Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Sci Signal ; 12(580)2019 05 07.
Article in English | MEDLINE | ID: mdl-31064884

ABSTRACT

Forkhead-associated (FHA) domains are modules that bind to phosphothreonine (pThr) residues in signaling cascades. The FHA-containing mycobacterial protein GarA is a central element of a phosphorylation-dependent signaling pathway that redirects metabolic flux in response to amino acid starvation or cell growth requirements. GarA acts as a phosphorylation-dependent ON/OFF molecular switch. In its nonphosphorylated ON state, the GarA FHA domain engages in phosphorylation-independent interactions with various metabolic enzymes that orchestrate nitrogen flow, such as 2-oxoglutarate decarboxylase (KGD). However, phosphorylation at the GarA N-terminal region by the protein kinase PknB or PknG triggers autoinhibition through the intramolecular association of the N-terminal domain with the FHA domain, thus blocking all downstream interactions. To investigate these different FHA binding modes, we solved the crystal structures of the mycobacterial upstream (phosphorylation-dependent) complex PknB-GarA and the downstream (phosphorylation-independent) complex GarA-KGD. Our results show that the phosphorylated activation loop of PknB serves as a docking site to recruit GarA through canonical FHA-pThr interactions. However, the same GarA FHA-binding pocket targets an allosteric site on nonphosphorylated KGD, where a key element of recognition is a phosphomimetic aspartate. Further enzymatic and mutagenesis studies revealed that GarA acted as a dynamic allosteric inhibitor of KGD by preventing crucial motions in KGD that are necessary for catalysis. Our results provide evidence for physiological phosphomimetics, supporting numerous mutagenesis studies using such approaches, and illustrate how evolution can shape a single FHA-binding pocket to specifically interact with multiple phosphorylated and nonphosphorylated protein partners.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Forkhead Transcription Factors/metabolism , Mycobacterium tuberculosis/metabolism , Signal Transduction , Amino Acid Sequence , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites/genetics , Carboxy-Lyases/chemistry , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Crystallography, X-Ray , Models, Molecular , Mutation , Mycobacterium tuberculosis/genetics , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary
2.
PLoS One ; 12(8): e0183198, 2017.
Article in English | MEDLINE | ID: mdl-28817634

ABSTRACT

Malaria, a disease endemic in many tropical and subtropical regions, is caused by infection of the erythrocyte by the apicomplexan parasite Plasmodium. Host-cell invasion is a complex process but two Plasmodium proteins, Apical Membrane Antigen 1 (AMA1) and the Rhoptry Neck protein complex (RON), play a key role. AMA1, present on the surface of the parasite, binds tightly to the RON2 component of the RON protein complex, which is inserted into the erythrocyte membrane during invasion. Blocking the AMA1-RON2 interaction with antibodies or peptides inhibits invasion, underlining its importance in the Plasmodium life cycle and as a target for therapeutic strategies. We describe the crystal structure of the complex formed between AMA1 from P. vivax (PvAMA1) and a peptide derived from the externally exposed region of P. vivax RON2 (PvRON2sp1), and of the heterocomplex formed between P. falciparum AMA1 (PfAMA1) and PvRON2sp1. Binding studies show that the affinity of PvRON2sp1 for PvAMA1 is weaker than that previously reported for the PfRON2sp1-PfAMA1 association. Moreover, while PvRON2sp1 shows strong cross-reactivity with PfAMA1, PfRON2sp1 displays no detectable interaction with PvAMA1. The structures show that the equivalent residues PvRON2-Thr2055 and PfRON2-Arg2041 largely account for this pattern of reactivity.


Subject(s)
Cross Reactions , Plasmodium falciparum/immunology , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Animals , Ligands , Protein Binding , Protozoan Proteins/metabolism
3.
PLoS One ; 10(4): e0123567, 2015.
Article in English | MEDLINE | ID: mdl-25886591

ABSTRACT

The malaria parasite Plasmodium knowlesi, previously associated only with infection of macaques, is now known to infect humans as well and has become a significant public health problem in Southeast Asia. This species should therefore be targeted in vaccine and therapeutic strategies against human malaria. Apical Membrane Antigen 1 (AMA1), which plays a role in Plasmodium merozoite invasion of the erythrocyte, is currently being pursued in human vaccine trials against P. falciparum. Recent vaccine trials in macaques using the P. knowlesi orthologue PkAMA1 have shown that it protects against infection by this parasite species and thus should be developed for human vaccination as well. Here, we present the crystal structure of Domains 1 and 2 of the PkAMA1 ectodomain, and of its complex with the invasion-inhibitory monoclonal antibody R31C2. The Domain 2 (D2) loop, which is displaced upon binding the Rhoptry Neck Protein 2 (RON2) receptor, makes significant contacts with the antibody. R31C2 inhibits binding of the Rhoptry Neck Protein 2 (RON2) receptor by steric blocking of the hydrophobic groove and by preventing the displacement of the D2 loop which is essential for exposing the complete binding site on AMA1. R31C2 recognizes a non-polymorphic epitope and should thus be cross-strain reactive. PkAMA1 is much less polymorphic than the P. falciparum and P. vivax orthologues. Unlike these two latter species, there are no polymorphic sites close to the RON2-binding site of PkAMA1, suggesting that P. knowlesi has not developed a mechanism of immune escape from the host's humoral response to AMA1.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Protozoan/chemistry , Membrane Proteins/chemistry , Plasmodium knowlesi/immunology , Protozoan Proteins/chemistry , Amino Acid Sequence , Animals , Antigens, Protozoan/immunology , Base Sequence , Crystallography, X-Ray , Membrane Proteins/immunology , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protozoan Proteins/immunology
4.
PLoS One ; 10(4): e0124400, 2015.
Article in English | MEDLINE | ID: mdl-25881166

ABSTRACT

Infection with Plasmodium knowlesi, a zoonotic primate malaria, is a growing human health problem in Southeast Asia. P. knowlesi is being used in malaria vaccine studies, and a number of proteins are being considered as candidate malaria vaccine antigens, including the Apical Membrane Antigen 1 (AMA1). In order to determine genetic diversity of the ama1 gene and to identify epitopes of AMA1 under strongest immune selection, the ama1 gene of 52 P. knowlesi isolates derived from human infections was sequenced. Sequence analysis of isolates from two geographically isolated regions in Sarawak showed that polymorphism in the protein is low compared to that of AMA1 of the major human malaria parasites, P. falciparum and P. vivax. Although the number of haplotypes was 27, the frequency of mutations at the majority of the polymorphic positions was low, and only six positions had a variance frequency higher than 10%. Only two positions had more than one alternative amino acid. Interestingly, three of the high-frequency polymorphic sites correspond to invariant sites in PfAMA1 or PvAMA1. Statistically significant differences in the quantity of three of the six high frequency mutations were observed between the two regions. These analyses suggest that the pkama1 gene is not under balancing selection, as observed for pfama1 and pvama1, and that the PkAMA1 protein is not a primary target for protective humoral immune responses in their reservoir macaque hosts, unlike PfAMA1 and PvAMA1 in humans. The low level of polymorphism justifies the development of a single allele PkAMA1-based vaccine.


Subject(s)
Antigens, Protozoan/genetics , Haplotypes/genetics , Malaria/genetics , Membrane Proteins/genetics , Mutation/genetics , Plasmodium knowlesi/isolation & purification , Polymorphism, Genetic/genetics , Protozoan Proteins/genetics , Selection, Genetic/genetics , Amino Acid Sequence , Humans , Malaria/parasitology , Molecular Sequence Data , Sequence Analysis, DNA , Sequence Homology, Amino Acid
5.
PLoS Pathog ; 8(6): e1002755, 2012.
Article in English | MEDLINE | ID: mdl-22737069

ABSTRACT

Members of the phylum Apicomplexa, which include the malaria parasite Plasmodium, share many features in their invasion mechanism in spite of their diverse host cell specificities and life cycle characteristics. The formation of a moving junction (MJ) between the membranes of the invading apicomplexan parasite and the host cell is common to these intracellular pathogens. The MJ contains two key parasite components: the surface protein Apical Membrane Antigen 1 (AMA1) and its receptor, the Rhoptry Neck Protein (RON) complex, which is targeted to the host cell membrane during invasion. In particular, RON2, a transmembrane component of the RON complex, interacts directly with AMA1. Here, we report the crystal structure of AMA1 from Plasmodium falciparum in complex with a peptide derived from the extracellular region of PfRON2, highlighting clear specificities of the P. falciparum RON2-AMA1 interaction. The receptor-binding site of PfAMA1 comprises the hydrophobic groove and a region that becomes exposed by displacement of the flexible Domain II loop. Mutations of key contact residues of PfRON2 and PfAMA1 abrogate binding between the recombinant proteins. Although PfRON2 contacts some polymorphic residues, binding studies with PfAMA1 from different strains show that these have little effect on affinity. Moreover, we demonstrate that the PfRON2 peptide inhibits erythrocyte invasion by P. falciparum merozoites and that this strong inhibitory potency is not affected by AMA1 polymorphisms. In parallel, we have determined the crystal structure of PfAMA1 in complex with the invasion-inhibitory peptide R1 derived by phage display, revealing an unexpected structural mimicry of the PfRON2 peptide. These results identify the key residues governing the interactions between AMA1 and RON2 in P. falciparum and suggest novel approaches to antimalarial therapeutics.


Subject(s)
Antigens, Protozoan/chemistry , Host-Parasite Interactions/physiology , Membrane Proteins/chemistry , Plasmodium falciparum/chemistry , Protozoan Proteins/chemistry , Amino Acid Sequence , Animals , Antigens, Protozoan/metabolism , Cell Membrane/metabolism , Crystallization , Membrane Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Plasmodium falciparum/metabolism , Polymorphism, Genetic , Protein Binding , Protein Structure, Quaternary , Protozoan Proteins/metabolism , Surface Plasmon Resonance
6.
Glycobiology ; 21(12): 1570-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21610193

ABSTRACT

Carbohydrates are likely to maintain significant conformational flexibility in antibody (Ab):carbohydrate complexes. As demonstrated herein for the protective monoclonal Ab (mAb) F22-4 recognizing the Shigella flexneri 2a O-antigen (O-Ag) and numerous synthetic oligosaccharide fragments thereof, the combination of molecular dynamics simulations and nuclear magnetic resonance saturation transfer difference experiments, supported by physicochemical analysis, allows us to determine the binding epitope and its various contributions to affinity without using any modified oligosaccharides. Moreover, the methods used provide insights into ligand flexibility in the complex, thus enabling a better understanding of the Ab affinities observed for a representative set of synthetic O-Ag fragments. Additionally, these complementary pieces of information give evidence to the ability of the studied mAb to recognize internal as well as terminal epitopes of its cognate polysaccharide antigen. Hence, we show that an appropriate combination of computational and experimental methods provides a basis to explore carbohydrate functional mimicry and receptor binding. The strategy may facilitate the design of either ligands or carbohydrate recognition domains, according to needed improvements of the natural carbohydrate:receptor properties.


Subject(s)
Antibodies, Monoclonal/chemistry , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , Oligosaccharides/chemistry
7.
PLoS Pathog ; 7(2): e1001276, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21347343

ABSTRACT

Obligate intracellular Apicomplexa parasites share a unique invasion mechanism involving a tight interaction between the host cell and the parasite surfaces called the moving junction (MJ). The MJ, which is the anchoring structure for the invasion process, is formed by secretion of a macromolecular complex (RON2/4/5/8), derived from secretory organelles called rhoptries, into the host cell membrane. AMA1, a protein secreted from micronemes and associated with the parasite surface during invasion, has been shown in vitro to bind the MJ complex through a direct association with RON2. Here we show that RON2 is inserted as an integral membrane protein in the host cell and, using several interaction assays with native or recombinant proteins, we define the region that binds AMA1. Our studies were performed both in Toxoplasma gondii and Plasmodium falciparum and although AMA1 and RON2 proteins have diverged between Apicomplexa species, we show an intra-species conservation of their interaction. More importantly, invasion inhibition assays using recombinant proteins demonstrate that the RON2-AMA1 interaction is crucial for both T. gondii and P. falciparum entry into their host cells. This work provides the first evidence that AMA1 uses the rhoptry neck protein RON2 as a receptor to promote invasion by Apicomplexa parasites.


Subject(s)
Antigens, Protozoan/metabolism , Apicomplexa/physiology , Host-Parasite Interactions/physiology , Protozoan Proteins/metabolism , Virus Internalization , Animals , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Apicomplexa/genetics , Apicomplexa/metabolism , Cells, Cultured , Chlorocebus aethiops , Connexins/metabolism , Conserved Sequence , Host-Parasite Interactions/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/physiology , Models, Biological , Models, Molecular , Parasites/genetics , Parasites/metabolism , Parasites/physiology , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Plasmodium falciparum/physiology , Protein Binding/genetics , Protein Interaction Domains and Motifs/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Toxoplasma/genetics , Toxoplasma/metabolism , Toxoplasma/physiology , Vero Cells
8.
J Struct Biol ; 169(3): 360-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19857576

ABSTRACT

Ammodytoxin A (AtxA) and its natural isoform AtxC from the venom of Vipera ammodytes ammodytes belong to group IIA-secreted phospholipases A(2) which catalyze the hydrolysis of glycerophospholipids and exhibit strong neurotoxic and anticoagulant effects. The two isoforms, which differ in sequence by only two amino acid residues (Phe124>Ile and Lys128>Glu), display significant differences in toxicity and anticoagulant properties and act on protein targets including neurotoxic proteic receptors and coagulation factor Xa with significantly different strengths of binding. In order to characterize the structural basis of these functional differences, we have determined the crystal structures of the two isoforms. Comparison of the structures shows that the mutation Lys128>Glu in AtxC could perturb interactions with FXa, resulting in lower anticoagulant activity, since the side chain of Glu128 is partly buried, making a stabilizing hydrogen bond with the main-chain nitrogen atom of residue Thr35. This interaction leads to a displacement of the main polypeptide chain at positions 127 and 128 (identified by mutagenesis as important for interaction with FXa), and a different orientation of the side chain of unmutated Lys127. The mutation Phe124>Ile in AtxC induces no significant conformational changes, suggesting that the differences in toxicity of the two isoforms are due essentially to differences in surface complementarity in the interaction of the toxin with the neurotoxic protein receptor. The crystal structures also reveal a novel dimeric quaternary association involving significant hydrophobic interactions between the N-terminal alpha-helices of two molecules of ammodytoxin related by crystallographic symmetry. Interactions at the dimer interface include important contributions from Met7, which is unique to ammodytoxin. Equilibrium sedimentation experiments are consistent with the crystallographic model. Competition experiments using SPR technology show complete inhibition of AtxA binding to FXa by calmodulin (CaM). The crystal structure shows that the C-terminal region, important for binding to FXa and CaM, is fully exposed and accessible for interaction with proteic receptors in both the monomeric and dimeric forms of ammodytoxin described here.


Subject(s)
Anticoagulants/chemistry , Isoenzymes/chemistry , Neurotoxins/chemistry , Phospholipases A2/chemistry , Viper Venoms/chemistry , Viperidae/metabolism , Amino Acid Sequence , Animals , Anticoagulants/metabolism , Calmodulin/metabolism , Crystallography, X-Ray , Factor Xa/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Molecular Sequence Data , Neurotoxins/genetics , Neurotoxins/metabolism , Phospholipases A2/genetics , Phospholipases A2/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Secondary , Sequence Homology, Amino Acid , Surface Plasmon Resonance , Viper Venoms/genetics , Viper Venoms/metabolism
9.
J Mol Biol ; 388(4): 839-50, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19328810

ABSTRACT

The use of carbohydrate-mimicking peptides to induce immune responses against surface polysaccharides of pathogenic bacteria offers a novel approach to vaccine development. Factors governing antigenic and immunogenic mimicry, however, are complex and poorly understood. We have addressed this question using the anti-lipopolysaccharide monoclonal antibody F22-4, which was raised against Shigella flexneri serotype 2a and shown to protect against homologous infection in a mouse model. In a previous crystallographic study, we described F22-4 in complex with two synthetic fragments of the O-antigen, the serotype-specific saccharide moiety of lipopolysaccharide. Here, we present a crystallographic and NMR study of the interaction of F22-4 with a dodecapeptide selected by phage display using the monoclonal antibody. Like the synthetic decasaccharide, the peptide binds to F22-4 with micromolar affinity. Although the peptide and decasaccharide use very similar regions of the antigen-binding site, indicating good antigenic mimicry, immunogenic mimicry by the peptide was not observed. The F22-4-antigen interaction is significantly more hydrophobic with the peptide than with oligosaccharides; nonetheless, all hydrogen bonds formed between the peptide and F22-4 have equivalents in the oligosaccharide complex. Two bridging water molecules are also in common, adding to partial structural mimicry. Whereas the bound peptide is entirely helical, its structure in solution, as shown by NMR, is helical in the central region only. Moreover, docking the NMR structure into the antigen-binding site shows that steric hindrance would occur, revealing poor complementarity between the major solution conformation and the antibody that could contribute to the absence of immunogenic mimicry.


Subject(s)
Antibodies , Molecular Mimicry , O Antigens , Peptides , Shigella flexneri/immunology , Animals , Antibodies/chemistry , Antibodies/immunology , Carbohydrate Sequence , Crystallography, X-Ray , Mice , Molecular Conformation , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , O Antigens/chemistry , O Antigens/immunology , Peptides/chemistry , Peptides/immunology , Protein Conformation , Thermodynamics
10.
J Mol Biol ; 366(5): 1523-37, 2007 Mar 09.
Article in English | MEDLINE | ID: mdl-17229439

ABSTRACT

Apical membrane antigen 1 (AMA1) has an important, but as yet uncharacterised, role in host cell invasion by the malaria parasite, Plasmodium. The protein, which is quite conserved between Plasmodium species, comprises an ectoplasmic region, a single transmembrane segment and a small cytoplasmic domain. The ectoplasmic region, which can induce protective immunity in animal models of human malaria, is a leading vaccine candidate that has entered clinical trials. The monoclonal antibody F8.12.19, raised against the recombinant ectoplasmic region of AMA1 from Plasmodium vivax, cross-reacts with homologues from Plasmodium knowlesi, Plasmodium cynomolgi, Plasmodium berghei and Plasmodium falciparum, as shown by immunofluorescence assays on mature schizonts. The binding of F8.12.19 to recombinant AMA1 from both P. vivax and P. falciparum was measured by surface plasmon resonance, revealing an apparent affinity constant that is about 100-fold weaker for the cross-reacting antigen when compared to the cognate antigen. Crystal structure analysis of Fab F8.12.19 complexed to AMA1 from P. vivax and P. falciparum shows that the monoclonal antibody recognises a discontinuous epitope located on domain III of the ectoplasmic region, the major component being a loop containing a cystine knot. The structures provide a basis for understanding the cross-reactivity. Antibody contacts are made mainly to main-chain and invariant side-chain atoms of AMA1; contact antigen residues that differ in sequence are located at the periphery of the antigen-binding site and can be accommodated at the interface between the two components of the complex. The implications for AMA1 vaccine development are discussed.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Protozoan/immunology , Membrane Proteins/immunology , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Amino Acid Sequence , Animals , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Base Sequence , Cross Reactions , Crystallography, X-Ray , Cysteine/chemistry , Epitopes , Fluorescent Antibody Technique, Indirect , Hydrogen Bonding , Membrane Proteins/chemistry , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Plasmodium vivax/chemistry , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Sequence Homology, Amino Acid , Surface Plasmon Resonance
11.
J Immunol ; 176(3): 1686-94, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16424198

ABSTRACT

Protection against reinfection with noncapsulated Gram-negative bacteria, such as Shigella, an enteroinvasive bacterium responsible for bacillary dysentery, is mainly achieved by Abs specific for the O-Ag, the polysaccharide part of the LPS, the major bacterial surface Ag. The use of chemically defined glycoconjugates encompassing oligosaccharides mimicking the protective determinants carried by the O-Ag, thus expected to induce an efficient anti-LPS Ab response, has been considered an alternative to detoxified LPS-protein conjugate vaccines. The aim of this study was to identify such functional oligosaccharide mimics of the S. flexneri serotype 2a O-Ag. Using protective murine mAbs specific for S. flexneri serotype 2a and synthetic oligosaccharides designed to analyze the contribution of each sugar residue of the branched pentasaccharide repeating unit of the O-Ag, we demonstrated that the O-Ag exhibited an immunodominant serotype-specific determinant. We also showed that elongating the oligosaccharide sequence improved Ab recognition. From these antigenicity data, selected synthetic oligosaccharides were assessed for their potential to mimic the O-Ag by analyzing their immunogenicity in mice when coupled to tetanus toxoid via single point attachment. Our results demonstrated that induction of an efficient serotype 2a-specific anti-O-Ag Ab response was dependent on the length of the oligosaccharide sequence. A pentadecasaccharide representing three biological repeating units was identified as a potential candidate for further development of a chemically defined glycoconjugate vaccine against S. flexneri 2a infection.


Subject(s)
Dysentery, Bacillary/prevention & control , Glycoconjugates/immunology , Molecular Mimicry/immunology , O Antigens/chemistry , Oligosaccharides/chemistry , Shigella Vaccines/chemical synthesis , Shigella flexneri/classification , Amino Acid Sequence , Animals , Carbohydrate Sequence , Drug Design , Dysentery, Bacillary/immunology , Glycoconjugates/chemistry , Immunoglobulin G/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , O Antigens/immunology , Oligosaccharides/immunology , Serotyping , Shigella Vaccines/immunology , Shigella flexneri/chemistry , Shigella flexneri/immunology , Vaccines, Conjugate/chemistry , Vaccines, Conjugate/immunology
12.
Mol Biochem Parasitol ; 144(1): 55-67, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16154214

ABSTRACT

Apical membrane antigen 1 (AMA1) is a membrane protein present in Plasmodium species and is probably common to all apicomplexan parasites. The recent crystal structure of the complete ectoplasmic region of AMA1 from Plasmodium vivax has shown that it comprises three structural domains and that the first two domains are based on the PAN folding motif. Here, we discuss the consequences of this analysis for the three-dimensional structure of AMA1 from other Plasmodium species and other apicomplexan parasites, and for the Plasmodium paralogue MAEBL. Many polar and apolar interactions observed in the PvAMA1 crystal structure are made by residues that are invariant or highly conserved throughout all Plasmodium orthologues; a subgroup of these residues is also present in other apicomplexan orthologues and in MAEBL. These interactions presumably play a key role in defining the protein fold. Previous studies have shown that the ectoplasmic region of AMA1 must be cleaved from the parasite surface for host-cell invasion to proceed. The cleavage site in the crystal structure is not readily accessible to proteases and we discuss possible consequences of this observation. The three-dimensional distribution of polymorphic sites in PfAMA1 shows that these are all on the surface and that their positions are significantly biased to one side of the ectoplasmic region. Of particular note, a flexible segment in domain II, comprising about 40 residues and devoid of polymorphism, carries an epitope recognized by an invasion-inhibitory monoclonal antibody and a T-cell epitope implicated in the human immune response to AMA1.


Subject(s)
Antigens, Protozoan/chemistry , Antigens, Surface/chemistry , Membrane Proteins/chemistry , Plasmodium/chemistry , Protozoan Proteins/chemistry , Amino Acid Sequence , Animals , Epitopes, T-Lymphocyte , Models, Molecular , Molecular Sequence Data , Polymorphism, Genetic , Protein Structure, Secondary , Protein Structure, Tertiary/genetics , Sequence Alignment
13.
Science ; 308(5720): 408-11, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15731407

ABSTRACT

Apical membrane antigen 1 from Plasmodium is a leading malaria vaccine candidate. The protein is essential for host-cell invasion, but its molecular function is unknown. The crystal structure of the three domains comprising the ectoplasmic region of the antigen from P. vivax, solved at 1.8 angstrom resolution, shows that domains I and II belong to the PAN motif, which defines a superfamily of protein folds implicated in receptor binding. We also mapped the epitope of an invasion-inhibitory monoclonal antibody specific for the P. falciparum ortholog and modeled this to the structure. The location of the epitope and current knowledge on structure-function correlations for PAN domains together suggest a receptor-binding role during invasion in which domain II plays a critical part. These results are likely to aid vaccine and drug design.


Subject(s)
Antigens, Protozoan/chemistry , Membrane Proteins/chemistry , Plasmodium vivax/immunology , Protozoan Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antigens, Protozoan/immunology , Binding Sites , Crystallization , Crystallography, X-Ray , Epitope Mapping , Epitopes , Heparin/metabolism , Malaria Vaccines , Membrane Proteins/immunology , Models, Molecular , Molecular Sequence Data , Plasmodium falciparum/chemistry , Plasmodium falciparum/immunology , Plasmodium vivax/chemistry , Protein Conformation , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Protozoan Proteins/immunology , Recombinant Proteins/chemistry , Sequence Alignment
14.
Acta Crystallogr D Biol Crystallogr ; 60(Pt 11): 2040-3, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15502321

ABSTRACT

Apical membrane antigen 1 (AMA1), a type 1 transmembrane protein present in the microneme organelles of Plasmodium, is a leading malaria-vaccine candidate. The ectoplasmic region of AMA1 from P. vivax has been expressed in Pichia pastoris and crystallized in two different forms: an orthorhombic form (space group P2(1)2(1)2(1), unit-cell parameters a = 54.1, b = 76.1, c = 103.9 A) and a monoclinic form (space group C2, unit-cell parameters a = 150.0, b = 53.8, c = 60.3 A, beta = 113.2 degrees ). Native data have been collected to 2.0 A resolution for the orthorhombic form and 1.8 A for the monoclinic form. A platinum derivative was prepared for the orthorhombic and monoclinic crystals using K(2)PtCl(4) and data were collected at several wavelengths to obtain phases by the MAD technique. A partial model has been built from the electron-density maps of both forms and refinement is in progress.


Subject(s)
Antigens, Protozoan/chemistry , Antigens, Protozoan/metabolism , Malaria Vaccines , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Plasmodium vivax/chemistry , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Amino Acid Motifs , Animals , Antigens, Protozoan/immunology , Antigens, Protozoan/isolation & purification , Crystallization , Crystallography, X-Ray , Gene Expression , Genetic Vectors/genetics , Membrane Proteins/immunology , Membrane Proteins/isolation & purification , Protozoan Proteins/immunology , Protozoan Proteins/isolation & purification
15.
Protein Sci ; 12(3): 577-85, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12592028

ABSTRACT

Maltose-binding protein (MBP or MalE) of Escherichia coli is the periplasmic receptor of the maltose transport system. MalE31, a defective folding mutant of MalE carrying sequence changes Gly 32-->Asp and Ile 33-->Pro, is either degraded or forms inclusion bodies following its export to the periplasmic compartment. We have shown previously that overexpression of FkpA, a heat-shock periplasmic peptidyl-prolyl isomerase with chaperone activity, suppresses MalE31 misfolding. Here, we have exploited this property to characterize the maltose transport activity of MalE31 in whole cells. MalE31 displays defective transport behavior, even though it retains maltose-binding activity comparable with that of the wild-type protein. Because the mutated residues are in a region on the surface of MalE not identified previously as important for maltose transport, we have solved the crystal structure of MalE31 in the maltose-bound state in order to characterize the effects of these changes. The structure was determined by molecular replacement methods and refined to 1.85 A resolution. The conformation of MalE31 closely resembles that of wild-type MalE, with very small displacements of the mutated residues located in the loop connecting the first alpha-helix to the first beta-strand. The structural and functional characterization provides experimental evidence that MalE31 can attain a wild-type folded conformation, and suggest that the mutated sites are probably involved in the interactions with the membrane components of the maltose transport system.


Subject(s)
Escherichia coli Proteins/chemistry , Maltose/metabolism , Periplasm/chemistry , Periplasmic Binding Proteins/chemistry , Protein Folding , Biological Transport , Cell Compartmentation , Cells, Cultured , Crystallization , Crystallography, X-Ray , Escherichia coli , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fluorescence , Inclusion Bodies , Mutagenesis, Site-Directed , Mutation , Periplasmic Binding Proteins/genetics , Periplasmic Binding Proteins/metabolism , Plasmids , Protein Binding , Protein Conformation , Proteolipids/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...