Next Article in Journal
The Role of Mitochondria in Obstructive Sleep Apnea: Implications for the Upper Airway Muscles
Previous Article in Journal
In Vitro Detoxification of Fumonisin B1 (FB1) into Hydrolyzed Fumonisin B1 (HFB1) by Lactobacillus spp. Isolated from Pig Caecum
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Differential Binding of ΔFN3 Proteins of Bifidobacterium longum GT15 and Bifidobacterium bifidum 791 to Cytokines Determined by Surface Plasmon Resonance and De Novo Molecular Modeling

by
Maria G. Alekseeva
1,
Sophia S. Borisevich
2,
Alfia R. Yusupova
1,
Diana A. Reznikova
1,3,
Dilara A. Mavletova
1,
Andrey A. Nesterov
1,
Margarita G. Ilyina
2,
Natalia I. Akimova
1,*,
Alexander A. Shtil
4,* and
Valery N. Danilenko
1
1
Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia
2
Synchrotron Radiation Facility—Siberian Circular Photon Source “SKlF” Boreskov Institute of Catalysis of Siberian Branch of the Russian Academy of Sciences, 630559 Koltsovo, Russia
3
Moscow Center for Advanced Studies, 123592 Moscow, Russia
4
Blokhin National Medical Research Center of Oncology, 115522 Moscow, Russia
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(21), 10560; https://doi.org/10.3390/ijms262110560
Submission received: 19 September 2025 / Revised: 17 October 2025 / Accepted: 27 October 2025 / Published: 30 October 2025
(This article belongs to the Section Molecular Immunology)

Abstract

Bifidobacteria, a genus of obligate anaerobes, comprise a major component of the intestinal microbiota. Importantly, bifidobacteria participate in immune reactions. These bacteria carry a species-specific operon in which the fn3 gene encodes a multifunctional protein FN3 that mediates bacterial adhesion to the intestinal epithelium and is capable of binding individual cytokines. Bioinformatics and biochemical approaches were used to study the possible interaction of recombinant ∆FN3 fragments of B. longum and B. bifidum strains with cytokines TNF-α, IL-6, IL-8, and IL-10. De novo molecular modeling generated, for the first time, the structural models of species-derived ∆FN3 proteins and revealed new tentative regions for differential cytokine binding. Combined treatment with ∆FN3 and TNF-α induced TNF-α mRNA abundance in the human monocytic cell line. Altogether, these findings provide structural evidence for the regulation of immune reactions by microbiota-derived proteins.

1. Introduction

Intestinal microbiota emerges as a key factor of homeostasis [1]. Commensal bacteria, in particular, the Bifidobacterium genus, gained momentum as functional modulators of a variety of physiological processes as well as in disease [1,2]. The B. longum infantis and B. bifidum species are the first bacteria that colonize the intestine of newborns [3,4], suggesting a principal role of microbiota in post-embryonic development. Of particular interest is a cross-talk between the intestinal microbiota and the immune system [5,6,7]. Liwen et al. (2018) demonstrated a role for bifidobacteria in the balance of cytokines TNF-α/IL-4 and IL-5/IL-17A as a tentative biomarker of asthma in children [8]. Furthermore, the addition of bifidobacteria to the cultured colon carcinoma cell line decreased the expression of TNF-α, IL-6, and IL-12 and concomitant elevation of IL-10 and IL-8 mRNAs [9]. Thus, the outcome of bifidobacteria–cytokine interactions may vary from pro- to anti-inflammatory depending on the specific context [10].
Mechanisms whereby bifidobacteria and cytokines interact deserve an in-depth investigation. In particular, these interactions presume direct binding of cytokines to bifidobacterial proteins. Can bifidobacteria provide candidate protein binders that physically target (selectively or promiscuously) mammalian cytokines and modulate their functions? Previously, we discovered a species-specific PFNA operon that determines a significant diversity of amino acid sequences between strains of different species of bifidobacteria. The operon contains five genes: pkb2, aaatp, duf58, tgm, and fn3 [11,12] that encode, respectively, the serine-threonine protein kinase [11,13,14]; MoxR-ATPase AAA-ATP, a protein with an unknown function that contains the annotated DUF58 domain, and a predicted transglutaminase [15]. The fn3 gene encodes a ~210 kDa membrane-bound FN3 protein. The functional roles of this protein in bifidobacteria are poorly understood. In B. bifidum strain S17, FN3 mediated the adhesion of bacteria to the intestinal epithelium [16]. Thus, FN3 proteins can mediate protein–protein interactions, and specifically, bind the recombinant cytokines in cell-free systems [15,17].
Bifidobacterial FN3 proteins contain two fibronectin type III domains (FNIII, FN3). These domains were initially identified in fibronectin, a component of the extracellular matrix [18,19,20], where they act as structural spacers or are involved in protein–protein interactions [21]. FN3 domains are structurally close to immunoglobulin domains with seven β-sheets, but the intradomain disulfide bond in FN3 is absent [22,23]. Nevertheless, FN3 domains are conformationally stable, thereby providing the protein binding motifs different from those engaged by antibodies [24]. Generally, FN3 domains share a consensus motif WSXWS (WS motif) in which the conservative residues WS…WS have been reported in the type I cytokine (e.g., IL-21) receptors [25]. Mutations in the WS motif affected interaction with cytokines [26,27]. The residue X plays a role in the spatial organization of protein–protein complexes and in signaling [25,28,29]. These data mechanistically attributed the WS motif in the binding of cytokines to their cognate receptors.
Our comparative analysis of nucleotide sequences of bifidobacterial FN3 domains revealed species-specific variations of the annotated WS motif: WSXPS, WSXES, WSXDS, or WSXYS. The most pronounced differences were found in B. angulatum and B. bifidum: the WS motif in the 2nd domain of FN3 proteins was absent [15,30]. One may hypothesize that the above stretches can be involved in the interaction of bifidobacteria with cytokines.
The FN3 protein in the B. longum GT15 strain carries two tandem domains. The region 3′ to the 2nd domain is the C-terminus [31]. Previously, we cloned the fn3 gene fragment encoding both FN3 domains and the C-terminal region (aa. 1494–1994). The protein expressed from this construct has been termed ΔFN3.1, where the symbol “Δ” denotes a truncated recombinant fragment encompassing two FN3 domains and the C-terminal region (aa. 1494–1994). This notation is used solely to distinguish the recombinant fragment from the full-length membrane-bound FN3 protein (~210 kDa) [17,31]. We demonstrated that ∆FN3.1 of B. longum GT15 can bind the recombinant TNF-α as determined in enzyme-linked immunosorbent assays [17]. This binding requires full-length ∆FN3.1, including the C-terminal region. The initial in silico analysis suggested that ΔFN3.1 can form a cavity for TNF-α binding [31]. Furthermore, treatment of B. longum GT15 with tumor necrosis factor α (TNF-α) modulated transcription of 176 operons (~1000 genes). Importantly, this treatment activated the expression of PFNA genes, including fn3 [32]. These results provided strong evidence in favor of the link between bifidobacterial FN3 proteins and cytokines, thereby justifying an in-depth analysis of structural and functional interactions between bifidobacteria and the immune system.
Recent advances in de novo protein structure prediction have expanded the toolbox for structural biology. Among the most powerful methods are deep learning-based approaches such as AlphaFold2 [33] and RoseTTAFold [34]. The former approach demonstrates an exceptional accuracy in predicting global folds of single and multi-domain proteins, especially when evolutionary information is abundant. RoseTTAFold provides superior modeling of inter-domain arrangements and flexible linkers due to its three-track neural network architecture that simultaneously processes sequence, distance, and coordinate information. In contrast, I-TASSER [35,36,37] relies on threading and fragment assembly, which may limit its performance for proteins lacking close homologs. Given the absence of experimentally resolved structures for bifidobacterial FN3 proteins, we employed a multi-platform strategy to ensure the robustness of our models and to cross-validate the predictions.
In the present study, we expanded the array of bifidobacterial strains and cloned the genes encoding ΔFN3 in B. angulatum и B. bifidum. Comparison of the binding of recombinant ΔFN3 proteins from different species of bifidobacteria to the cytokines TNF-α, IL-6, IL-8, and IL-10 demonstrated differential affinity to individual cytokines, as determined by the surface plasmon resonance technique. We, for the first time, predicted in silico tertiary structures of ΔFN3 fragments and identified new potential cytokine binding regions. In cell culture, the combined treatment with FN3 and TNF-α elevated the abundance of TNF-α mRNA, suggesting that the consequences of interactions between microbiota and the immune system should be considered with caution.

2. Results

2.1. Analysis of Amino Acid Sequences of Bifidobacterial ΔFN3 Fragments

The ΔFN3 protein in B. angulatum GT102 (NCBI AMK57067, locus_tag Bang102_000210; 1956 aa. residues; GenBank GCA_000966445.2) [30] contains two domains FN3 (residues 1454–1539 and 1544–1635). The ΔFN3 protein in B. bifidum 791 (NCBI KYJ84759, locus_tag APS66_RS03355; 1992 residues; GenBank GCA_022014355.1) also contains two domains FN3 (residues 1490–1576 and 1581–1665). The protein expressed from this DNA fragment was termed ΔFN3.1. The names of respective proteins in B. angulatum GT102 and B. bifidum 791 strains were ΔFN3.2 and ΔFN3.3. Amino acid sequence identities (similarities) were: 33 (49%) between ΔFN3.1 and ΔFN3.2; 39 (54%) between ΔFN3.1 and ΔFN3.3; and 32 (46%) between ΔFN3.2 and ΔFN3.3.
Alignment of amino acid sequences of bifidobacterial ΔFN3 proteins revealed motifs other than the previously described conservative WSXWS stretch. In B. longum, the 1st FN3 domain contains the WSXPS sequence in which the tryptophan residue is substituted with proline. In the 2nd domain, the respective motif is WSXES (glutamic acid instead of tryptophan). The most pronounced difference was detectable between the 2nd FN3 domains of B. angulatum: SGXXAS (W → S, S → G, W → Q, S → A) and B. bifidum: EGXPS (W → E, S → G, W → P).
Alignment of amino acid sequences of ΔFN3.1 B. longum GT15, ΔFN3.2 B. angulatum GT102, and ΔFN3.3 B. bifidum 791 revealed that the homologies in domains 1 were more pronounced than in domains 2 (Figure 1). Thirty-five core (conservative) residues were detected in the 1st domain compared to 16 in the 2nd domain. The C-terminal region is the most variable among bifidobacteria. Therefore, expanding the array of bifidobacterial strains to investigate the binding of species-specific FN3 proteins with cytokines, we cloned the genes encoding ΔFN3.2 B. angulatum and ΔFN3.3 B. bifidum, isolated the respective proteins, and compared their characteristics with the previously obtained ΔFN3.1.

2.2. Cloning and Expression of Genes Encoding ΔFN3.2 B. angulatum and ΔFN3.3 B. bifidum Isolation and Purification of Recombinant Proteins

Previously, we cloned the fn3 gene fragment (residues 1494–1994) of B. longum GT15 that encompasses two FN3 domains and a C-terminal region. The recombinant ΔFN3.1 was isolated [17,31]. In the present study, we cloned the genes encoding ΔFN3.2 B. angulatum and ΔFN3.3 B. bifidum. For PCR amplification of fragments of genomic DNAs of B. angulatum GT102 and B. bifidum 791, the oligonucleotides homologous to N- and C-terminal regions of the Δfn3.2 and Δfn3.3 genes, respectively, were used. The amplified fragments were cloned into the pET16b expression vector, followed by the procedures of protein expression and purification (see Section 4). SDS-PAGE showed ~56 kDa bands that corresponded to the calculated molecular masses of ΔFN3.2 and ΔFN3.3, including the His-Tag linker of the pET16b plasmid (Figure S1).
The criterion for selecting proteins for further research is the possibility of obtaining target proteins in preparative quantities in a native and homogeneous state. To test the possibility of purification of ΔFN3.2 B. angulatum and ΔFN3.3 B. bifidum under native conditions, pellets of E. coli BL21 (DE3) containing recombinant plasmids were lysed in a buffer (50 mM NaH2PO4, 5 mM Tris-HCl, 300 mM NaCl, 1 mM PMSF, 5 mM DTT (pH 8.0) containing lysozyme (1 mg/mL), 0.5% Triton X-100, and 20 mM 2-mercaptoethanol, sonicated with ultrasound and centrifuged to separate soluble fraction and the pellet. SDS-PAGE revealed that B. bifidum ΔFN3.2 is soluble, allowing for its purification under native conditions. The B. angulatum ΔFN3.3 protein was found in the pellet (inclusion bodies) and can be isolated only under denaturing conditions and subsequent re-folding. Thus, for further studies, ΔFN3.3 B. bifidum were used. The recombinant protein ΔFN3.3 of B. bifidum was purification in preparative quantities, the purity of the isolated protein was confirmed using SDS-PAGE (Figure S2).
Two approaches were used to investigate the physical interaction of ΔFN3.1 B. longum and ΔFN3.3 B. bifidum with cytokines: a kinetic analysis by surface plasmon resonance and in silico simulations. To conduct a comparative kinetic analysis of interactions with cytokines, a fresh recombinant protein ∆FN3.1 of B. longum GT15 was also purificated.

2.3. Interactions of ΔFN3.1 B. longum GT15 and ΔFN3.3 B. bifidum 791 with TNF-α, IL-6, IL-8, and IL-10

We determined the ability of ΔFN3.1 B. longum GT15 and ΔFN3.3 B. bifidum 791 to bind the recombinant TNF-α, IL-6, IL-8, and IL-10 using the surface plasmon resonance technique. These analyses were performed in neutral (pH 7.4) or alkaline (pH 8.0) solutions. These values were chosen based on the isoelectric points of ΔFN3.1 B. longum (pI 6.05) и ΔFN3.3 B. bifidum (pI 7.18). As shown in Table 1 and Figure S3, ΔFN3.1 B. longum GT15 and ΔFN3.3 B. bifidum 791 were avid TNF-α binders: dissociation constants were 13.1 nM and 58.2 nM, respectively. However, the efficacy of binding to interleukins was different. For ΔFN3.1 B. longum GT15, pH did not influence the binding to IL-8 (Table 2, Figure S4A,B). In contrast, at pH 7.4, the binding of ΔFN3.1 B. longum GT15 to IL-10 (Table 2, Figure S5A,B) was detectable (Kb = 62.2 nM). The ΔFN3.1 B. longum GT15 interacted with IL-6 neither at pH 8.0 nor at pH 7.4 (Table 2, Figure S6A,B). Furthermore, binding of ΔFN3.3 B. bifidum 791 to IL-8 (Figure S4C,D), IL-10 (Figure S5C,D), and IL-6 (Figure S6C,D) did not depend on pH (Table 3). Interactions of ΔFN3.3 B. bifidum 791 with IL-6 and IL-10 at either pH were below the level of detection. Thus, binding of ΔFN3.1 B. longum GT15 with IL-10 was pH-sensitive, probably due to conformational changes at alkaline conditions. ΔFN3.3 B. bifidum 791 showed no detectable interaction with IL-10.
These results indicated that ΔFN3.1 B. longum GT15 and ΔFN3.3 B. bifidum 791 differentially bind to TNF-α, IL-6, IL-8, and IL-10. We next performed a systematic analysis of protein–protein interactions using molecular modeling, aiming to identify critical parameters of these interactions.

2.4. Prediction of Tertiary Structures

Prior to our study, experimentally confirmed 3D structures of bifidobacterial ∆FN3.1 and ∆FN3.3 proteins were not received. Therefore, we performed de novo structural prediction using state-of-the-art deep learning servers: AlphaFold2, RoseTTAFold, I-TASSER, and IntFOLD5 [38,39]. Importantly, no homology modeling was performed, as suitable structural templates with high sequence identity were absent [31]. (Figure 2A). In so doing, we used data on eukaryotic FN3 domain-containing proteins resolved by X-ray crystallography. Presumably, the proteins are formed by two linear FN3 domains with the predominant anti-parallel β-sheets.
Tertiary structures of ΔFN3 proteins obtained with RoseTTAFold were used as references for comparison with the results of AlphaFold2 and IntFOLD5 (Table S1). For MD simulations and refinement of geometrical parameters, ∆FN3.1-AF2 and ∆FN3.1-IF5 models were chosen. The absence of black triangles in the prohibited areas reflects the absence of steric conflicts, that is, the models demonstrated high probability, similarly to RoseTTAFold.
Geometrical parameters of tertiary structures were refined for ∆FN3.1, predicted with RoseTTAFold (∆FN3.1-RF) and AlphaFold2 (∆FN3.1-AF2), as well as for ∆FN3.3, predicted with RoseTTAFold (∆FN3.3-RF) and IntFOLD5 (∆FN3.3-IF5). MD simulations lasted for 100 ns, followed by evaluation of RMSD fluctuations, clusterization, and Ramachandran map analyses of the most representative structures. MD simulations of ∆FN3.1-RF and ∆FN3.1-AF2 were completed successfully and reached the plateau (see RMSD curves in Figure S9A). The amplitude of RMSD fluctuations for ∆FN3.1-AF2 was substantially lower than for ∆FN3.1-RF, suggesting that the latter model is less stable. Evaluation of the spatial organization of representative frames using Ramachandran maps (Figure S9B) revealed a large number of sterically unfavorable conformations of the side chains in the amino acid residues in ∆FN3.1-RF compared to ∆FN3.1-AF2. Therefore, the latter model was considered in subsequent calculations.
The tertiary model of ∆FN3.3-RF achieved an equilibrium state neither within the initial 100 ns of MD simulations nor after an additional 100 ns (Figure S10C). To ensure that MD trajectories reached equilibrium, we applied the quantitative criteria: (i) the backbone RMSD plateaued (fluctuations ≤ 0.2 Å over the last 20 ns), (ii) the radius of gyration stabilized, and (iii) the Ramachandran outliers remained below 1%. For ∆FN3.3-RF, RMSD continued to drift beyond 100 ns (Figure S10C), indicating a failure to equilibrate; this model was excluded from further analysis. In contrast, ∆FN3.1-AF2 and ∆FN3.3-IF5 achieved stable RMSD plateaus (1.8 ± 0.3 Å and 10.5 ± 1.2 Å, respectively), confirming convergence. Clusterization of trajectory data was not performed. MD simulations of the ∆FN3.3-IF5 structure, followed by the clusterization procedure, generated a tertiary structure for molecular modeling stable for 100 ns. The RMSD curve (Figure S10B) calculated by Cα atoms reached a plateau and fluctuated within 8–12 Å. The Ramachandran maps (Figure S10C) revealed minor steric conflicts and irregular conformations in the loops and terminal zones. Nevertheless, in the clusterization procedure, the representative frames reflected the most stable conformations. The tertiary structure of ΔFN3.3 was chosen based on these data.
Thus, we focused on tertiary structures of ΔFN3.1 and ΔFN3.3 proteins obtained with AlphaFold2 and IntFOLD5, respectively. Of note, the models depicted in Figure 2B,C differed from the reported tertiary structures [31]. The reasons for these discrepancies remain to be elucidated; however, all three methodologies of structural prediction generated close results (Table S2).
Geometrical parameters of TNF-α, IL-8, IL-10, and IL-6 were taken from PDB. Among 37 structures [40], 4G3Y [41] and 5UUI [42] fit the parameters obtained by X-ray analyses with the best resolution. For interleukins, the best geometrical parameters corresponded to the PBD codes 1ALU [43]—IL-6, 5D14—IL-8, and 2H24 [44]—IL-10. Also, we used an AlphaFold2 methodology for the prediction of tertiary structures of the above proteins. Since these proteins are well studied, the results of the predictions were good (Table S2). Unlike for ΔFN3 proteins, different servers were unnecessary. Geometrical parameters of predicted tertiary structures of TNF-α, IL-8, IL-10, and IL-6 were refined during 100 ns MD simulations. The analysis produced the geometrical parameters of proteins that corresponded to the most representative clusterization frames (Figures S11 and S12).

2.5. Molecular Docking

2.5.1. FN3-TNF-α Interaction

The region(s) in TNF-α involved in the interactions with ∆FN3 proteins has not been identified [31]. For FN3 domain-containing eukaryotic proteins, the interactions take place in the cytokine receptor motif. We hypothesized that in the ΔFN3.1 protein of B. longum GT15, the following amino acid residues may be involved in binding to cytokines: Trp78, Ser79, Pro81, and Ser82 (the “cytokine receptor motif” in the 1st domain of FN3, annotated in NCBI); Trp174, Ser175, Glu177, Ser178 (the “cytokine receptor motif” in the 2nd domain of FN3, annotated in NCBI), and Ala43, Ala51, Thr111, Pro417, and Ala424, which we have identified in 203 sequenced genomes of B. longum subsp. longum [31]. For the B. bifidum 791 ΔFN3.3, it can be assumed that the residues involved in cytokine binding are located in the cytokine receptor motifs in the 1st and 2nd domains (Table 4). The amino acid sequences of ΔFN3.3 proteins of all B. bifidum genomes were identical (our unpublished data).
Protein–protein docking procedures generated 30 poses of the cytokine relative to FN3 proteins. In the first approximation, we selected optimal docking poses (Tables S3 and S4). Selection was based on energy parameters and the analysis of intermolecular interactions between the amino acid residues in the cytokine and FN3. Also, we considered the involvement of residues presented in Table 4. Then, two poses were chosen for MD refinement of the geometrical parameters of ΔFN3.1-TNF-α and ΔFN3.3-TNF-α complexes.
MD simulations of ΔFN3.1-TNF-α complexes in positions 24 and 5, as well as ΔFN3.3-TNF-α complexes in positions 21 and 14, were performed for 200 ns. RMSD fluctuations are given in Figure S8. The ΔFN3.1-TNF-α-pose5 equilibrated by 120 ns (Figure S13A), unlike ΔFN3.1-TNF-α-pose24 (Figure S13B). RMSD fluctuations were ~10 Å. A similar conclusion can be drawn from the analysis of ΔFN3.1-TNF-α MD curves (Figure S13C,D): in the ΔFN3.3-TNF-α-pose14, RMSD fluctuations of ΔFN3.3 and TNF-α were within 2–4 Å by the completion of simulations (Figure S13C), whereas in ΔFN3.3-TNF-α-pose21, the fluctuations were significant (Figure S13D).
Clusterization procedures produced a representative frame that corresponded to the most stable position of the system over the entire time of simulations. This frame contains similar RMSD values. In all cases, Ramachandran maps were good, with a few insignificant steric conflicts (Figure S14). Free energies of binding (ΔGbind) and dissociation constants (KD) were estimated for each complex. Since the binding regions of TNF-α and ΔFN3 are uncertain, we selected the complexes with KD values close to experimental values (Table S5).
During the entire time of simulation, TNF-α localized predominantly between epitopes I and II of ∆FN3.1 and formed intermolecular interactions such as hydrogen bonds, salt bridges, and π–π stacking (Figure 3A, Table S5). In the representative frame ∆FN3.1-TNF-α-pose24), the hydrogen bonds were found between the residues in TNF-α and epitope II; residues W174-Q123, D158-S147, and N173-Q123 were critically important (Table S5). In pose 5, the cytokine was bound to FN3. Intermolecular contacts between Y195 in TNF-α formed π–π cation stacking bonds with W174 (Figure 3B). This position of the cytokine relative to ∆FN3.1 was characterized by a smaller number of hydrogen bonds compared to pose24. However, the theoretically calculated KD value was closer to the experimental data.
Most probably, TNF-α interacts with ∆FN3.3 between the epitopes. Nevertheless, numerous intermolecular contacts with epitope III were registered (Table S5, Figure 3C). In pose14, the pairs of residues in the complexes form hydrogen bonds (Figure 3D). In pose21, the salt bridges were formed between K204-E219 in ∆FN3.3 and K216-E180 in the cytokine (Table S5). Values ΔGbind for TNF-α-∆FN3.3 complexes in pose14 were below those in pose21. Consequently, KD (∆FN3.3-TNF-α-pose14) was comparable with experimental data (Table S5, Figure 3D). We suggested that the main differences between KD values of TNF-α-FN3 complexes are dictated by the differential positioning of TNF-α relative to the surfaces of ∆FN3.1 and ∆FN3.3.

2.5.2. FN3–Interleukin Interactions

The potential binding site(s) of bifidobacterial ∆FN3 proteins with interleukins is also unknown. We selected optimal docking poses out of 30 for MD-assisted refinement of geometrical parameters of ∆FN3-interleukin complexes (Tables S6 and S7). For IL-8, two poses were selected in which the cytokine was located between the epitopes I and II (pose3) or II and III (pose11) in ΔFN3.1. A number of hydrogen bonds were found between the aa. residues (Table S6). In the majority of docking poses, the contacts of ∆FN3.3 with IL-8 involved the epitope V. In this case, we analyzed two opposite poses: #17 (IL-8 bound to the epitope V) and #19 (IL-8 bound to the epitope III; Table S5). In the latter pose, only a few hydrogen bridges were detected, unlike ΔFN3.3-IL-8 (pose17). The analysis of MD trajectories showed that both ΔFN3.1-IL-8 (pose3) and ΔFN3.1-IL-8 (pose11) reached an equilibrium by the end of the simulation (Figure S15A,B). Still, for ΔFN3.1-IL-8 (pose11) complexes, RMSD fluctuations of IL-8 atoms were more pronounced (~10 Å; Figure S15D) than in ΔFN3.1-IL-8 (pose3) complexes (Figure S9C). The interaction of IL-8 with the C-terminus of ΔFN3.3 (pose17) was more stable than with epitope III (pose19; Figure S15C,D). The Ramachandran maps that corresponded to the representative frames were satisfactory (Figure S16). The calculated values ΔGbind and KD suggested that the probable positioning of IL-8 on the surface of ΔFN3.1 and ΔFN3.3 corresponded to pose3 (Figure 4A) and pose17 (Figure 4B), respectively. The calculated KD values were in agreement with experimental values.
In the case of IL-10, we selected two poses: IL-10 binds to the C-terminus of ΔFN3.1 (pose16) or the cytokine is localized between the epitopes I and II (pose28). In the former scenario, more intermolecular hydrogen bonds and salt bridges were registered compared with pose28 (Table S6). One may suppose that IL-10 interacts with ΔFN3.3 via the C-terminus (Table S7). Each of four MD simulations showed an unstable positioning of the interleukin relative to the surface of ΔFN3 proteins (Figure S17). Representative frames with Ramachandran maps were obtained only for ΔFN3.1-IL-10 complexes in two poses (Figure S18). Values ΔGbind for both poses were comparable (Table S8). Nevertheless, KD values suggest that, predominantly, IL-10 makes contact with the epitope V at the C-terminus of ΔFN3.1 (Figure 3C).
According to the results of molecular docking, IL-6 preferentially binds to the C-termini of ΔFN3.1 and ΔFN3.3 (Tables S6 and S7), forming a small number of hydrogen bonds between the paired aa. residues. MD simulations revealed an unstable positioning of IL-6 relative to the surfaces of ΔFN3.1 or ΔFN3.3 (Figure S19). Since the experimental data showed a weak interaction of IL-6 with ΔFN3 proteins, we did not calculate ΔGbind and KD values.

2.6. Effects of ΔFN3 and TNF-α on Cytokine mRNA Abundance in THP-1 Cells

To obtain insight into the physiological significance of the binding of ΔFN3 to TNF-α, we studied the effects of the combination of these proteins on the expression of genes known to be regulated, at least in part, by TNF-α: IL-6, IL-8, and TNF-α. In so doing, we treated the THP-1 human monocytic leukemia cell line with ΔFN3 proteins alone or in combination with TNF-α for 3 h, followed by RT-PCR. Conditions of treatment were optimized in preliminary experiments. As shown in Figure 5, ΔFN3.1, ΔFN3.3, or TNF-α alone caused no significant increase in TNF-α mRNA except for relatively large (900 ng) amounts of ΔFN3 proteins. In contrast, the combinations of ΔFN3.1 or ΔFN3.3 with TNF-α elevated the abundance of TNF-α mRNA ~4–7-fold. The synergy was independent of the amounts of ΔFN3 proteins; that is, the fold increase of TNF-α mRNA was similar for each combination. These results strongly suggested that ΔFN3 can synergize with TNF-α in activating the gene encoding this cytokine. The effects of combinations were gene-specific: TNF-α alone was a strong inducer of the IL-8 mRNA, whereas no synergy with ΔFN3 proteins was detected (at least at the concentrations used in the experiments) (Figure S20). Also, no synergistic effect on the IL-6 mRNA was observed.

3. Discussion

To date, the main components of the human immune system have been well studied and described [45,46,47]. These components include effector cells, receptors localized on them, and signaling molecules called cytokines, which regulate and control the organism’s immune homeostasis. Cytokines are the most important cellular signaling molecules that mediate the organism’s immune response [48]. Cytokines are divided into pro-inflammatory and anti-inflammatory. Pro-inflammatory cytokines play an important role in protecting organisms from pathogens. However, an excessive inflammatory response can lead to the development of chronic inflammation and disruption of homeostasis, which can cause diseases such as cancer, diabetes, cardiovascular diseases, and Parkinson’s disease [49]. The main proinflammatory cytokines include L-6, IL-8, and TNF-α. Based on its functions, the immune system is divided into innate and adaptive [50]. The adaptive immune system is believed to have emerged and functions in response to various effector factors, including pathogenic bacteria, viruses, and effector molecules of different natures.
The adaptive immune system plays an important role in self-or-foe recognition. Pathogenic bacteria can be classified as foreign agents. But to what category should commensal bacteria of the gut microbiota, and particularly bifidobacteria, be classified? Intensive research over the past decade into the human and animal gut microbiome has enabled the identification and characterization of commensal, functionally beneficial bacteria using effector molecules, proteins, and cellular components that interact with the host organism and, directly or indirectly, with components of its immune system [51,52,53].
Thus, the gut microbiome is another important component of the immune system. In addition to the microbiome–brain axis, the microbiome–immune system axis has begun to emerge and be studied. Then came the matter of searching for receptor sites within elements of this immune system component that are capable of interacting with the ligands and signaling molecules (cytokines) of the organism`s immune system. These receptor domains have been identified and partially characterized in bifidobacteria [12,15,17].
Bifidobacteria comprise an ancient group of anaerobic bacteria [54]. These microbes are the only representatives of actinobacteria in the gastrointestinal tract [55], accounting for 99 species [56]. B. longum and B. bifidum are the primary residents of the child’s intestine that participate in the immune system development [3]. Bifidobacteria and their metabolites have been implicated in intestine–brain and intestine–immunity cross-talks [5,57].
The FN3 protein, initially identified by us as a product of the PFNA operon, can be a candidate mediator of immune regulation by bifidobacteria. ΔFN3 proteins demonstrate a high interspecies divergence of amino acid sequences. Also, ΔFN3 carries various motifs of cytokine receptors; these structural features substantiate the differential affinity to certain cytokines. We have shown that the conservative cytokine receptor motifs in bifidobacteria contain WS-PS, WS-ES, WS-DS, or WS-YS stretches. The most striking was the difference between B. angulatum and B. bifidum [15], namely, the absence of the WSXWS motif in the 2nd FN3 domain.
In the present study, we dissected FN3-cytokine interactions in more detail. Previously, we have used ELISA to show that the FN3 fragment (∆FN3.1) of the B. longum GT15 strain preferentially binds TNF-α [17]. The structural model of ΔFN3.1 generated with the trRosetta software «https://yanglab.qd.sdu.edu.cn/trRosetta/ (accessed on 14 December 2021)» included five epitopes with β-sheets as predominant folds. Two epitopes are formed by two FN3 domains, whereas another three epitopes are presented in the C-terminal region [31]. We determined the binding profiles of ΔFN3.1 B. longum GT15 and ΔFN3.3 B. bifidum 791 with TNF-α, IL-6, IL-8, and IL-10 using the surface plasmon resonance measurements. Kinetic analysis of ΔFN3-cytokine interaction proved that ΔFN3.1 and ΔFN3.3 can bind TNF-α and IL-8. Furthermore, ΔFN3.1 was capable of binding IL-10, whereas IL-6 interacted with neither ΔFN3.
We next performed de novo molecular modeling [34] and neuronal network technologies to generate, for the first time, the tertiary structures of ΔFN3.1 and ΔFN3.3. Based on these models, we provided novel evidence regarding tentative cytokine binding regions in ΔFN3 proteins. The results of calculated binding energies and dissociation constants of ΔFN3-cytokine complexes were in good agreement with experimental data.
Most importantly, our modeling demonstrated that cytokine-binding regions differed for individual ΔFN3 proteins. For ΔFN3.1 B. longum GT15, TNF-α interacted presumably with the 2nd FN3 domain; this interaction involved aa. residues of the cytokine receptor motif WSXWS (Figure S21). In complexes ΔFN3.1-IL8 pose3 (Table S8), the interaction involves W174 in the 2nd domain as well as the neighboring residues I172 and N173. In contrast, the above motif is absent in the 2nd domain of ΔFN3.3 B. bifidum 791; the binding involved other residues located in the 1st domain and the C-terminus. These findings, being not obviously predictable, expand our knowledge of mechanisms of interactions between microbiota and the immune system. Several protein regions in the participating proteins are involved, making these interactions non-incidental; mutational analysis will determine the significance of specific sites. One may hypothesize that individual regions provide more conformational opportunities that ultimately cooperate to form stable complexes. Thus, structural diversity of binding regions ensures the evolutionary conserved manner of microbiota–cytokine interactions.
Testing whether the affinity of ΔFN3 to cytokines in cell-free systems may be translated into the physiological effect, we observed that both ΔFN3.1 and ΔFN3.3 synergized with TNF-α in the activation of the TNF-α gene in the human monocyte cell line. We suggest that ΔFN3 binds TNF-α in the extracellular milieu, thereby facilitating the autoregulatory loop ΔFN3-TNF-α-TNF-α induction. The synergistic effect of the combination was specific for TNF-α since no additivity was registered for IL-8 or IL-6 mRNAs upon cell exposure to TNF-α together with either ΔFN3.1 or ΔFN3.3. These observations add complexity to the interpretation of the role of microbiota in immune regulation. TNF-α is a pleiotropic cytokine with established significance in a variety of immune reactions [58]. The gene encoding this factor carries two promoters with functionally opposite effects [59]. More studies are needed to clarify the mechanism(s) whereby the bifidobacterial proteins synergize with TNF-α to sustain TNF-α activation. One plausible explanation could be the acquisition of a specific confirmation of TNF-α in complexes with ΔFN3, making the cytokine a stronger inducer of the cognate gene. This assumption is in line with the above-mentioned conformational hypothesis: the more binding sites, the more variants of formation of functionally diverse protein–protein complexes. Finally, interference with the ΔFN3-TNF-α interaction with a small molecular weight compound or a peptide inhibitor might be considered for the disruption of the autoregulatory loop to attenuate stress-induced cytokine release.
Tumor necrosis factor TNF-α is a pleiotropic cytokine and one of the key modulators of the human immune system [58]. The data obtained do not allow us to definitively suggest the mechanisms of action that enhance TNF-α gene expression. According to the literature data, there is [59] a TNF-α expression pattern that takes into account the presence of two promoter regions, the preferential binding of which leads to opposite functional results. Further studies are required to establish the molecular mechanisms that determine the influence of the interaction of ΔFN3 proteins with TNF-α on the mechanism of regulation of its expression in certain effector immune cells of the human body.
IL-8 is an important inflammatory mediator that induces the expression of adhesion molecules on the vessel wall, in that way stimulating neutrophil binding, adhesion, and migration to tissue sites of injury, as well as inducing the formation of neutrophil extracellular traps (NETs) [60]. It is of great interest how the FN3.3 protein, through its interaction with IL-8, participates in these processes.
IL-6 is considered a major mediator of immune and inflammatory responses [48]. Furthermore, IL-6 is one of the key factors contributing to the virus-induced cytokine “storm.” IL-6 acts by binding to the membrane-bound IL-6R receptor and forming a complex with the membrane glycoprotein gp130. The latter step triggers the Jak/STAT and NFκB-mediated proinflammatory response in immune cells, such as macrophages, neutrophils, and T- and B-lymphocytes [61]. Tumor necrosis factor TNF-α is a pleiotropic cytokine and one of the key modulators of the human immune system.
The observed differences in cytokine-binding profiles between ∆FN3.1 (B. longum) and ∆FN3.3 (B. bifidum) may underlie species-specific immunomodulatory effects of bifidobacteria in the human gut. For instance, the ability of ∆FN3.1, but not of ∆FN3.3, to bind IL-10 (an anti-inflammatory cytokine), suggests that B. longum strains might contribute more effectively to the resolution of inflammation. Conversely, the stronger TNF-α binding by ∆FN3.1 could amplify pro-inflammatory signals, as demonstrated by upregulation of TNF-α mRNA in THP-1 cells. These findings highlight the potential of ∆FN3 proteins as strain-specific biomarkers for personalized probiotic selection and as scaffolds for engineered immunomodulators.

4. Materials and Methods

4.1. Bacterial Strains, Plasmid Vectors, Culture Media, and Conditions

We used the following strains: B. angulatum GT102 [30], B. bifidum 791, E. coli DH5a (F–, Φ 80 ΔlacZΔM15, Δ(lacZYA-argF), U169) (Promega, Madison, WI, USA) [62], and E. coli BL21(DE3) (F–, dcm, ompT, hsdS(r B–mB–), gal λ (DE3)) (Novagen, Madison, WI, USA). The expression vector pET16b (Novagen, Madison, WI, USA) [63] contains a His-Tag linker in the N-terminal region for protein isolation and purification. B. angulatum GT102 and B. bifidum 791 strains were cultured under anaerobic conditions (HiAnaerobic SystemeMark III, HiMedia, Maharashtra, Mumbai, India) in agar and MRS broth (HiMedia, Maharashtra, Mumbai, India) supplemented with cysteine (0.5 g/L) at 37 °C for 24–48 h. E. coli strains were propagated in Luria-Bertani (LB) broth [64]. Ampicillin (150 μg/mL) was used as a selective agent for the selection of plasmid-bearing cells. All reagents were from Amresco (Solon, OH, USA) unless specified otherwise.

4.2. DNA Manipulations

Genomic DNAs of B. angulatum GT102 and B. bifidum 791 were isolated using the GenElute Bacterial Genomic DNA Kit (Sigma-Aldrich, St. Louis, MO, USA). Isolation of plasmid DNA, obtaining competent E. coli cells, transformation, and analysis of recombinant plasmids were performed using standard methods [64]. DNA fragments carrying Δfn3 were amplified from genomic DNAs of B. angulatum GT102 and B. bifidum 791 strains using Phusion High Fidelity PCR Master Mix (Thermo Fisher Sci., Vilnius, Lithuania) on a mini-cycler PTC-0150 (MJ Research, Inc., Waltham, MA, USA). The following oligonucleotides were used for amplification: FN3B.ang-N: (5’-tcgtcatatgcccgacgccccgtcactgt-3’), FN3B.ang-C: (5’-gatcctcgagctaccgggaatacgtatgcaattc-3’), FN3B.bif-N(5’-tcgtcatatggacaagcccggcgcgccgc-3’), and FN3B.bif-C: (5’-gatcctcgagtcatggtcggtttgaggccag-3’) (all from Eurogen, Moscow, Russia). PCR-amplified fragments were cloned into the NdeI and XhoI restriction sites of the His-Tag-containing pET16b expression vector.

4.3. Expression in E. coli and Purification of Recombinant ΔFN3 Proteins

E. coli BL21 (DE3) cells containing the recombinant plasmid were grown in LB broth at 37 °C until they reached an OD600 of 0.6–0.8. The fn3 gene of B. angulatum GT102 and B. bifidum 791 was induced by the addition of 1 mM isopropyl-β-D-1-thiogalactopyranoside (IPTG) for 5 h at 28 °C. Cells were pelleted and frozen at −20 °C. To study the expression of fn3, cells were resuspended in a sample buffer containing 62.5 mM Tris-HCl, pH 6.8, 5% glycerol, 2% 2-mercaptoethanol, 0.1% SDS, and 0.001% bromophenol blue, then heated at 95 °C for 10 min and analyzed by 12.5% SDS-PAGE. Protein fractions of E. coli BL21 (DE3) cells containing an empty pET16b plasmid were used as negative controls.
Isolation and purification of ΔFN3.1 B. longum GT15 and ΔFN3.3 B. bifidum 791 were carried out as described [17]. Samples were dialyzed in a buffer containing 10 mM HEPES-NaOH, pH7.4, 150 mM NaCl, 10% glycerol, and 1 mM phenylmethylsulfonyl fluoride (PMSF). Protein concentration was measured on a Qubit 2.0 fluorimeter (Invitrogen, Waltham, MA, USA). Purified proteins were stored at −80 °C.

4.4. Interaction of ΔFN3 Proteins with TNF-α, IL-8, IL-6, and IL-10

For surface plasmon resonance analysis (imSPR-Pro system, iCLIEBIO, Dangjin-si, Chungcheongnam-do, Republic of Korea), recombinant TNF-α, IL-8, IL-6, and IL-10 (ProSpec, Rehovot, Israel) were immobilized on the sensor COOH-chip (iCLIEBIO). Four different concentrations of the analyte sample (ΔFN3.1 B. longum GT15 or ΔFN3.3 B. bifidum 791) were prepared by serial dilution in a running buffer containing 10 mM HEPES-NaOH pH8.0 (or pH 7.4), 150 mM NaCl, and 0.05% Tween-20. The ΔFN3.1 B. longum GT15 or ΔFN3.3 B. bifidum 791 samples were injected at a flow rate of 30 µL/min. The injection step included a 200 s association phase followed by a 600 s dissociation phase. Serum albumin (SibEnzyme, Novosibirsk, Russia) was used as a control for non-specific protein binding. Data were analyzed by globally fitting curves describing the simple 1:1 bimolecular model to the set of three sensorograms using BIAEvaluation software version 4.1 «http://biaevaluation.software.informer.com (accessed on 4 February 2024)». Sensorograms characterize the changes of the signal over time. Signals were measured in resonance units (RU) proportionally to the amount of the surface-bound substance. Initially, the analytes ∆FN3 B. longum GT15 and B. bifidum 791 were loaded onto the chip and allowed to interact with TNF-α, IL-6, IL-8, and IL-10 for 200 s. The sensorograms registered the responses over time. The rate of complex association Ka (1/Ms) was calculated. Next, the chips were washed with a running buffer, and the complexes were allowed to dissociate for up to 600 s. This process reflected a decreased response on the sensorograms. The dissociation rate Kd (1/s) and dissociation constant KD = Kd/Ka (M) were calculated.

4.5. Molecular Modeling Studies

4.5.1. Prediction of Tertiary Structure of ∆FN3 Proteins

Tertiary structures of ∆FN3.1 and ∆FN3.3 were predicted on the basis of AlphaFold2 [33], RoseTTAFold [34], I-TASSER [35,36,37], IntFold5 [38], and scoring functions [39]. In AlphaFold2 and RoseTTAFold, the accuracy of predictions is estimated as predicted Local Distance Difference Test (pLDDT) values that reflect local atomic distances [65]. The I-TASSER and IntFOLD5 tools use their evaluation functions to assess the accuracy of prediction, analogously to pLDDT: C-score (Confidence Score) [35,66,67] and GMQS (Global Model Quality Score) [38,68]. The algorithm is presented in Figure S22. Tertiary structures of TNF-α, IL-6, IL-8, and IL-10 were predicted using AlphaFold2 [33]. The geometrical parameters of each structure were refined by molecular dynamic (MD) simulations within 100 ns.
Selection of optimal structures among the array of predicted variants was based on scoring functions in AlphaFold2 [33], RoseTTAFold [34], I-TASSER [35,36,37], and IntFOLD5 [38]. Table S2 shows the quality of prediction for the best structures out of five variants. In all cases, the non-structured portions at the C-termini of ∆FN3.1 and ∆FN3.3 (aa. 475–503) were poorly predictable (red color). In general, the predictive results obtained with AlphaFold2, RoseTTAFold, and IntFOLD5 were similar. In contrast, the I-TASSER algorithm generated the structures with minimal relevance to the expected architecture of ΔFN3-containing proteins.
The analysis of Ramachandran maps (Figures S7 and S8) allowed us to estimate the geometrical probability of predicted ∆FN3 proteins. Top quality models were obtained with RoseTTAFold, AlphaFold2, and IntFOLD5: >90% amino acid residues were located in permissible areas (red and yellow) with a minimal number of prohibited conformations. I-TASSER generated the biggest number of geometrical alterations, making this algorithm not applicable for further analysis. Thus, we focused on predictions of ∆FN3.1 and ∆FN3.3 structures with RoseTTAFold. Recently, this server has been validated as preferred for the prediction of the full-size tertiary structure of poly(ADPribose) polymerase 1 [39].

4.5.2. Protein–Protein Docking

To study the affinity of TNF-α, IL-8, IL-10, and IL-6 to FN3 proteins, we used the protocol of protein–protein docking PIPER [69]. The protocol is based on the method of quick Fourier transformation with a novel potential DARS (Decoys As the Reference State). The docking algorithm presumes the rotation of the ligand (TNF-α, IL-8, IL-10, or IL-6) relative to the receptor (∆FN3 protein) fixed in the coordinate system; all possible orientations of proteins relative to each other are detectable. The systems were considered solid bodies without optimization of the protein–protein interface. Because the binding regions have not been identified, the positions were unlimited. The docking poses were ranged according to the weight coefficients of energy terms PIPER score and PIPER energy [70].

4.5.3. MD Simulations

Model systems of tertiary structures and protein–protein complexes were generated in the graphic milieu of the academic version of Maestro (Schrödinger Release 2024-1). The systems were placed into a cubic well with the buffer zone 15–25 Å filled with 0.15 M NaCl. Extra charges on proteins were neutralized with Na+ and Cl. TIP3P was used as a solvent. MD modeling was performed in an NPT ensemble at 310 K (37 °C). Total time of simulations was 50–200 ns, 2 fs increment of the integrator, 5000–10,000 trajectory frames. The force field was OPLS4 [71]. MD simulations were performed using Desmond [72]. The analysis included mean square deviation of atomic positions (RMSD) and clusterization of frames [73].

4.5.4. Estimation of Binding Energy and KD

Representative frames were aligned with geometrical parameters of statistically significant protein–protein complexes for which the free binding energies (∆Gbind) were estimated according to MM-GBSA methodology [74]. Frames were selected based on minimal RMSD values and maximal number of repetitive images. The binding of TNF-α and interleukins to the surface of FN3 proteins can be expressed as:
P + L e q P e q + L e q ,
where [P]eq are FN3 proteins, [L]eq are TNF-α or interleukins, [P+L]eq is the protein–protein complex in an equilibrated state. The value ∆Gbind depends on the dissociation constants as:
Δ G b i n d = R T ln K D ° ,
where R is universal gas constant = 8.314 J/(mol × K), T is temperature, K D ° is the dissociation constant:
K D ° = K i 1 = P + L P L .
The physical meaning of K D ° represents the standard constant of equilibrium, a value transformed into KD using the formula:
K D = K D ° R T P ν
where P is pressure (H/m2), ∆ν is the sum of stoichiometry parameters of reaction coefficients (1).

4.6. Detection of Cytokine mRNA by Reverse Transcription-Polymerase Chain Reaction

The THP-1 human monocytic leukemia cell line (American Type Culture Collection, Manassas, VA, USA) was propagated in RPMI-1640 supplemented with 2 mM L-glutamine (PanEco, Moscow, Russia), 10% fetal bovine serum (Atlanta Biol., Flowery Branch, GA, USA), 50 U/mL penicillin, and 50 µg/mL streptomycin (PanEco, Moscow, Russia) at 37 °C, 5% CO2 in a humidified atmosphere. Cells in the logarithmic phase of growth were used in experiments. Cells were plated into 6-well plates (FDCELL, China; 4 × 105/2 mL of culture medium) and treated with 300 ng of recombinant TNF-α (SCI STORE, Moscow, Russia) in the absence or presence of varying amounts of ΔFN3.1 or ΔFN3.3 for 3 h (Table S9). Cells were pelleted, washed with saline, and lysed in ExtractRNA reagent (Evrogen, Moscow, Russia). Total RNA was isolated according to the manufacturer’s instructions. Reverse transcription was performed using the MMLV RT kit (Evrogen, Moscow, Russia). PCR mixtures (25 µL) contained 13 ng cDNA template, primers (0.4 µM each), qPCRmix-HS SYBR (Evrogen, Moscow, Russia), and deionized water. Amplifications were carried out on a CFX96 (Bio-Rad, Hercules, CA, USA) at 95 °C—5 min, 95 °C 30 s, 62 °C 30 s, and 72 °C 30 s (40 cycles). To analyze RT-PCR data, the CFX Manager V 3.1 program (Bio-Rad) was used. The HPRT1 mRNA was taken as a reference [75]. Three biological replicates (each sample in triplicate) were analyzed; data were expressed as ∆∆Cq. Primers were designed using primer-BLAST [76] (Table S10).

4.7. Bioinformatic Analysis

Nucleotide and amino acid sequences were from NCBI «http://www.ncbi.nlm.nih.gov/ (accessed on 13 December 2021)» and UniProt «http://www.uniprot.org/ (accessed on 17 December 2021)» databases. BLAST «https://blast.ncbi.nlm.nih.gov/Blast.cgi (accessed on 13 December 2021) » and Clustal Omega «https://www.ebi.ac.uk/jdispatcher/msa/clustalo (accessed on 14 December 2021)» programs were used for sequence alignment. Molecular weights and the isoelectric points of newly isolated proteins were calculated based on Molbiol servers «http://molbiol.ru/scripts/01_18.html (accessed on 17 January 2022)».

5. Conclusions

Interactions of the intestinal microbiota with the immune system form an important axis in normal homeostasis as well as in disease. The differential ability of individual bifidobacterial proteins to bind cytokines sets the stage for personalized use of chemical and biotechnological instruments that regulate these interactions. Knowledge about the molecular determinants of microbiota–cytokine binding can be expanded to other commensal species. Bifidobacterial ΔFN3 proteins are perspective as prototypic modulators of microbiome-immunity cross-talk, in particular, for the design of chemical or peptide disruptors for prophylaxis and therapy.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms262110560/s1.

Author Contributions

Conceptualization, M.G.A., S.S.B., and V.N.D.; methodology, M.G.A., S.S.B., D.A.M., and A.A.S.; software, M.G.A. and S.S.B.; validation, M.G.A., S.S.B., A.R.Y., A.A.S., and V.N.D.; formal analysis, M.G.A., A.R.Y., M.G.I., and A.A.S.; investigation, M.G.A., D.A.M., A.A.N., and D.A.R.; resources, S.S.B. and V.N.D.; writing—original draft preparation, M.G.A. and N.I.A.; writing—review and editing, N.I.A., A.A.S., and V.N.D.; visualization, M.G.A., S.S.B., and N.I.A.; supervision, V.N.D.; project administration, V.N.D.; funding acquisition, V.N.D. All authors have read and agreed to the published version of the manuscript.

Funding

This work was carried out as part of the State Contract “Creation of biotechnological products for agriculture, food industry, and medicine” No. 125050605758-0.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article/Supplementary Materials. Further inquiries can be directed to the corresponding authors.

Acknowledgments

The analysis of the interaction of ΔFN3 proteins with cytokines by the surface plasmon resonance method was performed at the Center for Collective Use “Structural and Functional Studies of Proteins and RNA” of the Protein Institute of the Russian Academy of Sciences (No. 3678921, «https://protres.ru/ckp-ib-ran (accessed on 21 May 2024)». The authors are grateful to Venera Z. Nezametdinova for providing experimental materials and participating in the discussion of the results, and to Ekaterina S. Ivanova for assistance in experiments.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
C-scoreConfidence Score
DARSDecoys As the Reference State
GMQSGlobal Model Quality Score
MD simulations Molecular dynamic simulations
pLDDTPredicted Local Distance Difference Test

References

  1. Sharma, A.; Sharma, G.; Im, S.H. Gut microbiota in regulatory T cell generation and function: Mechanisms and health implications. Gut Microbes 2025, 17, 2516702. [Google Scholar] [CrossRef] [PubMed]
  2. Kumar, H.; Collado, M.C.; Wopereis, H.; Salminen, S.; Knol, J.; Roeselers, G. The Bifidogenic Effect Revisited-Ecology and Health Perspectives of Bifidobacterial Colonization in Early Life. Microorganisms 2020, 8, 1855. [Google Scholar] [CrossRef] [PubMed]
  3. Rabe, H.; Lundell, A.C.; Sjöberg, F.; Ljung, A.; Strömbeck, A.; Gio-Batta, M.; Maglio, C.; Nordström, I.; Andersson, K.; Nookaew, I.; et al. Neonatal gut colonization by Bifidobacterium is associated with higher childhood cytokine responses. Gut Microbes 2020, 12, 1847628. [Google Scholar] [CrossRef]
  4. Ennis, D.; Shmorak, S.; Jantscher-Krenn, E.; Yassour, M. Longitudinal quantification of Bifidobacterium longum subsp. infantis reveals late colonization in the infant gut independent of maternal milk HMO composition. Nat. Commun. 2024, 15, 894. [Google Scholar] [CrossRef]
  5. Shang, J.; Wan, F.; Zhao, L.; Meng, X.; Li, B. Potential Immunomodulatory Activity of a Selected Strain Bifidobacterium bifidum H3-R2 as Evidenced in vitro and in Immunosuppressed Mice. Front. Microbiol. 2020, 11, 2089. [Google Scholar] [CrossRef]
  6. Vinkler, M.; Fiddaman, S.R.; Těšický, M.; O’Connor, E.A.; Savage, A.E.; Lenz, T.L.; Smith, A.L.; Kaufman, J.; Bolnick, D.I.; Davies, C.S. Understanding the evolution of immune genes in jawed vertebrates. J. Evol. Biol. 2023, 36, 847–873. [Google Scholar] [CrossRef]
  7. Alessandri, G.; van Sinderen, D.; Ventura, M. The genus bifidobacterium: From genomics to functionality of an important component of the mammalian gut microbiota running title: Bifidobacterial adaptation to and interaction with the host. Comput. Struct. Biotechnol. J. 2021, 19, 1472–1487. [Google Scholar] [CrossRef]
  8. Liwen, Z.; Yu, W.; Liang, M.; Kaihong, X.; Baojin, C. A low abundance of Bifidobacterium but not Lactobacillius in the feces of Chinese children with wheezing diseases. Medicine 2018, 97, e12745. [Google Scholar] [CrossRef]
  9. Choi, Y.J.; Shin, S.H.; Shin, H.S. Immunomodulatory Effects of Bifidobacterium spp. and Use of Bifidobacterium breve and Bifidobacterium longum on Acute Diarrhea in Children. J. Microbiol. Biotechnol. 2022, 9, 1186–1194. [Google Scholar] [CrossRef]
  10. Lim, H.J.; Shin, H.S. Antimicrobial and Immunomodulatory Effects of Bifidobacterium Strains: A Review. J. Microbiol. Biotechnol. 2020, 30, 1793–1800. [Google Scholar] [CrossRef] [PubMed]
  11. Nezametdinova, V.Z.; Mavletova, D.A.; Alekseeva, M.G.; Chekalina, M.S.; Zakharevich, N.V.; Danilenko, V.N. Species-specific serine-threonine protein kinase Pkb2 of Bifidobacterium longum subsp. longum: Genetic environment and substrate specificity. Anaerobe 2018, 51, 26–35. [Google Scholar] [CrossRef] [PubMed]
  12. Dyachkova, M.S.; Chekalin, E.V.; Danilenko, V.N. Positive Selection in Bifidobacterium Genes Drives Species-Specific Host-Bacteria Communication. Front. Microbiol. 2019, 10, 2374. [Google Scholar] [CrossRef]
  13. Nezametdinova, V.Z.; Zakharevich, N.V.; Alekseeva, M.G.; Averina, O.V.; Mavletova, D.A.; Danilenko, V.N. Identification and characterization of the serine/threonine protein kinases in Bifidobacterium. Arch. Microbiol. 2014, 196, 125–136. [Google Scholar] [CrossRef]
  14. Zakharevich, N.V.; Averina, O.V.; Klimina, K.M.; Kudryavtseva, A.V.; Kasianov, A.S.; Makeev, V.J.; Danilenko, V.N. Complete Genome Sequence of Bifidobacterium longum GT15: Identification and Characterization of Unique and Global Regulatory Genes. Microb. Ecol. 2015, 70, 819–834. [Google Scholar] [CrossRef]
  15. Nezametdinova, V.Z.; Yunes, R.A.; Dukhinova, M.S.; Alekseeva, M.G.; Danilenko, V.N. The Role of the PFNA Operon of Bifidobacteria in the Recognition of Host’s Immune Signals: Prospects for the Use of the FN3 Protein in the Treatment of COVID-19. Int. J. Mol. Sci. 2021, 22, 9219. [Google Scholar] [CrossRef] [PubMed]
  16. Westermann, C.; Gleinser, M.; Corr, S.C.; Riedel, C.U. A Critical Evaluation of Bifidobacterial Adhesion to the Host Tissue. Front. Microbiol. 2016, 7, 1220. [Google Scholar] [CrossRef] [PubMed]
  17. Dyakov, I.N.; Mavletova, D.A.; Chernyshova, I.N.; Snegireva, N.A.; Gavrilova, M.V.; Bushkova, K.K.; Dyachkova, M.S.; Alekseeva, M.G.; Danilenko, V.N. FN3 protein fragment containing two type III fibronectin domains from B. longum GT15 binds to human tumor necrosis factor alpha in vitro. Anaerobe 2020, 65, 102247. [Google Scholar] [CrossRef]
  18. Henderson, B.; Nair, S.; Pallas, J.; Williams, M.A. Fibronectin: A multidomain host adhesin targeted by bacterial fibronectin-binding proteins. FEMS Microbiol. Rev. 2011, 35, 147–200. [Google Scholar] [CrossRef]
  19. Speziale, P.; Arciola, C.R.; Pietrocola, G. Fibronectin and Its Role in Human Infective Diseases. Cells 2019, 8, 1516. [Google Scholar] [CrossRef]
  20. Malagrinò, F.; Pennacchietti, V.; Santorelli, D.; Pagano, L.; Nardella, C.; Diop, A.; Toto, A.; Gianni, S. On the Effects of Disordered Tails, Supertertiary Structure and Quinary Interactions on the Folding and Function of Protein Domains. Biomolecules 2022, 12, 209. [Google Scholar] [CrossRef]
  21. Valk, V.; van der Kaaij, R.M.; Dijkhuizen, L. The evolutionary origin and possible functional roles of FNIII domains in two Microbacterium aurum B8. A granular starch degrading enzymes, and in other carbohydrate acting enzymes. Amylase 2017, 1, 1–11. [Google Scholar] [CrossRef]
  22. Sun, H.; Guo, Z.; Hong, H.; Zhang, Z.; Zhang, Y.; Wang, Y.; Le, S.; Chen, H. Free Energy Landscape of Type III Fibronectin Domain with Identified Intermediate State and Hierarchical Symmetry. Phys. Rev. Lett. 2023, 131, 218402. [Google Scholar] [CrossRef]
  23. Koide, A.; Koide, S. Use of Phage Display and Other Molecular Display Methods for the Development of Monobodies. Cold Spring Harb. Protoc. 2024, 5, 107982. [Google Scholar] [CrossRef]
  24. Zhu, N.; Smallwood, P.M.; Rattner, A.; Chang, T.H.; Williams, J.; Wang, Y.; Nathans, J. Utility of protein-protein binding surfaces composed of anti-parallel alpha-helices and beta-sheets selected by phage display. J. Biol. Chem. 2024, 300, 107283. [Google Scholar] [CrossRef] [PubMed]
  25. Siupka, P.; Hamming, O.T.; Kang, L.; Gad, H.H.; Hartmann, R. A conserved sugar bridge connected to the WSXWS motif has an important role for transport of IL-21R to the plasma membrane. Genes Immun. 2015, 6, 405–413. [Google Scholar] [CrossRef]
  26. Singh, S.; Chaturvedi, N.; Rai, G. De novo modeling and structural characterization of IL9 IL9 receptor complex: A potential drug target for hematopoietic stem cell therapy. Netw. Model. Anal. Health Inform. Bioinform. 2020, 9, 29. [Google Scholar] [CrossRef]
  27. Singh, S.; Rai, G. Structural Insights into the IL12:IL12 Receptor Complex Assembly by Molecular Modeling, Docking, and Molecular Dynamics Simulation. Comb. Chem. High Throughput Screen. 2022, 25, 677–688. [Google Scholar] [CrossRef] [PubMed]
  28. Olsen, J.G.; Kragelund, B.B. Who climbs the tryptophan ladder? On the structure and function of the WSXWS motif in cytokine receptors and thrombospondin repeats. Cytokine Growth Factor Rev. 2014, 25, 337–341. [Google Scholar] [CrossRef] [PubMed]
  29. Szabó, R.; Láng, O.; Láng, J.; Illyés, E.; Kőhidai, L.; Hudecz, F. Effect of SXWS/WSXWS peptides on chemotaxis and adhesion of the macrophage-like cell line J774. J. Mol. Recognit. 2015, 28, 253–260. [Google Scholar] [CrossRef]
  30. Zakharevich, N.V.; Nezametdinova, V.Z.; Averina, O.V.; Chekalina, M.S.; Alekseeva, M.G.; Danilenko, V.N. Complete Genome Sequence of Bifidobacterium angulatum GT102: Potential Genes and Systems of Communication with Host. Russ. J. Genet. 2019, 55, 847–864. [Google Scholar] [CrossRef]
  31. Alekseeva, M.G.; Dyakov, I.N.; Bushkova, K.K.; Mavletova, D.A.; Yunes, R.A.; Chernyshova, I.N.; Masalitin, I.A.; Koshenko, T.A.; Nezametdinova, V.Z.; Danilenko, V.N. Study of the binding of ΔFN3.1 fragments of the Bifidobacterium longum GT15 with TNFα and prevalence of domain-containing proteins in groups of bacteria of the human gut microbiota. Microbiome Res. Rep. 2023, 2, 10. [Google Scholar] [CrossRef]
  32. Veselovsky, V.A.; Dyachkova, M.S.; Menyaylo, E.A.; Polyaeva, P.S.; Olekhnovich, E.I.; Shitikov, E.A.; Bespiatykh, D.A.; Semashko, T.A.; Kasianov, A.S.; Ilina, E.N.; et al. Gene Networks Underlying the Resistance of Bifidobacterium longum to Inflammatory Factors. Front. Immunol. 2020, 11, 595877. [Google Scholar] [CrossRef]
  33. Jumper, J.; Evans, R.; Pritzel, A.; Green, T.; Figurnov, M.; Ronneberger, O.; Tunyasuvunakool, K.; Bates, R.; Žídek, A.; Potapenko, A.; et al. Highly accurate protein structure prediction with AlphaFold. Nature 2021, 596, 583–589. [Google Scholar] [CrossRef]
  34. Baek, M.; DiMaio, F.; Anishchenko, I.; Dauparas, J.; Ovchinnikov, S.; Lee, G.R.; Wang, J.; Cong, Q.; Kinch, L.N.; Schaeffer, R.D.; et al. Accurate prediction of protein structures and interactions using a 3-track neural network. Science 2021, 373, 871–876. [Google Scholar] [CrossRef]
  35. Zheng, W.; Zhang, C.; Li, Y.; Pearce, R.; Bell, E.W.; Zhang, Y. Folding non-homologous proteins by coupling deep-learning contact maps with I-TASSER assembly simulations. Cell Rep. Methods 2021, 1, 100014. [Google Scholar] [CrossRef]
  36. Zhang, C.; Freddolino, L.; Zhang, Y. COFACTOR: Improved protein function prediction by combining structure, sequence and protein–protein interaction information. Nucleic Acids Res. 2017, 45, 291–299. [Google Scholar] [CrossRef]
  37. Yang, J.; Zhang, Y. I-TASSER server: New development for protein structure and function predictions. Nucleic Acids Res. 2015, 43, 174–181. [Google Scholar] [CrossRef] [PubMed]
  38. McGuffin, L.J.; Edmunds, N.S.; Genc, A.G.; Alharbi, S.M.A.; Salehe, B.R.; Adiyaman, R. Prediction of protein structures, functions and interactions using the IntFOLD7, MultiFOLD and ModFOLDdock servers. Nucleic Acids Res. 2023, 51, 274–280. [Google Scholar] [CrossRef] [PubMed]
  39. Mustaev, E.; Khamitov, E.M. Predicting the PARP1 Tertiary Structure by Molecular Modeling Methods. J. Struct. Chem. 2025, 66, 898–910. [Google Scholar] [CrossRef]
  40. Berman, H.M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T.N.; Weissig, H.; Shindyalov, I.N.; Bourne, P.E. The Protein Data Bank. Nucleic Acids Res. 2000, 28, 235–242. [Google Scholar] [CrossRef]
  41. Liang, S.; Dai, J.; Hou, S.; Su, L.; Zhang, D.; Guo, H.; Hu, S.; Wang, H.; Rao, Z.; Guo, Y.; et al. Structural Basis for Treating Tumor Necrosis Factor α (TNFα)-associated Diseases with the Therapeutic Antibody Infliximab. J. Biol. Chem. 2013, 288, 13799–13807. [Google Scholar] [CrossRef]
  42. Carrington, B.; Myers, W.K.; Horanyi, P.; Calmianol, M.; Lawson, A.D.G. Natural Conformational Sampling of Human TNFα Visualized by Double Electron-Electron Resonance. Biophys. J. 2017, 113, 371–380. [Google Scholar] [CrossRef]
  43. Somers, W.; Stahl, M.; Seehra, J.S. Å crystal structure of interleukin 6: Implications for a novel mode of receptor dimerization and signaling. EMBO J. 1997, 16, 989–997. [Google Scholar] [CrossRef]
  44. Yoon, S.I.; Logsdon, N.J.; Sheikh, F.; Donnelly, R.P.; Walter, M.R. Conformational Changes Mediate Interleukin-10 Receptor 2 (IL-10R2) Binding to IL-10 and Assembly of the Signaling Complex. Protein Struct. Fold. 2006, 281, 35088–35096. [Google Scholar] [CrossRef] [PubMed]
  45. Daëron, M. The immune system as a system of relations. Front. Immunol. 2022, 13, 984678. [Google Scholar] [CrossRef]
  46. Wang, R.; Lan, C.; Benlagha, K.; Olsen, N.; Camara, S.; Miller, H.; Kubo, M.; Heegaard, S.; Lee, P.; Yang, L.; et al. The interaction of innate immune and adaptive immune system. MedComm 2024, 5, e714. [Google Scholar] [CrossRef] [PubMed]
  47. Quiroz, R.N.; Camacho, J.V.; Peñata, E.Z.; Lemus, Y.B.; López-Fernández, C.; Escorcia, L.G.; Fernández-Ponce, C.; Cobos, M.R.; Moreno, J.F.; Fiorillo-Moreno, O. Multiscale information processing in the immune system. Front. Immunol. 2025, 16, 1563992. [Google Scholar] [CrossRef] [PubMed]
  48. Liu, C.; Chu, D.; Kalantar-Zadeh, K.; George, J.; Young, H.A.; Liu, G. Cytokines: From clinical significance to quantification. Adv. Sci. 2021, 15, 2004433. [Google Scholar] [CrossRef]
  49. Mendes, V.; Galvão, I.; Vieira, A.T. Mechanisms by Which the Gut Microbiota Influences Cytokine Production and Modulates Host Inflammatory Responses. J. Interferon Cytokine Res. 2019, 39, 393–409. [Google Scholar] [CrossRef]
  50. Sun, L.; Wang, X.; Saredy, J.; Yuan, Z.; Yang, X.; Wang, H. Innate-adaptive immunity interplay and redox regulation in immune response. Redox Biol. 2020, 37, 101759. [Google Scholar] [CrossRef]
  51. Shukla, A.; Tangney, M. Bacterial exopolysaccharides and live biotherapeutics: Orchestrating immune modulation and therapeutic potential in the gut microbiome era. Biomed. Pharmacother. 2025, 191, 118510. [Google Scholar] [CrossRef]
  52. Campbell, C.; Kandalgaonkar, M.R.; Golonka, R.M.; Yeoh, B.S.; Vijay-Kumar, M.; Saha, P. Crosstalk between Gut Microbiota and Host Immunity: Impact on Inflammation and Immunotherapy. Biomedicines 2023, 11, 294. [Google Scholar] [CrossRef] [PubMed]
  53. Spencer, S.P.; Fragiadakis, G.K.; Sonnenburg, J.L. Pursuing human-relevant gut microbiota-immune interactions. Immunity 2019, 51, 225–239. [Google Scholar] [CrossRef]
  54. Gao, B.; Gupta, R.S. Phylogenetic Framework and Molecular Signatures for the Main Clades of the Phylum Actinobacteria. Microbiol. Mol. Biol. Rev. 2012, 76, 66–112. [Google Scholar] [CrossRef]
  55. Barka, E.A.; Vatsa, P.; Sanchez, L.; Gaveau-Vaillant, N.; Jacquard, C.; Meier-Kolthoff, J.P.; Klenk, H.P.; Clément, C.; Ouhdouch, Y.; van Wezel, G.P. Taxonomy, physiology, and natural products of Actinobacteria. Microbiol. Mol. Biol. Rev. 2015, 80, 1–43. [Google Scholar] [CrossRef]
  56. Duranti, S.; Longhi, G.; Ventura, M.; van Sinderen, D.; Turroni, F. Exploring the ecology of Bifidobacteria and their genetic adaptation to the mammalian gut. Microorganisms 2020, 9, 8. [Google Scholar] [CrossRef]
  57. Olajide, T.S.; Ijomone, O.M. Targeting gut microbiota as a therapeutic approach for neurodegenerative diseases. Neuroprotection 2025, 3, 120–130. [Google Scholar] [CrossRef] [PubMed]
  58. Mopuru, R.; Chaturvedi, S.; Burkholder, B.M. Relapsing Thrombotic Thrombocytopenic Purpura (TTP) in a Patient Treated with Infliximab for Chronic Uveitis. Ocul. Immunol. Inflamm. 2022, 30, 241–243. [Google Scholar] [CrossRef]
  59. El-Tahan, R.R.; Ghoneim, A.M.; El-Mashad, N. TNF-α gene polymorphisms and expression. Springerplus 2016, 5, 1508. [Google Scholar] [CrossRef] [PubMed]
  60. Asiri, A.; Hazeldine, J.; Moiemen, N.; Harrison, P. IL-8 Induces Neutrophil Extracellular Trap Formation in Severe Thermal Injury. Int. J. Mol. Sci. 2024, 25, 7216. [Google Scholar] [CrossRef]
  61. Copaescu, A.; Smibert, O.; Gibson, A.; Phillips, E.J.; Trubiano, J.A. The role of IL-6 and other mediators in the cytokine storm associated with SARS-CoV-2 infection. J. Allergy Clin. Immunol. 2020, 146, 518–534. [Google Scholar] [CrossRef]
  62. Inoue, H.; Nojima, H.; Okayama, H. High efficiency transformation of Escherichia coli with plasmids. Gene 1990, 96, 23–28. [Google Scholar] [CrossRef]
  63. Mierendorf, R.; Yeager, K.; Novy, R. Innovations. Newsl. Novagen Inc. 1994, 1, 1–3. [Google Scholar]
  64. Maniatis, T.; Fritsch, E.F.; Sambrook, J. Molecular Cloning: A Laboratory Manual; Cold Spring Harbor: New York, USA, 1984. [Google Scholar]
  65. Mariani, V.; Biasini, M.; Barbato, A.; Schwede, T. lDDT: A local superposition-free score for comparing protein structures and models using distance difference tests. Bioinformatics 2013, 29, 2722–2728. [Google Scholar] [CrossRef]
  66. Zheng, W.; Wuyun, Q.; Li, Y.; Liu, Q.; Zhou, X.; Peng, C.; Zhu, Y.; Freddolino, L.; Zhang, Y. Deep-learning-based single-domain and multidomain protein structure prediction with D-I-TASSER. Nat. Biotechnol. 2025, 23, 1–26. [Google Scholar] [CrossRef]
  67. Zhou, X.; Zheng, W.; Li, Y.; Pearce, R.; Zhang, C.; Bell, E.W.; Zhang, G.; Zhang, Y. I-TASSER-MTD: A deep-learning-based platform for multi-domain protein structure and function prediction. Nat. Protoc. 2022, 17, 2326–2353. [Google Scholar] [CrossRef]
  68. McGuffin, L.J.; Shuid, A.N.; Kempster, R.; Maghrabi, A.H.A.; Nealon, J.O.; Salehe, B.R.; Atkins, J.D.; Roche, D.B. Accurate template-based modeling in CASP12 using the IntFOLD4-TS, ModFOLD6, and ReFOLD methods. Proteins 2018, 86, 335–344. [Google Scholar] [CrossRef] [PubMed]
  69. Kozakov, D.; Brenke, R.; Comeau, S.R.; Vajda, S. PIPER: An FFT-Based Protein Docking Program with Pairwise Potentials. PROTEINS: Struct. Funct. Bioinform. 2006, 65, 392–406. [Google Scholar] [CrossRef] [PubMed]
  70. Kozakov, D.; Hall, D.R.; Xia, B.; Porter, K.A.; Padhorny, D.; Yueh, C.; Beglov, D.; Vajda, S. The ClusPro web server for protein-protein docking. Nat. Protoc. 2017, 12, 255–278. [Google Scholar] [CrossRef] [PubMed]
  71. Lu, C.; Wu, C.; Ghoreishi, D.; Chen, W.; Wang, L.; Damm, W.; Ross, G.A.; Dahlgren, M.K.; Russell, E.; Von Bargen, C.D.; et al. OPLS4: Improving Force Field Accuracy on Challenging Regimes of Chemical Space. J. Chem. Theory Comput. 2021, 17, 4291–4300. [Google Scholar] [CrossRef]
  72. Bowers, K.J.; Chow, E.; Xu, H.; Dror, R.O.; Eastwood, M.P.; Gregersen, B.A.; Klepeis, J.L.; Kolossvary, I.; Moraes, M.A.; Sacerdoti, F.D.; et al. Scalable Algorithms for Molecular Dynamics Simulations on Commodity Clusters. In Proceedings of the ACM/IEEE SC2006 Conference on High Performance Networking and Computing, Tampa, FL, USA, 11–17 November 2006. [Google Scholar]
  73. Frey, B.J.; Dueck, D. Clustering by Passing Messages Between Data Points. Science 2007, 315, 972–976. [Google Scholar] [CrossRef] [PubMed]
  74. Genheden, S.; Ryde, U. The MM/PBSA and MM/GBSA methods to estimate ligand-binding affinities. Expert Opin. Drug Discov. 2015, 10, 449–461. [Google Scholar] [CrossRef] [PubMed]
  75. Haimes, J.; Kelley, M. Demonstration of a ΔΔCq Calculation Method to Compute Relative Gene Expression from qPCR Data; A Horizon Discovery Group Company: Lafayette, CO, USA, 2015; pp. 1–4. [Google Scholar]
  76. Ye, J.; Coulouris, G.; Zaretskaya, I.; Cutcutache, I.; Rozen, S.; Madden, T.L. Primer-BLAST: A tool to design target-specific primers for polymerase chain reaction. BMC Bioinform. 2012, 13, 134. Available online: https://www.biomedcentral.com/1471-2105/13/134 (accessed on 24 April 2025). [CrossRef] [PubMed]
Figure 1. Alignment of amino acid sequences of ΔFN3.1 B. longum GT15, ΔFN3.2 B. angulatum GT102, and ΔFN3.3 B. bifidum 791. The conservative (core) residues are marked in gray, the annotated motifs of cytokine receptors are underlined. The FN3 protein domains are highlighted in the box. (*) - indicates matches of all three amino acids; (:) - two amino acids match, the third is similar; (.) - two amino acids match, the third is not similar.
Figure 1. Alignment of amino acid sequences of ΔFN3.1 B. longum GT15, ΔFN3.2 B. angulatum GT102, and ΔFN3.3 B. bifidum 791. The conservative (core) residues are marked in gray, the annotated motifs of cytokine receptors are underlined. The FN3 protein domains are highlighted in the box. (*) - indicates matches of all three amino acids; (:) - two amino acids match, the third is similar; (.) - two amino acids match, the third is not similar.
Ijms 26 10560 g001
Figure 2. Tertiary architecture of ∆FN3.1 и ∆FN3.3 domains. (A): a V-shaped module of two linear FN3 domains carrying a potential ligand binding site (arrow; proposed in [31]; (B,C): tertiary structures of ΔFN3.1-AF2 и FN3.3-IF5 predicted by AF2 (AplhaFold2) and IF5 (IntFOLD5), respectively, and visualized as the chains of antiparallel β-sheets with numbered epitopes I–V. The colored architecture of proteins shows the numbered amino acid residues.
Figure 2. Tertiary architecture of ∆FN3.1 и ∆FN3.3 domains. (A): a V-shaped module of two linear FN3 domains carrying a potential ligand binding site (arrow; proposed in [31]; (B,C): tertiary structures of ΔFN3.1-AF2 и FN3.3-IF5 predicted by AF2 (AplhaFold2) and IF5 (IntFOLD5), respectively, and visualized as the chains of antiparallel β-sheets with numbered epitopes I–V. The colored architecture of proteins shows the numbered amino acid residues.
Ijms 26 10560 g002
Figure 3. Results of MD simulations. Shown are the positions of TNF-α relative to ∆FN3.1 (A,B) and ∆FN3.3 (C,D) epitopes. Hydrogen bonds are rendered as dashed lines, π–π stacking interactions are depicted as blue lines. Colours indicated epitopes of ∆FN3 (I–V). The designations correspond to Figure 2.
Figure 3. Results of MD simulations. Shown are the positions of TNF-α relative to ∆FN3.1 (A,B) and ∆FN3.3 (C,D) epitopes. Hydrogen bonds are rendered as dashed lines, π–π stacking interactions are depicted as blue lines. Colours indicated epitopes of ∆FN3 (I–V). The designations correspond to Figure 2.
Ijms 26 10560 g003
Figure 4. Molecular models of ΔFN3-interleukin complexes. (A) molecular model illustrating interaction ∆FN3.1 with IL-8; (B) molecular model illustrating interaction ∆FN3.3 with IL-8; (C) molecular model illustrating interaction ∆FN3.1 with IL-10. Colour indicate epitopes of ∆FN3 (I–V). IL-8 is indicated in light green. IL-10 is indicated in mustard colour.
Figure 4. Molecular models of ΔFN3-interleukin complexes. (A) molecular model illustrating interaction ∆FN3.1 with IL-8; (B) molecular model illustrating interaction ∆FN3.3 with IL-8; (C) molecular model illustrating interaction ∆FN3.1 with IL-10. Colour indicate epitopes of ∆FN3 (I–V). IL-8 is indicated in light green. IL-10 is indicated in mustard colour.
Ijms 26 10560 g004
Figure 5. Synergistic combinations of ΔFN3 proteins and TNF-α in regard to TNF-α mRNA abundance. THP-1 cells were treated with indicated concentrations of ΔFN3.1, ΔFN3.3, or 300 ng TNF-α (alone or in combinations) for 3 h, followed by total RNA isolation and RT-PCR. Gene-specific signals were normalized on HPRT1 cDNA. Each sample was analyzed in triplicate. Shown are mean + S.E.M. of three biological replicates. The mRNA levels in untreated cells were taken as 1. Asterisks p < 0.01 (**), or p < 0.001 (***) between the groups ‘combination’ vs. each protein alone. An independent Student’s t-test was used for statistical analysis.
Figure 5. Synergistic combinations of ΔFN3 proteins and TNF-α in regard to TNF-α mRNA abundance. THP-1 cells were treated with indicated concentrations of ΔFN3.1, ΔFN3.3, or 300 ng TNF-α (alone or in combinations) for 3 h, followed by total RNA isolation and RT-PCR. Gene-specific signals were normalized on HPRT1 cDNA. Each sample was analyzed in triplicate. Shown are mean + S.E.M. of three biological replicates. The mRNA levels in untreated cells were taken as 1. Asterisks p < 0.01 (**), or p < 0.001 (***) between the groups ‘combination’ vs. each protein alone. An independent Student’s t-test was used for statistical analysis.
Ijms 26 10560 g005
Table 1. Parameters of binding of ΔFN3.1 B. longum GT15 and ΔFN3.3 B. bifidum 791 to TNF-α.
Table 1. Parameters of binding of ΔFN3.1 B. longum GT15 and ΔFN3.3 B. bifidum 791 to TNF-α.
TNFαKa (1/Ms) *Kd (1/s)KD (nM)
ΔFN3.13.43 × 10−5451 × 10−513.1 ± 0.6
ΔFN3.313.1 × 10−576.1 × 10−558.2 ± 2.9
AlbuminBelow the level of detection
* Binding parameters were calculated based on sensorograms using BIAEvaluation program Version 4.1. and Langmuir model (1:1). KD = Kd/Ka.
Table 2. Binding of ΔFN3.1 B. longum GT15 to IL-6, IL-8, and IL-10 at neutral and alkaline pH.
Table 2. Binding of ΔFN3.1 B. longum GT15 to IL-6, IL-8, and IL-10 at neutral and alkaline pH.
ΔFN3.1Ka (1/Ms)Ka (1/Ms)Ka (1/Ms)
pH 7.4
IL-6Below the level of detection
IL-8330 × 10−51.6 × 10−54.9 ± 0.2
IL-1022.5 × 10−5140 × 10−562.2 ± 3.1
pH 8.0
IL-6Below the level of detection
IL-8397 × 10−51.6 × 10−54.0 ± 0.2
IL-10Below the level of detection
Table 3. Binding of ΔFN3.3 B. bifidum 791 to IL-6, IL-8, and IL-10 at neutral and alkaline pH.
Table 3. Binding of ΔFN3.3 B. bifidum 791 to IL-6, IL-8, and IL-10 at neutral and alkaline pH.
ΔFN3.1Ka (1/Ms)Ka (1/Ms)Ka (1/Ms)
pH 7.4
IL-6Below the level of detection
IL-8878 × 10−52.0 × 10−52.3 ± 0.1
IL-10Below the level of detection
pH 8.0
IL-6Below the level of detection
IL-8879 × 10−51.1 × 10−51.2 ± 0.04
IL-10Below the level of detection
Table 4. Putative amino acid residues in FN3 domains.
Table 4. Putative amino acid residues in FN3 domains.
ProteinAmino Acid ResiduesLocation
ΔFN3.1Trp78, Ser79, Pro81, Ser82Cytokine receptor motif (FN3- domain I)
Trp174, Ser175, Glu177, Ser178Cytokine receptor motif (FN3- domain II)
Ala43, Ala51, Thr111, Pro417, Ala424[31]
ΔFN3.3Trp77, Ser78, Pro80, Ser81Cytokine receptor motif (FN3- domain I)
Glu172, Gly173, Pro175, Ser176Cytokine receptor motif (FN3- domain II)
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

Alekseeva, M.G.; Borisevich, S.S.; Yusupova, A.R.; Reznikova, D.A.; Mavletova, D.A.; Nesterov, A.A.; Ilyina, M.G.; Akimova, N.I.; Shtil, A.A.; Danilenko, V.N. Differential Binding of ΔFN3 Proteins of Bifidobacterium longum GT15 and Bifidobacterium bifidum 791 to Cytokines Determined by Surface Plasmon Resonance and De Novo Molecular Modeling. Int. J. Mol. Sci. 2025, 26, 10560. https://doi.org/10.3390/ijms262110560

AMA Style

Alekseeva MG, Borisevich SS, Yusupova AR, Reznikova DA, Mavletova DA, Nesterov AA, Ilyina MG, Akimova NI, Shtil AA, Danilenko VN. Differential Binding of ΔFN3 Proteins of Bifidobacterium longum GT15 and Bifidobacterium bifidum 791 to Cytokines Determined by Surface Plasmon Resonance and De Novo Molecular Modeling. International Journal of Molecular Sciences. 2025; 26(21):10560. https://doi.org/10.3390/ijms262110560

Chicago/Turabian Style

Alekseeva, Maria G., Sophia S. Borisevich, Alfia R. Yusupova, Diana A. Reznikova, Dilara A. Mavletova, Andrey A. Nesterov, Margarita G. Ilyina, Natalia I. Akimova, Alexander A. Shtil, and Valery N. Danilenko. 2025. "Differential Binding of ΔFN3 Proteins of Bifidobacterium longum GT15 and Bifidobacterium bifidum 791 to Cytokines Determined by Surface Plasmon Resonance and De Novo Molecular Modeling" International Journal of Molecular Sciences 26, no. 21: 10560. https://doi.org/10.3390/ijms262110560

APA Style

Alekseeva, M. G., Borisevich, S. S., Yusupova, A. R., Reznikova, D. A., Mavletova, D. A., Nesterov, A. A., Ilyina, M. G., Akimova, N. I., Shtil, A. A., & Danilenko, V. N. (2025). Differential Binding of ΔFN3 Proteins of Bifidobacterium longum GT15 and Bifidobacterium bifidum 791 to Cytokines Determined by Surface Plasmon Resonance and De Novo Molecular Modeling. International Journal of Molecular Sciences, 26(21), 10560. https://doi.org/10.3390/ijms262110560

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