range of 1.25–100 µg/mL (Figure 2C–F). Given the presence of IL6R in A549 media (Figure 1A), we conducted experiments in this cell line in the presence of GW280264X to obviate possible IL6 trans-signalling effects. sgp130Fc dose-dependently reduced IL6 and OSM-mediated cis-stimulation of STAT3 activation. The relationship between sgp130Fc concentrations and the levels of p-STAT3 was evaluated using semi-quantitative densitometry, which revealed IC50 values of 7.73 μg/mL and 1.06 μg/mL for sgp130Fc inhibition of IL6 and OSM cis-signalling, respectively (Figure 2D,F).
To address any potential bias arising from the effects of sgp130Fc in a single cell type, particularly a cancer cell line such as A549, we evaluated the IC50 values of sgp130Fc for IL6 cis-signalling in primary human hepatocytes and HSCs. Consistent with the results in A549 cells, a dose-dependent reduction in IL6-mediated phosphorylation of STAT3 was observed with sgp130Fc in both cell types with IC50 values of 3.83 μg/mL and 5.21 μg/mL in hepatocytes and HSCs, respectively (Figure 2G,H).
As mentioned in the introduction, sgp130Fc has pronounced therapeutic effects across murine disease models and also shows promise in human trials. On review of the literature, we observed that many of the murine POC studies used sgp130Fc at concentrations in excess of its IC50 values for IL6 or OSM cis-signalling, which was also true for clinical trials of olamkicept (Table 1). This suggests that sgp130Fc, at concentrations used in preclinical studies and clinical trials, most likely inhibits gp130 signalling beyond just IL6 trans-signalling.
We next examined the effects of sgp130Fc, as compared to a commercially available gp130 neutralising antibody (clone B-R3) [27]). In A549 cells treated with GW280264X, B-R3 inhibited IL6 and OSM signalling, as expected. sgp130Fc also inhibited both IL6 and OSM signalling with effects that were indistinguishable from those of B-R3 (Figure 3A–C).
To conclude, we present a simplified overview and interpretation of the findings of this study in a schematic, which illustrates the inhibitory effects of sgp130Fc on IL6 trans-signalling at low concentrations, and on IL6 family cytokine cis-signalling at high concentrations (Figure 3D).

3. Discussion

It is taught that IL6 cis-signalling is homeostatic, whereas IL6 trans-signalling amplifies inflammatory responses in autocrine and paracrine and is disease-causing [2]. As such, sgp130Fc is proposed, and has been shown, to have therapeutic benefits over combined IL6 cis- and trans-signalling inhibitors, such as tocilizumab [7]. However, it has also been reported that sgp130 might inhibit some aspects of OSM, CNTF and/or LIF signalling [1,5,6,12] and IL11 trans-signalling [28].
We show here that sgp130Fc inhibits gp130-dependent cis-signalling by several IL6 family cytokines in neoplastic (A549), stromal (hepatic stellate) and epithelial (hepatocyte) cells. This broad inhibitory effect across diverse cell types is independent of the presence, absence, or amount of sIL6R or sIL11RA, for IL6 or IL11 signalling, and was also seen for CT1 and OSM. While proponents of IL6 trans-signalling may argue that sub-picomolar levels of IL6R may still play a role, our findings also reveal that the sgp130Fc activity is not influenced by the presence of GW280264X, an inhibitor for IL6R shedding. The inhibitory effects of sgp130Fc were particularly robust for OSM, which does not signal in trans. This may be attributed to OSM’s direct and low-affinity binding to gp130 before interacting with its alpha receptors, potentially rendering it more susceptible to decoying [13,14,15,16].
The investigational drug olamkicept shows suggestive therapeutic effect (NCT03235752) at a dose of 600 mg, bi-weekly: maximum concentration (Cmax), 159 μg/mL [10,11]. These Cmax levels are approximately 100-fold higher than those reported as specific for IL6 trans-signalling [6] and, assuming equitable drug distribution in the extracellular space, are expected to inhibit IL6, IL11, OSM and CT1 cis-signalling. Similarly, the concentrations of sgp130Fc that have therapeutic effects in murine POC studies exceed the IC50 values of sgp130Fc for IL6 family cytokine cis-signalling.
Our data suggest the following: [1] sgp130Fc, at doses effective in treating both murine and human disease, is not specific for IL6 trans-signalling and [2] sgp130Fc effects on IL6 family cytokine cis-signalling are similar to those of a gp130 neutralising antibody. These findings raise important questions for the field of IL6 trans-signalling in general that are amplified by recent data indicating the non-specific nature of HyperIL6, a synthetic trans-signalling complex [29].
We thus propose global inhibition of gp130 cis-signalling (e.g., using bazedoxifene [30] or an antibody like B-R3 [27]) as a therapeutic approach for treating diseases in which sgp130Fc, an incomplete blocker of gp130 cis- and trans-signalling, has demonstrated efficacy in preclinical and clinical studies. The mouse model of cytokine storm syndrome (CSS) or sepsis may be particularly useful for testing this hypothesis, given the important roles of IL6 [17,31] and OSM [32] in CSS pathogenesis. Notably, genetic deletion of gp130 or administration of sgp130Fc has shown protective effects against CSS [17,33,34], gp130 is upregulated in humans and mice with CSS [35], and genetically enhanced gp130 signalling causes hypersensitivity and increased mortality in CSS [36].
Genetic perturbation of gp130 signalling during development and early life has well-documented detrimental effects in mice and humans [37,38]. Based on this observation, gp130 has been predominantly viewed as a protective factor. However, more recent studies suggest that inhibition of gp130 in adult mice can protect against various conditions, including hepatotoxicity, sepsis, osteoarthritis, inflammation, myeloma, and endothelial dysfunction, while also promoting tissue regeneration [9,34,39,40].
In light of our new findings, global or modular inhibition of gp130 signalling in adults could be considered as a therapeutic approach for human diseases where enhanced gp130 activity is specifically implicated. Such conditions include inflammatory bowel disease [41], solid cancers [42], myeloma [9], retinal disease [8], and autoimmune diseases [43]. It is important to consider potential on-target toxicities that may be associated with prolonged inhibition of gp130, such as infection risk, for long-term indications. Interestingly, sgp130Fc has a reassuring safety profile, including the absence of platelet toxicity, even when used at concentrations expected to inhibit OSM signalling, which is intriguing considering that direct targeting of OSM causes thrombocytopenia [44].

4. Materials and Methods

4.1. Antibodies

For neutralisation study: anti-gp130 (B-R3, ab34315, Abcam, Cambridge, UK), IgG control (clone 11E10, Genovac, Freiburg, Germany).
For immunoblotting: GAPDH (2118, CST, Danvers, MA, USA), phospho-STAT3 (4113, CST, Danvers, MA, USA), STAT3 (4904, CST, Danvers, MA, USA), mouse HRP (7076, CST, Danvers, MA, USA), rabbit HRP (7074, CST, Danvers, MA, USA). All primary antibodies were used at 1:1000 dilution in TBST, and secondary antibodies were diluted 1:2000 in TBST containing 3% bovine serum albumin (BSA).

4.2. Recombinant Proteins

Recombinant human (rh)CNTF (257-NT, R&D Systems, Minneapolis, MN, USA), rhCT1 (612-CD, R&D Systems, Minneapolis, MN, USA), rhIL6 (206-IL, R&D Systems, Minneapolis, MN, USA), rhIL11 (Z03108, Genscript, Minneapolis, MN, USA), rhLIF (7734-LF, R&D Systems, Minneapolis, MN, USA), rhOSM (PHC5015, ThermoFisher Scientific, San Francisco, CA, USA), sgp130Fc (671-GP, R&D Systems, Minneapolis, MN, USA). All lyophilized protein was dissolved in PBS with 15 min of gentle agitation.

4.3. Chemicals

Bovine serum albumin (BSA, A7906, Sigma-Aldrich, St. Louis, MO, USA), DMSO (D2650, Sigma-Aldrich, St. Louis, MO, USA), GW280264X (HY-115670, MedChemExpress, South Brunswick, NJ, USA), Poly-L-Lysine solution (P4707, Sigma-Aldrich, St. Louis, MO, USA).

4.4. Cell Culture

All cells used in this study were sourced commercially. Cells were grown and maintained at 37 °C and 5% CO2. The growth medium was renewed every 2–3 days and cells were passaged at 80% confluence, using standard trypsinization techniques. Cells were serum-starved overnight in basal media prior to stimulation. Cells were stimulated with either recombinant human CNTF, CT1, IL6, IL11, LIF, or OSM, at a concentration of 5 ng/mL in serum-free media for 15 min. Stimulated cells were compared to unstimulated cells that have been grown for the same duration under the same conditions, but without the stimulus (referred to as ‘baseline (BL)’ conditions).

4.4.1. A549

Human lung epithelial carcinoma cells (A549) that were isolated from a 58-year-old male (CCL-185, ATCC) were grown and maintained in DMEM (11995065, ThermoFisher Scientific, San Francisco, CA, USA) complete medium which contains 10% FBS (10500064, ThermoFisher Scientific, San Francisco, CA, USA) and 1% P/S (15140122, ThermoFisher Scientific, San Francisco, CA, USA).

4.4.2. Human Hepatocytes

Primary human hepatocytes used in this study were isolated from a 22-week-old foetus (5200, ScienCell, Carlsbad, CA, USA) and cultured as described previously [45]. Briefly, once hepatocytes were recovered from the initial thaw cycle, cells were seeded at a density 4 × 105 cells/well in hepatocyte medium (5201, ScienCell, Carlsbad, CA, USA) supplemented with 2% fetal bovine serum, 1% penicillin-streptomycin on a a poly-L-lysine-coated 6-well plate (2 µg/cm2, 0403, ScienCell, Carlsbad, CA, USA). Hepatocytes were used directly for downstream experiments within 48 h of seeding. All experiments with primary human hepatocytes were carried out at passage 2 (P2).

4.4.3. Human Hepatic Stellate Cells (HSC)

Primary HSC used in this study were isolated from a 50-year-old male (5300, ScienCell, Carlsbad, CA, USA) and cultured as described previously [46]. For experiments, we thawed and seeded P3 cells at a density of 2.5 × 105 cells/well on a poly-L-lysine-coated 6-well plate in stellate cells complete media (5301, ScienCell, Carlsbad, CA, USA). Within 24 h of seeding, HSC were serum-starved overnight prior to stimulation.

4.5. ADAM10/17 Inhibitor Experiments

GW280264X came pre-dissolved in 100% DMSO at a concentration of 10 mM, which we subsequently diluted to a stock concentration of 1 mM in 100% DMSO DMSO and kept as stock. Further dilution was performed to a final concentration of 1 µM i.e., 1 µL of GW280264X in 1 mL of cell culture media. Cells were treated with 1 µM of GW280264X throughout the overnight starvation, a 30-min incubation with sgp130, and a 15-min stimulation period; 0.1% DMSO was used as control.

4.6. sgp130/Anti-gp130 Experiment

sgp130Fc was dissolved in PBS to a stock concentration of 2.5 mg/mL; IgG and anti gp130 (B-R3) were dissolved in PBS to a stock concentration of 5 mg/mL. Cells were pre-incubated with either IgG (5 µg/mL), sgp130Fc (100 µg/mL) or B-R3 (5 µg/mL) for 30 min prior to cytokine stimulation.

4.7. IC50 Experiments

Cells were pre-incubated with either sgp130Fc (1.25, 2.5, 5, 10, 20, 40, 80, 100 µg/mL) an equivalent amount of PBS to 100 ug/mL of sgp130Fc (40 µL in 1 mL media) for 30 min and then stimulated with 5 ng/mL of various cytokines (as outlined in the text/figure/figure legends) for 15 min.

4.8. Immunoblotting

Western blots were carried out on total protein extracts from A549, hepatocytes, and HSCs. After stimulation, cells were rinsed twice in cold PBS. Cell lysates were homogenised in radioimmunoprecipitation assay (RIPA) Lysis and Extraction Buffer (89901, Thermo Fisher Scientific, San Francisco, CA, USA) containing protease inhibitor (A32965, Thermo Fisher Scientific, San Francisco, CA, USA), and phosphatase inhibitor (A32957, Thermo Fisher Scientific, San Francisco, CA, USA), followed by centrifugation to clear the lysate. Protein concentrations were determined by Bradford assay (5000006, Bio-Rad, Watford, UK). Equal amounts (5 µg) of protein lysates were separated by SDS-PAGE and transferred to PVDF membranes. Protein bands were visualised using the SuperSignal West Femto Maximum Sensitivity Substrate detection system (34096, Thermo Fisher Scientific, San Francisco, CA, USA) with the appropriate secondary antibodies: anti-mouse HRP or anti-rabbit HRP. Densitometry analyses were performed by ImageJ software. (version 1.53f51), in which the phospho STAT3 signal was normalised to its respective total STAT3 intensity. For IC50 experiment (Section 4.7), we considered STAT3 activation levels at baseline as maximal inhibition (100%) while STAT3 activation levels after stimulation with the respective recombinant protein (IL6/IL11;10 ng/mL) in the presence of PBS constituted minimum inhibition (0%).

4.9. Enzyme-Linked Immunosorbent Assay (ELISA)

The levels of IL6R, IL11RA, LIFR, and OSMR in an equal volume of cell culture supernatant were quantified using Human IL-6R alpha Quantikine ELISA kit (DR600, R&D Systems, Minneapolis, MN, USA), Human interleukin 11 receptor alpha (IL11RA) ELISA Kit (MBS453817, MyBioSource, San Diego, CA, USA), Human LIFR ELISA Kit (MBS2513036, MyBioSource, San Diego, CA, USA), or Human OSMR ELISA Kit (MBS2514091, MyBioSource, San Diego, CA, USA), respectively. All ELISAs were performed according to the manufacturer’s protocol.

4.10. Statistical Analysis

All statistical analyses were performed using GraphPad Prism software (version 9). Data were tested for normality with Shapiro-Wilk tests and all were found to follow normal distribution. One-way ANOVA with Tukey’s correction was used when several conditions were compared to each other. The criterion for statistical significance was set at p < 0.05.

5. Conclusions

In summary, the conventional notion of sgp130 as “a decoy protein that exclusively blocks IL6 proinflammatory trans-signalling” [11] appears invalid at the concentrations of sgp130Fc used in most rodent studies and in human clinical trials. Instead, the strong therapeutic effects of sgp130Fc more likely reflect its more extensive inhibition of IL6, IL11, OSM and CT1 cis-signalling, which would explain its superiority over anti-IL6 or anti-IL6R monotherapy [7,8,9]. An implication of this interpretation is that compounds, whether small or large molecules, that have global or modular inhibition of gp130, which is superior or more extensive to that achieved with sgp130Fc, could offer a new therapeutic approach for treating human disease.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms25031363/s1.

Author Contributions

A.A.W. and S.A.C. conceived, designed, and funded the study, analysed the data, and prepared the manuscript (writing and editing). A.A.W. performed the experiments. All authors have read and agreed to the published version of the manuscript.

Funding

This research is supported by the National Medical Research Council (NMRC) MOH-OFIRG21nov-0006 (A.A.W.) and NMRC MOH-STaR21nov-0003 (S.A.C.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data are provided in the manuscript and Supplementary Materials.

Acknowledgments

The authors would like to acknowledge the technical expertise and support of Sivakumar Viswanathan, Jessie Tan, and Benjamin George.

Conflicts of Interest

A.A.W. and S.A.C. are co-inventors on patents relating to IL11 biology. S.A.C. is a co-founder and shareholder of Enleofen Bio Pte Ltd. and VVB Bio Pte Ltd.

References

  1. Taga, T.; Hibi, M.; Hirata, Y.; Yamasaki, K.; Yasukawa, K.; Matsuda, T.; Hirano, T.; Kishimoto, T. Interleukin-6 triggers the association of its receptor with a possible signal transducer, gp130. Cell 1989, 58, 573–581. [Google Scholar] [CrossRef] [PubMed]
  2. Rose-John, S.; Jenkins, B.J.; Garbers, C.; Moll, J.M.; Scheller, J. Targeting IL-6 trans-signalling: Past, present and future prospects. Nat. Rev. Immunol. 2023, 23, 666–681. [Google Scholar] [CrossRef] [PubMed]
  3. Schumacher, N.; Meyer, D.; Mauermann, A.; Von Der Heyde, J.; Wolf, J.; Schwarz, J.; Knittler, K.; Murphy, G.; Michalek, M.; Garbers, C.; et al. Shedding of Endogenous Interleukin-6 Receptor (IL-6R) Is Governed by A Disintegrin and Metalloproteinase (ADAM) Proteases while a Full-length IL-6R Isoform Localizes to Circulating Microvesicles. J. Biol. Chem. 2015, 290, 26059–26071. [Google Scholar] [CrossRef] [PubMed]
  4. Garbers, C.; Jänner, N.; Chalaris, A.; Moss, M.L.; Floss, D.M.; Meyer, D.; Koch-Nolte, F.; Rose-John, S.; Scheller, J. Species specificity of ADAM10 and ADAM17 proteins in interleukin-6 (IL-6) trans-signaling and novel role of ADAM10 in inducible IL-6 receptor shedding. J. Biol. Chem. 2011, 286, 14804–14811. [Google Scholar] [CrossRef] [PubMed]
  5. Narazaki, M.; Yasukawa, K.; Saito, T.; Ohsugi, Y.; Fukui, H.; Koishihara, Y.; Yancopoulos, G.D.; Taga, T.; Kishimoto, T. Soluble forms of the interleukin-6 signal-transducing receptor component gp130 in human serum possessing a potential to inhibit signals through membrane-anchored gp130. Blood 1993, 82, 1120–1126. [Google Scholar] [CrossRef] [PubMed]
  6. Jostock, T.; Müllberg, J.; Özbek, S.; Atreya, R.; Blinn, G.; Voltz, N.; Fischer, M.; Neurath, M.F.; Rose-John, S. Soluble gp130 is the natural inhibitor of soluble interleukin-6 receptor transsignaling responses. Eur. J. Biochem. 2001, 268, 160–167. [Google Scholar] [CrossRef] [PubMed]
  7. George, M.J.; Jasmin, N.H.; Cummings, V.T.; Richard-Loendt, A.; Launchbury, F.; Woollard, K.; Turner-Stokes, T.; Diaz, A.I.G.; Lythgoe, M.; Stuckey, D.J.; et al. Selective Interleukin-6 Trans-Signaling Blockade Is More Effective Than Panantagonism in Reperfused Myocardial Infarction. JACC Basic Transl. Sci. 2021, 6, 431–443. [Google Scholar] [CrossRef]
  8. Wang, X.; Miller, E.B.; Goswami, M.; Zhang, P.; Ronning, K.E.; Karlen, S.J.; Zawadzki, R.J.; Pugh, E.N., Jr.; Burns, M.E. Rapid monocyte infiltration following retinal detachment is dependent on non-canonical IL6 signaling through gp130. J. Neuroinflammat. 2017, 14, 121. [Google Scholar] [CrossRef]
  9. Burger, R.; Guenther, A.; Klausz, K.; Staudinger, M.; Peipp, M.; Penas, E.M.M.; Rose-John, S.; Wijdenes, J.; Gramatzki, M. Due to interleukin-6 type cytokine redundancy only glycoprotein 130 receptor blockade efficiently inhibits myeloma growth. Haematologica 2017, 102, 381–390. [Google Scholar] [CrossRef]
  10. Zhang, S.; Chen, B.; Wang, B.; Chen, H.; Li, Y.; Cao, Q.; Zhong, J.; Shieh, M.-J.; Ran, Z.; Tang, T.; et al. Effect of Induction Therapy With Olamkicept vs Placebo on Clinical Response in Patients With Active Ulcerative Colitis: A Randomized Clinical Trial. JAMA 2023, 329, 725–734. [Google Scholar] [CrossRef]
  11. Schreiber, S.; Aden, K.; Bernardes, J.P.; Conrad, C.; Tran, F.; Höper, H.; Volk, V.; Mishra, N.; Blase, J.I.; Nikolaus, S.; et al. Therapeutic Interleukin-6 Trans-signaling Inhibition by Olamkicept (sgp130Fc) in Patients With Active Inflammatory Bowel Disease. Gastroenterology 2021, 160, 2354–2366.e11. [Google Scholar] [CrossRef] [PubMed]
  12. Hui, W.; Bell, M.; Carroll, G. Soluble glycoprotein 130 (gp130) attenuates OSM- and LIF-induced cartilage proteoglycan catabolism. Cytokine 2000, 12, 151–155. [Google Scholar] [CrossRef] [PubMed]
  13. Auguste, P.; Guillet, C.; Fourcin, M.; Olivier, C.; Veziers, J.; Pouplard-Barthelaix, A.; Gascan, H. Signaling of type II oncostatin M receptor. J. Biol. Chem. 1997, 272, 15760–15764. [Google Scholar] [CrossRef] [PubMed]
  14. Deller, M.C.; Hudson, K.R.; Ikemizu, S.; Bravo, J.; Jones, E.Y.; Heath, J.K. Crystal structure and functional dissection of the cytostatic cytokine oncostatin M. Structure 2000, 8, 863–874. [Google Scholar] [CrossRef] [PubMed]
  15. Mosley, B.; De Imus, C.; Friend, D.; Boiani, N.; Thoma, B.; Park, L.S.; Cosman, D. Dual oncostatin M (OSM) receptors. Cloning and characterization of an alternative signaling subunit conferring OSM-specific receptor activation. J. Biol. Chem. 1996, 271, 32635–32643. [Google Scholar] [CrossRef]
  16. Gearing, D.P.; Comeau, M.R.; Friend, D.J.; Gimpel, S.D.; Thut, C.J.; McGourty, J.; Brasher, K.K.; King, J.A.; Gillis, S.; Mosley, B.; et al. The IL-6 Signal Transducer, gp130: An Pncostatin M Receptor and Affinity Converter for the LIF Receptor. Science 1992, 255, 1434–1437. [Google Scholar] [CrossRef]
  17. Barkhausen, T.; Tschernig, T.; Rosenstiel, P.; van Griensven, M.; Vonberg, R.-P.; Dorsch, M.; Mueller-Heine, A.; Chalaris, A.; Scheller, J.; Rose-John, S.; et al. Selective blockade of interleukin-6 trans-signaling improves survival in a murine polymicrobial sepsis model. Crit. Care Med. 2011, 39, 1407–1413. [Google Scholar] [CrossRef]
  18. Qin, M.; Xin, Y.; Bian, Y.; Yang, X.; Xi, T.; Xiong, J. Phosphorylation-Induced Ubiquitination and Degradation of PXR through CDK2-TRIM21 Axis. Cells 2022, 11, 264. [Google Scholar] [CrossRef]
  19. Schafer, S.; Viswanathan, S.; Widjaja, A.A.; Lim, W.-W.; Moreno-Moral, A.; DeLaughter, D.M.; Ng, B.; Patone, G.; Chow, K.; Khin, E.; et al. IL-11 is a crucial determinant of cardiovascular fibrosis. Nature 2017, 552, 110–115. [Google Scholar] [CrossRef]
  20. Diehl, K.; Hull, R.; Morton, D.; Pfister, R.; Rabemampianina, Y.; Smith, D.; Vidal, J.; Van De Vorstenbosch, C. A good practice guide to the administration of substances and removal of blood, including routes and volumes. J. Appl. Toxicol. 2001, 21, 15–23. [Google Scholar] [CrossRef]
  21. Atreya, R.; Mudter, J.; Finotto, S.; Müllberg, J.; Jostock, T.; Wirtz, S.; Schütz, M.; Bartsch, B.; Holtmann, M.; Becker, C.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  22. Nowell, M.A.; Williams, A.S.; Carty, S.A.; Scheller, J.; Hayes, A.J.; Jones, G.W.; Richards, P.J.; Slinn, S.; Ernst, M.; Jenkins, B.J.; et al. Therapeutic targeting of IL-6 trans signaling counteracts STAT3 control of experimental inflammatory arthritis. J. Immunol. 2009, 182, 613–622. [Google Scholar] [CrossRef] [PubMed]
  23. Matsumoto, S.; Hara, T.; Mitsuyama, K.; Yamamoto, M.; Tsuruta, O.; Sata, M.; Scheller, J.; Rose-John, S.; Kado, S.-I.; Takada, T. Essential roles of IL-6 trans-signaling in colonic epithelial cells, induced by the IL-6/soluble-IL-6 receptor derived from lamina propria macrophages, on the development of colitis-associated premalignant cancer in a murine model. J. Immunol. 2010, 184, 1543–1551. [Google Scholar] [CrossRef] [PubMed]
  24. Braun, G.S.; Nagayama, Y.; Maruta, Y.; Heymann, F.; van Roeyen, C.R.; Klinkhammer, B.M.; Boor, P.; Villa, L.; Salant, D.J.; Raffetseder, U.; et al. IL-6 Trans-Signaling Drives Murine Crescentic GN. J. Am. Soc. Nephrol. 2016, 27, 132–142. [Google Scholar] [CrossRef] [PubMed]
  25. Li, X.; Wu, X.; Chen, X.; Peng, S.; Chen, S.; Zhou, G.; Wei, Y.; Lu, X.; Zhou, C.; Ye, Y.; et al. Selective blockade of interleukin 6 trans-signaling depresses atrial fibrillation. Heart Rhythm. 2023, 20, 1759–1770. [Google Scholar] [CrossRef] [PubMed]
  26. Robinson, R.; Srinivasan, M.; Shanmugam, A.; Ward, A.; Ganapathy, V.; Bloom, J.; Sharma, A.; Sharma, S. Interleukin-6 trans-signaling inhibition prevents oxidative stress in a mouse model of early diabetic retinopathy. Redox Biol. 2020, 34, 101574. [Google Scholar] [CrossRef] [PubMed]
  27. Chevalier, S.; Fourcin, M.; Robledo, O.; Wijdenes, J.; Pouplard-Barthelaix, A.; Gascan, H. Interleukin-6 family of cytokines induced activation of different functional sites expressed by gp130 transducing protein. J. Biol. Chem. 1996, 271, 14764–14772. [Google Scholar] [CrossRef] [PubMed]
  28. Lokau, J.; Nitz, R.; Agthe, M.; Monhasery, N.; Aparicio-Siegmund, S.; Schumacher, N.; Wolf, J.; Möller-Hackbarth, K.; Waetzig, G.H.; Grötzinger, J.; et al. Proteolytic Cleavage Governs Interleukin-11 Trans-signaling. Cell Rep. 2016, 14, 1761–1773. [Google Scholar] [CrossRef]
  29. Dong, J.; Viswanathan, S.; Adami, E.; Schafer, S.; Kuthubudeen, F.F.; Widjaja, A.A.; Cook, S.A. The pro-regenerative effects of HyperIL6 in drug induced liver injury are unexpectedly due to competitive inhibition of IL11 signaling. Elife 2021, 26, 10. [Google Scholar] [CrossRef] [PubMed]
  30. Wei, J.; Ma, L.; Lai, Y.H.; Zhang, R.; Li, H.; Li, C.; Lin, J. Bazedoxifene as a novel GP130 inhibitor for Colon Cancer therapy. J. Exp. Clin. Cancer Res. 2019, 38, 63. [Google Scholar] [CrossRef]
  31. Hamilton, F.W.; Thomas, M.; Arnold, D.; Palmer, T.; Moran, E.; Mentzer, A.J.; Maskell, N.; Baillie, K.; Summers, C.; Hingorani, A.; et al. Therapeutic potential of IL6R blockade for the treatment of sepsis and sepsis-related death: A Mendelian randomisation study. PLoS Med. 2023, 20, e1004174. [Google Scholar] [CrossRef] [PubMed]
  32. Salim, S.Y.; AlMalki, N.; Macala, K.F.; Wiedemeyer, A.; Mueller, T.F.; Churchill, T.A.; Bourque, S.L.; Khadaroo, R.G. Oncostatin M Receptor Type II Knockout Mitigates Inflammation and Improves Survival from Sepsis in Mice. Biomedicines 2023, 11, 483. [Google Scholar] [CrossRef] [PubMed]
  33. Klein, C.; Medina, E.; Sander, L.; Dierssen, U.; Roskams, T.; Mueller, W.; Trautwein, C.; Goldmann, O. Contribution of interleukin-6/gp 130 signaling in hepatocytes to the inflammatory response in mice infected with Streptococcus pyogenes. J. Infect. Dis. 2007, 196, 755–762. [Google Scholar] [CrossRef] [PubMed]
  34. Kang, S.; Onishi, S.; Ling, Z.; Inoue, H.; Zhang, Y.; Chang, H.; Zhao, H.; Wang, T.; Okuzaki, D.; Matsuura, H.; et al. Gp130-HIF1α axis-induced vascular damage is prevented by the short-term inhibition of IL-6 receptor signaling. Proc. Natl. Acad. Sci. USA 2024, 121, e2315898120. [Google Scholar] [CrossRef] [PubMed]
  35. Marsik, C.; Halama, T.; Cardona, F.; Schlifke, I.; Mittermayer, F.; Jilma, B. Endotoxemia enhances expression of the signaling receptor (GP130) on protein and molecular level. Clin. Immunol. 2005, 114, 293–298. [Google Scholar] [CrossRef] [PubMed]
  36. Greenhill, C.J.; Rose-John, S.; Lissilaa, R.; Ferlin, W.; Ernst, M.; Hertzog, P.J.; Mansell, A.; Jenkins, B.J. IL-6 trans-signaling modulates TLR4-dependent inflammatory responses via STAT3. J. Immunol. 2011, 186, 1199–1208. [Google Scholar] [CrossRef] [PubMed]
  37. Chen, Y.H.; van Zon, S.; Adams, A.; Schmidt-Arras, D.; Laurence, A.D.J.; Uhlig, H.H. The Human GP130 Cytokine Receptor and Its Expression-an Atlas and Functional Taxonomy of Genetic Variants. J. Clin. Immunol. 2023, 44, 30. [Google Scholar] [CrossRef]
  38. Fasnacht, N.; Müller, W. Conditional gp130 deficient mouse mutants. Semin. Cell Dev. Biol. 2008, 19, 379–384. [Google Scholar] [CrossRef]
  39. Dong, J.; Lim, W.-W.; Shekeran, S.G.; Tan, J.; Lim, S.Y.; Goh, J.W.T.; George, B.L.; Schafer, S.; Cook, S.A.; Widjaja, A.A. Hepatocyte Specific gp130 Signalling Underlies APAP Induced Liver Injury. Int. J. Mol. Sci. 2022, 23, 7089. [Google Scholar] [CrossRef]
  40. Shkhyan, R.; Flynn, C.; Lamoure, E.; Sarkar, A.; Van Handel, B.; Li, J.; York, J.; Banks, N.; Van der Horst, R.; Liu, N.Q.; et al. Inhibition of a signaling modality within the gp130 receptor enhances tissue regeneration and mitigates osteoarthritis. Sci. Transl. Med. 2023, 15, eabq2395. [Google Scholar] [CrossRef]
  41. Nayar, S.; Morrison, J.K.; Giri, M.; Gettler, K.; Chuang, L.S.; Walker, L.A.; Ko, H.M.; Kenigsberg, E.; Kugathasan, S.; Merad, M.; et al. A myeloid-stromal niche and gp130 rescue in NOD2-driven Crohn’s disease. Nature 2021, 593, 275–281. [Google Scholar] [CrossRef] [PubMed]
  42. Omokehinde, T.; Johnson, R.W. GP130 Cytokines in Breast Cancer and Bone. Cancers 2020, 12, 326. [Google Scholar] [CrossRef]
  43. Silver, J.S.; Hunter, C.A. gp130 at the nexus of inflammation, autoimmunity, and cancer. J. Leukoc. Biol. 2010, 88, 1145–1156. [Google Scholar] [CrossRef] [PubMed]
  44. Denton, C.P.; del Galdo, F.; Khanna, D.; Vonk, M.C.; Chung, L.; Johnson, S.R.; Varga, J.; Furst, D.E.; Temple, J.; Zecchin, C.; et al. Biological and clinical insights from a randomised phase II study of an anti-oncostatin M monoclonal antibody in systemic sclerosis. Rheumatology 2022, 62, 234–242. [Google Scholar] [CrossRef] [PubMed]
  45. Dong, J.; Viswanathan, S.; Adami, E.; Singh, B.K.; Chothani, S.P.; Ng, B.; Lim, W.W.; Zhou, J.; Tripathi, M.; Ko, N.S.J.; et al. Hepatocyte-specific IL11 cis-signaling drives lipotoxicity and underlies the transition from NAFLD to NASH. Nat. Commun. 2021, 12, 66. [Google Scholar] [CrossRef] [PubMed]
  46. Widjaja, A.A.; Singh, B.K.; Adami, E.; Viswanathan, S.; Dong, J.; D’agostino, G.A.; Ng, B.; Lim, W.W.; Tan, J.; Paleja, B.S.; et al. Inhibiting Interleukin 11 Signaling Reduces Hepatocyte Death and Liver Fibrosis, Inflammation, and Steatosis in Mouse Models of Nonalcoholic Steatohepatitis. Gastroenterology 2019, 157, 777–792.e14. [Google Scholar] [CrossRef]
Figure 1. sgp130Fc inhibits IL6, IL11, OSM, and CT1-induced STAT3 activation. (A) ELISA of IL6R, IL11RA, LIFR, and OSMR in the conditioned supernatant of A549, primary human hepatocytes (HEP), and primary human hepatic stellate cells (HSC) 17 h after plating. Data are shown as box-and-whisker plots with median (middle line), 25th–75th percentiles (box) and min-max percentiles (whiskers). Western blots of p-STAT3 and STAT3 in CNTF, CT1, IL6, IL11, LIF (5 ng/mL)-stimulated (B) A549, (C) HEP, (D) HSC (15 min) and (BD) immunoblots of p-STAT3, STAT3, GAPDH in OSM (5 ng/mL)-stimulated A549, HEP, HSC (15 min) in the presence or absence of sgp130Fc (5 µg/mL). (AD) n = 4 biological replicates. BL: baseline. n.d. not detected.
Figure 1. sgp130Fc inhibits IL6, IL11, OSM, and CT1-induced STAT3 activation. (A) ELISA of IL6R, IL11RA, LIFR, and OSMR in the conditioned supernatant of A549, primary human hepatocytes (HEP), and primary human hepatic stellate cells (HSC) 17 h after plating. Data are shown as box-and-whisker plots with median (middle line), 25th–75th percentiles (box) and min-max percentiles (whiskers). Western blots of p-STAT3 and STAT3 in CNTF, CT1, IL6, IL11, LIF (5 ng/mL)-stimulated (B) A549, (C) HEP, (D) HSC (15 min) and (BD) immunoblots of p-STAT3, STAT3, GAPDH in OSM (5 ng/mL)-stimulated A549, HEP, HSC (15 min) in the presence or absence of sgp130Fc (5 µg/mL). (AD) n = 4 biological replicates. BL: baseline. n.d. not detected.
Ijms 25 01363 g001
Figure 2. Dose-dependent inhibition of IL6 and OSM cis-signalling by sgp130Fc. (A) Western blots of STAT3 activation status (p-STAT3/STAT3) in A549 and HEP in the following conditions: without stimulus (BL), IL6 + PBS, or IL6 + sgp130Fc in the presence of either DMSO or GW280264X. (B) ELISA of IL6R in the supernatant of A549 after 17 h incubation with either DMSO or GW280264X. Data are shown as box-and-whisker plots with median (middle line), 25th–75th percentiles (box) and min-max percentiles (whiskers); 2-tailed Student’s t-test. (C) Western blots and (D) densitometry analyses of p-STAT3/STAT3 in GW280264X-treated A549 following stimulation with IL6 in the presence of different concentrations of sgp130Fc. (E) Western blots of p-STAT3, STAT3, and GAPDH, and (F) densitometry analyses of p-STAT3/STAT3 in OSM-stimulated A549 in the presence of increasing concentrations of sgp130Fc. (G) Western blots and (H) densitometry analyses of p-STAT3/STAT3 in IL6-stimulated HEP and HSC in the presence of increasing concentrations of sgp130Fc. (D,F,H) Data are shown as mean ± s.d. (AH) n = 4 biological replicates; DMSO (0.1%), GW280264X (1 µM), IL6 (5 ng/mL), OSM (5 ng/mL), sgp130Fc (5 µg/mL, unless otherwise specified). BL: baseline. n.d. not detected.
Figure 2. Dose-dependent inhibition of IL6 and OSM cis-signalling by sgp130Fc. (A) Western blots of STAT3 activation status (p-STAT3/STAT3) in A549 and HEP in the following conditions: without stimulus (BL), IL6 + PBS, or IL6 + sgp130Fc in the presence of either DMSO or GW280264X. (B) ELISA of IL6R in the supernatant of A549 after 17 h incubation with either DMSO or GW280264X. Data are shown as box-and-whisker plots with median (middle line), 25th–75th percentiles (box) and min-max percentiles (whiskers); 2-tailed Student’s t-test. (C) Western blots and (D) densitometry analyses of p-STAT3/STAT3 in GW280264X-treated A549 following stimulation with IL6 in the presence of different concentrations of sgp130Fc. (E) Western blots of p-STAT3, STAT3, and GAPDH, and (F) densitometry analyses of p-STAT3/STAT3 in OSM-stimulated A549 in the presence of increasing concentrations of sgp130Fc. (G) Western blots and (H) densitometry analyses of p-STAT3/STAT3 in IL6-stimulated HEP and HSC in the presence of increasing concentrations of sgp130Fc. (D,F,H) Data are shown as mean ± s.d. (AH) n = 4 biological replicates; DMSO (0.1%), GW280264X (1 µM), IL6 (5 ng/mL), OSM (5 ng/mL), sgp130Fc (5 µg/mL, unless otherwise specified). BL: baseline. n.d. not detected.
Ijms 25 01363 g002
Figure 3. Comparison of the inhibitory effects of sgp130Fc versus a gp130 neutralising antibody on IL6 or OSM cis-signalling.(A) Western blots of p-STAT3, STAT3 (for IL6 and OSM), and GAPDH (for OSM) and (B) densitometry analyses of p-STAT3/STAT3 in GW280264X-treated A549 following stimulation with IL6, or OSM in the presence of either IgG, sgp130Fc, or a neutralising antibody against gp130 (clone B-R3). (C) Levels of IL6R, and OSMR protein in the supernatant from GW280264X-treated A549 as quantified by ELISA. (D) Schematic showing our proposed interpretation of the comparative effects between low versus high concentrations of sgp130Fc on the inhibition of gp130-mediated cis- and/or trans-signalling. (AC) n = 4 biological replicates; B-R3 (5 µg/mL), GW280264X (1 µM), IgG (5 µg/mL), IL6 (5 ng/mL), IL11 (5 ng/mL), OSM (5 ng/mL), sgp130Fc (100 µg/mL). (B,C) Data are shown as box-and-whisker plots with median (middle line), 25th–75th percentiles (box) and min-max percentiles (whiskers). (B) One-way ANOVA with Tukey’s correction. BL: baseline; FC: fold change. n.d. not detected.
Figure 3. Comparison of the inhibitory effects of sgp130Fc versus a gp130 neutralising antibody on IL6 or OSM cis-signalling.(A) Western blots of p-STAT3, STAT3 (for IL6 and OSM), and GAPDH (for OSM) and (B) densitometry analyses of p-STAT3/STAT3 in GW280264X-treated A549 following stimulation with IL6, or OSM in the presence of either IgG, sgp130Fc, or a neutralising antibody against gp130 (clone B-R3). (C) Levels of IL6R, and OSMR protein in the supernatant from GW280264X-treated A549 as quantified by ELISA. (D) Schematic showing our proposed interpretation of the comparative effects between low versus high concentrations of sgp130Fc on the inhibition of gp130-mediated cis- and/or trans-signalling. (AC) n = 4 biological replicates; B-R3 (5 µg/mL), GW280264X (1 µM), IgG (5 µg/mL), IL6 (5 ng/mL), IL11 (5 ng/mL), OSM (5 ng/mL), sgp130Fc (100 µg/mL). (B,C) Data are shown as box-and-whisker plots with median (middle line), 25th–75th percentiles (box) and min-max percentiles (whiskers). (B) One-way ANOVA with Tukey’s correction. BL: baseline; FC: fold change. n.d. not detected.
Ijms 25 01363 g003
Table 1. Examples of pre-clinical and clinical studies where sgp130Fc was given at doses that exceeded its half-maximal inhibitory (IC50) concentrations for IL6 or OSM cis-signalling. Dose per mouse reports published values or is estimated based on an average adult mouse mass of 25 g. Estimated maximum concentrations for mouse studies were calculated based on the assumption that sgp130Fc is confined and equally distributed across the extracellular fluid spaces (interstitial fluid and plasma), which represents ~20% of the body weight of an adult mouse [20]. i.p.: intraperitoneal; i.v.: intravenous, d: day.
Table 1. Examples of pre-clinical and clinical studies where sgp130Fc was given at doses that exceeded its half-maximal inhibitory (IC50) concentrations for IL6 or OSM cis-signalling. Dose per mouse reports published values or is estimated based on an average adult mouse mass of 25 g. Estimated maximum concentrations for mouse studies were calculated based on the assumption that sgp130Fc is confined and equally distributed across the extracellular fluid spaces (interstitial fluid and plasma), which represents ~20% of the body weight of an adult mouse [20]. i.p.: intraperitoneal; i.v.: intravenous, d: day.
Disease Model or Clinical TrialDose, Route, FrequencyMax Dose Per IndividualEstimated Maximum Concentration
(μg/mL)
IC50 for IL6 or OSM cis-
Signalling Exceeded
Reference
Murine Studies
Polymicrobial sepsis10 mg/kg, i.p., 7 d250 μg50 Y[17]
Colitis-500 μg 100Y[21]
Arthritis2.5 mg/kg, i.p., 7 d62.5 μg12.5Y[22]
Colon cancer-500 μg100Y[23]
Nephritis3 mg/kg, i.p., 3.5 d 75 μg15Y[24]
Myocardial infarction0.5 mg/kg, i.p. 12.5 μg2.5Y[7]
Atrial Fibrillation0.5 mg/kg, i.p., 3.5 d12.5 μg2.5Y[25]
Diabetic retinopathy5 mg/kg, i.p., 3.5 d125 μg25Y[26]
Human Studies
Patients with inflammatory bowel disease 600 mg, i.v., 14 d600 mg>100Y[11]
Patients with ulcerative colitis300 mg or 600 mg, i.v., 14 d600 mg159Y[10]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Widjaja, A.A.; Cook, S.A. Nonspecific Inhibition of IL6 Family Cytokine Signalling by Soluble gp130. Int. J. Mol. Sci. 2024, 25, 1363. https://doi.org/10.3390/ijms25031363

AMA Style

Widjaja AA, Cook SA. Nonspecific Inhibition of IL6 Family Cytokine Signalling by Soluble gp130. International Journal of Molecular Sciences. 2024; 25(3):1363. https://doi.org/10.3390/ijms25031363

Chicago/Turabian Style

Widjaja, Anissa A., and Stuart A. Cook. 2024. "Nonspecific Inhibition of IL6 Family Cytokine Signalling by Soluble gp130" International Journal of Molecular Sciences 25, no. 3: 1363. https://doi.org/10.3390/ijms25031363

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop