Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Mol Cell ; 80(4): 607-620.e12, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33113344

ABSTRACT

Aberrant mitophagy has been implicated in a broad spectrum of disorders. PINK1, Parkin, and ubiquitin have pivotal roles in priming mitophagy. However, the entire regulatory landscape and the precise control mechanisms of mitophagy remain to be elucidated. Here, we uncover fundamental mitophagy regulation involving PINK1 and a non-canonical role of the mitochondrial Tu translation elongation factor (TUFm). The mitochondrion-cytosol dual-localized TUFm interacts with PINK1 biochemically and genetically, which is an evolutionarily conserved Parkin-independent route toward mitophagy. A PINK1-dependent TUFm phosphoswitch at Ser222 determines conversion from activating to suppressing mitophagy. PINK1 modulates differential translocation of TUFm because p-S222-TUFm is restricted predominantly to the cytosol, where it inhibits mitophagy by impeding Atg5-Atg12 formation. The self-antagonizing feature of PINK1/TUFm is critical for the robustness of mitophagy regulation, achieved by the unique kinetic parameters of p-S222-TUFm, p-S65-ubiquitin, and their common kinase PINK1. Our findings provide new mechanistic insights into mitophagy and mitophagy-associated disorders.


Subject(s)
Drosophila melanogaster/growth & development , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Mitophagy , Peptide Elongation Factor Tu/metabolism , Protein Kinases/metabolism , Animals , Cytosol/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , HeLa Cells , Humans , Male , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Peptide Elongation Factor Tu/genetics , Phosphorylation , Protein Interaction Domains and Motifs , Protein Kinases/genetics , Protein Transport , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
2.
Proc Natl Acad Sci U S A ; 112(12): 3710-5, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25751889

ABSTRACT

Bone morphogenetic proteins (BMPs) belong to the TGF-ß family, whose 33 members regulate multiple aspects of morphogenesis. TGF-ß family members are secreted as procomplexes containing a small growth factor dimer associated with two larger prodomains. As isolated procomplexes, some members are latent, whereas most are active; what determines these differences is unknown. Here, studies on pro-BMP structures and binding to receptors lead to insights into mechanisms that regulate latency in the TGF-ß family and into the functions of their highly divergent prodomains. The observed open-armed, nonlatent conformation of pro-BMP9 and pro-BMP7 contrasts with the cross-armed, latent conformation of pro-TGF-ß1. Despite markedly different arm orientations in pro-BMP and pro-TGF-ß, the arm domain of the prodomain can similarly associate with the growth factor, whereas prodomain elements N- and C-terminal to the arm associate differently with the growth factor and may compete with one another to regulate latency and stepwise displacement by type I and II receptors. Sequence conservation suggests that pro-BMP9 can adopt both cross-armed and open-armed conformations. We propose that interactors in the matrix stabilize a cross-armed pro-BMP conformation and regulate transition between cross-armed, latent and open-armed, nonlatent pro-BMP conformations.


Subject(s)
Bone Morphogenetic Protein 7/chemistry , Growth Differentiation Factors/chemistry , Transforming Growth Factor beta/chemistry , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Growth Differentiation Factor 2 , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Signal Transduction
3.
Biochem Biophys Res Commun ; 489(3): 353-359, 2017 07 29.
Article in English | MEDLINE | ID: mdl-28526409

ABSTRACT

Platelet Derived Growth Factor receptors (PDGFRs), members of receptor tyrosine kinase superfamily, play essential roles in early hematopoiesis, angiogenesis and organ development. Dysregulation of PDGF receptor signaling under pathological conditions associates with cancers, vascular diseases, and fibrotic diseases. Therefore, they are attractive targets in drug development. Like any other membrane proteins with a single-pass transmembrane domain, the high-resolution structural information of the full-length PDGF receptors is still not resolved. It is caused, at least in part, by the technical challenges in the expression and purification of the functional, full-length PDGF receptors. Herein, we reported our experimental details in expression and purification of the full-length PDGFRß from mammalian cells. We found that purified PDGFRß remained in two different oligomeric states, presumably the monomer and the dimer, with basal kinase activity in detergent micelles. Addition of PDGF-B promoted dimerization and elevated kinase activity of the receptor, suggesting that purified receptors were functional.


Subject(s)
Receptor, Platelet-Derived Growth Factor beta/isolation & purification , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Cells, Cultured , Cricetinae , Gene Expression , HEK293 Cells , Humans , Receptor, Platelet-Derived Growth Factor beta/genetics
4.
Proc Natl Acad Sci U S A ; 109(12): 4586-91, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-22393018

ABSTRACT

Integrin α(X)ß(2) functions as complement receptor for iC3b and mediates recognition and phagocytosis of pathogens. We used negative-stain EM to examine the α(X)ß(2) interaction with iC3b. EM class averages of α(X)ß(2) in complex with iC3b define the binding sites on both the integrin and iC3b. iC3b contains C3c and thioester domain moieties linked by a long flexible linker. The binding site is on the key ring of the C3c moiety, at the interface between the MG3 and MG4 domains. Similar complexes are seen between α(X)ß(2) and the C3c fragment. α(X)ß(2) binds through the α(X) αI domain, on the face known to bear the metal ion-dependent adhesion site, at the opposite end of the αI domain from its site of insertion in the ß-propeller domain.


Subject(s)
Complement C3b/chemistry , Integrin alphaXbeta2/metabolism , Animals , Binding Sites , CHO Cells , Cell Adhesion , Chromatography/methods , Complement C3/chemistry , Complement System Proteins , Cricetinae , Humans , Ions , Ligands , Microscopy, Electron/methods , Models, Biological , Phagocytosis , Protein Structure, Tertiary
5.
Proc Natl Acad Sci U S A ; 109(20): 7817-22, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22547819

ABSTRACT

Circumsporozoite (CS) protein is the major surface component of Plasmodium falciparum sporozoites and is essential for host cell invasion. A vaccine containing tandem repeats, region III, and thrombospondin type-I repeat (TSR) of CS is efficacious in phase III trials but gives only a 35% reduction in severe malaria in the first year postimmunization. We solved crystal structures showing that region III and TSR fold into a single unit, an "αTSR" domain. The αTSR domain possesses a hydrophobic pocket and core, missing in TSR domains. CS binds heparin, but αTSR does not. Interestingly, polymorphic T-cell epitopes map to specialized αTSR regions. The N and C termini are unexpectedly close, providing clues for sporozoite sheath organization. Elucidation of a unique structure of a domain within CS enables rational design of next-generation subunit vaccines and functional and medicinal chemical investigation of the conserved hydrophobic pocket.


Subject(s)
Malaria Vaccines/chemistry , Malaria, Falciparum/prevention & control , Models, Molecular , Plasmodium falciparum , Protein Folding , Protozoan Proteins/chemistry , Sporozoites/chemistry , Amino Acid Sequence , Chromatography, High Pressure Liquid , Crystallography , HEK293 Cells , Humans , Mass Spectrometry , Molecular Sequence Data , Protozoan Proteins/genetics , Scattering, Small Angle , Sequence Alignment
6.
J Biol Chem ; 287(45): 38244-53, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-22988250

ABSTRACT

We study a mechanism by which dimerization of the EGF receptor (EGFR) cytoplasmic domain is transmitted to the ectodomain. Therapeutic and other small molecule antagonists to the kinase domain that stabilize its active conformation, but not those that stabilize an inactive conformation, stabilize ectodomain dimerization. Inhibitor-induced dimerization requires an asymmetric kinase domain interface associated with activation. EGF and kinase inhibitors stimulate formation of identical dimer interfaces in the EGFR transmembrane domain, as shown by disulfide cross-linking. Disulfide cross-linking at an interface in domain IV in the ectodomain was also stimulated similarly; however, EGF but not inhibitors stimulated cross-linking in domain II. Inhibitors similarly induced noncovalent dimerization in nearly full-length, detergent-solubilized EGFR as shown by gel filtration. EGFR ectodomain deletion resulted in spontaneous dimerization, whereas deletion of exons 2-7, in which extracellular domains III and IV are retained, did not. In EM, kinase inhibitor-induced dimers lacked any well defined orientation between the ectodomain monomers. Fab of the therapeutic antibody cetuximab to domain III confirmed a variable position and orientation of this domain in inhibitor-induced dimers but suggested that the C termini of domain IV of the two monomers were in close proximity, consistent with dimerization in the transmembrane domains. The results provide insights into the relative energetics of intracellular and extracellular dimerization in EGFR and have significance for physiologic dimerization through the asymmetric kinase interface, bidirectional signal transmission in EGFR, and mechanism of action of therapeutics.


Subject(s)
ErbB Receptors/chemistry , Phosphotransferases/chemistry , Protein Multimerization , Protein Structure, Tertiary , Animals , Binding Sites/genetics , Blotting, Western , Cell Line , Cross-Linking Reagents/chemistry , Disulfides/chemistry , Epidermal Growth Factor/pharmacology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gefitinib , Humans , Microscopy, Electron , Mutation , Phosphotransferases/antagonists & inhibitors , Phosphotransferases/metabolism , Protein Conformation/drug effects , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology
7.
Front Immunol ; 14: 1259386, 2023.
Article in English | MEDLINE | ID: mdl-38149245

ABSTRACT

Introduction: Since the outbreak of SARS-CoV-2, vaccines have demonstrated their effectiveness in resisting virus infection, reducing severity, and lowering the mortality rate in infected individuals. However, due to the rapid and ongoing mutations of SARS-CoV-2, the protective ability of many available vaccines has been challenged. Therefore, there is an urgent need for vaccines capable of eliciting potent broadly neutralizing antibodies against various SARS-CoV-2 variants. Methods: In this study, we developed a novel subunit vaccine candidate for SARS-CoV-2 by introducing a series of shielding glycans to the Fc-fused receptor-binding domain (RBD) of the prototypic spike protein. This approach aims to mask non-neutralizing epitopes and focus the immune response on crucial neutralizing epitopes. Results: All modified sites were confirmed to be highly glycosylated through mass spectrometry analysis. The binding affinity of the glycan-shielded RBD (gsRBD) to the human ACE2 receptor was comparable to that of the wildtype RBD (wtRBD). Immunizing mice with gsRBD when combined with either Freund's adjuvant or aluminum adjuvant demonstrated that the introduction of the glycan shield did not compromise the antibody-inducing ability of RBD. Importantly, the gsRBD significantly enhanced the generation of neutralizing antibodies against SARS-CoV-2 pseudoviruses compared to the wtRBD. Notably, it exhibited remarkable protective activity against Beta (B.1.351), Delta (B.1.617.2), and Omicron (B.1.1.529), approximately 3-fold, 7- fold, and 17-fold higher than wtRBD, respectively. Discussion: Our data proved this multiple-epitope masking strategy as an effective approach for highly active vaccine production.


Subject(s)
Antibodies, Neutralizing , COVID-19 , Humans , Animals , Mice , SARS-CoV-2/genetics , COVID-19 Vaccines , COVID-19/prevention & control , Epitopes , Polysaccharides
8.
Biochemistry ; 51(44): 8814-28, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23106217

ABSTRACT

Many questions about the significance of structural features of integrin α(V)ß(3) with respect to its mechanism of activation remain. We have determined and re-refined crystal structures of the α(V)ß(3) ectodomain linked to C-terminal coiled coils (α(V)ß(3)-AB) and four transmembrane (TM) residues in each subunit (α(V)ß(3)-1TM), respectively. The α(V) and ß(3) subunits with four and eight extracellular domains, respectively, are bent at knees between the integrin headpiece and lower legs, and the headpiece has the closed, low-affinity conformation. The structures differ in the occupancy of three metal-binding sites in the ßI domain. Occupancy appears to be related to the pH of crystallization, rather than to the physiologic regulation of ligand binding at the central, metal ion-dependent adhesion site. No electron density was observed for TM residues and much of the α(V) linker. α(V)ß(3)-AB and α(V)ß(3)-1TM demonstrate flexibility in the linker between their extracellular and TM domains, rather than the previously proposed rigid linkage. A previously postulated interface between the α(V) and ß(3) subunits at their knees was also not supported, because it lacks high-quality density, required rebuilding in α(V)ß(3)-1TM, and differed markedly between α(V)ß(3)-1TM and α(V)ß(3)-AB. Together with the variation in domain-domain orientation within their bent ectodomains between α(V)ß(3)-AB and α(V)ß(3)-1TM, these findings are compatible with the requirement for large structural changes, such as extension at the knees and headpiece opening, in conveying activation signals between the extracellular ligand-binding site and the cytoplasm.


Subject(s)
Integrin alphaVbeta3/chemistry , Protein Structure, Tertiary , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Cricetinae , Crystallization , Crystallography, X-Ray , HEK293 Cells , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation
9.
Nanoscale ; 14(19): 7110-7122, 2022 May 19.
Article in English | MEDLINE | ID: mdl-35535618

ABSTRACT

The increasing demand for convenient, miniaturized and multifunctional antibodies necessitates the development of novel antigen-recognition molecules for biological and medical studies. Nanobodies, the functional variable regions of camelid heavy-chain-only antibodies, as a new tool, complement the conventional antibodies and are in the stage of rapid development. The outstanding advantages of nanobodies include a stable structure, easy production, excellent water solubility, high affinity toward antigens and low immunogenicity. With promising application potential, nanobodies are now increasingly applied to various studies, including protein structure analysis, microscopic imaging, medical diagnosis, and drug development. The approval of the first nanobody drug Caplacizumab by the FDA disclosed the therapeutic potential of nanobodies. The outbreak of COVID-19 accelerated the development of nanobody drugs in non-injectable and bispecific biotherapeutic applications. Herein, we reviewed recent studies on the nanobody structure, screening and their applications in protein structure analysis and nanobody drugs, especially on non-injectable nanobody and bispecific nanobody development.


Subject(s)
COVID-19 Drug Treatment , Single-Domain Antibodies , Antibodies , Antigens , Diagnostic Imaging , Humans , Single-Domain Antibodies/chemistry
10.
J Mol Biol ; 434(16): 167709, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35777468

ABSTRACT

As a member of PDGF/VEGF (Platelet-derived growth factor/ Vascular endothelial growth factor) growth factors, PDGF-D regulates blood vessel development, wound healing, innate immunity, and organogenesis. Unlike PDGF-A and PDGF-B, PDGF-D has an additional CUB (Complement C1r/C1s, Uegf, Bmp1) domain at the N-terminus of its growth factor domain, and thus it is secreted in a latent, inactive complex, which needs to be proteolytically activated for its biological activities. However, how the CUB domain contributes to the latency and activation of the growth factor remains elusive. In this study, we modeled the dimeric structure of PDGF-D pro-complex and studied the inhibitory functions of PDGF-D prodomain on PDGF-B and PDGF-D signaling. In our model, the growth factor domain of PDGF-D forms a VEGF-D-like dimer through their ß1 and ß3 interactions. The hinge and CUB domains of PDGF-D bind at the opposite sides of the growth factor domain and exclude the PDGFR-ß (PDGF Receptor ß) D2 and D3 domains from recognizing the growth factor. In addition, we verified that PDGF-D prodomain could inhibit both PDGF-B and PDGF-D mediated PDGFR-ß transphosphorylation in a dose-dependent manner. However, PDGF-D prodomain could only inhibit the proliferation of NIH 3T3 cells stimulated by PDGF-D but not by PDGF-B, indicating its differential inhibitory activities toward PDGF-B and PDGF-D signaling.


Subject(s)
Lymphokines , Platelet-Derived Growth Factor , Receptor, Platelet-Derived Growth Factor beta , Animals , Cell Proliferation/drug effects , Humans , Lymphokines/chemistry , Lymphokines/metabolism , Lymphokines/pharmacology , Mice , NIH 3T3 Cells , Platelet-Derived Growth Factor/chemistry , Platelet-Derived Growth Factor/metabolism , Platelet-Derived Growth Factor/pharmacology , Protein Domains , Protein Multimerization , Receptor, Platelet-Derived Growth Factor beta/chemistry , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, Platelet-Derived Growth Factor beta/pharmacology , Signal Transduction , Vascular Endothelial Growth Factor D/chemistry
11.
Nature ; 438(7071): 1181-5, 2005 Dec 22.
Article in English | MEDLINE | ID: mdl-16372014

ABSTRACT

Chromodomains are modules implicated in the recognition of lysine-methylated histone tails and nucleic acids. CHD (for chromo-ATPase/helicase-DNA-binding) proteins regulate ATP-dependent nucleosome assembly and mobilization through their conserved double chromodomains and SWI2/SNF2 helicase/ATPase domain. The Drosophila CHD1 localizes to the interbands and puffs of the polytene chromosomes, which are classic sites of transcriptional activity. Other CHD isoforms (CHD3/4 or Mi-2) are important for nucleosome remodelling in histone deacetylase complexes. Deletion of chromodomains impairs nucleosome binding and remodelling by CHD proteins. Here we describe the structure of the tandem arrangement of the human CHD1 chromodomains, and its interactions with histone tails. Unlike HP1 and Polycomb proteins that use single chromodomains to bind to their respective methylated histone H3 tails, the two chromodomains of CHD1 cooperate to interact with one methylated H3 tail. We show that the human CHD1 double chromodomains target the lysine 4-methylated histone H3 tail (H3K4me), a hallmark of active chromatin. Methylammonium recognition involves two aromatic residues, not the three-residue aromatic cage used by chromodomains of HP1 and Polycomb proteins. Furthermore, unique inserts within chromodomain 1 of CHD1 block the expected site of H3 tail binding seen in HP1 and Polycomb, instead directing H3 binding to a groove at the inter-chromodomain junction.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Histones/chemistry , Histones/metabolism , Amino Acid Sequence , Animals , Chromatin/chemistry , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/chemistry , Crystallography, X-Ray , DNA Helicases , Drosophila Proteins/chemistry , Humans , Lysine/metabolism , Methylation , Models, Molecular , Molecular Sequence Data , Polycomb Repressive Complex 1 , Protein Structure, Tertiary , Structure-Activity Relationship
12.
Biochemistry ; 47(39): 10314-23, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18771282

ABSTRACT

Cellular signaling mediated by the epidermal growth factor receptor (EGFR or ErbB) family of receptor tyrosine kinases plays an important role in regulating normal and oncogenic cellular physiology. While structures of isolated EGFR extracellular domains and intracellular protein tyrosine kinase domains have suggested mechanisms for growth factor-mediated receptor dimerization and allosteric kinase domain activation, understanding how the transmembrane and juxtamembrane domains contribute to transmembrane signaling requires structural studies on intact receptor molecules. In this report, recombinant EGFR constructs containing the extracellular, transmembrane, juxtamembrane, and kinase domains are overexpressed and purified from human embryonic kidney 293 cell cultures. The oligomerization state, overall structure, and functional stability of the purified EGF-bound receptor are characterized in detergent micelles and phospholipid bilayers. In the presence of EGF, catalytically active EGFR dimers can be isolated by gel filtration in dodecyl maltoside. Visualization of the dimeric species by negative stain electron microscopy and single particle averaging reveals an overall structure of the extracellular domain that is similar to previously published crystal structures and is consistent with the C-termini of domain IV being juxtaposed against one another as they enter the transmembrane domain. Although detergent-soluble preparations of EGFR are stable as dimers in the presence of EGF, they exhibit differential functional stability in Triton X-100 versus dodecyl maltoside. Furthermore, the kinase activity can be significantly stabilized by reconstituting purified EGF-bound EGFR dimers in phospholipid nanodiscs or vesicles, suggesting that the environment around the hydrophobic transmembrane and amphipathic juxtamembrane domains is important for stabilizing the tyrosine kinase activity in vitro.


Subject(s)
ErbB Receptors/chemistry , ErbB Receptors/metabolism , Phospholipids/pharmacology , Cell Line , DNA/genetics , Detergents , Dimerization , Drug Stability , ErbB Receptors/genetics , Humans , Image Processing, Computer-Assisted , Kidney/embryology , Micelles , Microscopy, Electron , Nanostructures , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
13.
Sci Rep ; 7: 42580, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28198446

ABSTRACT

An unprecedented epidemic of Zika virus (ZIKV) infection had spread to South and Central America. ZIKV infection was recently confirmed by CDC (the Centers for Disease Control and Prevention) to cause neonatal microcephaly, which posed a significant public health emergency of international concern. No specific vaccines or drugs are currently available to fight ZIKV infection. ZIKV nonstructural protein 1 (NS1) plays an essential role in viral replication and immune evasion. We determined the crystal structure of ZIKV NS1172-352, which forms a head-to-head, symmetric dimer with a unique 14-stranded ß-ladder conserved among flaviviruses. The assembly of the ß-ladder dimer is concentration dependent. Strikingly, one pathogenic mutation T233A (NCBI accession no. KU527068), found in the brain tissue of infected fetus with neonatal microcephaly, is located at the dimer interface. Thr233, a unique residue found in ZIKV but not in other flaviviruses, organizes a central hydrogen bonding network at NS1 dimer interface. Mutation of Thr233 to Ala disrupts this elaborated interaction network, and destabilizes the NS1 dimeric assembly in vitro. In addition, our structural comparison of epitopes for protective antibody 22NS1, targeting West Nile Virus NS1, could potentially be valuable in understanding its anti-virus specificities and in the development of antibodies against ZIKV.


Subject(s)
Microcephaly/etiology , Mutation , Viral Nonstructural Proteins/genetics , Zika Virus Infection/complications , Zika Virus Infection/virology , Zika Virus/physiology , Amino Acid Sequence , Antibodies, Viral/immunology , Epitopes/immunology , Humans , Infant, Newborn , Microcephaly/diagnosis , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Stability , Structure-Activity Relationship , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/metabolism , Zika Virus Infection/immunology
14.
J Mol Biol ; 342(2): 585-92, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15327957

ABSTRACT

GM2-activator protein (GM2-AP) is a lipid transfer protein that has the ability to stimulate the enzymatic processing of gangliosides as well as T-cell activation through lipid presentation. Our previous X-ray crystallographic studies of GM2-AP have revealed a large lipid binding pocket as the central overall feature of the structure with non-protein electron density within this pocket suggesting bound lipid. To extend these studies, we present here the 2A crystal structure of GM2-AP complexed with platelet activating factor (PAF). PAF is a potent phosphoacylglycerol whose toxic patho-physiological effects can be inhibited by GM2-AP. The structure shows an ordered arrangement of two bound lipids and a fatty acid molecule. One PAF molecule binds in an extended conformation within the hydrophobic channel that has an open and closed conformation, and was seen to contain bound phospholipid in the low pH apo structure. The second molecule is submerged inside the pocket in a U-shaped conformation with its head group near the single polar residue S141. It was refined as lyso-PAF as it lacks electron density for the sn-2 acetate group. The alkyl chains of PAF interact through van der Waals' contacts, while the head groups bind in different environments with their phosphocholine moieties in contact with aromatic rings (Y137, F80). The structure has revealed further insights into the lipid binding properties of GM2-AP, suggesting an unexpected unique mode of lipid packaging that may explain the efficiency of GM2-AP in inhibiting the detrimental biological effects of PAF.


Subject(s)
Lipids/chemistry , Platelet Activating Factor/chemistry , Proteins/chemistry , Binding Sites , Crystallography, X-Ray , G(M2) Activator Protein , Lipid Metabolism , Models, Molecular , Platelet Activating Factor/metabolism , Protein Structure, Tertiary , Proteins/metabolism
15.
J Cell Biol ; 196(1): 131-46, 2012 Jan 09.
Article in English | MEDLINE | ID: mdl-22232704

ABSTRACT

The lymphocyte homing receptor integrin α(4)ß(7) is unusual for its ability to mediate both rolling and firm adhesion. α(4)ß(1) and α(4)ß(7) are targeted by therapeutics approved for multiple sclerosis and Crohn's disease. Here, we show by electron microscopy and crystallography how two therapeutic Fabs, a small molecule (RO0505376), and mucosal adhesion molecule-1 (MAdCAM-1) bind α(4)ß(7). A long binding groove at the α(4)-ß(7) interface for immunoglobulin superfamily domains differs in shape from integrin pockets that bind Arg-Gly-Asp motifs. RO0505376 mimics an Ile/Leu-Asp motif in α(4) ligands, and orients differently from Arg-Gly-Asp mimics. A novel auxiliary residue at the metal ion-dependent adhesion site in α(4)ß(7) is essential for binding to MAdCAM-1 in Mg(2+) yet swings away when RO0505376 binds. A novel intermediate conformation of the α(4)ß(7) headpiece binds MAdCAM-1 and supports rolling adhesion. Lack of induction of the open headpiece conformation by ligand binding enables rolling adhesion to persist until integrin activation is signaled.


Subject(s)
Cell Adhesion , Integrins/chemistry , Amino Acid Motifs , Crystallography, X-Ray , HEK293 Cells , Humans , Integrins/physiology , Models, Biological , Protein Structure, Tertiary
16.
Nat Struct Mol Biol ; 18(9): 984-9, 2011 Aug 07.
Article in English | MEDLINE | ID: mdl-21822280

ABSTRACT

To our knowledge, no structural study to date has characterized, in an intact receptor, the coupling of conformational change in extracellular domains through a single-pass transmembrane domain to conformational change in cytoplasmic domains. Here we examine such coupling, and its unexpected complexity, using nearly full-length epidermal growth factor receptor (EGFR) and negative-stain EM. The liganded, dimeric EGFR ectodomain can couple both to putatively active, asymmetrically associated kinase dimers and to putatively inactive, symmetrically associated kinase dimers and monomers. Inhibitors that stabilize the active or inactive conformation of the kinase active site, as well as mutations in the kinase dimer interface and a juxtamembrane phosphorylation site, shift the equilibrium among the three kinase association states. This coupling of one conformation of an activated receptor ectodomain to multiple kinase-domain arrangements reveals previously unanticipated complexity in transmembrane signaling and facilitates regulation of receptor function in the juxtamembrane and cytoplasmic environments.


Subject(s)
ErbB Receptors/chemistry , Catalytic Domain , Dimerization , ErbB Receptors/genetics , ErbB Receptors/ultrastructure , Humans , Ligands , Mutation , Protein Structure, Tertiary
17.
Mol Cell Biol ; 30(22): 5432-43, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20837704

ABSTRACT

The mechanisms by which signals are transmitted across the plasma membrane to regulate signaling are largely unknown for receptors with single-pass transmembrane domains such as the epidermal growth factor receptor (EGFR). A crystal structure of the extracellular domain of EGFR dimerized by epidermal growth factor (EGF) reveals the extended, rod-like domain IV and a small, hydrophobic domain IV interface compatible with flexibility. The crystal structure and disulfide cross-linking suggest that the 7-residue linker between the extracellular and transmembrane domains is flexible. Disulfide cross-linking of the transmembrane domain shows that EGF stimulates only moderate association in the first two α-helical turns, in contrast to association throughout the membrane over five α-helical turns in glycophorin A and integrin. Furthermore, systematic mutagenesis to leucine and phenylalanine suggests that no specific transmembrane interfaces are required for EGFR kinase activation. These results suggest that linkage between ligand-induced dimerization and tyrosine kinase activation is much looser than was previously envisioned.


Subject(s)
ErbB Receptors/chemistry , ErbB Receptors/metabolism , Ligands , Protein Structure, Quaternary , Protein Structure, Secondary , Signal Transduction/physiology , Amino Acid Sequence , Cell Line , Cross-Linking Reagents/chemistry , Cysteine/chemistry , Cysteine/genetics , Dimerization , Disulfides/chemistry , Enzyme Activation , ErbB Receptors/genetics , Humans , Models, Molecular , Molecular Sequence Data , Protein Binding , Sequence Alignment , X-Ray Diffraction
18.
Proc Natl Acad Sci U S A ; 104(39): 15358-63, 2007 Sep 25.
Article in English | MEDLINE | ID: mdl-17881562

ABSTRACT

The Ig superfamily (IgSF) intercellular adhesion molecule-1 (ICAM-1) equilibrates between monomeric and dimeric forms on the cell surface, and dimerization enhances cell adhesion. A crystal structure of ICAM-1 IgSF domains (D) 3-5 revealed a unique dimerization interface in which D4s of two protomers fuse through edge beta-strands to form a single super beta-sandwich domain. Here, we describe a crystal structure at 2.7-A resolution of monomeric ICAM-1 D3-D5, stabilized by the monomer-specific Fab CA7. CA7 binds to D5 in a region that is buried in the dimeric interface and is distal from the dimerization site in D4. In monomeric ICAM-1 D3-D5, a 16-residue loop in D4 that is disordered in the dimeric structure could clearly be traced as a BC loop, a short C strand, and a CE meander with a cis-Pro followed by a solvent-exposed, flexible four-residue region. Deletions of 6 or 10 residues showed that the C-strand is essential for monomer stability, whereas a distinct six-residue deletion showed little contribution of the CE meander. Mutation of two inward-pointing Leu residues in edge beta-strand E to Lys increased monomer stability, confirming the hypothesis that inward-pointing charged side chains on edge beta-strands are an important design feature to prevent beta-supersheet formation. Overall, the studies reveal that monomer-dimer transition is associated with a surprisingly large, physiologically relevant, IgSF domain rearrangement.


Subject(s)
Cell Membrane/metabolism , Immunoglobulin Subunits/chemistry , Intercellular Adhesion Molecule-1/chemistry , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray , Immunoglobulin Fab Fragments/chemistry , Molecular Conformation , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid
19.
Biochemistry ; 44(41): 13510-21, 2005 Oct 18.
Article in English | MEDLINE | ID: mdl-16216074

ABSTRACT

GM2-activator protein (GM2AP) is a lysosomal lipid transfer protein with important biological roles in ganglioside catabolism, phospholipid metabolism, and T-cell activation. Previous studies of crystal structures of GM2AP complexed with the physiological ligand GM2 and platelet activating factor (PAF) have shown binding at two specific locations within the spacious apolar pocket and an ordering effect of endogenous resident lipids. To investigate the structural basis of phospholipid binding further, GM2AP was cocrystallized with phosphatidylcholine (PC), known to interact with GM2AP. Analysis of three crystal forms revealed binding of single chain lipids and fatty acids only and surprisingly not intact PC. The regions of best defined electron density are consistent with the presence of lyso-PC and oleic acid, which constitute deacylation products of PC. Their acyl tails are in stacking contact with shorter, less well-defined stretches of electron density that may represent resident fatty acids. The GM2AP associated hydrolytic activity that generates lyso-PC was further confirmed by mass spectrometry and enzymatic assays. In addition, we report the structures of (i) mutant Y137S, assessing the role of Tyr137 in lipid transfer via the hydrophobic cleft, and (ii) apo-mouse GM2AP, revealing a hydrophobic pocket with a constricted opening. Our structural results provide new insights into the biological functions of GM2AP. The combined effect of hydrolytic and lipid transfer properties has profound implications in cellular signaling.


Subject(s)
G(M2) Activator Protein/chemistry , G(M2) Activator Protein/metabolism , Phosphatidylcholines/chemistry , Phospholipases A/metabolism , Animals , Binding Sites , Crystallization , G(M2) Activator Protein/genetics , Humans , Lysophosphatidylcholines/biosynthesis , Lysophosphatidylcholines/chemistry , Mice , Models, Structural , Phospholipases A/analysis , Tyrosine/chemistry , X-Ray Diffraction
20.
Mol Cell ; 11(4): 1093-100, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12718893

ABSTRACT

The nuclear receptor FXR is the sensor of physiological levels of enterohepatic bile acids, the end products of cholesterol catabolism. Here we report crystal structures of the FXR ligand binding domain in complex with coactivator peptide and two different bile acids. An unusual A/B ring juncture, a feature associated with bile acids and no other steroids, provides ligand discrimination and triggers a pi-cation switch that activates FXR. Helix 12, the activation function 2 of the receptor, adopts the agonist conformation and stabilizes coactivator peptide binding. FXR is able to interact simultaneously with two coactivator motifs, providing a mechanism for enhanced binding of coactivators through intermolecular contacts between their LXXLL sequences. These FXR complexes provide direct insights into the design of therapeutic bile acids for treatment of hyperlipidemia and cholestasis.


Subject(s)
Bile Acids and Salts/metabolism , DNA-Binding Proteins/chemistry , Hepatocytes/metabolism , Liver/metabolism , Transcription Factors/chemistry , Amino Acid Sequence/physiology , Animals , Bile Acids and Salts/agonists , Bile Acids and Salts/therapeutic use , Binding Sites/genetics , Cations/metabolism , Chemical Engineering , Humans , Hyperlipidemias/drug therapy , Ligands , Molecular Conformation , Molecular Sequence Data , Molecular Structure , Protein Structure, Tertiary/physiology , Receptors, Cytoplasmic and Nuclear
SELECTION OF CITATIONS
SEARCH DETAIL