1. Introduction
Granulocyte-macrophage colony-stimulating factor (GM-CSF; CSF-2) is a pleiotropic 23 kDa short-chain four-helical cytokine (SCOP [
1] ID 4000852 and 3001717) belonging along with IL-3 and IL-5 to the β common family of cytokines [
2,
3,
4]. Basal levels of serum GM-CSF are of 1-4 pM but may rise rapidly in response to infection [
5,
6]. GM-CSF is normally expressed in lung, urinary bladder, pancreas, bone marrow and some other tissues [
7], and is produced at sites of tissue inflammation [
5,
8]. GM-CSF is produced by T-cells, natural killer cells, alveolar cells type 2, granulocytes, ductal cells, basal respiratory cells, pancreatic endocrine cells, ionocytes, endothelial cells, Langerhans cells [
7], other cells under various stimuli, and tumor cells [
9,
10]. GM-CSF regulates activity of macrophages, including monocytes and microglia, dendritic cells, lymphocytes, granulocytes, endothelial cells, pneumocytes type I, mesenchymal cells,
etc. [
6,
11,
12]. Thus, GM-CSF modulates activity of the innate immune cells serving as a bridge to the activation of adaptive immunity, thereby globally affecting host immunity under pathological conditions [
9].
GM-CSF is a colony-stimulating factor along with M-CSF (CSF-1) and G-CSF (CSF-3), the growth factors that favor formation of myeloid colonies from bone marrow precursors [
5,
13]. Some other cytokines, such as IL-3, IL-5, and IL-34, are also involved in myelopoiesis [
5]. G-CSF and M-CSF/IL-34 signal through the homodimeric receptors G-CSF-R and CSF-1-R, respectively [
5]. In contrast, GM-CSF, IL-3, and IL-5 signal via the heterodimeric complex consisting of the cytokine-specific α chain (GM-CSF receptor subunit α, CD116, for GM-CSF) and a shared signal-transducing Cytokine receptor common subunit β (CD131) [
5]. Erythropoietin is able to signal in the both manners [
14]. CD116 has two isoforms and a truncated, soluble form [
11]. The hexameric complex of GM-CSF with its receptors contains two intertwined CD131 chains in the center, surrounded by two CD116 chains with two GM-CSF molecules wrapped by the CD116 and CD131 chains [
4,
11]. Dimerization of the hexamer brings the cytoplasmic domains of the CD131 chains into proximity, which is thought to be crucial for the subsequent signaling events [
4]. The GM-CSF signaling pathways include JAK-STAT5, PI3K-AKT, MEK-ERK, NF-κB, β-catenin/Tcf4, JMJD3/IRF-4, IFN-γ-R/IRF-1, and AKT-mTOR [
4,
5,
8,
15,
16,
17]. Sulfated glycosaminoglycans, heparan sulfate proteoglycans, including Syndecan-2, are implicated in modulation of GM-CSF signaling [
18,
19]. The sophisticated signaling system of GM-CSF contributes to its multifactorial impact on host immune responses, and its involvement in pathogenesis of many diseases, including inflammatory arthritis, osteoarthritis, multiple sclerosis, inflammatory bowel disease, asthma, interstitial lung disease, pulmonary alveolar proteinosis, COVID-19, aortic aneurysm, obesity and its associated meta-inflammation, cancers, and other disorders [
5,
11,
15,
20,
21].
Elevated GM-CSF expression drives production of pro-inflammatory cytokines/chemokines (IL-1, IL-6, TNF, CCL2, IL-8, CCL17,
etc.), excessive inflammation, chemotaxis, and tissue damage [
22]. Therefore, several monoclonal antibodies that neutralize either GM-CSF (Otilimab, Namilumab, Gimsilumab, Plonmarlimab, and Lenzilumab) or CD116 (Mavrilimumab)/CD131 (CSL311) are currently in clinical trials for treatment of multiple sclerosis, rheumatoid arthritis, osteoarthritis, psoriatic arthritis, plaque psoriasis, asthma, COVID-19, lymphoma and leukemia [
4,
15,
23].
Meanwhile, GM-CSF suppresses autoimmune diabetes and autoimmune thyroiditis, systemic lupus erythematosus, myasthenia gravis, inflammatory bowel disease, atherosclerosis, some cancers,
etc. [
8,
9,
24]. Recombinant human GM-CSF with substitution at L23, known as sargramostim (Leukine
®), is clinically used for neutrophil recovery and reduction of the incidence of the infections following induction chemotherapy in older patients with acute myeloid leukemia; mobilization of hematopoietic progenitor cells into peripheral blood for collection by leukapheresis and transplantation; acceleration of myeloid reconstitution following bone marrow or peripheral blood progenitor cell transplantation; treatment of delayed neutrophil recovery or graft failure after bone marrow transplantation; treatment of the patients acutely exposed to myelosuppressive doses of radiation. Another recombinant GM-CSF, molgramostim, has been used in the clinical trials for cystic fibrosis, bronchiectasis, pulmonary alveolar proteinosis, acute respiratory distress syndrome, COVID-19, Mycobacterium and Hepatitis B infections, and several cancers. GM-CSF alone or in combination with chemotherapy, monoclonal antibodies/cancer vaccines is being tested in clinical trials for numerous cancers [
9].
GM-CSF incorporated into vaccines (GM-CSF-secreting cancer cell vaccines, GM-CSF-fused tumor-associated antigen protein-based vaccines, GM-CSF-based DNA vaccines, and GM-CSF-producing oncolytic viruses) stimulates potent antitumor responses, probably by promoting differentiation and activation of dendritic cells [
4,
9]. The clinically approved GM-CSF-based cancer vaccines include Sipuleucel-T (Provenge
®), a cell-based cancer immunotherapy for treatment of prostate cancer, and Talimogene laherparepvec (IMLYGIC
®), an oncolytic herpes virus for advanced melanoma treatment. Many investigational GM-CSF-based cancer vaccines are currently in trials: cancer-specific GVAX vaccines, Pexa-Vec
®, Vigil
®, ONCOS-102, OrienX010, MVX-ONCO-1, and STINGVAX.
Therefore, both inhibition and stimulation of GM-CSF activity are valuable therapeutic options depending on the specific disorder and its stage. The global GM-CSF market is valued at US
$1.5 billion in 2021 and is expected to reach US
$3.4 billion in 2029 [
25], in part due to the presence of the numerous drug candidates undergoing clinical trials. For this reason, knowledge of the factors affecting GM-CSF activity is important for both the development and effective use of the drugs targeting GM-CSF signaling.
Despite the vast knowledge of the mechanisms of GM-CSF signaling and its contribution to cellular homeostasis in both health and disease, the regulation of GM-CSF activity by extracellular soluble proteins remains largely unexplored. We recently showed that the members of the S100 family of Ca
2+-binding proteins, S100P and S100A6, specifically interact with GM-CSF and many other four-helical cytokines under physiological
in vitro conditions [
26,
27]. The S100 protein family includes over 20 regulatory multifunctional proteins consisting of two Ca
2+-binding motifs linked by a flexible ‘hinge’ region: a canonical C-terminal EF-hand and a low-affinity N-terminal pseudo-EF-hand [
28,
29]. Ca
2+ binding typically alters structure of a S100 protein, resulting in changes in its affinity for binding partners such as soluble regulatory proteins, receptor/membrane proteins, nucleic acids and lipids, thereby implicating S100 proteins in multiple physiological processes [
30,
31,
32]. When S100 proteins are released into the extracellular space, some of them interact with cell surface receptors (RAGE, TLR4, ErbB1, ErbB3, ErbB4, IL-10R, integrin β1, neuroplastin-β, 5-HT1B, 4-HT4, SIRL-1, ALCAM, EMMPRIN, CD33, CD36, CD68, CD69, and CD146 [
26]), acting in a cytokine-like manner [
29,
32,
33]. In addition, extracellular forms of certain S100 proteins have been shown to bind specific cytokines, in some cases exerting distinct cellular effects [
26,
27,
34,
35,
36,
37,
38,
39,
40,
41,
42,
43,
44,
45,
46,
47]. Such a broad network of S100 interactions seems to contribute to their involvement in the progression of many socially significant diseases (oncological, neurological, inflammatory, autoimmune, cardiovascular and respiratory diseases), and suggests the use of S100 proteins as diagnostic and therapeutic targets [
29,
32,
33,
48,
49,
50,
51].
In this work, we reveal new aspects of regulation of GM-CSF activity by S100 proteins: we explored selectivity of GM-CSF interaction with S100 proteins, conformational prerequisites for the GM-CSF ‒ S100 interactions, and the cellular effects they exert.
2. Materials and Methods
2.1. Materials
Recombinant tag-free human S100A1/A3/A4/A5/A6/A7/A8/A9/A10/A11/A12/A13/ A14/A15/A16/B/P proteins were expressed in
E. coli and purified as described in [
41,
42,
46,
47]. Recombinant tag-free human S100A7L2/G/Z proteins and GM-CSF were produced in
E. coli as described below. Human S100P mutants F89A and Δ42–47 (lacks PGFLQS sequence in the ‘hinge’ reion) were prepared according to [
47]. The protein concentrations were determined spectrophotometrically according to [
52]. Catalytic light chain of bovine enteropeptidase was purchased from New England Biolabs Inc. (Ipswich, MA, USA).
Phosphate, imidazole, HEPES, Tris, sodium chloride, sodium hydroxide, SDS, DTT, glycerol, L-arginine, urea, as well as reduced and oxidized L-glutathione were purchased from PanReac AppliChem (Barcelona, Spain). IPTG and PMSF were purchased from Helicon (Moscow, Russia). Calcium chloride, EDTA, TWEEN 20 and glutaric aldehyde were from Sigma–Aldrich Co. (Burlington, MA, USA). Silver staining of electrophoresis gels was carried out using PlusOneTM Silver Staining Kit (Protein) (Amersham Biosciences, Inc., Amersham, UK). Molecular mass marker for SDS-PAGE and SnakeSkinTM dialysis tubing (MWCO of 3.5 kDa) were from Thermo Fisher Scientific, Inc. (Waltham, MA, USA). HiPrepTM 26/60 Sephacryl® S-100 HR was from GE HealthCare (Chicago, IL, USA). Bio-Scale Mini Profinity IMAC cartridge was from Bio-Rad Laboratories, Inc. (Hercules, CA, USA). NAP-5 column was bought from Cytiva (Marlborough, MA, USA). All water solutions were prepared using ultrapure water (Millipore Simplicity 185 system).
Human leukemia monocytic cell line THP-1 was from ATCC (Manassas, VA, USA). RPMI 1640 medium, L-glutamine, penicillin and streptomycin were purchased from PanEco Ltd. (Moscow, Russia). Fetal bovine serum was from Biosera (Cholet, France). MTT was bought from Dia-M (Moscow, Russia). Bovine pancreatic ribonuclease was from Thermo Fisher Scientific Inc. (Waltham, MA, USA). ELISA kit for LPS was from Cloud-Clone Corp. (Wuhan, China).
2.2. Preparation of GM-CSF
The nucleotide sequence (NCBI reference sequence: NM_000758.4) encoding human GM-CSF chain 18-144 (UniProt ID P04141 lacking a signal peptide) was cloned into pET-32mod vector (uses E. coli thioredoxin as a fusion partner; the fusion protein is cleaved by bovine enteropeptidase [
53]) between NdeI and XhoI restriction sites. E. coli BL21 (DE3) cells carrying pLacIRARE plasmid were transformed with the resulting plasmid and grown in 2 L of 2xYT medium with 100 μg/mL ampicillin at 30 °C, shaking at 180 rpm, until optical density at 600 nm reached 1 AU. Expression of the thioredoxin‒GM-CSF chimera was triggered by 1 mM IPTG. The cells were grown for 5 h, harvested by centrifugation at 5000× g for 15 min at 4 °C, resuspended in 50 mL of 50 mM phosphate, 20% glycerol, 1 mM PMSF, 1 M NaCl, 0.5% TWEEN 20, pH 8.0 buffer, and disintegrated by a French press. The lysate was centrifuged at 30,000× g for 30 min at 4 °C. The pellet containing inclusion bodies of the thioredoxin‒GM-CSF chimera was dissolved in 20 mM Tris-HCl, 200 mM NaCl, 1 mM EDTA, 0.5% TWEEN 20, pH 7.0 buffer; the solution was stirred evenly for 15 min, followed by centrifugation at 6,900× g for 15 min at 4 °C. The last steps were repeated again. The pellet was solubilized in 10 mL of 8 M urea solution in 50 mM Tris-HCl, 10 mM DTT, pH 8.0 buffer. The solution was stirred evenly for 2 h at 4 °C, followed by centrifugation at 6,900× g for 15 min at 4 °C. The supernatant was diluted with 50 mM Tris-HCl, 2.7 M urea, pH 8.0 buffer (#1) to reach urea concentration of 5.3 M, followed by dialysis (MWCO of 3.5 kDa) against buffer #1 for 2 h and second dialysis against 50 mM Tris-HCl, 1 M urea, 0.4 M L-arginine, 3 mM reduced L-glutathione, 0.9 mM oxidized L-glutathione, pH 8.0 buffer (#2) overnight at 4 °C. Next day, the solution was dialyzed for 2 h against buffer #2 diluted with distilled water by 50%, then dialyzed against 1 L of 50 mM Tris-HCl, 250 mM NaCl, 0.1 M L-arginine, 3 mM reduced L-glutathione, 0.9 mM oxidized L-glutathione, pH 8.0 buffer for 2 h at 4 °C.
The insoluble material was removed by centrifugation (6,900× g for 15 min at 4 °C) and the supernatant was dialyzed twice against 2 L of distilled water for 2 h at 4 °C. The final dialysis was performed against 1 L of 50 mM phosphate, 150 mM NaCl, pH 8.0 buffer overnight at 4 °C. 20% glycerol, 1 M urea and 0.5% TWEEN 20 were added to the resulting solution, followed by stirring for 30 min and centrifugation at 6,900× g for 15 min at room temperature. The supernatant was loaded onto a Bio-Scale Mini Profinity IMAC Ni-charged column equilibrated with 50 mM phosphate, 1 M NaCl, 20% glycerol, 0.5% TWEEN 20, pH 8.0 buffer (A). The column was washed with an excess of buffer A. The protein was eluted with 50 mM phosphate, 500 mM imidazole, pH 8.0 buffer. The fractions containing thioredoxin‒GM-CSF chimera were collected and dialyzed against 20 mM Tris-HCl, 50 mM NaCl, 5% glycerol, pH 8.0 buffer (B) overnight at 4 °C. 2 mM CaCl2 was added to the resulting solution. The thioredoxin‒GM-CSF chimera was cleaved by incubation with light chain of bovine enteropeptidase (the eznyme to chimera molar ratio of 1:1.9×106) for 24 h at room temperature with stirring, centrifugation (6,900× g for 15 min at 4 °C) and one more digestion at the same conditions.
The His-tagged thioredoxin was removed from the sample via passage through the Bio-Scale Mini Profinity IMAC column. The GM-CSF sample was further purified using HiPrep
TM 26/60 Sephacryl
® S-100 HR gel filtration column equilibrated with 20 mM Tris-HCl, 150 mM NaCl, pH 8.0 buffer (flow rate of 1 mL/min). The fractions corresponding to GM-CSF were collected, concentrated to a concentration of 1.5 mg/mL and stored at −70 °C. The mass of the GM-CSF sample corresponds to residues 18-144 of UniProt ID P04141, which was confirmed by electrospray ionization mass spectrometry according to [
54]. Functional activity of the GM-CSF sample was verified using an MTT assay as described below.
2.3. Exression and Purification of S100A7L2/G/Z Proteins
The nucleotide sequences encoding human S100A7L2, S100G, S100Z proteins (Uniprot entries Q5SY68, P29377, Q8WXG8, respectively) were codon optimized for expression in E. coli. The syntesized S100 genes were cloned into pHUE vector [
55] between the SacII and NotI restriction sites. The ubiquitin-S100 chimera was produced in E. coli BL21 (DE3) cells carrying pLacIRARE plasmid, purified and cleaved by ubiquitin specific peptidase 2, followed by purification of the tag-free S100 protein according to [
42] with the following modifications. The cleavage was performed in 20 mM Tris-HCl, 1 mM DTT, pH 8.2 buffer. The subsequent dialysis step was skipped. Purification of S100Z protein using the anion exchange column was carried out at a pH value of 9.7. The final purification of the S100 proteins using the gel filtration column was performed in 10 mM phosphate, 150 mM NaCl, 1 mM 2-ME, pH 7.5 buffer. The purified S100 proteins were stored in 1:1 (v/v) mixture of PBS-glycerol with 1 mM 2-ME at −20 °C.
2.4. Surface Plasmon Resonance Studies
SPR measurements of GM-CSF interaction with S100 proteins at 25 °C were carried out using ProteOn™ XPR36 system (Bio-Rad Laboratories, Inc., Hercules, CA, USA) and amine coupling of a ligand (GM-CSF or S100 protein) to ProteOn GLH sensor chip mainly as described in [
27]. When using S100 proteins as a ligand, the surface of the sensor chip was flushed with 0.5% SDS solution to ensure dissociation of the S100 dimer. The running buffer was 10 mM HEPES, 150 mM NaCl, 0.05% TWEEN 20, 1 mM CaCl
2 or 5 mM EDTA, pH 7.4. The ligand was regenerated by passage of 20 mM EDTA pH 8.0 solution for 200 s. The double-referenced SPR sensograms were described within a heterogeneous ligand model (1) (GM-CSF serves as a ligand, L; A - analyte) or one-site binding model (S100P as a ligand).
L1 + A |
→ ← |
L1A |
L2 + A |
→ ← |
L2A |
|
(1) |
|
kd1 Kd1
|
|
|
kd2 Kd2
|
|
|
The corresponding kinetic and equilibrium dissociation constants, kd and Kd, respectively, were evaluated using Bio-Rad ProteOn Manager™ v.3.1 software (Bio-Rad Laboratories, Inc.). The kd/Kd values were estimated for 5 analyte concentrations, followed by their averaging (standard deviations are indicated).
2.5. Fluorimetric Studies
Fluorescence measurements were performed using a Cary Eclipse spectrofluorometer (Varian, Inc., Palo Alto, CA, USA) mainly according to [
43]. 0.57 µM GM-CSF solution in 10 mM HEPES-NaOH, 150 mM NaCl, 1 mM CaCl
2, pH 7.4 buffer was titrated at 25 °C by stock solution of S100A4 in the same buffer. Tryptophan fluorescence of GM-CSF was excited at 295 nm (S100A4 lacks Trp residues). Excitation and emission monochromator bandwidths were 5 nm and 10 nm, respectively. The fluorescence spectra were smoothed by a log-normal function [
56] using LogNormal software (IBI RAS, Pushchino, Russia). The dependence of fluorescence emission intensity at 360 nm on S100A4 to GM-CSF molar ratio was described by a one-site binding model using FluoTitr v.1.42 software (IBI RAS, Pushchino, Russia).
2.6. Chemical Crosslinking
6.7 mg/mL S100A4 in 20 mM Tris-HCl, 10 mM DTT, 20 mM EDTA, pH 8.0 buffer was loaded onto a NAP-5 desalting column and eluted with 10 mM HEPES, 150 mM NaCl, pH 7.4 buffer (A) for calcium removal. 4 µM GM-CSF, S100A4, and their mixtures in various molar ratios were treated with 0.02% glutaric aldehyde at 25 °C for 17 h (buffer A with/without 1 mM CaCl2 for Ca2+-loaded/depleted S100A4, respectively). The reaction was quenched by addition of SDS-PAGE sample loading buffer, followed by SDS-PAGE (15%) with silver staining and gel scanning using Molecular Imager PharosFX Plus System (Bio-Rad Laboratories, Inc., Hercules, CA, USA).
2.7. Structural Modeling of the GM-CSF -‒ S100A4 Complex
The models of tertiary structure of the complex between GM-CSF and S100A4 were built using ClusPro docking server [
57] (accessed on September 15, 2023), based on the crystal structures of human GM-CSF (PDB [
58] entry 1CSG, chain A) and Ca
2+-bound human S100A4 dimer (PDB ID 2Q91, chains A, B). Selection of the contact residues and the best model for visualization was carried out according to [
27,
41]. The structural models were drawn using the PyMOL v.2.5.0 software [
59] (accessed on September 15, 2023).
2.8. Cell Viability Studies
GM-CSF and S100A4/A6/P samples were controlled for undetectable LPS levels (protein concentration of 20,000 ng/mL), using the ELISA kit for LPS according to the manufacturer’s instructions.
THP-1 cells were cultured at 37 °C in a humidified 5% CO2 atmosphere in RPMI 1640 medium supplemented with 10% heat-inactivated fetal bovine serum, 2 mM L-glutamine, 100 U/mL penicillin and 100 μg/mL streptomycin. 500 μL of the cell suspension (4×105 cells per mL) in the serum-free media were treated with 20 ng/mL GM-CSF or 20/100/500/700/1,000/2,000 ng/mL of S100A4/A6/P or their mixture, and incubated at 37 °C in 5% CO2 atmosphere for 30 min. Then, 50 μL of the cell suspension (2×104 cells per well) was seeded in quadruplicate in 96-well plates, and 50 μL of the growth medium supplemented with 10% fetal bovine serum was added, followed by cultivation for the next 48 h. After that 10 μL of 5 mg/mL MTT stock solution was added to the cells, and the plates were kept under the same conditions for 3 h. 250 μL of DMSO per well was then added to dissolve the formazan crystals. The absorbance at 590 nm was measured using a FilterMax F5 microplate reader (Thermo Fisher Scientific Inc., Waltham, MA, USA). The experiments were repeated at least four times and the result were averaged.
2.9. Search of the Diseases Associated with GM-CSF and S100A4/A6/P Proteins
The data on the diseases associated with human GM-CSF (UniProt ID P04141), S100A4 (UniProt ID P26447), S100A6 (UniProt ID P06703), S100P (UniProt ID P25815) were collected from the human disease databases DisGeNET v.7.0 [
60] (accessed on September 5, 2023) and Open Targets Platform v.20.09 [
61] (accessed on September 6, 2023) as described in [
43]. The DisGeNET entries were manually curated; false positive records were removed.
4. Conclusions
Examination of the 18 members of S100 protein family for their affinity to GM-CSF shows that in addition to S100A6 [
26] and S100P proteins [
27], another promiscuous S100 protein [
67,
68], S100A4, binds GM-CSF
in vitro with very similar
Kd values laying in the micromolar region (
Table 1) and resembling those reported for S100A4 interaction with GST-RAGE fusion [
62]. Importantly, the lowest estimate of the
Kd value is close to the serum S100A4 levels observed in some pathological conditions [
71], suggesting a potential (patho)physiological role for the S100A4 ‒ GM-CSF interaction. This conclusion is supported by the effective suppression of the GM-CSF/S100A4-induced changes in viability of THP-1 cells by the extracellular S100A4 (
Figure 4).
The S100A4 binding to GM-CSF requires Ca
2+ binding to S100A4 and its multimerization. Whereas S100A6 binding to GM-CSF follows the same regularities, S100P monomerization causes only a slight decrease in its affinity for GM-CSF (
Table 1). Meanwhile, the response of S100A4/A6/P proteins to their monomerization contrasts with that observed for S100P interactions with IL-11 and IFN-β [
43,
47,
66], in which S100P monomerization increases its affinity for the four-helical cytokines by 1.4-2.2 orders of magnitude. These facts demonstrate that the difference in the affinities of dimeric and monomeric forms of a S100 protein to a four-helical cytokine largely depends on the specific cytokine.
The molecular docking data evidence presence of a conserved S100A4/A6/P-binding site in GM-CSF molecule, which overlaps with the residues known to recognize GM-CSF receptors (
Figure 3D). These predictions are consistent with the inhibitory effect of extracellular S100A4/A6 proteins on the GM-CSF-induced suppression of viability of THP-1 cells (
Figure 4 and
Figure 5). The predicted contact residues of S100A4/A6/P dimers are less conserved (
Figure 3E). Nevertheless, the predicted involvement of the ‘hinge’ region and residue F89 of S100P protein in GM-CSF binding was verified by mutagenesis.
Although both GM-CSF and S100A4/A6 suppress viability of THP-1 cells, their combined action inhibits their individual effects (
Figure 4 and
Figure 5) in accord with the modeling results, similarly to the effects described for S100A1/A4/B/P ‒ IFN-β interactions (MCF-7 cells [
42,
43,
44]), and for S100A12/A13 binding to soluble TNF (Huh-7 cells [
40]). Since S100A6 and S100P interact with 71-73% of the four-helical cytokines studied to date [
26,
27], the S100 proteins may act as poorly selective inhibitors of their activity. At the same time, S100 binding was shown to promote cytokine signaling in some cases, including activation of the amphiregulin-induced proliferation of fibroblasts by S100A4 [
34], and FGF2-medited stimulation of FGFR1 in myoblasts by S100B [
39,
78]. For this reason, larger-scale studies are needed to establish the functional consequences of the S100-cytokine interactions. Meanwhile, the revealed ability of S100A4/A6/P proteins to interact with GM-CSF and limit its functional activity (S100A4/A6) should be taken into consideration in the clinical use of recombinant GM-CSF and design of the drugs targeting GM-SCF or its receptors.
Author Contributions
Conceptualization, S.E.P.; validation, A.S.K., V.A.R., A.A.V., M.Y.Z., L.A.B., E.I.D., M.P.S., V.N.U., E.A.P., S.E.P.; formal analysis, A.S.K., V.A.R., A.A.V., M.Y.Z., L.A.B., E.I.D., M.P.S., S.E.P.; investigation, A.S.K., V.A.R., A.A.V., M.Y.Z., L.A.B., E.I.D., M.E.P., A.S.S., E.A.L., M.P.S.; data curation, A.S.K., V.A.R., A.A.V., M.Y.Z., L.A.B., E.I.D., M.P.S., S.E.P.; writing—original draft preparation, A.S.K., V.A.R., A.A.V., M.Y.Z., E.I.D., M.E.P., A.S.S., E.A.P., S.E.P.; writing—review and editing, V.A.R., V.N.U., E.A.P., S.E.P.; supervision, E.A.P., S.E.P.; project administration, S.E.P.; funding acquisition, S.E.P. All authors have read and agreed to the published version of the manuscript.