Next Article in Journal
Anti-Amnesia-like Effect of Pinus densiflora Extract by Improving Apoptosis and Neuroinflammation on Trimethyltin-Induced ICR Mice
Previous Article in Journal
Objective Physical Function in the Alzheimer’s Disease Continuum: Association with Cerebrospinal Fluid Biomarkers in the ALBION Study
Previous Article in Special Issue
Molecular Relay Stations in Membrane Nanotubes: IRSp53 Involved in Actin-Based Force Generation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Heterologous Interactions with Galectins and Chemokines and Their Functional Consequences

Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota Health Sciences Center, 6-155 Jackson Hall, Minneapolis, MN 55455, USA
Int. J. Mol. Sci. 2023, 24(18), 14083; https://doi.org/10.3390/ijms241814083
Submission received: 12 August 2023 / Revised: 11 September 2023 / Accepted: 12 September 2023 / Published: 14 September 2023
(This article belongs to the Special Issue Protein-Protein Interactions in Cellular Function)

Abstract

:
Extra- and intra-cellular activity occurs under the direction of numerous inter-molecular interactions, and in any tissue or cell, molecules are densely packed, thus promoting those molecular interactions. Galectins and chemokines, the focus of this review, are small, protein effector molecules that mediate various cellular functions—in particular, cell adhesion and migration—as well as cell signaling/activation. In the past, researchers have reported that combinations of these (and other) effector molecules act separately, yet sometimes in concert, but nevertheless physically apart and via their individual cell receptors. This view that each effector molecule functions independently of the other limits our thinking about functional versatility and cooperation, and, in turn, ignores the prospect of physiologically important inter-molecular interactions, especially when both molecules are present or co-expressed in the same cellular environment. This review is focused on such protein-protein interactions with chemokines and galectins, the homo- and hetero-oligomeric structures that they can form, and the functional consequences of those paired interactions.

1. Introduction

Galectins and chemokines have been known, essentially since their discovery, to form homodimers, as well as some higher order homo-oligomers. However, at the turn of the millennium, two CC chemokines (macrophage inflammatory proteins 1α and 1β) were reported to be secreted simultaneously from cells as heterodimers [1]. At that time, those authors thought it was a unique phenomenon with chemokines, and the observation was reported without experimental validation of its functional significance [1]. However, shortly thereafter it was discovered that multiple chemokines indeed cross-react inter-molecularly as CXC-CXC and CC-CC chemokines, as well as CC-CXC paired chemokines, and with functional significance [2,3,4]. This was quickly followed by further experimental confirmation of the functional significance of paired chemokines [5,6]. In these instances, NMR and molecular modeling was used to demonstrate their physical interactions as heterodimers that modulate their respective functions. The next observation was that galectins too could cross-interact with each other as galectin-galectin heterodimers [7,8]. Finally, because some chemokines and galectins were found to be present in the same cellular micro-environment, this concept was extended to show that members from each family could interact as galectin-chemokine heterodimers with functional consequences [9]. Since these initial reports, a number of other research labs have picked up on this concept and have started to investigate such inter-molecular interactions in other systems.
Due to growing interest in this area, this review is focused on chemokines and galectins, their structures and interactions, and consequent functional effects. We first present some general background on chemokines and galectins, followed by experimental validation of their structural interactions as homo- and hetero-dimers (both within each family and then inter-family as chemokine–galectin heterodimers), and finally the functional consequences thereof.

2. Chemokines

Chemokines are a family of small, highly conserved proteins (~8 kDa to ~12 kDa) involved in many biological processes, including chemotaxis [10], leukocyte degranulation [11], hematopoiesis [12], and angiogenesis [13,14]. Chemokines function by interacting with cell surface G-protein coupled receptors (GPCRs), and are usually categorized into sub-families that are based on the sequential positioning of the first two of four highly conserved cysteine residues: CXC, CC, and CX3C [15]. The C chemokine sub-family is the exception with only one N-terminal cysteine residue. In the largest subfamilies, CC and CXC, the first two cysteines are adjacent (CC motif) or separated by one amino acid residue (CXC motif). C-type chemokines lack the first and third of these cystines, and CX3C chemokines have three amino acids between the first two cysteine residues. Even though sequence identity between chemokines varies from about 20% to 90%, their sequences overall are highly conserved, and all chemokines adopt the same monomer fold that consists of a flexible N-terminus and loop, followed by a three-stranded antiparallel β-sheet on to which is folded a C-terminal α-helix, e.g., [16]. The highly conserved cysteine residues (four in CXC and CC chemokines) pair up to form disulfide bridges that are crucial to maintaining structural integrity and binding to their respective GPCRs [17].
Chemokine monomers usually associate to form oligomers, primarily dimers, but some are also known to form tetramers [18,19] and higher order species, e.g., [20,21]. Despite their highly conserved monomer structures, chemokines form different types of homodimer structures, depending upon which sub-family they are associated [16]. In general, homodimer structures are determined primarily by the composition of amino acid residues that are present at the particular inter-subunit interface [4]. Within each chemokine sub-family (i.e., CC or CXC), homodimer structures are essentially the same, albeit quite different from those in another family. Figure 1A,B shows homodimer structures of CXC chemokine CXCL8 (Interleukin-8) [22] and CC chemokine CCL5 (RANTES) [23]. The more globular CXC-type dimer (Figure 1A) is formed by interactions between β1 strands from each monomer subunit that extend the three stranded anti-parallel β-sheet from each monomer into a six-stranded β-sheet, on top of which are folded the two C-terminal α-helices that run in an antiparallel fashion. On the other hand, CC-type chemokines (Figure 1B) form elongated end-to end-type dimers through contacts between their short N-terminal β-strands (labeled βN) with the two C-terminal helices running almost perpendicular to each other on opposite sides of the molecule. Nevertheless, some CC-type dimer structures such as CCL5 have been reported to differ in the relative orientation of some secondary structure elements (e.g., C-terminal α-helices), although this may be related to differences in structural dynamics and/or crystal lattice effects [21,24]. Chemokines can also oligomerize to tetramers with CXCL4 (platelet factor-4) [25], where two CXC-type dimers associate to form a β-sandwich with the β-sheet of one dimer lying on top of the β-sheet of the other. Tetramers have also been observed with CXCL7 [18], and their higher order structures can vary considerably, e.g., CXCL10 [26] that forms both CC- and CXC-type homodimer topologies. Some CC-type chemokines even associate as higher order oligomers, e.g., CCL5 [20] and CCL27 [21].
Chemokine monomers, homodimers and higher order homo-oligomers exist in a complex equilibrium where distinct oligomer structures co-exist and interconvert within a dynamic distribution [20,21,27,28,29,30,31]. This is exemplified by NMR studies on CXCL4 [32,33], low affinity CXCL4 [34], and CXCL7 (platelet basic protein) [18] and its N-terminal degradation products CTAP-III (connective tissue-activating peptide III) and NAP-2 (neutrophil-activating peptide-2) [35]. The weighting factor with oligomer populations is dictated by thermodynamic stability and the amino acid composition/conformation of their inter-subunit interfaces [4]. Some chemokines form strong oligomers and others form much weaker ones or remain as monomers/dimers. This equilibrium can be perturbed by changing solution conditions (e.g., lower pH, buffer type, ionic strength, presence of ligands such as heparin), e.g., with CXCL4 [32,33,36], CXCL7 [37], CXCL12 [38,39], and CCL11 (exotoxin) [40]). The oligomer subunit exchange rate provides a reasonable explanation as to why some chemokines cannot be crystallized or why their structures cannot be solved by NMR.
Chemokines are also known to bind specifically and strongly to GAGs (glycosaminoglycans) [30,41,42,43]. CCL5 homodimers bind GAGs [44,45,46] with binding affinities depending on the type of GAG and its sulfation pattern [46]. Similar observations have been made for CCL2 [47] and CCL11 [41]. Overall, the highly negatively charged GAGs interact electrostatically with positively charged residues in these chemokines. Contrary to some CXCL4/GAG binding models that focus on clusters of lysines in the chemokine C-terminal α-helix, Mayo et al. [48] used NMR and site-directed mutagenesis to demonstrate that the loop containing Arg20, Arg22, His23 and Thr25, as well as Lys46 and Arg49, also plays a significant role in GAG/heparin binding. GAG binding can even induce chemokine oligomerization [49], as exemplified with CCL5 [30,50,51]. GAGs can also affect chemokine structure [30,47], structural dynamics [44], and chemokine receptor dimerization, e.g., CCR2 [52]. For example, Rek et al. [30] found that there is a structural transition in CCL5 upon GAG binding, and Verkaar et al. [53] discovered that GAGs affect chemokine cooperativity. Furthermore, Mikhailov et al. [54] showed early on that heparin dodecasaccharide binding to CXCL4 induces higher order oligomer formation that is dependent upon the chemokine-to-GAG molar ratio.

2.1. Chemokine Heterodimers

Because chemokine monomer structures are highly conserved (see Figure 1A), heterodimer structures, similarly to homodimer structures, are determined primarily by amino acid residues at the particular inter-subunit interface of the interacting chemokine monomers [4]. Therefore, monomers of different chemokines may be swapped if the arrangement and composition of residues at a given monomer-monomer interface in the heterodimer make for a more energetically favorable state relative to that in either homodimer. Modeled structures of CXCL4 and CXCL8 homodimers, as well as of the CXCL4/CXCL8 heterodimer, are shown in Figure 2A–C. These structures were guided by NMR HSQC chemical shift and intensity changes from 15N-labeled CXCL4 and CCL5, followed by manual docking and energy minimization using MD simulations. Positively charged lysine and arginine residues (shown in dark blue) electrostatically interact with negatively charged glutamate residues (shown in dark red) to thermodynamically stabilize both homo- and hetero-dimers.
Guan et al. [1] showed that CC chemokines CCL3/4 and CCL2/8 (macrophage inflammatory protein 1β (MIP-1β) and 1α (MIP-1α), respectively) are secreted together from activated human monocytes and peripheral blood lymphocytes, thereby forming heterodimers in vitro, yet only speculating (but not experimentally proving) that this heterodimer pair effects intracellular signaling by binding to, and activating, its CCR5 cell receptor [1]. However, we now know that even chemokines that are not simultaneously secreted from cells do form heterodimers. For example, at least three members of the CXC sub-family: CXCL4 [3,32], its N-terminal chimera PF4-M2 [25], and CXCL8 [3], were shown early on to exchange subunits and form heterodimers with similar equilibrium dissociation constants (KD) as observed for homodimers [2,3]. Moreover, CXCL4/CXCL8 heterodimers were shown to be functionally significant in vitro in cell-based assays [3].
Nesmelova et al. [4] investigated the energetic basis for CC and CXC heterodimer formation by using molecular mechanics and the Poisson-Boltzmann surface area (MM-PBSA) approach to calculate binding free energies and to predict which pairs of CXC and CC chemokines would likely form in solution. This study indicated that heterodimers within and between members of CXC and CC sub-families can occur. Calculations were performed to assess also which type of heterodimer might form, i.e., CXC-type vs. CC-type heterodimers. In this regard, CXCL4 can make thermodynamically favorable interactions with CXCL1, CXCL7, and CXCL8, as well as CXCL1/L8, CXCL7/L8 and CXCL1/L7, with all hetero-pairs forming only CXC-type dimers. CC chemokine CCL2 also favorably paired with CCL5 and CCL8, with CXC-type heterodimers being favored with the CCL2/CCL8 pair. Several CXC/CC mixed chemokine pairs were also examined, with CCL2/CXCL4 and CCL2/CXCL8 favoring CXC-type heterodimers, and CCL5/CXCL4 greatly favoring CC-type heterodimers, and CXCL8/CCL5 forming both equally well.
Von Hundelshausen et al. [6] used this in silico approach [4] with CXCL4 and CCL5, in which CXCL4 and CCL5 monomers were initially docked as both CXC- and CC-type heterodimers, with the CC-type heterodimer being more energetically favored. Moreover, the formation of the CXCL4/CCL5 heterodimer was validated experimentally both in vitro and in vivo [5,6]. More recently, Brandhofer et al. [55] demonstrated that heterocomplexes also form between atypical chemokine MIF (macrophage migration inhibitory factor) and CXCL4L1 and regulate inflammation and thrombus formation in patients with peripheral artery disease.
Chemokine heterodimers, similarly to their homodimer counterparts, are also stabilized by binding to GAGs [56], as exemplified with CCR2 ligands CCL2 (MCP-1), CCL8 (MCP-2), CCL7 (MCP-3), CCL13 (MCP-4), and CCL11 (eotaxin) [52]. In particular, CCL2 and CCL8 form strong CC-type heterodimers, whereas CCL2/CCL13, CCL2/CCL11 and CCL8/CCL13 heterodimers are only moderately stable, and CCL7 does not form heterodimers with any other CCR2 chemokine ligand. In addition, the heparin pentasaccharide Arixtra promotes formation of CCL8/CCL11 and CCL2/CCL11 heterodimers, which otherwise do not form or form only weakly.

2.2. Functional Considerations

Chemokines play a significant role in biology and are involved in many pathologic disorders, including cancer, HIV/AIDS, and atherosclerosis [57,58,59]. About fifty chemokines are involved in various aspects of cell interactions and communication within the immune system. In general, chemokines trigger their functional activities by binding to cell surface G-protein coupled receptors (GPCRs) [60,61,62,63]. Determining high resolution structures of chemokine ligand/receptor complexes is highly challenging, and few structures are presently known. However, Handel et al., for example, have reported on a complex formed between chemokine MCP1 and its receptor CCR2 [64], and more recently, on an X-ray crystal structure of chemokine vMIPII and its receptor CXCR4 showing that it is the vMIPII monomer, and not the homodimer, that binds to CXCR4 [65]. In studies such as these, structural biology plays a major role in delineating how chemokine ligands interact with their GPCR receptors, which in turn relates to how chemokines trigger cell signaling. Because GPCRs are difficult to purify and work with, studies have generally focused on identifying residues in the chemokine ligands responsible for activating GPCRs. For example, the Glu-Leu-Arg tripeptide at the N-terminus of some CXC chemokines is crucial for interactions with GPCRs. CXC chemokines with the ELR motif (ELR1: CXCL1, 2, 3, 5, 6, 7, 8) promote angiogenesis and those lacking the ELR motif (ELR2: CXCL4, 9, 10) are reported to be angiostatic [14,66]. Residues within the N-loop between the two N-terminal cysteines, as well as in the helix, can also be involved in GPCR binding [19,67]. Binding interactions vary from chemokine to chemokine and the specific GPCR.
Chemokine heterodimers also mediate some cellular responses [1,2,3,4,5,6,28,68,69,70]. For example, CXCL8/CXCL4 heterodimers enhance CXCL4-induced endothelial cell proliferation and CXCL8-induced migration of Baf3 cells [3], whereas the co-presence of CXCL4 and CXCL8 attenuates a CXCL8-mediated rise in intracellular calcium in the amyeloid progenitor cell line and enhances CXCL8-induced migration of pro-B-cells (Baf/3) [2,3]. Heterodimerization between members of the CXC and CC sub-families has been reported with CXCL4 and CCL5 [5,6,70], as well as with CCL21 (secondary lymphoid tissue chemokine) and CXCL13 (B cell attracting chemokine-1) [28]. The functional result is that hetero-dimerization dramatically modulates chemokine-mediated activities in vitro and in vivo. Chemokine heterodimerization can affect the overall signaling response of GPCRs, thereby providing a general mechanism for regulating chemokine function. The recent synthesis and in vitro/in vivo testing of a covalently-linked CXCL4/CCL5 heterodimer has validated the functional relevance of chemokine heterodimers in GPCR-mediated signal transduction [6,71]. Another covalently-linked heterodimer (termed OHD 4–12) was produced by Nguyen et al. [72] between CXCL4 and CXCL12. OHD 4–12 was shown to inhibit CXCL12-driven migration of triple-negative MDA-MB-231 breast cancer cells, and to bind to and activate CXCR4 receptors [72].
As mentioned above, GAGs are essential to chemokine function in vivo [51,73,74]. In fact, GAGs also affect chemokine heterodimer function, as exemplified with chondroitin sulfate that appears to induce CCL5/CXCL4 heterodimers and promote atherosclerosis [5,70]. The possibility of functionally relevant, structurally distinct oligomer conformations as a result of GAG/chemokine interactions has been postulated [44,75]. Mikhailov et al. [54] showed that a heparin-based dodecasaccharide binding to CXCL4 induces the formation of higher order oligomers which depends on the molar ratio of chemokine:GAG, and thereby can lead to the pathological disorder of thrombocytopenia. Even though there is evidence that chemokine heterodimers impact biological activity, this does not exclude the occurrence of individual chemokines working in concert on their respective GPCRs to elicit synergistic effects. In fact, Gouwy et al. [76,77,78] demonstrated that blocking one of two chemokine receptors negates synergistic interactions, suggesting that synergy requires each chemokine to bind to its respective cell receptor leading to intra-cellular signaling. It is likely that there are several ways to obtain synergy with these chemo-attractants [79].
Whereas initial reports of chemokine heterodimers [1,2,3,4] were viewed as somewhat controversial in terms of their biological relevance, the heterodimer concept has been validated experimentally and presents a novel paradigm for designing chemokine antagonists [3,5,6]. For example, Koenen et al. [5] reported on the use of CCL5-derived peptides, e.g., CKEY, that function as chemokine heterodimer agonists. The term “chemokine interactome” was recently introduced to promote the chemokine heterodimer concept and present further empirical evidence as to which CXC and CC chemokines interact physically with each other [6]. This large sampling of chemokines demonstrated that not all of them can associate as heterodimers, which imparts selectivity to interactions between mixed chemokines. Moreover, the GAG chondroitin sulfate appears to induce a specific CCL5/CXCL4 heterodimer that enhances atherosclerotic development [5,70]. Other chemokine-derived peptides are also effective chemokine antagonists [6]. However, it was the design and synthesis of two covalently-linked CXCL4/CCL5 [71] and CXCL4/CXCL12 heterodimers [72] that has provided the most compelling evidence in vitro and in vivo that validates the biological relevance of chemokine heterodimers.

3. Galectins

Aside from chemokines, another class of effector molecules that stands out from other regulators is the family of galectins [80,81]. As a sub-family of lectins, galectins have a high level of sequence similarity in their carbohydrate recognition domains (CRDs) along with a preference (albeit non-exclusive) for binding β-galactosides [80]. Even though there are ~14–16 known galectins [82,83,84], galectin-1 (Gal-1) is by far the most studied [85], primarily because it was the first one reported [86]. Galectins are separated into three groups: prototype (Gal-1, 2, 5, 7, 10, 11, 13, 14), chimera (Gal-3), and tandem repeat (Gal-4, 6, 8, 9, 12) [87,88]. Whereas prototype galectins have one CRD, tandem repeat galectins have two homologous CRDs linked by a flexible peptide linker. Gal-3 is the only chimera-type galectin with its conserved CRD tethered to a very long, dynamic, proline-rich N-terminal tail (NT) comprised of multiple PGAY-tetrapeptide repeats. Transient interactions between the Gal-3 CRD and its NT [89] are reported to have a significant effect on glycan binding and various Gal-3-mediated cell-based activities [90]. Moreover, Zhao et al. [90] demonstrated that mutation of any single Gal-3 NT proline (especially P37, P55, P60, P64, and P67) dramatically and differentially inhibits Gal-3–mediated cellular activities (e.g., cell migration, activation, endocytosis, and hemagglutination), as well as glycoprotein-binding-induced Gal-3-mediated oligomerization (via liquid–liquid phase separation, LLPS), a fundamental process required for various activities. Some of the more recently identified galectins show some unusual behavior. For example, recombinant Gal-16 has a pseudo ligand binding site that plays a role in regulating c-Rel-mediated lymphocyte activity [91]; recombinant Gal-14 stands out as an important effector molecule in embryonic development [92], and actin binding to recombinant Gal-13 occurs independently of the galectin canonical ligand binding site [93]. In this regard, Gal-13, -14 and -16 are highly conserved compared to other galectins, are mostly expressed in the placenta of anthropoids (particularly at the maternal-fetal interface), and do not readily bind lactose unless specific residues within the canonical sugar binding site are mutated, e.g., Gal-13 R53 and H57 changed to H53 and R57, and Gal-16 R55 substituted as N55 [91,92,93].
The CRD structures of numerous galectins, usually bound to the disaccharide lactose, have been reported (search the Protein Data Bank, PDB). All galectin CRDs fold as β-sandwiches formed from two β-sheets, one with six anti-parallel β-strands (the sugar binding S-face) and the other with five anti-parallel β-strands (the opposing F-face) (see Figure 3). Their conserved folded structures result from their amino acid sequences being highly conserved [94], another commonality shared with chemokines. Gal-1 is unique, because it has six cysteine residues that need to be oxidized into three disulfide bridges for the lectin to be active in binding carbohydrates [95,96]. There are, however, no known structures of wild type tandem repeat galectins due to the presence of their flexible linker or of full length Gal-3 due to the presence of its long, dynamic N-terminal tail.
Galectins normally bind to β-galactosides or β-galactoside-containing glycans on the cell surface or within the extracellular matrix. Binding occurs at a site in their CRD that is formed by highly conserved residues in the six-stranded β-sheet S-face (Figure 3B) [94]. Upon binding the β-galactoside lactose, for example, CRD residues within the carbohydrate binding site generally do not vary much in terms of their conformations. The major change occurs with the orientation of the highly conserved tryptophan ring that is disordered in the ligand-free state and highly ordered in the lactose bound state as it interacts with the galactose ring. Although the conformation may not change much, internal motions or dynamics can, as evidenced with Gal-1 where lactose binding modulates structural dynamics, increases conformational entropy, and occurs with apparent negative cooperativity [97]. Lactose binding to Gal-7 also induces long-range, allosteric effects through the protein resulting in increased homodimer stability and positive ligand binding cooperativity [98]. With Gal-7, proline isomerization impacts its structural dynamics and glycan affinity [99].
Galectins bind to many carbohydrate ligands, e.g., N-acetyllactosamine octasac-charide [100], N-acetyllactosamine disaccharide [101] and GalNAc-Man-related glycans [102]. In each instance, the non-reducing end of the sugar binds in a similar way, in particular via its interaction with the conserved tryptophan mentioned above. However, with N-acetyllactosamine octasaccharide, the remainder of its longer oligosaccharide chain points out into solution, thus possibly creating a scenario in which a second galectin dimer binds to the free end of the octamer to promote extended polymerization, something that has yet to experimentally verified.
Although β-galactosides are the generally-accepted recognition elements to which galectins bind, this is not always the case, because α-linked digalactosides for example have also been shown to bind to Gal-1, albeit more weakly [103]. Moreover, natural galectin-binding glycans on the cell surface are much more complicated and may determine and/or differentiate galectin function. For example, Gal-3 binds to endothelial cell adhesion molecule CD146 in a somewhat non-conventional way with non-canonical interactions between glycans on CD146 and the lectin’s CRD β-sandwich F-face [104], and Gal-3 binds selectively to the terminal non-reducing end of β(1–4)-galactans with affinity increasing with chain length. Moreover, polysaccharides with a larger glycan footprint bind Gal-1 in a more extensive fashion through most of the CRD S-face [105], and an α-galactomannan (i.e., Davanat) binds Gal-1 at a non-canonical site on its CRD [106,107]. Furthermore, β(1–4)-polymannan and α-branched mannan also bind to Gal-3 at a non-canonical site on the F-face of its CRD, and apparently to a lesser extent to the canonical carbohydrate binding site on the CRD S-face [108]. Polysaccharides may also interact with the N-terminal tail (NT) of Gal-3, a process that is likely modulated by NT proline isomerization [109], and macromolecular assemblies of complex polysaccharides can interact with Gal-3 with synergestic effects on function [110]. In addition, Zheng et al. [111] showed that negatively-charged homogalacturonan-derived oligosaccharides bind to the Gal-3 CRD canonical site in a “topsy-turvy” fashion with their reducing end residue (as opposed to the non-reducing end residues as in lactose) interacting with the conserved tryptophan. This scenario may well be how galectins bind to heparan sulfate-based glycoconjugates on the surfaces of cells, as further exemplified by the binding of keratan sulfate and related saccharides to Gal-1, -3, -7 and -9N [112].
Polysaccharide branching also affects galectin binding. Gal-1 and -7 show greater binding affinities when the oligosaccharide is branched, whereas Gal-8 shows decreased affinity for branched oligosaccharides [113]. The variance of galectin binding to branched or non-branched glycans may provide for different galectins to modulate their biological functions, which can be quite different, E.g., Gal-1 induces T-cell apoptosis, whereas Gal-3 can suppress apoptosis and increase T-cell proliferation [114]. Thus, the function of any one galectin can be multi-faceted, and galectin dimerization and the more varied and complicated nature of glycans in situ, as well as interactions with other biomolecules, will play critical roles in differentiating galectin function.
Similarly to chemokines, galectin monomers can also associate as homodimers (especially prototype galectins), and in some cases, higher oligomer states. Regardless of their having similar monomer folds, galectins, similarly to chemokines, can form different types of homodimers via different surface interactions. Because the folded structures of all galectin monomers are essentially the same, the formation of any one type of homodimer structure is thermodynamically explained by the chemical composition of the set of residues at the monomer-monomer interface. Additionally, as with chemokines, when free energies/residue are in toto greater for one dimer type over another, then that one will dominate. For example, the Gal-1 homodimer is formed primarily via hydrophobic interactions between the N- and C-termini of two monomers (Figure 3A), whereas the Gal-7 homodimer is formed primarily by electrostatic interactions between residues from the two CRD β-sandwich F-faces (Figure 3B). A given homodimer type may be functionally important over another, and possibly explain why different galectins bind differently to complex glycans than to simple disaccharides (e.g., lactose) that are usually employed to investigate galectin-carbohydrate binding and function.
In solution, Gal-1 forms symmetric homodimers [97], whereas the crystal structure of Gal-1 shows the presence of asymmetric dimers, possibly the result of crystal lattice force-based distortion. In situ, it is more likely that symmetric Gal-1 homodimers form. In solution, Gal-3 appears either as a monomer [115] or homodimer [116], and although Ahmad et al. reported that Gal-3 precipitates from solution in a pentameric state (via interactions among their N-Terminal tails (NTs)) upon binding synthetic carbohydrates [117], this is generally unconfirmed in other studies. Nevertheless, the NT of Gal-3 has been reported to mediate higher order oligomerization [118], and recently, it was shown that Gal-3 in the presence of various glycoproteins (i.e., CD146, CD45, CD71, and CD7) oligomerizes through its NT (albeit not as a pentamer) to initiate liquid–liquid phase separation (LLPS), a fundamental process required for these cell activities [90].
The oligomerization state of a galectin can differ with varying solution conditions and thereby affect function, as exemplified with Gal-5 and -7 that behave as monomers [119,120], yet Gal-5 is active at hemagglutination. Gal-7 in crystals is a homodimer, whereas Gal-7 in solution is observed in a dynamic equilibrium between monomers and homodimers [121]. On the other hand, prototype Gal-10 spontaneously forms Charcot-Leyden crystals [122], and Gal-13 homodimers are linked via disulfide bonds. A natural form of Gal-1 (Gal-1β) that lacks the initial six residues, remains monomeric [123] and promotes axonal regeneration, but not Jurkat cell death, unlike Gal-1 homodimers that promote both [123]. In this instance, the homodimeric state is important to function, and changes to that state can be consequential to function. Even though the galectin oligomer state can be crucial to function, many papers often do not discuss it when explaining function.
Whereas some galectins have intracellular functions, they are usually most active extracellularly on the cell surface and within the extracellular matrix. In their oligomeric state, galectins normally function by cross-linking glycans on the cell surface [124] where they bind numerous glycoconjugates to trigger some function, e.g., cell-cell and cell-matrix interactions, proliferation, apoptosis, adhesion, migration, immunity, and inflammation [125]. Galectins are perhaps most generally associated with immune cells and cells in the cardiovascular system, and some galectins are associated with cell growth and differentiation. In this regard, galectins have been described as “exquisite modulators of the immune response” [84]. As broad-spectrum effectors of leukocyte activity and migration [126,127,128], galectins serve as bridges between cells and cell surface glycoconjugates to form lattices in membranes [129,130,131,132,133,134] and to act as sensors of damage- and pathogen-associated effects [135,136]. Cellular expression of galectins varies by cell type and activation state, with any given cell expressing at least one galectin [137]. For example, Gal-1 binds glycoconjugates in the extracellular matrix and on endothelial cells [138] and T lymphocytes [139]. Gal-3 also binds glycoconjugates in the extracellular matrix, as well as CD43 and CD45 on leukocytes, and can be induced by inflammatory mediators [140], e.g., CXC chemokine CXCL8 (interleukin-8, IL8) [141]. Neutrophils from peripheral blood are not responsive to Gal-3, yet neutrophils harvested from an inflammatory site are [142], suggesting glycan modifications are at play. In addition, while Gal-1-induces T-cell apoptosis, Gal-3 may suppress it and enhance T-cell proliferation [114]. Gal-3 is likely the most promiscuous lectin, because it displays diverse biological activities from cell adhesion, apoptosis, and immune regulation. Overall, galectins play various roles to control and mediate different biological functions.

3.1. Galectin-Galectin Heterodimers

Because galectins, similarly to chemokines, are small, protein effector molecules with highly conserved monomeric structures that form different types of homodimers, Miller et al. first proposed that galectins too could form heterodimers between/among themselves with functional significance [7]. As with chemokines, this is especially true when galectins are temporally and spatially co-expressed. The report of physical interactions between two C-type tissue lectins dectin-3 (MCL) and dectin-2 or Mincle [143,144] supports this concept. Using resin-bound Gal-7 in an affinity chromatography-based assay, Miller et al. [7] found that the CRDs of Gal-1 and Gal-3 bound to Gal-7, thus demonstrating hybrid formation between these paired galectins. Moreover, tandem-repeat Gal-8 also associated with Gal-7 on the resin, as did Gal-1 variants where either two CRDs were covalently linked by a Gly-Gly spacer or with the Gal-8 linker sequence (i.e., Gal-1 [GG] or Gal-1 [8S] [145,146]). Since secreted galectins bind cell surface receptors, Miller et al. [7] used a cell-based assay with mixtures of galectins and exploited the lack of strong binding of the truncated Gal-3 CRD. The presence of unlabeled Gal-1 or -7 increased cell surface association of labeled Gal-3 CRD, an increase that depends on binding of the unlabeled lectin. Co-incubation of labeled Gal-3 CRD with a label-free Gal-1 mutant was inhibited from binding lactose by an E71Q mutant and thus failed to enhance the signal.
Puled-field gradient (PFG) NMR experiments have demonstrated the formation of galectin heterodimers, as opposed to higher order oligomers [7]. For the 15N-labeled Gal-3 CRD, the diffusion coefficient, D, was 1.39 × 10−6 cm2/s, a value that is very close to that expected for a monomeric galectin CRD. However, when 15N-labeled Gal-3 CRD was paired with unlabeled Gal-7, the 15N-PFG derived D value decreased to 0.98 × 10−6 cm2/s, a value close to that observed for a prototype galectin homodimer [97]. Thus, the monomeric Gal-3 CRD was shown to interact with Gal-7 CRD to form a Gal-3:Gal-7 heterodimer. HSQC NMR was also used to validate their physical interaction and to provide insight as to how they interact. By mixing unlabeled Gal-7 with 15N-labeled Gal-3, and vice-versa, Miller et al. [7] used HSQC NMR experiments to show that a subset of residues in both Gal-3 CRD and Gal-7 were spectrally perturbed, thus allowing for identification of residues at the heterodimer interface of each galectin. This in turn allowed a structural model of the Gal-3/Gal-7 heterodimer to be developed [7]. The most perturbed residues are within the S-face β-sheet and inter-connecting loops. Using this information, key residues from interacting surfaces between Gal-3 and Gal-7 identified in these HSQC studies, were manually aligned or docked, and molecular dynamics (MD) simulations and energy minimization were performed to optimize the resulting heterodimer structure shown in Figure 4.
Similarly to previous findings with chemokine heterodimers, NMR spectral effects depend on the protein concentration, with effects being more clearly observed when galectin concentrations were kept relatively low to promote some dissociation of their homodimeric states [7]. When Gal-1 CRDs are not strongly associated, as with Gal-1 [8S], hybrid interactions with Gal-7 or the truncated Gal-3 CRD occur more readily than with wild type Gal-1. Furthermore, tandem-repeat Gal-8, with its two monomeric CRDs, also readily associates with resin-bound Gal-7, unlike wild type Gal-1. In toto, these NMR data demonstrate that mixed galectins indeed can form heterodimers and not higher order oligomeric species [7].

3.2. Functional Considerations

The concept of galectin-galectin heterodimers presents a new paradigm in the field. At present, there is only one report that has investigated the biological relevance of galectin heterodimers. Hopefully others will follow.
Dings et al. used cell-based assays to demonstrate that combinations of Gal-1, Gal-3 and/or Gal-7 function synergistically in agglutination, cell surface binding, and endothelial cell proliferation and migration [8]. When combined at various molar ratios, Gal-1 and Gal-7 promote the agglutination of red blood cells and leukocytes significantly more than either of these galectins do alone when adjusted for differences in concentration [8]. FACs analysis with FITC-labeled Gal-1 and mouse splenocytes shows that addition of unlabeled Gal-7 enhances binding of FITC-labeled Gal-1 to the surfaces of CD4+ and CD8+ leukocytes, as well as to CD31+ endothelial cells [8]. This can only occur if these two galectins physically associate as hetero-oligomers. FACs analysis also shows that various combinations of Gal-1, Gal-7, and truncated Gal-3 CRD induce apoptosis in these mouse splenocyte cells [8]. Furthermore, Gal-1 and Gal-7 have an effect on mouse 2H11 endothelial cell proliferation and migration. In this regard, various combinations of Gal-1 and Gal-7 attenuate endothelial cell growth compared to either galectin alone. In the wound healing assay used to assess cell migration, combinations of Gal-1 and Gal-7 reduce the extent of endothelial cell migration considerably more than either galectin alone [8]. These effects occur synergistically, and likely would not occur unless pairs of galectins physically associated as heterodimers.
The generation of galectin CRD heterodimers can offer a way to make new functional species in distinct microenvironments, as in protease-rich inflamed tissues that turn full-length Gal-3 into truncated Gal-3 CRD. Non-covalently linked heterodimers are likely to form in vivo under certain circumstances and thereby promote differential functions, representing a physiologic way to achieve further versatility.

4. Chemokine–Galectin Heterodimers

Cell migration and activation are mediated by both chemokines and galectins, players that are crucial to these physiological events. The expression of galectins and chemokines is correlated by a galectin-dependent enhancement of chemokine production and secretion, a phenomenon that is found in various cell types, e.g., monocytes [147] or in activated pancreatic stellate cells (that secreted the chemokine CXCL12 promoting pancreatic cancer cell invasiveness) [148,149], rheumatoid synovial fibroblasts [150], osteoarthritic chondrocytes [151,152], endothelial cells [153,154], and bone marrow-derived dendritic cells [155]. Collectively, these functional similarities, along with structural data on chemokine–chemokine and galectin–galectin heterodimers, provided the incentive to validate the fundamental hypothesis that galectins and chemokines interact cross-family to form heterodimers. The first step was to establish an “interactome” of chemokines that associate with Gal-1 and Gal-3. With NMR resonance assignments and structural characterizations in hand for both galectins [156,157] and for the chemokine CXCL12 [158,159], the proof-of-principle was solidly established up to the level of mapping contact sites [9]. This in turn opened the way to explore the potential for this type of interaction and to elicit functional effects in vitro and in vivo, with the perspective of a full-scale network analysis.
Gal-1 and -3, similarly to some chemokines, are upregulated during inflammation and are among the most abundant and best studied galectins in inflammatory diseases. Under inflammatory conditions, metalloproteinases cleave the N-terminal region of Gal-3, resulting in free Gal-3 CRD [160,161,162]. Therefore, the encounter of these galectins with chemokines becomes highly likely and focuses interest on demonstrating heterodimerization in vitro. Solid-phase and surface plasmon resonance-based assays showed that Gal-1 and Gal-3 (as well as the Gal-3 CRD) bind to specific chemokines from CC-, CXC-, XC- and CX3C-families [9]. Surface plasmon resonance was used to show that KD values for CXCL12 binding to Gal-3 and Gal-3 CRD are 80 nM and 34 nM, respectively. Chemokine-galectin binding showed remarkable selectivity, and the analysis of HSQC spectra indicated that the binding epitope on Gal-1 or Gal-3 does not involve the canonical sugar-binding site on the CRD S-face [9], and moreover, allowed us to model the galectin–chemokine heterodimer, a model validated by mutagenesis studies [9].
Figure 5 shows an in silico-generated model of the CXCL12-Gal-3 CRD heterodimer. The largest Gal-3 contact region with CXCL12 involves Gal-3 CRD β-strands β7–β9 (colored orange) on the CRD F-face that opposes the canonical sugar-binding S-face of the CRD β-sandwich (colored green, labeled β4–β6). Even though the carbohydrate binding site on Gal-3 is not directly involved in the interaction with CXCL12, it is indirectly affected in that some sugar binding residues, such as N160, are perturbed enough that carbohydrate binding is influenced. This finding has implications for Gal-3 activity, in that Gal-3 can still bind β-galactosides when CXCL12 is bound. In terms of CXCL12, some residues within its β-sheet (β1–β3) and C-terminal helix interact with the Gal-3 CRD. CXCL12 β1-strand is involved in GAG binding, and CXCL12 binding to heparin attenuates CXCL12-Gal-3 heterodimerization, a finding that has implications for GAG-regulated biology of CXCL12 [9]. In principle, a chemokine dimer can be presented by a GAG and simultaneously to its receptor [163]. A prerequisite for Gal-3-CXCL12 heterodimer formation in vivo would likely be that Gal-3 binds as well to GAGs, and indeed there are reports that it does [112,164], an observation requiring that the dimer interface does not overlap with the GAG/receptor binding site.
Gal-3 is expressed by tumor-associated macrophages that are responsible for reducing the mobility of other leukocyte subsets, especially CD8+ T cells [165]. In this respect, the binding of Gal-3 to chemokines appears to be mechanistically relevant [9,166]. Indirectly, Gal-3 inhibits chemokine-dependent migration of CXCR3-expressing lymphocytes by preventing diffusion of IFNγ and in turn the upregulation of chemokines CXCL9, CXCL10, and CXCL11. Recently, evidence for direct biological effects from Gal-3–chemokine heterodimers has been reported with the inhibition of CXCL12- or CCL26-mediated chemotaxis (CXCL12-induced T-cell and neutrophil migration and CCL26-induced eosinophil migration) being attenuated by heterodimer formation with full-length Gal-3 or its CRD [9].
Gal-3/CXCL12 heterodimers have been detected on the cell surface, and a model of the Gal-3/CXCL12 heterodimer bound to CXCR4 within the membrane shows that Gal-3 binds CXCL12 with no steric hindrance to CXCL12 in binding its receptor CXCR4 [167]. This model stands somewhat in contrast to the X-ray crystal structure of viral chemokine vMIPII bound to CXCR4, which shows that it is the chemokine monomer, and not the homodimer, that binds to CXCR4 [65]. Whereas chemokine ligand CXCL12 indeed binds receptor CXCR4 as a monomer, it is the CXCL12/Gal-3 heterodimer that is modeled to bind to CXCR4 via direct interactions with CXCL12 and not with Gal-3 CRD [167]. These CXCL12/Gal-3 complexes occur at concentrations that are lower than the actual KD values would predict. The reason for this is likely that cell surface structures and molecules, such as GAGs and the glycocalyx, facilitate heteromerization. This model implies that Gal-3 can influence CXCL12 signaling by forming a ternary complex with CXCR4 that induces a conformational change in the GPCR [167]. With chemokine heterodimers inducing chemokine receptor heterodimerization, it is possible that a galectin–chemokine heterodimer can bring together a pair of chemokine and galectin receptors. Although speculative, the formation of a chemokine–galectin heterodimer could have wide-ranging biological consequences, opening a new, broad area for study.

5. Conclusions

As small effector molecules, chemokines and galectins are crucial to various biological functions. Additionally, although they can function within their individual families as monomers or homodimers, it is now evident that they can also cross-interact within each of their respective families, as well as between family members as chemokine-galectin heterodimers. The formation of these novel heterodimers has been shown to be functionally significant, to differentiate and expand on their known activities, and to fine-tune those activities, especially when both are present in the same cellular environment. Some of these are summarized in Table 1.
These reports on chemokine and galectin heterodimers have increased our understanding of the functional versatility and cooperation among galectins and chemokines. Moreover, due to the growing interest in this area, a number of labs have initiated their own investigations on these and other small protein effector molecules. The heterodimer paradigm among these small effector molecules is compounded by several observations, namely some chemokine and galectin concentrations are simultaneously elevated at some cellular/tissue sites, e.g., inflammation; both chemokines and galectins can interact with each other within and between their families in diverse manners, and while galectins engage with cell surface glycans, chemokines engage with cell-surface GAGs. Overall, this provides direction to additional structural and functional studies of chemokine and galectin heterodimers, along with production of stable heterodimers that will enhance the physiological significance of these new classes of molecular hybrids and possibly their use as pharmaceutical drugs in clinical practice. However, when new paradigms are initially reported, such as galectin and chemokine heterodimers, there is always some controversy and level of unacceptance. This is particularly true in terms of biological relevance with the new paradigm. Nevertheless, skepticism is a sound scientific practice that then prompts investigators to delve into the new paradigm and experiment further to prove or disprove it.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Guan, E.; Wang, J.; Norcross, M.A. Identification of Human Macrophage Inflammatory Proteins 1α and 1β as a Native Secreted Heterodimer. J. Biol. Chem. 2001, 276, 12404–12409. [Google Scholar] [CrossRef] [PubMed]
  2. Dudek, A.Z.; Nesmelova, I.; Mayo, K.H.; Verfaillie, C.M.; Pitchford, E.; Slungaard, A. Platelet Factor 4 Promotes Adhesion of Hematopoietic Progenitor Cells and Binds IL-8: Novel Mechanisms for Modulation of Hematopoiesis. Blood 2003, 101, 4687–4694. [Google Scholar] [CrossRef] [PubMed]
  3. Nesmelova, I.; Sham, Y.; Dudek, A.Z.; van Eijk, L.I.; Wu, G.; Slungaard, A.; Mortari, F.; Griffioen, A.W.; Mayo, K.H. Platelet Factor 4 and Interleukin-8 CXC Chemokine Heterodimer Formation Modulates Function at the Quaternary Structural Level. J. Biol. Chem. 2005, 280, 4948–4958. [Google Scholar] [CrossRef]
  4. Nesmelova, I.V.; Sham, Y.; Gao, J.; Mayo, K.H. CXC-chemokines associate with CC-chemokines to form mixed heterodimers: RANTES and PF4 monomers associate as CC-type heterodimers. J. Biol. Chem. 2008, 283, 24155–24166. [Google Scholar] [CrossRef]
  5. Koenen, R.; von Hundelhausen, P.; Nesmelova, I.V.; Zernecke, A.; Liehn, E.A.; Sarabi, A.; Kramp, B.K.; Piccinini, A.; Paludan, S.R.; Kowalska, M.A.; et al. Disrupting functional interactions between platelet chemokines inhibits atherosclerosis in hyperlipidemic mice. Nat. Med. 2009, 15, 97–103. [Google Scholar] [CrossRef] [PubMed]
  6. von Hundelshausen, P.; Agten, S.; Eckardt, V.; Schmitt, M.; Blanchet, X.; Neideck, C.; Ippel, H.; Bidzhekov, K.; Wichapong, K.; Faussner, A.; et al. Chemokine interactome mapping enables tailored intervention in acute and chronic inflammation. Sci. Transl. Med. 2017, 9, 384–392. [Google Scholar] [CrossRef]
  7. Miller, M.C.; Ludwig, A.K.; Wichapong, K.; Kaltner, H.; Kopitz, J.; Gabius, H.-J.; Mayo, K.H. Adhesion/growth-regulatory galectins tested in combination: Evidence for synergistic functional cooperation and subunit swapping to form heterodimers. Biochem. J. 2018, 475, 1003–1018. [Google Scholar] [CrossRef]
  8. Dings, R.P.M.; Kumar, N.; Mikkelson, S.; Gabius, H.-J.; Mayo, K.H. Stimulating cellular galectins networks by mixing galectins in vitro reveals synergistic activity. Biochem. Biophys. Rep. 2021, 28, 101–116. [Google Scholar]
  9. Eckardt, V.; Miller, M.C.; Blanchet, X.; Duan, R.; Leberzammer, J.; Duchene, J.; Soehnlein, O.; Megens, R.T.; Ludwig, A.K.; Dregni, A.; et al. Chemokines and galectins form heterodimers to modulate inflammation. EMBO Rep. 2020, 21, e47852. [Google Scholar] [CrossRef]
  10. Baggiolini, M. Chemokines and leukocyte traffic. Nature 1998, 392, 565–568. [Google Scholar] [CrossRef]
  11. Mackay, C.R. Chemokines: Immunology’s high impact factors. Nat. Immunol. 2001, 2, 95–101. [Google Scholar] [CrossRef] [PubMed]
  12. Youn, B.S.; Mantel, C.; Broxmeyer, H.E. Chemokines, chemokine receptors and hematopoiesis. Immunol. Rev. 2000, 177, 150–174. [Google Scholar] [CrossRef] [PubMed]
  13. Koch, A.E.; Polverini, P.J.; Kunkel, S.L.; Harlow, L.A.; DiPietro, L.A.; Elner, V.M.; Elner, S.G.; Strieter, R.M. Interleukin-8 as a macrophage-derived mediator of angiogenesis. Science 1992, 258, 1798–1801. [Google Scholar] [CrossRef] [PubMed]
  14. Belperio, J.A.; Keane, M.P.; Arenberg, D.A.; Addison, C.L.; Ehlert, J.E.; Burdick, M.D.; Strieter, R.M. CXC Chemokines in Angiogenesis. J. Leukoc. Biol. 2000, 68, 1–8. [Google Scholar] [CrossRef] [PubMed]
  15. Zlotnik, A.; Yoshie, O. Chemokines: A new classification system and their role in immunity. Immunity 2000, 12, 121–127. [Google Scholar] [CrossRef] [PubMed]
  16. Clore, G.M.; Gronenborn, A.M. Three-dimensional structures of α- and β-chemokines. FASEB J. 1995, 9, 57–62. [Google Scholar] [CrossRef] [PubMed]
  17. Thomas, M.A.; Buelow, B.J.; Nevins, A.M.; Jones, S.E.; Peterson, F.C.; Gundry, R.L.; Grayson, M.H.; Volkman, B.F. Structure-function analysis of CCL28 in the development of post-viral asthma. J. Biol. Chem. 2015, 290, 4528–4536. [Google Scholar] [CrossRef]
  18. Yang, Y.; Mayo, K.H.; Daly, T.; Barry, J.K.; La Rosa, G.J. Subunit Association and Structural Analysis of Platelet Basic Protein and Related Proteins Investigated by 1H-NMR Spectroscopy and Circular Dichroism. J. Biol. Chem. 1994, 269, 20110–20118. [Google Scholar] [CrossRef]
  19. Clark-Lewis, I.; Kim, K.S.; Rajarathnam, K.; Gong, J.H.; Dewald, B.; Moser, B.; Baggiolini, M.; Sykes, B.D. Structure-activity relationships of chemokines. J. Leukoc. Biol. 1995, 57, 703–711. [Google Scholar] [CrossRef]
  20. Wang, X.; Watson, C.; Sharp, J.S.; Handel, T.M.; Prestegard, J.H. Oligomeric structure of the chemokine CCL5/RANTES from NMR, MS, and SAXS data. Structure 2011, 19, 1138–1148. [Google Scholar] [CrossRef]
  21. Jansma, A.L.; Kirkpatrick, J.P.; Hsu, A.R.; Handel, T.M.; Nietlispach, D. NMR analysis of the structure, dynamics, and unique oligomerization properties of the chemokine CCL27. J. Biol. Chem. 2010, 285, 14424–14437. [Google Scholar] [CrossRef] [PubMed]
  22. Clore, G.M.; Appella, E.; Yamada, M.; Matsushima, K.; Gronenborn, A.M. Three dimensional structure of interleukin 8 in solution. Biochemistry 1990, 29, 1689–1696. [Google Scholar] [CrossRef] [PubMed]
  23. Liang, W.G.; Triandafillou, C.G.; Huang, T.Y.; Zulueta, M.M.; Banerjee, S.; Dinner, A.R.; Hung, S.C.; Tang, W.J. Crystal structure of CC chemokine 5 (CCL5). Proc. Natl. Acad. Sci. USA 2016, 113, 5000–5005. [Google Scholar] [CrossRef] [PubMed]
  24. Skelton, N.J.; Aspiras, F.; Ogez, J.; Schall, T.J. Proton NMR assignments and solution conformation of RANTES, a chemokine of the CC type. Biochemistry 1995, 34, 5329–5342. [Google Scholar] [CrossRef]
  25. Mayo, K.H.; Roongta, V.; Barker, S.; Milius, R.; Ilyina, E.; Quinlan, C.; La Rosa, G.; Daly, T. NMR Solution Structure of the 32 kD Tetrameric Platelet Factor-4 ELR-Motif N-terminal Chimer: A Symmetric Tetramer. Biochemistry 1995, 34, 11399–11409. [Google Scholar] [CrossRef]
  26. Swaminathan, G.J.; Holloway, D.E.; Colvin, R.A.; Campanella, K.; Papageorgiou, A.C.; Luster, A.D.; Acharya, K.R. Crystal Structures of Oligomeric Forms of the IP-10/CXCL10 Chemokine. Structure 2003, 11, 521–532. [Google Scholar] [CrossRef]
  27. Zhang, X.; Chen, L.; Bancroft, D.P.; Lai, C.K.; Maione, T.E. Crystal structure of recombinant human platelet factor 4. Biochemistry 1994, 33, 8361–8366. [Google Scholar] [CrossRef]
  28. Paoletti, S.; Petkovic, V.; Sebastiani, S.; Danelon, M.G.; Uguccioni, M.; Gerber, B.O. A rich chemokine environment strongly enhances leukocyte migration and activities. Blood 2005, 105, 3405–3412. [Google Scholar] [CrossRef]
  29. Campanella, G.S.V.; Grimm, J.; Manice, L.A.; Colvin, R.A.; Medoff, B.D.; Woitkiewicz, G.R.; Weissleder, R.; Luster, A.D. Oligomerization of CXCL10 Is Necessary for Endothelial Cell Presentation and In Vivo Activity. J. Immunol. 2006, 177, 6991–6998. [Google Scholar] [CrossRef]
  30. Rek, A.; Brandner, B.; Geretti, E.; Kungl, A.J. A biophysical insight into the RANTES–glycosaminoglycan interaction. Biochim. Biophys. Acta 2009, 1794, 577–582. [Google Scholar] [CrossRef]
  31. Ren, M.; Guo, Q.; Guo, L.; Lenz, M.; Qian, F.; Koenen, R.R.; Xu, H.; Schilling, A.B.; Weber, C.; Ye, R.D.; et al. Polymerization of MIP-1 chemokine (CCL3 and CCL4) and clearance of MIP1 by insulin-degrading enzyme. EMBO J. 2010, 29, 3952–3966. [Google Scholar] [CrossRef] [PubMed]
  32. Mayo, K.H.; Chen, M.-J. Human Platelet Factor 4 Monomer-Dimer-Tetramer Equilibria Investigated by NMR Spectroscopy. Biochemistry 1989, 28, 9469–9478. [Google Scholar] [CrossRef]
  33. Chen, M.J.; Mayo, K.H. Human Platelet Factor 4 Subunit Association-Dissociation Thermodynamics and Kinetics. Biochemistry 1991, 30, 6402–6411. [Google Scholar] [CrossRef]
  34. Mayo, K.H. Low Affinity Platelet Factor 4 1H-NMR Derived Aggregate Equilibria Indicate Physiological Preference for Monomers over Dimers and Tetramers. Biochemistry 1991, 30, 925–934. [Google Scholar] [CrossRef] [PubMed]
  35. Young, H.; Roongta, V.; Daly, T.J.; Mayo, K.H. NMR Structure and Dynamics of Monomeric Neutrophil Activating Peptide-2. Biochem. J. 1999, 338, 591–598. [Google Scholar] [CrossRef] [PubMed]
  36. Yang, Y.; Mayo, K.H. Alcohol-Induced Protein Folding Transitions in Platelet Factor- 4: The O-State. Biochemistry 1993, 32, 8661–8671. [Google Scholar] [CrossRef]
  37. Yang, Y.; Barker, S.; Chen, M.-J.; Mayo, K.H. Effect of Low Molecular Weight Aliphatic Alcohols and Related Compounds on Platelet Factor-4 Subunit Association. J. Biol. Chem. 1993, 268, 9223–9229. [Google Scholar] [CrossRef]
  38. Veldkamp, C.T.; Ziarek, J.J.; Su, J.; Basnet, H.; Lennertz, R.; Weiner, J.J.; Peterson, F.C.; Baker, J.E.; Volkman, B.F. Monomeric structure of the cardio-protective chemokine SDF-1/CXCL12. Protein Sci. 2009, 18, 1359–1369. [Google Scholar] [CrossRef]
  39. Veldkamp, C.T.; Peterson, F.C.; Pelzek, A.J.; Volkman, B.F. The monomer-dimer equilibrium of stromal cell-derived factor-1 (CXCL12) is altered by pH, phosphate, sulfate, and heparin. Protein Sci. 2005, 14, 1071–1081. [Google Scholar] [CrossRef]
  40. Crump, M.P.; Rajarathnam, K.; Kim, K.-S.; Clark-Lewis, I.; Sykes, B.D. Solution Structure of Eotaxin, a Chemokine That Selectively Recruits Eosinophils in Allergic Inflammation. J. Biol. Chem. 1998, 273, 22471–22479. [Google Scholar] [CrossRef]
  41. Ellyard, J.I.; Simson, L.; Bezos, A.; Johnston, K.; Freeman, C.; Parish, C.R. Eotaxin Selectively Binds Heparin. J. Biol. Chem. 2007, 282, 15238–15247. [Google Scholar] [CrossRef]
  42. Proudfoot, A.E. The BBXB Motif of RANTES Is the Principal Site for Heparin Binding and Controls Receptor Selectivity. J. Biol. Chem. 2001, 276, 10620–10626. [Google Scholar] [CrossRef] [PubMed]
  43. Sheng, G.J.; Oh, Y.I.; Chang, S.-K.; Hsieh-Wilson, L.C. Tunable Heparan Sulfate Mimetics for Modulating Chemokine Activity. J. Am. Chem. Soc. 2013, 135, 10898–10901. [Google Scholar] [CrossRef] [PubMed]
  44. Salanga, C.L.; Handel, T.M. Chemokine oligomerization and interactions with receptors and glycosaminoglycans: The role of structural dynamics in function. Exp. Cell Res. 2011, 317, 590–601. [Google Scholar] [CrossRef]
  45. Proudfoot, A.E. Chemokines and Glycosaminoglycans. Front. Immunol. 2015, 6, 246–258. [Google Scholar] [CrossRef] [PubMed]
  46. Shaw, J.P.; Johnson, Z.; Borlat, F.; Zwahlen, C.; Kungl, A.; Roulin, K.; Harrenga, A.; Wells, T.N.; Proudfoot, A.E. The X-ray Structure of RANTES Heparin-Derived Disaccharides Allows the Rational Design of Chemokine Inhibitors. Structure 2004, 12, 2081–2093. [Google Scholar] [CrossRef] [PubMed]
  47. Seo, Y.; Andaya, A.; Bleiholder, C.; Leary, J.A. Differentiation of CC vs. CXC Chemokine Dimers with GAG Octasaccharide Binding Partners: An Ion Mobility Mass Spectrometry Approach. J. Am. Chem. Soc. 2013, 135, 4325–4332. [Google Scholar] [CrossRef]
  48. Mayo, K.H.; Ilyina, E.; Roongta, V.; Dundas, M.; Joseph, J.; Lai, C.K.; Maione, T.; Daly, T. Heparin Binding to Platelet Factor-4. An NMR and Site-Directed Mutagenesis Study: Arginine Residues Crucial for Binding. Biochem. J. 1995, 312, 357–365. [Google Scholar] [CrossRef]
  49. Dyer, D.P.; Salanga, C.L.; Volkman, B.F.; Kawamura, T.; Handel, T.M. The dependence of chemokine-glycosaminoglycan interactions on chemokine oligomerization. Glycobiology 2016, 26, 312–326. [Google Scholar] [CrossRef]
  50. Hoogewerf, A.J.; Kuschert, G.S.; Proudfoot, A.E.; Borlat, F.; Clark-Lewis, I.; Power, C.A.; Wells, T.N. Glycosaminoglycans mediate cell surface oligomerization of chemokines. Biochemistry 1997, 36, 13570–13578. [Google Scholar] [CrossRef]
  51. Proudfoot, A.E.; Handel, T.M.; Johnson, Z.; Lau, E.K.; Li Wang, P.; Clark-Lewis, I.; Borlat, F.; Wells, T.N.; Kosco-Vilbois, M.H. Glycosaminoglycan binding and oligomerization are essential for the in vivo activity of certain chemokines. Proc. Natl. Acad. Sci. USA 2003, 100, 1885–1890. [Google Scholar] [CrossRef] [PubMed]
  52. Crown, S.E.; Yu, Y.; Sweeney, M.D.; Leary, J.A.; Handel, T.M. Heterodimerization of CCR2 chemokines and regulation by glycosaminoglycan binding. J. Biol. Chem. 2006, 281, 25438–25446. [Google Scholar] [CrossRef] [PubMed]
  53. Verkaar, F.; van Offenbeek, J.; van der Lee, M.M.C.; van Lith, L.H.C.J.; Watts, A.O.; Rops, A.L.W.M.M.; Aguilar, D.C.; Ziarek, J.J.; van der Vlag, J.; Handel, T.M.; et al. Chemokine cooperativity is caused by competitive glycosaminoglycan binding. J. Immunol. 2014, 192, 3908–3914. [Google Scholar] [CrossRef] [PubMed]
  54. Mikhailov, D.V.; Young, H.; Linhardt, R.J.; Mayo, K.H. Heparin Dodecasaccharide Binding to Platelet Factor-4 and Growth-related Protein-α: Induction of a Partially Folded State and Implications for Heparin-Induced Thromnocytopenia. J. Biol. Chem. 1999, 274, 25317–25329. [Google Scholar] [CrossRef]
  55. Brandhofer, M.; Hoffmann, A.; Blanchet, X.; Siminkovitch, E.; Rohlfing, A.-K.; El Bounkari, O.; Nestele, J.A.; Bild, A.; Kontos, C.; Hille, K.; et al. Heterocomplexes between the Atypical Chemokine MIF and the CXC-Motif Chemokine CXCL4L1 Regulate Inflammation and Thrombus Formation. Cell. Mol. Life Sci. 2022, 79, 512. [Google Scholar] [CrossRef] [PubMed]
  56. Jansma, A.; Handel, T.M.; Hamel, D.J. Chapter 2. Homo- and hetero-oligomerization of chemokines. Methods Enzymol. 2009, 461, 31–50. [Google Scholar]
  57. Koenen, R.R.; Weber, C. Therapeutic targeting of chemokine interactions in atherosclerosis. Nat. Rev. Drug Discov. 2010, 9, 141–153. [Google Scholar] [CrossRef]
  58. Raman, D.; Sobolik-Delmaire, T.; Richmond, A. Chemokines in health and disease. Exp. Cell Res. 2011, 317, 575–589. [Google Scholar] [CrossRef]
  59. Viola, A.; Luster, A.D. Chemokines and Their Receptors: Drug Targets in Immunity and Inflammation. Ann. Rev. Pharmacol. Toxicol. 2008, 48, 171–197. [Google Scholar] [CrossRef]
  60. Allen, S.J.; Crown, S.E.; Handel, T.M. Chemokine: Receptor structure, interactions, and antagonism. Annu. Rev. Immunol. 2007, 25, 787–820. [Google Scholar] [CrossRef]
  61. Park, S.H.; Das, B.B.; Casagrande, F.; Tian, Y.; Nothnagel, H.J.; Chu, M.; Kiefer, H.; Maier, K.; DeAngelis, A.A.; Marassi, F.M.; et al. Structure of the chemokine receptor CXCR1 in phospholipid bilayers. Nature 2012, 491, 779–783. [Google Scholar] [CrossRef] [PubMed]
  62. Thelen, M.; Stein, J.V. How chemokines invite leukocytes to dance. Nat. Immun. 2008, 9, 953–959. [Google Scholar] [CrossRef] [PubMed]
  63. Wu, B.; Chien, E.Y.; Mol, C.D.; Fenalti, G.; Liu, W.; Katritch, V.; Abagyan, R.; Brooun, A.; Wells, P.; Bi, F.C.; et al. Structures of the CXCR4 Chemokine GPCR with Small-Molecule and Cyclic Peptide Antagonists. Science 2010, 330, 1066–1071. [Google Scholar] [CrossRef] [PubMed]
  64. Hemmerich, S.; Paavola, C.; Bloom, A.; Bhakta, S.; Freedman, R.; Grunberger, D.; Krstenansky, J.; Lee, S.; McCarley, D.; Mulkins, M.; et al. Identification of residues in the monocyte chemotactic protein-1 that contact the MCP-1 receptor, CCR2. Biochemistry 1999, 38, 13013–13025. [Google Scholar] [CrossRef]
  65. Qin, L.; Kufareva, I.; Holden, L.G.; Wang, C.; Zheng, Y.; Zhao, C.; Fenalti, G.; Wu, H.; Han, G.W.; Cherezov, V.; et al. Crystal structure of the chemokine receptor CXCR4 in complex with a viral chemokine. Science 2015, 347, 1117–11122. [Google Scholar] [CrossRef] [PubMed]
  66. Strieter, R.M.; Polverini, P.J.; Kunkel, S.L.; Arenberg, D.A.; Burdick, M.D.; Kasper, J.; Dzuiba, J.; VanDamme, J.; Walz, A.; Marriott, D. The functional role of the ELR motif in CXC chemokine-mediated angiogenesis. J. Biol. Chem. 1995, 270, 27348–27357. [Google Scholar] [CrossRef] [PubMed]
  67. Blanpain, C.; Doranz, B.J.; Bondue, A.; Govaerts, C.; De Leener, A. The core domain of chemokines bind CCR5 extracellular domains while their amino terminus interacts with the transmembrane helix bundle. J. Biol. Chem. 2002, 278, 5179–5187. [Google Scholar] [CrossRef]
  68. Gouwy, M. Synergy between proinflammatory ligands of G protein-coupled receptors in neutrophil activation and migration. J. Leukoc. Biol. 2004, 76, 185–194. [Google Scholar] [CrossRef]
  69. Gouwy, M.; Struyf, S.; Noppen, S.; Schutyser, E.; Springael, J.-Y.; Parmentier, M.; Proost, P.; VanDamme, J. Synergy between Co-produced CC and CXC Chemokines in Monocyte Chemotaxis through Receptor-Mediated Events. Mol. Pharmacol. 2008, 74, 485–495. [Google Scholar] [CrossRef]
  70. von Hundelshausen, P.; Koenen, R.R.; Sack, M.; Mause, S.F.; Adriaens, W.; Proudfoot, A.E.; Hackeng, T.M.; Weber, C. Heterophilic interactions of platelet factor 4 and RANTES promote monocyte arrest on endothelium. Blood 2005, 105, 924–930. [Google Scholar] [CrossRef]
  71. Agten, S.M.; Koenen, R.; Ippel, H.; Eckert, V.; von Hundelshausen, P.; Mayo, K.H.; Weber, C.; Hackeng, T.M. Probing Functional Heteromeric Chemokine Protein-Protein Interactions through Conformation-assisted Oxime-Linkage. Angewante Chem. 2016, 55, 14963–14966. [Google Scholar] [CrossRef] [PubMed]
  72. Nguyen, K.T.P.; Volkman, B.; Dréau, D.; Nesmelova, I.V. A new obligate CXCL4-CXCL12 heterodimer for studying chemokine heterodimer activities and mechanisms. Sci. Rep. 2022, 12, 17204. [Google Scholar] [CrossRef] [PubMed]
  73. Stevens, R.L.; Colombo, M.; Gonzales, J.J.; Hollander, W.; Schmid, K. The glycosaminoglycans of the human artery and their changes in atherosclerosis. J. Clin. Investig. 1976, 58, 470–477. [Google Scholar] [CrossRef] [PubMed]
  74. Taylor, K.R.; Gallo, R.L. Glycosaminoglycans and their proteoglycans: Host-associated molecular patterns for initiation and modulation of inflammation. FASEB J. 2006, 20, 9–22. [Google Scholar] [CrossRef]
  75. Handel, T.M.; Johnson, Z.; Crown, S.E.; Lau, E.K.; Proodfoot, A.E. Regulation of protein function by glycosaminoglycans—As exemplified by chemokines. Ann. Rev. Biochem. 2005, 74, 385–410. [Google Scholar] [CrossRef]
  76. Gouwy, M.; Struyf, S.; Berghmans, N.; Vanormelingen, C.; Schols, D.; Van Damme, J. CXCR4 and CCR5 ligands cooperate in monocyte and lymphocyte migration and in inhibition of dual-tropic (R5/X4) HIV-1 infection. Eur. J. Immunol. 2011, 41, 963–973. [Google Scholar] [CrossRef]
  77. Gouwy, M.; Schiraldi, M.; Struyf, S.; Van Damme, J.; Uguccioni, M. Possible mechanisms involved in chemokine synergy fine tuning the inflammatory response. Immunol. Lett. 2012, 145, 10–14. [Google Scholar] [CrossRef]
  78. Gouwy, M.; Struyf, S.; Leutenez, L.; Pörtner, N.; Sozzani, S.; Van Damme, J. Chemokines and other GPCR ligands synergize in receptor-mediated migration of monocyte-derived immature and mature dendritic cells. Immunobiology 2014, 219, 218–229. [Google Scholar] [CrossRef]
  79. Mortier, A.; Van Damme, J.; Proost, P. Overview of the mechanisms regulating chemokine activity and availability. Immunol. Lett. 2012, 145, 2–9. [Google Scholar] [CrossRef]
  80. Barondes, S.H.; Castronovo, V.; Cooper, D.N.; Cummings, R.D.; Drickamer, K.; Feizi, T.; Gitt, M.A.; Hirabayashi, J.; Hughes, C.; Kasai, K. Galectins: A family of animal β-galactoside-binding lectins. Cell 1994, 76, 597–598. [Google Scholar] [CrossRef]
  81. Barondes, S.H. Galectins: A personal overview. Trends Glycosci. Glycotechnol. 1998, 9, 1–7. [Google Scholar] [CrossRef]
  82. Cooper, D.N.; Barondes, S.H. God must love galectins; he made so many of them. Glycobiology 1990, 9, 979–984. [Google Scholar] [CrossRef] [PubMed]
  83. Cooper, D.N. Galectinomics: Finding themes in complexity. Biochim. Biophys. Acta 2002, 1572, 209–231. [Google Scholar] [CrossRef] [PubMed]
  84. Cooper, D.; Iqbal, A.J.; Gittens, B.R.; Cervone, C.; Perretti, M. The effect of galectins on leukocyte trafficking in inflammation: Sweet or sour? Ann. N.Y. Acad. Sci. 2012, 1253, 181–192. [Google Scholar] [CrossRef] [PubMed]
  85. Camby, I.; Le Mercier, M.; Lefranc, F.; Kiss, R. Galectin-1: A small protein with major functions. Glycobiology 2006, 16, 137–157. [Google Scholar] [CrossRef] [PubMed]
  86. Teichberg, V.I.; Silman, I.; Beitsch, D.D.; Resheff, G. A β-D-galactoside binding protein from electric organ tissue of Electrophorus electricus. Proc. Natl. Acad. Sci. USA 1975, 72, 1383–1387. [Google Scholar] [CrossRef]
  87. Hirabayashi, J.; Kasai, K. The family of metazoan metal-independent β-galactoside-binding lectins: Structure, function and molecular evolution. Glycobiology 1993, 3, 297–304. [Google Scholar] [CrossRef]
  88. Kasai, K.; Hirabayashi, J. Galectins: A family of animal lectins that decipher glycocodes. J. Biochem. 1996, 119, 1–8. [Google Scholar] [CrossRef]
  89. Ippel, H.; Miller, M.C.; Vértesy, S.; Zheng, Y.; Cañada, F.J.; Suylen, D.; Umemoto, K.; Romano, C.; Hackeng, T.; Tai, G.; et al. Intra- and intermolecular interactions of human galectin-3: Assessment by full-assignment-based NMR. Glycobiology 2016, 26, 888–903. [Google Scholar] [CrossRef]
  90. Zhao, Z.; Xu, X.; Cheng, H.; Miller, M.C.; He, Z.; Gu, H.; Zhang, Z.; Raz, A.; Mayo, K.H.; Tai, G.; et al. Galectin-3 N-terminal tail prolines modulate cell activity and glycan-mediated oligomerization/phase separation. Proc. Natl. Acad. Sci. USA 2021, 118, e2021074118. [Google Scholar] [CrossRef]
  91. Si, Y.; Yao, Y.; Ayala, G.; Li, X.; Han, Q.; Zhang, W.; Tai, G.; Mayo, K.H.; Zhou, Y.; Su, J. Human galectin-16 has a pseudo ligand binding site and plays a role in regulating c-Rel-mediated lymphocyte activity. Biochim. Biophys. Acta 2021, 1865, e129755. [Google Scholar] [CrossRef] [PubMed]
  92. Si, Y.; Li, Y.; Yang, T.; Li, X.; Ayala, G.J.; Mayo, K.H.; Tai, G.; Su, J.; Zhou, Y. Structure-function studies of galectin-14, an important effector molecule in embryology. FEBS J. 2021, 288, 1041–1055. [Google Scholar] [CrossRef] [PubMed]
  93. Li, X.; Yao, Y.; Liu, T.; Gu, K.; Han, Q.; Zhang, W.; Ayala, G.J.; Liu, Y.; Na, H.; Yu, J.; et al. Actin binding to galectin-13/placental protein-13 occurs independently of the galectin canonical ligand binding site. Glycobiology 2021, 31, 1219–1229. [Google Scholar] [CrossRef] [PubMed]
  94. Kadoya, T.; Horie, H. Structural and functional studies of galectin-1: A novel axonal regeneration-promoting activity for oxidized galectin-1. Curr. Drug Targets 2005, 6, 375–383. [Google Scholar] [CrossRef] [PubMed]
  95. Hirabayashi, J.; Kasai, K. Effect of amino acid substitution by sited-directed mutagenesis on the carbohydrate recognition and stability of human 14-kDa β-galactoside-binding lectin. J. Biol. Chem. 1991, 266, 23648–23653. [Google Scholar] [CrossRef]
  96. Tracey, B.M.; Feizi, T.; Abbott, W.M.; Carruthers, R.A.; Green, B.N.; Lawson, A.M. Subunit molecular mass assignment of 14,654 Da to the soluble β-galactoside-binding lectin from bovine heart muscle and demonstration of intramolecular disulfide bonding associated with oxidative inactivation. J. Biol. Chem. 1992, 267, 10342–10347. [Google Scholar] [CrossRef]
  97. Nesmelova, I.V.; Ermakova, E.; Daragan, V.A.; Pang, M.; Menéndez, M.; Lagartera, L.; Solís, D.; Baum, L.G.; Mayo, K.H. Lactose binding to galectin-1 modulates structural dynamics, increases conformational entropy, and occurs with apparent negative cooperativity. J. Mol. Biol. 2010, 397, 1209–1230. [Google Scholar] [CrossRef]
  98. Ermakova, E.; Miller, M.C.; Nesmelova, I.V.; Lopez-Merino, L.; Berbís, M.A.; Nesmelov, Y.; Lagartera, L.; Daragan, V.A.; André, S.; Cañada, F.J.; et al. Lactose Binding to Human Galectin-7 (p53-induced gene 1) Induces Long-range Effects through the Protein Resulting in Increased Dimer Stability and Evidence for Positive Cooperativity. Glycobiology 2013, 23, 508–523. [Google Scholar] [CrossRef]
  99. Miller, M.C.; Nesmelova, I.V.; Daragan, V.A.; Ippel, H.; Michalak, M.; Dregni, A.; Kaltner, H.; Kopitz, J.; Gabius, H.-J.; Mayo, K.H. Identification of Pro4 prolyl peptide bond isomerization in human galectin-7 and its impact on structural dynamics and glycan affinity. Biochem. J. 2020, 477, 3147–3165. [Google Scholar] [CrossRef]
  100. Bourne, Y.; Bolgiano, B.; Liao, D.I.; Strecker, G.; Cantau, P.; Herzberg, O.; Feizi, T.; Cambillau, C. Crosslinking of mammalian lectin (galectin-1) by complex biantennary saccharides. Nat. Struct. Biol. 1994, 1, 863–870. [Google Scholar] [CrossRef]
  101. Nagae, M.; Nishi, N.; Murata, T.; Usui, T.; Nakamura, T.; Wakatsuki, S.; Kato, R. Crystal structure of the galectin-9 N-terminal carbohydrate recognition domain from Mus musculus reveals the basic mechanism of carbohydrate recognition. J. Biol. Chem. 2006, 281, 35884–35893. [Google Scholar] [CrossRef] [PubMed]
  102. Pinho, S.S.; Alves, I.; Gaifem, J.; Rabinovich, G.A. Immune regulatory networks coordinated by glycans and glycan-binding proteins in autoimmunity and infection. Cell. Molecul. Immun. 2023. [Google Scholar] [CrossRef]
  103. Miller, M.C.; Ribeiro, J.P.; Roldós, V.; Martín-Santamaría, S.; Cañada, F.J.; Nesmelova, I.A.; André, S.; Pang, M.; Klyosov, A.A.; Baum, L.G.; et al. Structural aspects of binding of α-linked digalactosides to human galectin-1. Glycobiology 2011, 21, 1627–1641. [Google Scholar] [CrossRef] [PubMed]
  104. Zhang, Z.; Miller, M.C.; Xu, X.; Zheng, Y.; Song, C.; Zhang, F.; Zhou, Y.; Tai, G.; Mayo, K.H. NMR-based insight into galectin-3 binding to endothelial cell adhesion molecule CD146: Evidence for non-cannonical interactions with the lectin’s CRD b-sandwich F-face. Glycobiology 2019, 29, 608–618. [Google Scholar] [CrossRef] [PubMed]
  105. Miller, M.; Nesmelova, I.V.; Klyosov, A.; Platt, D.; Mayo, K.H. The carbohydrate binding domain on galectin-1 is more extensive for a complex glycan than for simple saccharides: Implications for galectin-glycan interactions at the cell surface. Biochem. J. 2009, 421, 211–221. [Google Scholar] [CrossRef]
  106. Miller, M.; Klyosov, A.; Mayo, K.H. The α-galactomannan Davanat binds galectin-1 at asite different from the conventional galectin carbohydrate binding site. Glycobiology 2009, 19, 1034–1045. [Google Scholar] [CrossRef]
  107. Miller, M.C.; Klyosov, A.; Mayo, K.H. Structural Features for α-galactomannan binding to galectin-1. Glycobiology 2012, 22, 543–551. [Google Scholar] [CrossRef]
  108. Miller, M.C.; Ippel, H.; Suylen, D.; Klyosov, A.A.; Traber, P.G.; Hackeng, T.; Mayo, K.H. Binding of Polysaccharides to Human Galectin-3 at a Non-Canonical Site in its Carbohydrate Recognition Domain. Glycobiology 2016, 26, 88–99. [Google Scholar] [CrossRef]
  109. Miller, M.C.; Zheng, Y.; Yan, J.; Zhou, Y.; Tai, G.; Mayo, K.H. Novel polysaccharide binding to the N-terminal tail of galection-3 is likely modulated by proline isomerization. Glycobiology 2017, 27, 1038–1051. [Google Scholar] [CrossRef]
  110. Zhang, T.; Miller, M.C.; Lan, Y.; Zheng, Y.; Liu, F.; Zhao, D.; Su, J.; Mayo, K.H.; Tai, G.; Zhou, Y. Macromolecular assemblies of complex polysaccharides with Galectin-3 and their synergestic effects on function. Biochem. J. 2017, 474, 3849–3868. [Google Scholar] [CrossRef]
  111. Zheng, Y.; Su, J.; Miller, M.C.; Zhang, T.; Mayzel, M.; Tai, G.; Mayo, K.H.; Zhou, Y. Topsy-turvy binding of negatively-charged homogalacturonan oligosaccharides to galectin-3. Glycobiology 2021, 31, 341–350. [Google Scholar] [CrossRef]
  112. Miller, M.C.; Cai, C.; Wichapong, K.; Bhaduri, S.; Pohl, N.L.B.; Linhardt, R.J.; Gabius, H.-J.; Mayo, K.H. Structural insight into the binding of human galectins to corneal keratan sulfate, its desulfated form and related saccharides. Sci. Rep. 2020, 10, 15708–15713. [Google Scholar] [CrossRef] [PubMed]
  113. Hirabayashi, J.; Hashidate, T.; Arata, Y.; Nishi, N.; Nakamura, T.; Hirashima, M.; Urashima, T.; Oka, T.; Futai, M.; Muller, W.E.; et al. Oligosaccharide specificity of galectins: A search by frontal affinity chromatography. Biochim. Biophys. Acta 2002, 1572, 232–254. [Google Scholar] [CrossRef] [PubMed]
  114. Yang, R.Y.; Hsu, D.K.; Liu, F.T. Expression of galectin-3 modulates T-cell growth and apoptosis. Proc. Natl. Acad. Sci. USA 1996, 93, 6737–6742. [Google Scholar] [CrossRef] [PubMed]
  115. Massa, S.M.; Cooper, D.N.; Leffler, H.; Barondes, S.H. L-29, an endogenous lectin, binds to glycoconjugate ligands with positive cooperativity. Biochemistry 1993, 32, 260–267. [Google Scholar] [CrossRef]
  116. Kuklinski, S.; Probstmeier, R. Homophilic binding properties of galectin-3: Involvement of the carbohydrate recognition domain. J. Neurochem. 1998, 70, 814–823. [Google Scholar] [CrossRef] [PubMed]
  117. Ahmad, N.; Gabius, H.-J.; Andre, S.; Kaltner, H.; Sabesan, S.; Roy, R.; Liu, B.; Macaluso, F.; Brewer, C.F. Galectin-3 precipitates as a pentamer with synthetic multivalent carbohydrates and forms heterogeneous cross-linked complexes. J. Biol. Chem. 2004, 279, 10841–10847. [Google Scholar] [CrossRef] [PubMed]
  118. Rini, J.M. Lectin structure. Annu. Rev. Biophys. Biomol. Struct. 1995, 24, 551–577. [Google Scholar] [CrossRef]
  119. Gitt, M.A.; Wiser, M.F.; Leffler, H.; Herrmann, J.; Xia, Y.R.; Massa, S.M.; Cooper, D.N.; Lusis, A.J.; Barondes, S.H. Sequence and mapping of galectin-5, a β-galactoside-binding lectin, found in rat erythrocytes. J. Biol. Chem. 1995, 270, 5032–5038. [Google Scholar] [CrossRef]
  120. Madsen, P.; Rasmussen, H.H.; Flint, T.; Gromov, P.; Kruse, T.A.; Honore, B.; Vorum, H.; Celis, J.E. Cloning, expression, and chromosome mapping of human galectin-7. J. Biol. Chem. 1995, 270, 5823–5829. [Google Scholar] [CrossRef]
  121. Leonidas, D.D.; Vatzaki, E.H.; Vorum, H.; Celis, J.E.; Madsen, P.; Acharya, K.R. Structural basis for the recognition of carbohydrates by human galectin-7. Biochemistry 1998, 37, 13930–13940. [Google Scholar] [PubMed]
  122. Leonidas, D.D.; Elbert, B.L.; Zhou, Z.; Leffler, H.; Ackerman, S.J.; Acharya, K.R. Crystal structure of human Charcot-Leyden crystal protein, an eosinophil lysophospholipase, identifies it as a new member of the carbohydrate-binding family of galectins. Structure 1995, 3, 1379–1393. [Google Scholar] [PubMed]
  123. Miura, T.; Takahashi, M.; Horie, H.; Kurushima, H.; Tsuchimoto, D.; Sakumi, K.; Nakabeppu, Y. Galectin-1β, a natural monomeric form of galectin-1 lacking its six amino-terminal residues promotes axonal regeneration but not cell death. Cell Death Differ. 2004, 11, 1076–1083. [Google Scholar] [CrossRef]
  124. Brewer, F.C. Binding and cross-linking properties of galectins. Biochim. Biophys. Acta 2002, 1572, 255–262. [Google Scholar] [CrossRef]
  125. Liu, F.T.; Rabinovich, G.A. Galectins as modulators of tumour progression. Nat. Rev. Cancer 2005, 5, 29–41. [Google Scholar]
  126. Thiemann, S.; Baum, L.G. The road less traveled: Regulation of leukocyte migration across vascular and lymphatic endothelium by galectins. J. Clin. Immunol. 2011, 31, 2–9. [Google Scholar] [PubMed]
  127. Thiemann, S.; Baum, L.G. Galectins and immune responses-just how do they do those things they do? Annu. Rev. Immunol. 2016, 34, 243–264. [Google Scholar] [CrossRef]
  128. Liu, F.T.; Yang, R.Y.; Hsu, D.K. Galectins in acute and chronic inflammation. Ann. N.Y. Acad. Sci. 2012, 1253, 80–91. [Google Scholar]
  129. Kaltner, H.; Toegel, S.; Caballero, G.G.; Manning, J.C.; Ledeen, R.W.; Gabius, H.-J. Galectins: Their network and roles in immunity/tumor growth control. Histochem. Cell Biol. 2017, 147, 239–256. [Google Scholar]
  130. Demetriou, M.; Nabi, I.R.; Dennis, J.W. Galectins as adaptors: Linking glycosylation and metabolism with extracellular cues. Trends Glycosci. Glycotechnol. 2018, 30, 167–177. [Google Scholar]
  131. Kasai, K. Galectins: Quadruple-faced proteins. Trends Glycosci. Glycotechnol. 2018, 30, 221–223. [Google Scholar] [CrossRef]
  132. de Jong, C.; Gabius, H.-J.; Baron, W. The emerging role of galectins in (re)myelination and its potential for developing new approaches to treat multiple sclerosis. Cell. Mol. Life Sci. 2020, 77, 1289–1317. [Google Scholar] [PubMed]
  133. Caballero, G.; Kaltner, H.; Kutzner, T.J.; Ludwig, A.K.; Manning, J.C.; Schmidt, S.; Sinowatz, F.; Gabius, H.-J. How galectins have become multifunctional proteins. Histol. Histopathol. 2020, 35, 509–539. [Google Scholar]
  134. Timoshenko, A.V.; Gorudko, I.V.; Maslakova, O.V.; André, S.; Kuwabara, I.; Liu, F.-T.; Kaltner, H.; Gabius, H.-J. Analysis of selected blood and immune cell responses to carbohydrate-dependent surface binding of proto- and chimera-type galectins. Mol. Cell. Biochem. 2003, 250, 139–149. [Google Scholar] [PubMed]
  135. Hong, M.-H.; Weng, I.C.; Liu, F.-T. Galectins as intracellular regulators of cellular responses through the detection of damaged endocytic vesicles. Trends Glycosci. Glycotechnol. 2018, 30, 179–184. [Google Scholar]
  136. Sato, S. Cytosolic galectins and their release and roles as carbohydrate-binding proteins in host–pathogen interaction. Trends Glycosci. Glycotechnol. 2018, 30, 199–209. [Google Scholar]
  137. Leffler, H. Galectins structure and function—A synopsis. Results Probl. Cell Differ. 2001, 33, 57–83. [Google Scholar]
  138. Neri, D.; Bicknell, R. Tumour vascular targeting. Nat. Rev. Cancer 2005, 5, 436–446. [Google Scholar]
  139. Perillo, N.L.; Pace, K.E.; Seilhamer, J.J.; Baum, L.G. Apoptosis of T cells mediated by galectin- 1. Nature 1995, 378, 736–739. [Google Scholar] [CrossRef]
  140. Cherayil, B.J.; Weiner, S.J.; Pillai, S. The Mac-2 antigen is a galactose-specific lectin that binds IgE. J. Exp. Med. 1989, 170, 1959–1972. [Google Scholar] [CrossRef]
  141. Gil, C.D.; La, M.; Perretti, M.; Oliani, S.M. Interaction of human neutrophils with endothelial cells regulates the expression of endogenous proteins annexin 1, galectin-1 and galectin-3. Cell Biol. Int. 2006, 30, 338–344. [Google Scholar] [CrossRef] [PubMed]
  142. Karlsson, A.; Follin, P.; Leffler, H.; Dahlgren, C. Galectin-3 activates the NADPH-oxidase in exudated but not peripheral blood neutrophils. Blood 1998, 91, 3430–3438. [Google Scholar] [CrossRef] [PubMed]
  143. Zhu, L.L.; Zhao, X.Q.; Jiang, C.; You, Y.; Chen, X.P.; Jiang, Y.Y.; Jia, X.M.; Lin, X. C-type lectin receptors dectin-3 and dectin-2 form a heterodimeric pattern-recognition receptor for host defense against fungal infection. Immunity 2013, 39, 324–334. [Google Scholar] [CrossRef]
  144. Ostrop, J.; Lang, R. Contact, collaboration, and conflict: Signal integration of Syk-coupled C-type lectin receptors. J. Immunol. 2017, 198, 1403–1414. [Google Scholar] [CrossRef] [PubMed]
  145. Vértesy, S.; Michalak, M.; Miller, M.C.; Schnölzer, M.; André, S.; Kopitz, J.; Mayo, K.H.; Gabius, H.-J. Structural significance of galectin design: Impairment of homodimer stability by linker insertion and partial reversion by ligand presence. Prot. Eng. Des. Sel. 2015, 28, 199–210. [Google Scholar] [CrossRef] [PubMed]
  146. Zhang, S.; Moussodia, R.O.; Murzeau, C.; Sun, H.J.; Klein, M.L.; Vértesy, S.; André, S.; Roy, R.; Gabius, H.-J.; Percec, V. Dissecting molecular aspects of cell interactions using glycodendrimersomes with programmable glycan presentation and engineered human lectins. Angew. Chem. Int. Ed. 2015, 54, 4036–4040. [Google Scholar] [CrossRef] [PubMed]
  147. Papaspyridonos, M.; McNeill, E.; de Bono, J.P.; Smith, A.; Burnand, K.G.; Channon, K.M.; Greaves, D.R. Galectin-3 is an amplifier of inflammation in atherosclerotic plaque progression through macrophage activation and monocyte chemoattraction. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 433–440. [Google Scholar] [CrossRef]
  148. Masamune, A.; Satoh, M.; Hirabayashi, J.; Kasai, K.; Satoh, K.; Shimosegawa, T. Galectin-1 induces chemokine production and proliferation in pancreatic stellate cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2006, 290, 729–736. [Google Scholar] [CrossRef]
  149. Qian, D.; Lu, Z.; Xu, Q.; Wu, P.; Tian, L.; Zhao, L.; Cai, B.; Yin, J.; Wu, Y.; Staveley-O’Carroll, K.F.; et al. Galectin-1-driven upregulation of SDF-1 in pancreatic stellate cells promotes pancreatic cancer metastasis. Cancer Lett. 2017, 397, 43–51. [Google Scholar] [CrossRef]
  150. Filer, A.; Bik, M.; Parsonage, G.N.; Fitton, J.; Trebilcock, E.; Howlett, K.; Cook, M.; Raza, K.; Simmons, D.L.; Thomas, A.M.; et al. Galectin 3 induces a distinctive pattern of cytokine and chemokine production in rheumatoid synovial fibroblasts via selective signaling pathways. Arthritis Rheumatol. 2009, 60, 1604–1614. [Google Scholar] [CrossRef]
  151. Toegel, S.; Weinmann, D.; Andre, S.; Walzer, S.M.; Bilban, M.; Schmidt, S.; Chiari, C.; Windhager, R.; Krall, C.; Bennani-Baiti, I.M.; et al. Galectin-1 couples glycobiology to inflammation in osteoarthritis through the activation of an NF-κB-regulated gene network. J. Immunol. 2016, 196, 1910–1921. [Google Scholar] [CrossRef] [PubMed]
  152. Weinmann, D.; Kenn, M.; Schmidt, S.; Schmidt, K.; Walzer, S.M.; Kubista, B.; Windhager, R.; Schreiner, W.; Toegel, S.; Gabius, H.-J. Galectin-8 induces functional disease markers in human osteoarthritis and cooperates with galectins-1 and -3. Cell. Mol. Life Sci. 2018, 75, 4187–4205. [Google Scholar] [CrossRef] [PubMed]
  153. Chen, C.; Duckworth, C.A.; Fu, B.; Pritchard, D.M.; Rhodes, J.M.; Yu, L.G. Circulating galectins -2, -4 and -8 in cancer patients make important contributions to the increased circulation of several cytokines and chemokines that promote angiogenesis and metastasis. Br. J. Cancer 2014, 110, 741–752. [Google Scholar] [CrossRef]
  154. Cattaneo, V.; Tribulatti, M.V.; Carabelli, J.; Carestia, A.; Schattner, M.; Campetella, O. Galectin-8 elicits pro-inflammatory activities in the endothelium. Glycobiology 2014, 24, 966–973. [Google Scholar] [CrossRef] [PubMed]
  155. Carabelli, J.; Quattrocchi, V.; D’Antuono, A.; Zamorano, P.; Tribulatti, M.V.; Campetella, O. Galectin-8 activates dendritic cells and stimulates antigen-specific immune response elicitation. J. Leukoc. Biol. 2017, 102, 1237–1247. [Google Scholar] [CrossRef] [PubMed]
  156. Nesmelova, I.V.; Pang, M.; Baum, L.G.; Mayo, K.H. 1H, 13C, and 15N backbone and side-chain chemical shift assignments for the 29 kDa human galectin-1 protein dimer. Biomol. NMR Assign. 2008, 2, 203–205. [Google Scholar] [CrossRef]
  157. Ippel, H.; Miller, M.C.; Berbis, M.A.; Suylen, D.; André, S.; Hackeng, T.M.; Cañada, F.J.; Weber, C.; Gabius, H.-J.; Jiménez-Barbero, J.; et al. 1H, 13C, and 15N backbone and side-chain chemical shift assignments for the 36 proline-containing, full length 29 kDa human chimera-type galectin-3. Biomol. NMR Assign. 2015, 9, 59–63. [Google Scholar] [CrossRef]
  158. Crump, M.P.; Gong, J.H.; Loetscher, P.; Rajarathnam, K.; Amara, A.; Arenzana-Seisdedos, F.; Virelizier, J.L.; Baggiolini, M.; Sykes, B.D.; Clark-Lewis, I. Solution structure and basis for functional activity of stromal cell-derived factor-1; dissociation of CXCR4 activation from binding and inhibition of HIV-1. EMBO J. 1997, 16, 6996–7007. [Google Scholar] [CrossRef]
  159. Gozansky, E.K.; Louis, J.M.; Caffrey, M.; Clore, G.M. Mapping the binding of the N-terminal extracellular tail of the CXCR4 receptor to stromal cell-derived factor-1α. J. Mol. Biol. 2005, 345, 651–658. [Google Scholar] [CrossRef]
  160. Hsu, D.K.; Zuberi, R.I.; Liu, F.T. Biochemical and biophysical characterization of human recombinant IgE-binding protein, an S-type animal lectin. J. Biol. Chem. 1992, 267, 14167–14174. [Google Scholar] [CrossRef]
  161. Herrmann, J.; Turck, C.W.; Atchison, R.E.; Huflejt, M.E.; Poulter, L.; Gitt, M.A.; Burlingame, A.L.; Barondes, S.H.; Leffler, H. Primary structure of the soluble lactose binding lectin L-29 from rat and dog and interaction of its non-collagenous proline-, glycine-tyrosine-rich sequence with bacterial and tissue collagenase. J. Biol. Chem. 1993, 268, 26704–26711. [Google Scholar] [CrossRef] [PubMed]
  162. Ochieng, J.; Fridman, R.; Nangia-Makker, P.; Kleiner, D.E.; Liotta, L.A.; Stetler-Stevenson, W.G.; Raz, A. Galectin-3 is a novel substrate for human matrix metalloproteinases-2 and -9. Biochemistry 1994, 33, 14109–14114. [Google Scholar] [CrossRef] [PubMed]
  163. Rajarathnam, K.; Desai, U.R. Structural insights into how proteoglycans determine chemokine–CXCR1/CXCR2 interactions: Progress and challenges. Front. Immunol. 2020, 11, 660–671. [Google Scholar] [CrossRef] [PubMed]
  164. Talaga, M.L.; Fan, N.; Fueri, A.L.; Brown, R.K.; Bandyopadhyay, P.; Dam, T.K. Multitasking human lectin galectin-3 interacts with sulfated glycosaminoglycans and chondroitin sulfate proteoglycans. Biochemistry 2016, 55, 4541–4551. [Google Scholar] [CrossRef]
  165. Peranzoni, E.; Lemoine, J.; Vimeux, L.; Feuillet, V.; Barrin, S.; Kantari-Mimoun, C.; Bercovici, N.; Guerin, M.; Biton, J.; Ouakrim, H.; et al. Macrophages impede CD8 T cells from reaching tumor cells and limit the efficacy of anti-PD-1 treatment. Proc. Natl. Acad. Sci. USA 2018, 115, 4041–4050. [Google Scholar] [CrossRef]
  166. Gordon-Alonso, M.; Hirsch, T.; Wildmann, C.; van der Bruggen, P. Galectin-3 captures interferon-γ in the tumor matrix reducing chemokine gradient production and T-cell tumor infiltration. Nat. Commun. 2017, 8, 793–798. [Google Scholar] [CrossRef]
  167. von Hundelshausen, P.; Wichapong, K.; Gabius, H.-J.; Mayo, K.H. The marriage of chemokines and galectins as functional heterodimers. Cell. Mol. Life Sci. 2021, 78, 8073–8095. [Google Scholar] [CrossRef]
Figure 1. (A) CXC chemokine CXCL8 homodimer (Interleukin-8, PDB access code 1IL8) and (B) CC chemokine CCL5 homodimer (RANTES, PDB access code 5COY) are shown. C-terminal helices are colored yellow, and the remainder of the sequences are colored green. The illustrations of structures shown in this figure were made by Dr. Michelle C. Miller.
Figure 1. (A) CXC chemokine CXCL8 homodimer (Interleukin-8, PDB access code 1IL8) and (B) CC chemokine CCL5 homodimer (RANTES, PDB access code 5COY) are shown. C-terminal helices are colored yellow, and the remainder of the sequences are colored green. The illustrations of structures shown in this figure were made by Dr. Michelle C. Miller.
Ijms 24 14083 g001
Figure 2. Modeled structures of the CXCL4/CXCL8 heterodimer based on chemical shift perturbations in HSQC spectra and in silico molecular mechanics and dynamics simulations. The CXCL4 (platelet factor 4) homodimer is shown in blue (A), and the CXCL8 (interleukin-8) homodimer is shown in red (B), with the heterodimer formed from a monomer subunit from each shown in (C). Positively charged lysine and arginine residues are shown in dark blue as labeled, and negatively charged glutamate residues are shown in dark red as labeled. Electrostatic interactions among these residues in particular stabilize both homodimers and heterodimers. The illustrations of structures shown here were made by Dr. Irina Nesmelova.
Figure 2. Modeled structures of the CXCL4/CXCL8 heterodimer based on chemical shift perturbations in HSQC spectra and in silico molecular mechanics and dynamics simulations. The CXCL4 (platelet factor 4) homodimer is shown in blue (A), and the CXCL8 (interleukin-8) homodimer is shown in red (B), with the heterodimer formed from a monomer subunit from each shown in (C). Positively charged lysine and arginine residues are shown in dark blue as labeled, and negatively charged glutamate residues are shown in dark red as labeled. Electrostatic interactions among these residues in particular stabilize both homodimers and heterodimers. The illustrations of structures shown here were made by Dr. Irina Nesmelova.
Ijms 24 14083 g002
Figure 3. Galectin homodimers. Two different types of prototype galectin homodimers are illustrated. (A) Gal-1 homodimer (PDB lGZW). (B) Gal-7 homodimer (PDB lBKZ). The sugar binding S-face and opposing F-face are indicated in (B), and molecules of bound lactose are shown as ball-and-stick structures in both (A,B).
Figure 3. Galectin homodimers. Two different types of prototype galectin homodimers are illustrated. (A) Gal-1 homodimer (PDB lGZW). (B) Gal-7 homodimer (PDB lBKZ). The sugar binding S-face and opposing F-face are indicated in (B), and molecules of bound lactose are shown as ball-and-stick structures in both (A,B).
Ijms 24 14083 g003
Figure 4. Model of the Gal-3/Gal-7 heterodimer. Using HSQC-derived information, in silico studies were performed by manually aligning and docking key residues from the Gal-3 CRD with those from Gal-7 to form the heterodimer, followed by MD simulations and energy minimization to thermodynamically optimize interactions. This figure has been re-printed from Miller et al. [7] with permission of the publisher.
Figure 4. Model of the Gal-3/Gal-7 heterodimer. Using HSQC-derived information, in silico studies were performed by manually aligning and docking key residues from the Gal-3 CRD with those from Gal-7 to form the heterodimer, followed by MD simulations and energy minimization to thermodynamically optimize interactions. This figure has been re-printed from Miller et al. [7] with permission of the publisher.
Ijms 24 14083 g004
Figure 5. Modeled structure of the CXCL12 and Gal-3 CRD heterodimer based on NMR chemical shift and intensity changes from HSQC experiments with CXCL12 and Gal-3 CRD. Analyses of the HSQC spectra were used to manually dock the two species and energy minimization the complex using MD simulations as discussed in [6]. This modeled structure was produced by Dr. Kanin Wichapong.
Figure 5. Modeled structure of the CXCL12 and Gal-3 CRD heterodimer based on NMR chemical shift and intensity changes from HSQC experiments with CXCL12 and Gal-3 CRD. Analyses of the HSQC spectra were used to manually dock the two species and energy minimization the complex using MD simulations as discussed in [6]. This modeled structure was produced by Dr. Kanin Wichapong.
Ijms 24 14083 g005
Table 1. Some heterodimers with functional consequences.
Table 1. Some heterodimers with functional consequences.
HeterodimerFunctional EffectsReferences
CXCL4–CXCL8Enhance EC proliferation, Baf3 cell migration[3]
CCL3/4, CCL2/8Activate CCR5 intra-cellular signaling[1]
CXCL4–CCL5Modulate GPCR-mediated signal transduction[4,5,6]
CXCL4L1-MIFRegulate inflammation and thrombus formation[55]
Gal-7-Gal-1 or Gal-3Increase EC and leukocyte cell surface binding[7]
Enhance hemagglutination and leukocyte apoptosis
Attenuate EC growth and migration
Gal-3-CXCL12Inhibit CXCR3-expressing lymphocyte migration[9,166]
Enhance upregulation of CXCL9, L10, L11
Inhibit CXCL12-induced T-cell, neutrophil migration
Gal-3-CCL26Inhibit CCL26-induced eosinophil migration[9]
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

Mayo, K.H. Heterologous Interactions with Galectins and Chemokines and Their Functional Consequences. Int. J. Mol. Sci. 2023, 24, 14083. https://doi.org/10.3390/ijms241814083

AMA Style

Mayo KH. Heterologous Interactions with Galectins and Chemokines and Their Functional Consequences. International Journal of Molecular Sciences. 2023; 24(18):14083. https://doi.org/10.3390/ijms241814083

Chicago/Turabian Style

Mayo, Kevin H. 2023. "Heterologous Interactions with Galectins and Chemokines and Their Functional Consequences" International Journal of Molecular Sciences 24, no. 18: 14083. https://doi.org/10.3390/ijms241814083

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