Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
J Immunol ; 211(5): 755-766, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37417746

ABSTRACT

Signal-transducing adaptor protein-2 (STAP-2) is an adaptor protein that contains pleckstrin and Src homology 2-like domains, as well as a proline-rich region in its C-terminal region. Our previous study demonstrated that STAP-2 positively regulates TCR signaling by associating with TCR-proximal CD3ζ ITAMs and the lymphocyte-specific protein tyrosine kinase. In this study, we identify the STAP-2 interacting regions of CD3ζ ITAMs and show that the STAP-2-derived synthetic peptide (iSP2) directly interacts with the ITAM sequence and blocks the interactions between STAP-2 and CD3ζ ITAMs. Cell-penetrating iSP2 was delivered into human and murine T cells. iSP2 suppressed cell proliferation and TCR-induced IL-2 production. Importantly, iSP2 treatment suppressed TCR-mediated activation of naive CD4+ T cells and decreased immune responses in CD4+ T cell-mediated experimental autoimmune encephalomyelitis. It is likely that iSP2 is a novel immunomodulatory tool that modulates STAP-2-mediated activation of TCR signaling and represses the progression of autoimmune diseases.


Subject(s)
Adaptor Proteins, Signal Transducing , Signal Transduction , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Immunity , Receptors, Antigen, T-Cell/metabolism , Peptide Fragments/pharmacology
2.
J Biol Chem ; 299(1): 102724, 2023 01.
Article in English | MEDLINE | ID: mdl-36410436

ABSTRACT

Signal-transducing adaptor family member-2 (STAP-2) is an adaptor protein that regulates various intracellular signals. We previously demonstrated that STAP-2 binds to epidermal growth factor receptor (EGFR) and facilitates its stability and activation of EGFR signaling in prostate cancer cells. Inhibition of this interaction may be a promising direction for cancer treatment. Here, we found that 2D5 peptide, a STAP-2-derived peptide, blocked STAP-2-EGFR interactions and suppressed EGFR-mediated proliferation in several cancer cell lines. 2D5 peptide inhibited tumor growth of human prostate cancer cell line DU145 and human lung cancer cell line A549 in murine xenograft models. Additionally, we determined that EGFR signaling and its stability were decreased by 2D5 peptide treatment during EGF stimulation. In conclusion, our study shows that 2D5 peptide is a novel anticancer peptide that inhibits STAP-2-mediated activation of EGFR signaling and suppresses prostate and lung cancer progression.


Subject(s)
Adaptor Proteins, Signal Transducing , Lung Neoplasms , Peptides , Prostatic Neoplasms , Animals , Humans , Male , Mice , Adaptor Proteins, Signal Transducing/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Lung Neoplasms/metabolism , Prostatic Neoplasms/metabolism , Signal Transduction , A549 Cells , Cell Line, Tumor , Peptides/pharmacology
3.
Biochem J ; 479(20): 2261-2278, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36305710

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is a carcinogenic virus that latently infects B cells and causes malignant tumors in immunocompromised patients. KSHV utilizes two viral E3 ubiquitin ligases, K3 and K5, in KSHV-infected cells to mediate the polyubiquitination-dependent down-regulation of several host membrane proteins involved in the immune system. Although K3 and K5 are members of the same family and have similar structural topologies, K3 and K5 have different substrate specificities. Hence, K5 may have a different substrate recognition mode than K3; however, the molecular basis of substrate recognition remains unclear. Here, we investigated the reason why human CD8α, which is known not to be a substrate for both K3 and K5, is not recognized by them, to obtain an understanding for molecular basis of substrate specificity. CD8α forms a disulfide-linked homodimer under experimental conditions to evaluate the viral ligase-mediated down-regulation. It is known that two interchain disulfide linkages in the stalk region between each CD8α monomer (Cys164-Cys164 and Cys181-Cys181) mediate homodimerization. When the interchain disulfide linkage of Cys181-Cys181 was eliminated, CD8α was down-regulated by K5 with a functional RING variant (RINGv) domain via polyubiquitination at the cytoplasmic tail. Aspartic acid, located at the stalk/transmembrane interface of CD8α, was essential for K5-mediated down-regulation of the CD8α mutant without a Cys181-Cys181 linkage. These results suggest that disulfide linkage near the stalk/transmembrane interface critically inhibits substrate targeting by K5. Accessibility to the extracellular juxtamembrane stalk region of membrane proteins may be important for substrate recognition by the viral ubiquitin ligase K5.


Subject(s)
Herpesvirus 8, Human , Immediate-Early Proteins , Humans , Ubiquitin/metabolism , Immediate-Early Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Membrane Proteins/metabolism , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Disulfides/metabolism
4.
Int J Mol Sci ; 24(1)2023 Jan 03.
Article in English | MEDLINE | ID: mdl-36614280

ABSTRACT

Iron-sulfur (Fe-S) clusters are essential cofactors for enzyme activity. These Fe-S clusters are present in structurally diverse forms, including [4Fe-4S] and [3Fe-4S]. Type-identification of the Fe-S cluster is indispensable in understanding the catalytic mechanism of enzymes. However, identifying [4Fe-4S] and [3Fe-4S] clusters in particular is challenging because of their rapid transformation in response to oxidation-reduction events. In this study, we focused on the relationship between the Fe-S cluster type and the catalytic activity of a tRNA-thiolation enzyme (TtuA). We reconstituted [4Fe-4S]-TtuA, prepared [3Fe-4S]-TtuA by oxidizing [4Fe-4S]-TtuA under strictly anaerobic conditions, and then observed changes in the Fe-S clusters in the samples and the enzymatic activity in the time-course experiments. Electron paramagnetic resonance analysis revealed that [3Fe-4S]-TtuA spontaneously transforms into [4Fe-4S]-TtuA in minutes to one hour without an additional free Fe source in the solution. Although the TtuA immediately after oxidation of [4Fe-4S]-TtuA was inactive [3Fe-4S]-TtuA, its activity recovered to a significant level compared to [4Fe-4S]-TtuA after one hour, corresponding to an increase of [4Fe-4S]-TtuA in the solution. Our findings reveal that [3Fe-4S]-TtuA is highly inactive and unstable. Moreover, time-course analysis of structural changes and activity under strictly anaerobic conditions further unraveled the Fe-S cluster type used by the tRNA-thiolation enzyme.


Subject(s)
Iron-Sulfur Proteins , Iron , Iron/chemistry , Electron Spin Resonance Spectroscopy , Oxidation-Reduction , Sulfur/chemistry , RNA, Transfer/chemistry , Iron-Sulfur Proteins/metabolism
5.
J Virol ; 94(17)2020 08 17.
Article in English | MEDLINE | ID: mdl-32581091

ABSTRACT

Measles virus (MeV) is a highly immunotropic and contagious pathogen that can even diminish preexisting antibodies and remains a major cause of childhood morbidity and mortality worldwide despite the availability of effective vaccines. MeV is one of the most extensively studied viruses with respect to the mechanisms of JAK-STAT antagonism. Of the three proteins translated from the MeV P gene, P and V are essential for inactivation of this pathway. However, the lack of data from direct analyses of the underlying interactions means that the detailed molecular mechanism of antagonism remains unresolved. Here, we prepared recombinant MeV V protein, which is responsible for human JAK-STAT antagonism, and a panel of variants, enabling the biophysical characterization of V protein, including direct V/STAT1 and V/STAT2 interaction assays. Unambiguous direct interactions between the host and viral factors, in the absence of other factors such as Jak1 or Tyk2, were observed, and the dissociation constants were quantified for the first time. Our data indicate that interactions between the C-terminal region of V and STAT2 is 1 order of magnitude stronger than that of the N-terminal region of V and STAT1. We also clarified that these interactions are completely independent of each other. Moreover, results of size exclusion chromatography demonstrated that addition of MeV-V displaces STAT2-core, a rigid region of STAT2 lacking the N- and C-terminal domains, from preformed complexes of STAT2-core/IRF-associated domain (IRF9). These results provide a novel model whereby MeV-V can not only inhibit the STAT2/IRF9 interaction but also disrupt preassembled interferon-stimulated gene factor 3.IMPORTANCE To evade host immunity, many pathogenic viruses inactivate host Janus kinase signal transducer and activator of transcription (STAT) signaling pathways using diverse strategies. Measles virus utilizes P and V proteins to counteract this signaling pathway. Data derived largely from cell-based assays have indicated several amino acid residues of P and V proteins as important. However, biophysical properties of V protein or its direct interaction with STAT molecules using purified proteins have not been studied. We have developed novel molecular tools enabling us to identify a novel molecular mechanism for immune evasion whereby V protein disrupts critical immune complexes, providing a clear strategy by which measles virus can suppress interferon-mediated antiviral gene expression.


Subject(s)
Interferon-Stimulated Gene Factor 3, gamma Subunit/chemistry , Measles virus/metabolism , Phosphoproteins/chemistry , STAT2 Transcription Factor/chemistry , Viral Proteins/chemistry , Binding Sites , Gene Expression , Humans , Immune Evasion , Immunity, Innate , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Janus Kinases/metabolism , Measles virus/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , Protein Domains , Protein Interaction Domains and Motifs , STAT1 Transcription Factor/chemistry , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Signal Transduction , Viral Proteins/genetics , Viral Proteins/metabolism , Zinc Fingers
6.
J Immunol ; 203(12): 3386-3394, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31694909

ABSTRACT

Human leukocyte Ig-like receptors (LILR) LILRB1 and LILRB2 are immune checkpoint receptors that regulate a wide range of physiological responses by binding to diverse ligands, including HLA-G. HLA-G is exclusively expressed in the placenta, some immunoregulatory cells, and tumors and has several unique isoforms. However, the recognition of HLA-G isoforms by LILRs is poorly understood. In this study, we characterized LILR binding to the ß2-microglobulin (ß2m)-free HLA-G1 isoform, which is synthesized by placental trophoblast cells and tends to dimerize and multimerize. The multimerized ß2m-free HLA-G1 dimer lacked detectable affinity for LILRB1, but bound strongly to LILRB2. We also determined the crystal structure of the LILRB1 and HLA-G1 complex, which adopted the typical structure of a classical HLA class I complex. LILRB1 exhibits flexible binding modes with the α3 domain, but maintains tight contacts with ß2m, thus accounting for ß2m-dependent binding. Notably, both LILRB1 and B2 are oriented at suitable angles to permit efficient signaling upon complex formation with HLA-G1 dimers. These structural and functional features of ligand recognition by LILRs provide novel insights into their important roles in the biological regulations.


Subject(s)
HLA-G Antigens/chemistry , Models, Molecular , Protein Conformation , Receptors, Immunologic/chemistry , Binding Sites , HLA-G Antigens/genetics , HLA-G Antigens/immunology , Humans , Ligands , Molecular Dynamics Simulation , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Protein Isoforms , Receptors, Immunologic/metabolism , Structure-Activity Relationship , beta 2-Microglobulin/chemistry , beta 2-Microglobulin/metabolism
7.
J Biol Chem ; 294(4): 1250-1256, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30504218

ABSTRACT

Venomous snakes have endogenous proteins that neutralize the toxicity of their venom components. We previously identified five small serum proteins (SSP-1-SSP-5) from a highly venomous snake belonging to the family Viperidae as inhibitors of various toxins from snake venom. The endogenous inhibitors belong to the prostate secretory protein of 94 amino acids (PSP94) family. SSP-2 interacts with triflin, which is a member of the cysteine-rich secretory protein (CRISP) family that blocks smooth muscle contraction. However, the structural basis for the interaction and the biological roles of these inhibitors are largely unknown. Here, we determined the crystal structure of the SSP-2-triflin complex at 2.3 Å resolution. A concave region centrally located in the N-terminal domain of triflin is fully occupied by the terminal ß-strands of SSP-2. SSP-2 does not bind tightly to the C-terminal cysteine-rich domain of triflin; this domain is thought to be responsible for its channel-blocker function. Instead, the cysteine-rich domain is tilted 7.7° upon binding to SSP-2, and the inhibitor appears to sterically hinder triflin binding to calcium channels. These results help explain how an endogenous inhibitor prevents the venomous protein from maintaining homeostasis in the host. Furthermore, this interaction also sheds light on the binding interface between the human homologues PSP94 and CRISP-3, which are up-regulated in prostate and ovarian cancers.


Subject(s)
Blood Proteins/chemistry , Blood Proteins/metabolism , Prostatic Secretory Proteins/metabolism , Snake Venoms/chemistry , Snake Venoms/metabolism , Viperidae/metabolism , Amino Acid Sequence , Animals , Calcium Channels/chemistry , Calcium Channels/metabolism , Crystallography, X-Ray , Models, Molecular , Prostatic Secretory Proteins/chemistry , Protein Conformation , Sequence Homology
8.
Biochem Biophys Res Commun ; 529(2): 507-512, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32703459

ABSTRACT

Lyssavirus P protein is a multifunctional protein that interacts with numerous host-cell proteins. The C-terminal domain (CTD) of P is important for inhibition of JAK-STAT signaling enabling the virus to evade host immunity. Several regions on the surface of rabies virus P are reported to interact with host factors. Among them, an extended, discrete hydrophobic patch of P CTD is notable. Although structures of P CTD of two strains of rabies virus, and of mokola virus have been solved, the structure of P CTD for Duvenhage virus, which is functionally divergent from these species for immune evasion function, is not known. Here, we analyze the structures of P CTD of Duvenhage and of a distinct rabies virus strain to gain further insight on the nature and potential function of the hydrophobic surface. Molecular contacts in crystals suggest that the hydrophobic patch is important to intermolecular interactions with other proteins, which differ between the lyssavirus species.


Subject(s)
Lyssavirus/chemistry , Rhabdoviridae Infections/virology , Viral Proteins/chemistry , Crystallography, X-Ray , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Protein Conformation , Protein Domains
9.
Protein Expr Purif ; 172: 105631, 2020 08.
Article in English | MEDLINE | ID: mdl-32213313

ABSTRACT

CD1d is a major histocompatibility complex (MHC) class I-like glycoprotein and binds to glycolipid antigens that are recognized by natural killer T (NKT) cells. To date, our understanding of the structural basis for glycolipid binding and receptor recognition of CD1d is still limited. Here, we established a preparation method for the ectodomain of human and mouse CD1d using a silkworm-baculovirus expression system. The co-expression of human and mouse CD1d and ß2-microglobulin (ß2m) in the silkworm-baculovirus system was successful, but the yield of human CD1d was low. A construct of human CD1d fused with ß2m via a flexible GS linker as a single polypeptide was prepared to improve protein yield. The production of this single-chained complex was higher (50 µg/larva) than that of the co-expression complex. Furthermore, differential scanning calorimetry revealed that the linker made the CD1d complex more stable and homogenous. These results suggest that the silkworm-baculovirus expression system is useful for structural and biophysical studies of CD1d in several aspects including low cost, easy handling, biohazard-free, rapid, and high yielding.


Subject(s)
Antigens, CD1d , Baculoviridae , Gene Expression , Animals , Antigens, CD1d/biosynthesis , Antigens, CD1d/chemistry , Antigens, CD1d/genetics , Antigens, CD1d/isolation & purification , Bombyx , Humans , Mice , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
10.
Nucleic Acids Res ; 46(6): 3232-3244, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29471537

ABSTRACT

Archaea and eukaryotes have ribosomal P stalks composed of anchor protein P0 and aP1 homodimers (archaea) or P1•P2 heterodimers (eukaryotes). These P stalks recruit translational GTPases to the GTPase-associated center in ribosomes to provide energy during translation. The C-terminus of the P stalk is known to selectively recognize GTPases. Here we investigated the interaction between the P stalk and elongation factor 2 by determining the structures of Pyrococcus horikoshii EF-2 (PhoEF-2) in the Apo-form, GDP-form, GMPPCP-form (GTP-form), and GMPPCP-form bound with 11 C-terminal residues of P1 (P1C11). Helical structured P1C11 binds to a hydrophobic groove between domain G and subdomain G' of PhoEF-2, where is completely different from that of aEF-1α in terms of both position and sequence, implying that such interaction characteristic may be requested by how GTPases perform their functions on the ribosome. Combining PhoEF-2 P1-binding assays with a structural comparison of current PhoEF-2 structures and molecular dynamics model of a P1C11-bound GDP form, the conformational changes of the P1C11-binding groove in each form suggest that in response to the translation process, the groove has three states: closed, open, and release for recruiting and releasing GTPases.


Subject(s)
Archaeal Proteins/metabolism , Peptide Elongation Factor 2/metabolism , Pyrococcus horikoshii/metabolism , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Amino Acid Sequence , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Guanosine Diphosphate/chemistry , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Peptide Elongation Factor 2/chemistry , Peptide Elongation Factor 2/genetics , Protein Binding , Protein Conformation , Pyrococcus horikoshii/genetics , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Ribosomes/chemistry , Sequence Homology, Amino Acid
11.
J Infect Dis ; 217(11): 1740-1749, 2018 05 05.
Article in English | MEDLINE | ID: mdl-29529215

ABSTRACT

Rabies virus (RABV) is the causative agent of fatal neurological disease. Cellular attachment is the initial and essential step for viral infections. Although extensive studies have demonstrated that RABV uses various target cell molecules to mediate infection, no specific molecule has been identified as an attachment factor for RABV infection. Here we demonstrate that cellular heparan sulfate (HS) supports RABV adhesion and subsequent entry into target cells. Enzymatic removal of HS reduced cellular susceptibility to RABV infection, and heparin, a highly sulfated form of HS, blocked viral adhesion and infection. The direct binding between RABV glycoprotein and heparin was demonstrated, and this interaction was shown to require HS N- and 6-O-sulfation. We also revealed that basic amino acids in the ectodomain of RABV glycoprotein serve as major determinants for the RABV-HS interaction. Collectively, our study highlights a previously undescribed role of HS as an attachment factor for RABV infection.


Subject(s)
Heparan Sulfate Proteoglycans/metabolism , Rabies virus/pathogenicity , Rabies/pathology , A549 Cells , Cell Line , Cell Line, Tumor , Glycoproteins/metabolism , Humans , Rabies virus/metabolism , Virus Internalization
12.
J Biol Chem ; 292(51): 21128-21136, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29046357

ABSTRACT

Before entering host cells, herpes simplex virus-1 uses its envelope glycoprotein B to bind paired immunoglobulin-like type 2 receptor α (PILRα) on immune cells. PILRα belongs to the Siglec (sialic acid (SA)-binding immunoglobulin-like lectin)-like family, members of which bind SA. PILRα is the only Siglec member to recognize not only the sialylated O-linked sugar T antigen (sTn) but also its attached peptide region. We previously determined the crystal structure of PILRα complexed with the sTn-linked glycopeptide of glycoprotein B, revealing the simultaneous recognition of sTn and peptide by the receptor. However, the contribution of each glycopeptide component to PILRα binding was largely unclear. Here, we chemically synthesized glycopeptide derivatives and determined the thermodynamic parameters of their interaction with PILRα. We show that glycopeptides with different sugar units linking SA and peptides (i.e. "GlcNAc-type" and "deoxy-GlcNAc-type" glycopeptides) have lower affinity and more enthalpy-driven binding than the wild type (i.e. GalNAc-type glycopeptide). The crystal structures of PILRα complexed with these glycopeptides highlighted the importance of stereochemical positioning of the O4 atom of the sugar moiety. These results provide insights both for understanding the unique O-glycosylated peptide recognition by the PILRα and for the rational design of herpes simplex virus-1 entry inhibitors.


Subject(s)
Membrane Glycoproteins/metabolism , Models, Molecular , Peptide Fragments/metabolism , Receptors, Immunologic/metabolism , Viral Envelope Proteins/metabolism , Acetylgalactosamine/analogs & derivatives , Acetylgalactosamine/chemistry , Acetylgalactosamine/metabolism , Binding Sites , Calorimetry , Crystallography, X-Ray , Escherichia coli/metabolism , Humans , Kinetics , Ligands , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/genetics , Polysaccharides/chemical synthesis , Polysaccharides/chemistry , Polysaccharides/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics , Recombinant Proteins , Stereoisomerism , Thermodynamics , Viral Envelope Proteins/chemical synthesis , Viral Envelope Proteins/chemistry
13.
J Biol Chem ; 291(7): 3124-34, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26635368

ABSTRACT

Constitutional heterozygous loss-of-function mutations in the SPRED1 gene cause a phenotype known as Legius syndrome, which consists of symptoms of multiple café-au-lait macules, axillary freckling, learning disabilities, and macrocephaly. Legius syndrome resembles a mild neurofibromatosis type 1 (NF1) phenotype. It has been demonstrated that SPRED1 functions as a negative regulator of the Ras-ERK pathway and interacts with neurofibromin, the NF1 gene product. However, the molecular details of this interaction and the effects of the mutations identified in Legius syndrome and NF1 on this interaction have not yet been investigated. In this study, using a yeast two-hybrid system and an immunoprecipitation assay in HEK293 cells, we found that the SPRED1 EVH1 domain interacts with the N-terminal 16 amino acids and the C-terminal 20 amino acids of the GTPase-activating protein (GAP)-related domain (GRD) of neurofibromin, which form two crossing α-helix coils outside the GAP domain. These regions have been shown to be dispensable for GAP activity and are not present in p120(GAP). Several mutations in these N- and C-terminal regions of the GRD in NF1 patients and pathogenic missense mutations in the EVH1 domain of SPRED1 in Legius syndrome reduced the binding affinity between the EVH1 domain and the GRD. EVH1 domain mutations with reduced binding to the GRD also disrupted the ERK suppression activity of SPRED1. These data clearly demonstrate that SPRED1 inhibits the Ras-ERK pathway by recruiting neurofibromin to Ras through the EVH1-GRD interaction, and this study also provides molecular basis for the pathogenic mutations of NF1 and Legius syndrome.


Subject(s)
Cafe-au-Lait Spots/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Models, Molecular , Mutation, Missense , Neurofibromatosis 1/genetics , Neurofibromin 1/metabolism , Point Mutation , Adaptor Proteins, Signal Transducing , Amino Acid Transport System A , Cafe-au-Lait Spots/metabolism , Cafe-au-Lait Spots/physiopathology , Epidermal Growth Factor/metabolism , Female , Genes, Reporter , Genetic Association Studies , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Kinetics , MAP Kinase Signaling System , Male , Membrane Proteins/chemistry , Membrane Proteins/genetics , Neurofibromatosis 1/metabolism , Neurofibromatosis 1/physiopathology , Neurofibromin 1/chemistry , Neurofibromin 1/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins p21(ras)/agonists , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
14.
Proc Natl Acad Sci U S A ; 111(24): 8877-82, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24889612

ABSTRACT

Paired Ig-like type 2 receptor α (PILRα) recognizes a wide range of O-glycosylated mucin and related proteins to regulate broad immune responses. However, the molecular characteristics of these recognitions are largely unknown. Here we show that sialylated O-linked sugar T antigen (sTn) and its attached peptide region are both required for ligand recognition by PILRα. Furthermore, we determined the crystal structures of PILRα and its complex with an sTn and its attached peptide region. The structures show that PILRα exhibits large conformational change to recognize simultaneously both the sTn O-glycan and the compact peptide structure constrained by proline residues. Binding and functional assays support this binding mode. These findings provide significant insight into the binding motif and molecular mechanism (which is distinct from sugar-recognition receptors) by which O-glycosylated mucin proteins with sTn modifications are recognized in the immune system as well as during viral entry.


Subject(s)
Membrane Glycoproteins/chemistry , Mucins/chemistry , Peptides/chemistry , Polysaccharides/chemistry , Receptors, Immunologic/chemistry , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray , Glycosylation , HEK293 Cells , Humans , Immune System , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis , Protein Binding , Protein Conformation , Recombinant Fusion Proteins/chemistry , Sequence Homology, Amino Acid , Surface Plasmon Resonance
15.
Eur J Immunol ; 45(6): 1605-13, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25826155

ABSTRACT

Emerging evidence has revealed the pivotal roles of C-type lectin-like receptors (CTLRs) in the regulation of a wide range of immune responses. Human natural killer cell receptor-P1A (NKRP1A) is one of the CTLRs and recognizes another CTLR, lectin-like transcript 1 (LLT1) on target cells to control NK, NKT and Th17 cells. The structural basis for the NKRP1A-LLT1 interaction was limitedly understood. Here, we report the crystal structure of the ectodomain of LLT1. The plausible receptor-binding face of the C-type lectin-like domain is flat, and forms an extended ß-sheet. The residues of this face are relatively conserved with another CTLR, keratinocyte-associated C-type lectin, which binds to the CTLR member, NKp65. A LLT1-NKRP1A complex model, prepared using the crystal structures of LLT1 and the keratinocyte-associated C-type lectin-NKp65 complex, reasonably satisfies the charge consistency and the conformational complementarity to explain a previous mutagenesis study. Furthermore, crystal packing and analytical ultracentrifugation revealed dimer formation, which supports a complex model. Our results provide structural insights for understanding the binding modes and signal transduction mechanisms, which are likely to be conserved in the CTLR family, and for further rational drug design towards regulating the LLT1 function.


Subject(s)
Lectins, C-Type/chemistry , Models, Molecular , NK Cell Lectin-Like Receptor Subfamily B/chemistry , Protein Conformation , Protein Interaction Domains and Motifs , Receptors, Cell Surface/chemistry , Amino Acid Sequence , Binding Sites , Crystallization , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Ligands , Molecular Sequence Data , Mutation , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Protein Binding , Protein Multimerization , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Sequence Alignment
16.
Biol Pharm Bull ; 39(11): 1897-1902, 2016.
Article in English | MEDLINE | ID: mdl-27803463

ABSTRACT

Herpes simplex virus type 1 (HSV-1) is a causative agent for a variety of diseases. Although antiherpetic drugs such as acyclovir have been developed to inhibit virus replication through interaction with DNA kinases, their continuous administration leads to an increase in the frequency of drug-resistant HSV-1, which is an important clinical issue that requires urgent solution. Recently, we reported that the sialylated O-linked sugar T antigen (sTn) and its attached peptide region (O-glycosylated sTn peptide) derived from the HSV-1 glycoprotein B (gB) protein inhibited HSV-1 infection by specifically targeting paired immunoglobulin-like type 2 receptor alpha (PILRα) in vitro. In this study, to further identify novel inhibitors of gB-mediated HSV-1 infection in vitro, we established a cell-based fusion assay for rapid drug screening. Chinese hamster ovary (CHO) cells were transfected with expression plasmids for HSV-1 gB, gD, gH, and gL, and T7 RNA polymerase, and were designated as the effector cells. The CHO-K1 cells stably expressing PILRα were transfected with the expression plasmid for firefly luciferase under the T7 promoter, and were designated as the target cells. The effector and target cells were co-cultured, and luminescence was measured when both cells were successfully fused. Importantly, we found that cell-to-cell fusion was specifically inhibited by O-glycosylated sTn peptide in a dose dependent manner. Our results suggested that this virus-free cell-based fusion assay system could be a useful and promising approach to identify novel inhibitors of gB-mediated HSV-1 infection, and will aid in the development of antiviral therapeutic strategies for HSV-1-associated diseases.


Subject(s)
Antigens, Viral, Tumor/chemistry , Antiviral Agents/pharmacology , Herpesvirus 1, Human , Peptides/pharmacology , Viral Envelope Proteins/genetics , Animals , Biological Assay , CHO Cells , Cell Fusion , Coculture Techniques , Cricetinae , Cricetulus , DNA-Directed RNA Polymerases/genetics , Herpes Simplex/drug therapy , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/pathogenicity , Luciferases, Firefly/genetics , Membrane Glycoproteins/genetics , Receptors, Immunologic/genetics , Viral Proteins/genetics
17.
Proc Natl Acad Sci U S A ; 110(43): 17438-43, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24101491

ABSTRACT

Mincle [macrophage inducible Ca(2+)-dependent (C-type) lectin; CLEC4E] and MCL (macrophage C-type lectin; CLEC4D) are receptors for the cord factor TDM (trehalose-6,6'-dimycolate), a unique glycolipid of mycobacterial cell-surface components, and activate immune cells to confer adjuvant activity. Although it is known that receptor-TDM interactions require both sugar and lipid moieties of TDM, the mechanisms of glycolipid recognition by Mincle and MCL remain unclear. We here report the crystal structures of Mincle, MCL, and the Mincle-citric acid complex. The structures revealed that these receptors are capable of interacting with sugar in a Ca(2+)-dependent manner, as observed in other C-type lectins. However, Mincle and MCL uniquely possess shallow hydrophobic regions found adjacent to their putative sugar binding sites, which reasonably locate for recognition of fatty acid moieties of glycolipids. Functional studies using mutant receptors as well as glycolipid ligands support this deduced binding mode. These results give insight into the molecular mechanism of glycolipid recognition through C-type lectin receptors, which may provide clues to rational design for effective adjuvants.


Subject(s)
Cord Factors/chemistry , Lectins, C-Type/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Immunologic/chemistry , Amino Acid Sequence , Animals , Binding Sites/genetics , Calcium/chemistry , Calcium/metabolism , Citric Acid/chemistry , Citric Acid/metabolism , Cord Factors/metabolism , Crystallography, X-Ray , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Ligands , Mice , Molecular Sequence Data , Mutation , Protein Binding , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Sequence Homology, Amino Acid , Surface Plasmon Resonance
18.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 12): 3090-8, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25478828

ABSTRACT

eIF5B and eIF1A are two translation-initiation factors that are universally conserved among all kingdoms. They show a unique interaction in eukaryotes which is important for ribosomal subunit joining. Here, the structures of two isolated forms of yeast eIF5B and of the eIF5B-eIF1A complex (eIF1A and eIF5B do not contain the respective N-terminal domains) are reported. The eIF5B-eIF1A structure shows that the C-terminal tail of eIF1A binds to eIF5B domain IV, while the core domain of eIF1A is invisible in the electron-density map. Although the individual domains in all structures of eIF5B or archaeal IF5B (aIF5B) are similar, their domain arrangements are significantly different, indicating high structural flexibility, which is advantageous for conformational change during ribosomal subunit joining. Based on these structures, models of eIF5B, eIF1A and tRNAi(Met) on the 80S ribosome were built. The models suggest that the interaction between the eIF1A C-terminal tail and eIF5B helps tRNAi(Met) to bind to eIF5B domain IV, thus preventing tRNAi(Met) dissociation, stabilizing the interface for subunit joining and providing a checkpoint for correct ribosome assembly.


Subject(s)
Eukaryotic Initiation Factor-1/chemistry , Eukaryotic Initiation Factors/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Crystallography, X-Ray , Eukaryotic Initiation Factor-1/metabolism , Eukaryotic Initiation Factors/metabolism , Models, Molecular , Protein Conformation , Ribosomes/chemistry , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
19.
J Virol ; 87(4): 2253-63, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23236061

ABSTRACT

Pol283-8-specific, HLA-B*51:01-restricted, cytotoxic T cells (CTLs) play a critical role in the long-term control of HIV-1 infection. However, these CTLs select for the reverse transcriptase (RT) I135X escape mutation, which may be accumulating in circulating HIV-1 sequences. We investigated the selection of the I135X mutation by CTLs specific for the same epitope but restricted by HLA-B*52:01. We found that Pol283-8-specific, HLA-B*52:01-restricted CTLs were elicited predominantly in chronically HIV-1-infected individuals. These CTLs had a strong ability to suppress the replication of wild-type HIV-1, though this ability was weaker than that of HLA-B*51:01-restricted CTLs. The crystal structure of the HLA-B*52:01-Pol283-8 peptide complex provided clear evidence that HLA-B*52:01 presents the peptide similarly to HLA-B*51:01, ensuring the cross-presentation of this epitope by both alleles. Population level analyses revealed a strong association of HLA-B*51:01 with the I135T mutant and a relatively weaker association of HLA-B*52:01 with several I135X mutants in both Japanese and predominantly Caucasian cohorts. An in vitro viral suppression assay revealed that the HLA-B*52:01-restricted CTLs failed to suppress the replication of the I135X mutant viruses, indicating the selection of these mutants by the CTLs. These results suggest that the different pattern of I135X mutant selection may have resulted from the difference between these two CTLs in the ability to suppress HIV-1 replication.


Subject(s)
Epitopes, T-Lymphocyte/immunology , HIV-1/immunology , HIV-1/pathogenicity , Immune Evasion , Selection, Genetic , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , Asian People , Epitopes, T-Lymphocyte/genetics , HIV Infections/immunology , HIV Infections/virology , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , HIV-1/genetics , HLA-B51 Antigen/immunology , HLA-B51 Antigen/metabolism , HLA-B52 Antigen/immunology , HLA-B52 Antigen/metabolism , Humans , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Protein Binding , White People
20.
Biosci Biotechnol Biochem ; 78(4): 680-6, 2014.
Article in English | MEDLINE | ID: mdl-25036967

ABSTRACT

The avirulence gene AVR-Pia of Magnaporthe oryzae, which induces a hypersensitive reaction in rice cultivars containing the resistance gene Pia, was expressed in Escherichia coli. AVR-Pia protein was collected as inclusion bodies, denatured, and refolded. Finally, recombinant AVR-Pia (rAVR-Pia) protein was purified by column chromatography. Infiltration of rAVR-Pia triggered cell browning in the leaves of rice cultivar Aichiasahi (Pia), with accumulation of H2O2 and induction of PR1a expression in rice. On the other hand, these reactions were not observed in Shin-2 (pia) leaves after the same treatment. This observation indicated that rAVR-Pia had the function of an avirulence protein. rAVR-Pia was used for immunization of a rabbit, and anti-AVR-Pia antiserum was prepared. The specificity of this antibody was appraised by detecting native AVR-Pia in the inoculated leaf sheath extract using Western blotting in combination with immunoprecipitation. Native AVR-Pia was successfully detected, and its molecular weight was estimated to be 7.4 kDa, indicating signal peptide cleavage. Additionally, secreted native AVR-Pia was quantified as 3.7 ng/g rice sheath.


Subject(s)
Fungal Proteins/biosynthesis , Fungal Proteins/isolation & purification , Genetic Engineering , Magnaporthe/genetics , Animals , Blotting, Western , Escherichia coli/genetics , Fungal Proteins/immunology , Fungal Proteins/metabolism , Immunoassay , Protein Refolding , Rabbits , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL