Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Nat Struct Mol Biol ; 31(4): 591-597, 2024 Apr.
Article En | MEDLINE | ID: mdl-38287195

Cell-surface receptor complexes mediated by pro-inflammatory interleukin (IL)-12 and IL-23, both validated therapeutic targets, are incompletely understood due to the lack of structural insights into their complete extracellular assemblies. Furthermore, there is a paucity of structural details describing the IL-12-receptor interaction interfaces, in contrast to IL-23-receptor complexes. Here we report structures of fully assembled mouse IL-12/human IL-23-receptor complexes comprising the complete extracellular segments of the cognate receptors determined by electron cryo-microscopy. The structures reveal key commonalities but also surprisingly diverse features. Most notably, whereas IL-12 and IL-23 both utilize a conspicuously presented aromatic residue on their α-subunit as a hotspot to interact with the N-terminal Ig domain of their high-affinity receptors, only IL-12 juxtaposes receptor domains proximal to the cell membrane. Collectively, our findings will help to complete our understanding of cytokine-mediated assemblies of tall cytokine receptors and will enable a cytokine-specific interrogation of IL-12/IL-23 signaling in physiology and disease.


Interleukin-12 , Signal Transduction , Humans , Animals , Mice , Signal Transduction/physiology , Interleukin-23 , Cytokines/metabolism , Receptors, Cell Surface
2.
Cell Rep ; 41(3): 111490, 2022 10 18.
Article En | MEDLINE | ID: mdl-36261006

Interleukin-27 (IL-27) uniquely assembles p28 and EBI3 subunits to a heterodimeric cytokine that signals via IL-27Rα and gp130. To provide the structural framework for receptor activation by IL-27 and its emerging therapeutic targeting, we report here crystal structures of mouse IL-27 in complex with IL-27Rα and of human IL-27 in complex with SRF388, a monoclonal antibody undergoing clinical trials with oncology indications. One face of the helical p28 subunit interacts with EBI3, while the opposite face nestles into the interdomain elbow of IL-27Rα to juxtapose IL-27Rα to EBI3. This orients IL-27Rα for paired signaling with gp130, which only uses its immunoglobulin domain to bind to IL-27. Such a signaling complex is distinct from those mediated by IL-12 and IL-23. The SRF388 binding epitope on IL-27 overlaps with the IL-27Rα interaction site explaining its potent antagonistic properties. Collectively, our findings will facilitate the mechanistic interrogation, engineering, and therapeutic targeting of IL-27.


Interleukin-27 , Humans , Mice , Animals , Cytokine Receptor gp130/metabolism , Receptors, Cytokine/metabolism , Interleukin-12 , Cytokines , Antibodies, Monoclonal/pharmacology , Epitopes , Interleukin-23
3.
J Biol Chem ; 298(5): 101908, 2022 05.
Article En | MEDLINE | ID: mdl-35398099

Human Interleukin-18 (IL-18) is an omnipresent proinflammatory cytokine of the IL-1 family with central roles in autoimmune and inflammatory diseases and serves as a staple biomarker in the evaluation of inflammation in physiology and disease, including the inflammatory phase of COVID-19. The sequestration of IL-18 by its soluble decoy receptor IL-18-Binding Protein (IL-18BP) is critical to the regulation of IL-18 activity. Since an imbalance in expression and circulating levels of IL-18 is associated with disease, structural insights into how IL-18BP outcompetes binding of IL-18 by its cognate cell-surface receptors are highly desirable; however, the structure of human IL-18BP in complex with IL-18 has been elusive. Here, we elucidate the sequestration mechanism of human IL-18 mediated by IL-18BP based on the crystal structure of the IL-18:IL-18BP complex. These detailed structural snapshots reveal the interaction landscape leading to the ultra-high affinity of IL-18BP toward IL-18 and identify substantial differences with respect to previously characterized complexes of IL-18 with IL-18BP of viral origin. Furthermore, our structure captured a fortuitous higher-order assembly between IL-18 and IL-18BP coordinated by a disulfide-bond distal to the binding surface connecting IL-18 and IL-18BP molecules from different complexes, resulting in a novel tetramer with 2:2 stoichiometry. This tetrapartite assembly was found to restrain IL-18 activity more effectively than the canonical 1:1 complex. Collectively, our findings provide a framework for innovative, structure-driven therapeutic strategies and further functional interrogation of IL-18 in physiology and disease.


Intercellular Signaling Peptides and Proteins , Interleukin-18/metabolism , COVID-19/immunology , Humans , Inflammation , Neoplasms/immunology
4.
J Biol Chem ; 298(2): 101574, 2022 02.
Article En | MEDLINE | ID: mdl-35007536

The glucocorticoid (GC) receptor (GR) is essential for normal development and in the initiation of inflammation. Healthy GRdim/dim mice with reduced dimerization propensity due to a point mutation (A465T) at the dimer interface of the GR DNA-binding domain (DBD) (here GRD/D) have previously helped to define the functions of GR monomers and dimers. Since GRD/D retains residual dimerization capacity, here we generated the dimer-nullifying double mutant GRD+L/D+L mice, featuring an additional mutation (I634A) in the ligand-binding domain (LBD) of GR. These mice are perinatally lethal, as are GRL/L mice (these mice have the I634A mutation but not the A465T mutation), displaying improper lung and skin formation. Using embryonic fibroblasts, high and low doses of dexamethasone (Dex), nuclear translocation assays, RNAseq, dimerization assays, and ligand-binding assays (and Kd values), we found that the lethal phenotype in these mice is due to insufficient ligand binding. These data suggest there is some correlation between GR dimerization potential and ligand affinity. We conclude that even a mutation as subtle as I634A, at a position not directly involved in ligand interactions sensu stricto, can still influence ligand binding and have a lethal outcome.


Dexamethasone , Point Mutation , Receptors, Glucocorticoid , Animals , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Ligands , Mice , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
5.
Nature ; 600(7887): 143-147, 2021 12.
Article En | MEDLINE | ID: mdl-34646012

Anaplastic lymphoma kinase (ALK)1 and the related leukocyte tyrosine kinase (LTK)2 are recently deorphanized receptor tyrosine kinases3. Together with their activating cytokines, ALKAL1 and ALKAL24-6 (also called FAM150A and FAM150B or AUGß and AUGα, respectively), they are involved in neural development7, cancer7-9 and autoimmune diseases10. Furthermore, mammalian ALK recently emerged as a key regulator of energy expenditure and weight gain11, consistent with a metabolic role for Drosophila ALK12. Despite such functional pleiotropy and growing therapeutic relevance13,14, structural insights into ALK and LTK and their complexes with cognate cytokines have remained scarce. Here we show that the cytokine-binding segments of human ALK and LTK comprise a novel architectural chimera of a permuted TNF-like module that braces a glycine-rich subdomain featuring a hexagonal lattice of long polyglycine type II helices. The cognate cytokines ALKAL1 and ALKAL2 are monomeric three-helix bundles, yet their binding to ALK and LTK elicits similar dimeric assemblies with two-fold symmetry, that tent a single cytokine molecule proximal to the cell membrane. We show that the membrane-proximal EGF-like domain dictates the apparent cytokine preference of ALK. Assisted by these diverse structure-function findings, we propose a structural and mechanistic blueprint for complexes of ALK family receptors, and thereby extend the repertoire of ligand-mediated dimerization mechanisms adopted by receptor tyrosine kinases.


Anaplastic Lymphoma Kinase/chemistry , Anaplastic Lymphoma Kinase/metabolism , Cytokines/chemistry , Cytokines/metabolism , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Anaplastic Lymphoma Kinase/classification , Anaplastic Lymphoma Kinase/genetics , Binding Sites , Enzyme Activation , Epidermal Growth Factor/chemistry , Glycine , HEK293 Cells , Humans , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Mutation , Protein Binding , Protein Domains , Protein Multimerization , Substrate Specificity
6.
Nat Commun ; 12(1): 3050, 2021 05 24.
Article En | MEDLINE | ID: mdl-34031427

Clathrin-mediated endocytosis (CME) is the gatekeeper of the plasma membrane. In contrast to animals and yeasts, CME in plants depends on the TPLATE complex (TPC), an evolutionary ancient adaptor complex. However, the mechanistic contribution of the individual TPC subunits to plant CME remains elusive. In this study, we used a multidisciplinary approach to elucidate the structural and functional roles of the evolutionary conserved N-terminal Eps15 homology (EH) domains of the TPC subunit AtEH1/Pan1. By integrating high-resolution structural information obtained by X-ray crystallography and NMR spectroscopy with all-atom molecular dynamics simulations, we provide structural insight into the function of both EH domains. Both domains bind phosphatidic acid with a different strength, and only the second domain binds phosphatidylinositol 4,5-bisphosphate. Unbiased peptidome profiling by mass-spectrometry revealed that the first EH domain preferentially interacts with the double N-terminal NPF motif of a previously unidentified TPC interactor, the integral membrane protein Secretory Carrier Membrane Protein 5 (SCAMP5). Furthermore, we show that AtEH/Pan1 proteins control the internalization of SCAMP5 via this double NPF peptide interaction motif. Collectively, our structural and functional studies reveal distinct but complementary roles of the EH domains of AtEH/Pan1 in plant CME and connect the internalization of SCAMP5 to the TPLATE complex.


Adaptor Proteins, Signal Transducing/chemistry , Calcium-Binding Proteins/chemistry , Endocytosis , Plant Proteins/chemistry , Protein Binding , Adaptor Proteins, Signal Transducing/genetics , Arabidopsis Proteins , Calcium-Binding Proteins/genetics , Cell Membrane/metabolism , Crystallography, X-Ray , Membrane Proteins/chemistry , Molecular Dynamics Simulation , Plant Proteins/genetics , Plants, Genetically Modified , Protein Domains , Protein Transport , Sequence Alignment , Nicotiana/genetics
7.
Acta Crystallogr D Struct Biol ; 76(Pt 12): 1244-1255, 2020 Dec 01.
Article En | MEDLINE | ID: mdl-33263330

Structural studies of glycoproteins and their complexes provide critical insights into their roles in normal physiology and disease. Most glycoproteins contain N-linked glycosylation, a key post-translation modification that critically affects protein folding and stability and the binding kinetics underlying protein interactions. However, N-linked glycosylation is often an impediment to yielding homogeneous protein preparations for structure determination by X-ray crystallography or other methods. In particular, obtaining diffraction-quality crystals of such proteins and their complexes often requires modification of both the type of glycosylation patterns and their extent. Here, we demonstrate the benefits of producing target glycoproteins in the GlycoDelete human embryonic kidney 293 cell line that has been engineered to produce N-glycans as short glycan stumps comprising N-acetylglucosamine, galactose and sialic acid. Protein fragments of human Down syndrome cell-adhesion molecule and colony-stimulating factor 1 receptor were obtained from the GlycoDelete cell line for crystallization. The ensuing reduction in the extent and complexity of N-glycosylation in both protein molecules compared with alternative glycoengineering approaches enabled their productive deployment in structural studies by X-ray crystallography. Furthermore, a third successful implementation of the GlycoDelete technology focusing on murine IL-12B is shown to lead to N-glycosylation featuring an immature glycan in diffraction-quality crystals. It is proposed that the GlycoDelete cell line could serve as a valuable go-to option for the production of homogeneous glycoproteins and their complexes for structural studies by X-ray crystallography and cryo-electron microscopy.


Glycoproteins/chemistry , Polysaccharides/chemistry , Cryoelectron Microscopy/methods , Crystallography, X-Ray/methods , Glycosylation , HEK293 Cells , Humans , Protein Processing, Post-Translational
8.
Nature ; 568(7753): 571-575, 2019 04.
Article En | MEDLINE | ID: mdl-30944476

Across different kingdoms of life, ATP citrate lyase (ACLY, also known as ACL) catalyses the ATP-dependent and coenzyme A (CoA)-dependent conversion of citrate, a metabolic product of the Krebs cycle, to oxaloacetate and the high-energy biosynthetic precursor acetyl-CoA1. The latter fuels pivotal biochemical reactions such as the synthesis of fatty acids, cholesterol and acetylcholine2, and the acetylation of histones and proteins3,4. In autotrophic prokaryotes, ACLY is a hallmark enzyme of the reverse Krebs cycle (also known as the reductive tricarboxylic acid cycle), which fixates two molecules of carbon dioxide in acetyl-CoA5,6. In humans, ACLY links carbohydrate and lipid metabolism and is strongly expressed in liver and adipose tissue1 and in cholinergic neurons2,7. The structural basis of the function of ACLY remains unknown. Here we report high-resolution crystal structures of bacterial, archaeal and human ACLY, and use distinct substrate-bound states to link the conformational plasticity of ACLY to its multistep catalytic itinerary. Such detailed insights will provide the framework for targeting human ACLY in cancer8-11 and hyperlipidaemia12,13. Our structural studies also unmask a fundamental evolutionary relationship that links citrate synthase, the first enzyme of the oxidative Krebs cycle, to an ancestral tetrameric citryl-CoA lyase module that operates in the reverse Krebs cycle. This molecular transition marked a key step in the evolution of metabolism on Earth.


ATP Citrate (pro-S)-Lyase/chemistry , ATP Citrate (pro-S)-Lyase/metabolism , Citric Acid Cycle , Evolution, Molecular , ATP Citrate (pro-S)-Lyase/genetics , Biocatalysis , Chlorobium/enzymology , Chlorobium/genetics , Crystallography, X-Ray , Humans , Methanosarcinales/enzymology , Methanosarcinales/genetics , Models, Molecular
9.
Biochem Pharmacol ; 165: 240-248, 2019 07.
Article En | MEDLINE | ID: mdl-30885765

The four core members of the Interleukin-12 (IL-12) family of cytokines, IL-12, IL-23, IL-27 and IL-35 are heterodimers which share α- and ß-cytokine subunits. All four cytokines are immune modulators and have been proposed to play divergent roles in inflammatory arthritis. In recent years additional combinations of α- and ß-cytokine subunits belonging to the IL-12 family have been proposed to form novel cytokines such as IL-39. However, the actual extent of the combinatorial potential of the cytokine subunits in the human IL-12 family is not known. Here, we identify several combinations of subunits that form secreted heterodimeric assemblies based on a systematic orthogonal approach. The heterodimers are detected in the conditioned media harvested from mammalian cell cultures transfected with unfused pairs of cytokine subunits. While certain previously reported subunit combinations could not be recapitulated, our approach showed robustly that all four of the canonical members could be secreted. Furthermore, we provide evidence for the interaction between Cytokine Receptor Like Factor 1 (CRLF1) and Interleukin-12 subunit alpha (p35). Similar to IL-27 and IL-35 this novel heterodimer is not abundantly secreted rendering isolation from the conditioned medium very challenging, unlike IL-12 and IL-23. Our findings set the stage for fine-tuning approaches towards the biochemical reconstitution of IL-12 family cytokines for biochemical, cellular, and structural studies.


Interleukin-12/chemistry , Interleukin-23/biosynthesis , Interleukins/chemistry , Recombinant Fusion Proteins/chemistry , HEK293 Cells , Humans , Interleukin-12/biosynthesis , Interleukin-23/chemistry , Interleukins/biosynthesis , Protein Multimerization , Protein Subunits/biosynthesis , Protein Subunits/chemistry , Recombinant Fusion Proteins/biosynthesis
10.
Clin Immunol ; 206: 15-22, 2019 09.
Article En | MEDLINE | ID: mdl-30196070

Psoriatic arthritis (PsA) is a chronic inflammatory arthritis of unknown etiology, and currently the cellular and molecular interactions that dictate its pathogenesis remain elusive. A role of the interleukin-23 (IL-23)/IL-23R (IL-23 receptor) interaction in the development of psoriasis and PsA is well established. As IL-23 regulates the differentiation and activation of innate and adaptive immunity, it pertains to a very complex pathophysiology involving a plethora of effectors and transducers. In this review, we will discuss recent advances on the cellular and molecular pathophysiological mechanisms that regulate the initiation and progression of PsA as well as new therapeutic approaches for IL-23/IL-23R targeted therapeutics.


Arthritis, Psoriatic/drug therapy , Arthritis, Psoriatic/physiopathology , Dermatologic Agents/pharmacology , Interleukin-23/antagonists & inhibitors , Signal Transduction/drug effects , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Dermatologic Agents/therapeutic use , Humans , Ustekinumab/pharmacology , Ustekinumab/therapeutic use
11.
Nat Commun ; 9(1): 5340, 2018 12 17.
Article En | MEDLINE | ID: mdl-30559399

Activated invariant natural killer T (iNKT) cells rapidly produce large amounts of cytokines, but how cytokine mRNAs are induced, stabilized and mobilized following iNKT activation is still unclear. Here we show that an endoplasmic reticulum stress sensor, inositol-requiring enzyme 1α (IRE1α), links key cellular processes required for iNKT cell effector functions in specific iNKT subsets, in which TCR-dependent activation of IRE1α is associated with downstream activation of p38 MAPK and the stabilization of preformed cytokine mRNAs. Importantly, genetic deletion of IRE1α in iNKT cells reduces cytokine production and protects mice from oxazolone colitis. We therefore propose that an IRE1α-dependent signaling cascade couples constitutive cytokine mRNA expression to the rapid induction of cytokine secretion and effector functions in activated iNKT cells.


Cytokines/genetics , Endoplasmic Reticulum Stress/physiology , Endoribonucleases/genetics , Lymphocyte Activation/immunology , Natural Killer T-Cells/immunology , Protein Serine-Threonine Kinases/genetics , Animals , Cells, Cultured , Colitis/genetics , Gene Deletion , Mice , Mice, Knockout , Oxazolone/toxicity , RNA, Messenger/genetics , Signal Transduction , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Immunity ; 48(1): 45-58.e6, 2018 01 16.
Article En | MEDLINE | ID: mdl-29287995

Interleukin-23 (IL-23), an IL-12 family cytokine, plays pivotal roles in pro-inflammatory T helper 17 cell responses linked to autoimmune and inflammatory diseases. Despite intense therapeutic targeting, structural and mechanistic insights into receptor complexes mediated by IL-23, and by IL-12 family members in general, have remained elusive. We determined a crystal structure of human IL-23 in complex with its cognate receptor, IL-23R, and revealed that IL-23R bound to IL-23 exclusively via its N-terminal immunoglobulin domain. The structural and functional hotspot of this interaction partially restructured the helical IL-23p19 subunit of IL-23 and restrained its IL-12p40 subunit to cooperatively bind the shared receptor IL-12Rß1 with high affinity. Together with structural insights from the interaction of IL-23 with the inhibitory antibody briakinumab and by leveraging additional IL-23:antibody complexes, we propose a mechanistic paradigm for IL-23 and IL-12 whereby cognate receptor binding to the helical cytokine subunits primes recruitment of the shared receptors via the IL-12p40 subunit.


Interleukin-12 Receptor beta 1 Subunit/metabolism , Interleukin-23/metabolism , Receptors, Interleukin/metabolism , Animals , Calorimetry/methods , Cell Line , Humans , Interferometry/methods , Interleukin-12 Subunit p40/metabolism , Male , Mice , Protein Binding/physiology , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology
13.
Nat Commun ; 7: 13228, 2016 11 07.
Article En | MEDLINE | ID: mdl-27819269

Subversion of the host immune system by viruses is often mediated by molecular decoys that sequester host proteins pivotal to mounting effective immune responses. The widespread mammalian pathogen parapox Orf virus deploys GIF, a member of the poxvirus immune evasion superfamily, to antagonize GM-CSF (granulocyte macrophage colony-stimulating factor) and IL-2 (interleukin-2), two pleiotropic cytokines of the mammalian immune system. However, structural and mechanistic insights into the unprecedented functional duality of GIF have remained elusive. Here we reveal that GIF employs a dimeric binding platform that sequesters two copies of its target cytokines with high affinity and slow dissociation kinetics to yield distinct complexes featuring mutually exclusive interaction footprints. We illustrate how GIF serves as a competitive decoy receptor by leveraging binding hotspots underlying the cognate receptor interactions of GM-CSF and IL-2, without sharing any structural similarity with the cytokine receptors. Our findings contribute to the tracing of novel molecular mimicry mechanisms employed by pathogenic viruses.


Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interleukin-2/immunology , Parapoxvirus/immunology , Viral Proteins/immunology , Crystallography, X-Ray , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Interleukin-2/chemistry , Interleukin-2/metabolism , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/immunology , Multiprotein Complexes/metabolism , Parapoxvirus/metabolism , Poxviridae Infections/immunology , Poxviridae Infections/metabolism , Poxviridae Infections/virology , Protein Binding , Viral Proteins/chemistry , Viral Proteins/metabolism
14.
Nat Commun ; 5: 5237, 2014 Oct 30.
Article En | MEDLINE | ID: mdl-25354530

Protein scaffolds can provide a promising alternative to antibodies for various biomedical and biotechnological applications, including therapeutics. Here we describe the design and development of the Alphabody, a protein scaffold featuring a single-chain antiparallel triple-helix coiled-coil fold. We report affinity-matured Alphabodies with favourable physicochemical properties that can specifically neutralize human interleukin (IL)-23, a pivotal therapeutic target in autoimmune inflammatory diseases such as psoriasis and multiple sclerosis. The crystal structure of human IL-23 in complex with an affinity-matured Alphabody reveals how the variable interhelical groove of the scaffold uniquely targets a large epitope on the p19 subunit of IL-23 to harness fully the hydrophobic and hydrogen-bonding potential of tryptophan and tyrosine residues contributed by p19 and the Alphabody, respectively. Thus, Alphabodies are suitable for targeting protein-protein interfaces of therapeutic importance and can be tailored to interrogate desired design and binding-mode principles via efficient selection and affinity-maturation strategies.


Interleukin-23/antagonists & inhibitors , Peptides/chemistry , Amino Acid Sequence , Animals , Cell Line , Drug Evaluation, Preclinical , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Peptides/therapeutic use , Psoriasis/prevention & control
15.
Nat Struct Mol Biol ; 21(4): 375-82, 2014 Apr.
Article En | MEDLINE | ID: mdl-24632570

Thymic stromal lymphopoietin (TSLP), a cytokine produced by epithelial cells at barrier surfaces, is pivotal for the development of widespread chronic inflammatory disorders such as asthma and atopic dermatitis. The structure of the mouse TSLP-mediated signaling complex reveals how TSLP establishes extensive interfaces with its cognate receptor (TSLPR) and the shared interleukin 7 receptor α-chain (IL-7Rα) to evoke membrane-proximal receptor-receptor contacts poised for intracellular signaling. Binding of TSLP to TSLPR is a mechanistic prerequisite for recruitment of IL-7Rα to the high-affinity ternary complex, which we propose is coupled to a structural switch in TSLP at the crossroads of the cytokine-receptor interfaces. Functional interrogation of TSLP-receptor interfaces points to putative interaction hotspots that could be exploited for antagonist design. Finally, we derive the structural rationale for the functional duality of IL-7Rα and establish a consensus for the geometry of ternary complexes mediated by interleukin 2 (IL-2)-family cytokines.


Cytokines/chemistry , Signal Transduction/immunology , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Cytokines/immunology , Cytokines/metabolism , Humans , Interleukin-2/metabolism , Interleukin-2/physiology , Mice , Models, Immunological , Molecular Sequence Data , Protein Structure, Tertiary , Receptors, Interleukin-7/metabolism , Receptors, Interleukin-7/physiology , Thymic Stromal Lymphopoietin
16.
Biol Chem ; 395(4): 413-24, 2014 Apr.
Article En | MEDLINE | ID: mdl-24231122

OrysaEULD1A is one of the five EUL genes in rice (Oryza sativa) encoding a putative carbohydrate-binding protein belonging to the family of Euonymus related lectins (EUL). The OrysaEULD1A sequence comprises two highly similar EUL domains (91% sequence similarity and 72% sequence identity) separated by a 23 amino acid linker sequence and preceded by a 19 amino acid N-terminal sequence. In the present study, the full-length protein OrysaEULD1A as well as its individual domains OrysaEULD1A domain 1 and 2 were expressed in Pichia pastoris. After purification of the recombinant proteins, their carbohydrate-binding specificity was analyzed and compared. Interestingly, all recombinant lectins showed clear specificity towards galactosylated structures. Furthermore, all recombinant proteins agglutinated red blood cells, indicating that the full-length protein OrysaEULD1A and its domains are true lectins. These results taken together with data previously reported for single-domain EUL proteins indicate that although the amino acids--responsible for the formation of the carbohydrate-binding site--are identical for all EUL proteins in rice, these lectins show different carbohydrate specificities. This promiscuity of the carbohydrate-binding site can be attributed to gene divergence.


Oryza/genetics , Pichia/metabolism , Plant Lectins/chemistry , Plant Lectins/metabolism , Amino Acid Sequence , Molecular Sequence Data , Pichia/genetics , Plant Lectins/classification , Plant Lectins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Sequence Alignment
...