Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 298(12): 102601, 2022 12.
Article in English | MEDLINE | ID: mdl-36265588

ABSTRACT

MqnA, the only chorismate dehydratase known so far, catalyzes the initial step in the biosynthesis of menaquinone via the futalosine pathway. Details of the MqnA reaction mechanism remain unclear. Here, we present crystal structures of Streptomyces coelicolor MqnA and its active site mutants in complex with chorismate and the product 3-enolpyruvyl-benzoate, produced during heterologous expression in Escherichia coli. Together with activity studies, our data are in line with dehydration proceeding via substrate assisted catalysis, with the enol pyruvyl group of chorismate acting as catalytic base. Surprisingly, structures of the mutant Asn17Asp with copurified ligand suggest that the enzyme converts to a hydrolase by serendipitous positioning of the carboxyl group. All complex structures presented here exhibit a closed Venus flytrap fold, with the enzyme exploiting the characteristic ligand binding properties of the fold for specific substrate binding and catalysis. The conformational rearrangements that facilitate complete burial of substrate/product, with accompanying topological changes to the enzyme surface, could foster substrate channeling within the biosynthetic pathway.


Subject(s)
Bacterial Proteins , Chorismate Mutase , Nucleosides , Streptomyces coelicolor , Catalysis , Chorismate Mutase/metabolism , Escherichia coli/metabolism , Ligands , Nucleosides/metabolism , Streptomyces coelicolor/enzymology , Bacterial Proteins/metabolism
2.
Ann Neurol ; 84(2): 315-328, 2018 08.
Article in English | MEDLINE | ID: mdl-30014603

ABSTRACT

OBJECTIVE: Autoantibodies against myelin oligodendrocyte glycoprotein (MOG) occur in a proportion of patients with inflammatory demyelinating diseases of the central nervous system (CNS). We analyzed their pathogenic activity by affinity-purifying these antibodies (Abs) from patients and transferring them to experimental animals. METHODS: Patients with Abs to MOG were identified by cell-based assay. We determined the cross-reactivity to rodent MOG and the recognized MOG epitopes. We produced the correctly folded extracellular domain of MOG and affinity-purified MOG-specific Abs from the blood of patients. These purified Abs were used to stain CNS tissue and transferred in 2 models of experimental autoimmune encephalomyelitis. Animals were analyzed histopathologically. RESULTS: We identified 17 patients with MOG Abs from our outpatient clinic and selected 2 with a cross-reactivity to rodent MOG; both had recurrent optic neuritis. Affinity-purified Abs recognized MOG on transfected cells and stained myelin in tissue sections. The Abs from the 2 patients recognized different epitopes on MOG, the CC' and the FG loop. In both patients, these Abs persisted during our observation period of 2 to 3 years. The anti-MOG Abs from both patients were pathogenic upon intrathecal injection in 2 different rat models. Together with cognate MOG-specific T cells, these Abs enhanced T-cell infiltration; together with myelin basic protein-specific T cells, they induced demyelination associated with deposition of C9neo, resembling a multiple sclerosis type II pathology. INTERPRETATION: MOG-specific Abs affinity purified from patients with inflammatory demyelinating disease induce pathological changes in vivo upon cotransfer with myelin-reactive T cells, suggesting that these Abs are similarly pathogenic in patients. Ann Neurol 2018;84:315-328.


Subject(s)
Autoantibodies/blood , Brain/metabolism , Brain/pathology , Myelin-Oligodendrocyte Glycoprotein/blood , Adult , Aged , Animals , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Guinea Pigs , Humans , Inflammation/blood , Inflammation/diagnosis , Male , Middle Aged , Rats , Rats, Inbred Lew , Young Adult
3.
Ann Clin Transl Neurol ; 2(3): 295-301, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25815356

ABSTRACT

We present histological, MRI, and clinical features of an adult patient with relapsing encephalomyelitis and antibodies against myelin oligodendrocyte glycoprotein (MOG). Furthermore, we report molecular details of the recognized epitope that is specific for human MOG. A brain biopsy revealed multiple sclerosis (MS)-type II pathology. Some features overlapped with both MS and neuromyelitis optica spectrum disorders (NMOSD), whereas others were distinct from both MS and NMOSD. Immunoadsorption and rituximab induced clinical stabilization. This case contributes a new, so far missing link in the emerging spectrum of MOG-antibody-associated encephalomyelitis.

4.
Proc Natl Acad Sci U S A ; 111(17): 6281-6, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24733933

ABSTRACT

Drosophila Toll receptors are involved in embryonic development and the immune response of adult flies. In both processes, the only known Toll receptor ligand is the human nerve growth factor-like cystine knot protein Spätzle. Here we present the crystal structure of a 1:1 (nonsignaling) complex of the full-length Toll receptor ectodomain (ECD) with the Spätzle cystine knot domain dimer. The ECD is divided into two leucine-rich repeat (LRR) domains, each of which is capped by cysteine-rich domains. Spätzle binds to the concave surface of the membrane-distal LRR domain, in contrast to the flanking ligand interactions observed for mammalian Toll-like receptors, with asymmetric contributions from each Spätzle protomer. The structure allows rationalization of existing genetic and biochemical data and provides a framework for targeting the immune systems of insects of economic importance, as well as a variety of invertebrate disease vectors.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/immunology , Immunity, Innate , Signal Transduction , Toll-Like Receptors/metabolism , Animals , Drosophila Proteins/chemistry , Humans , Models, Molecular , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Toll-Like Receptors/chemistry
5.
Proc Natl Acad Sci U S A ; 110(51): 20587-92, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24297905

ABSTRACT

Receptor tyrosine kinases participate in several signaling pathways through small G proteins such as Ras (rat sarcoma). An important component in the activation of these G proteins is Son of sevenless (SOS), which catalyzes the nucleotide exchange on Ras. For optimal activity, a second Ras molecule acts as an allosteric activator by binding to a second Ras-binding site within SOS. This allosteric Ras-binding site is blocked by autoinhibitory domains of SOS. We have reported recently that Ras activation also requires the actin-binding proteins ezrin, radixin, and moesin. Here we report the mechanism by which ezrin modulates SOS activity and thereby Ras activation. Active ezrin enhances Ras/MAPK signaling and interacts with both SOS and Ras in vivo and in vitro. Moreover, in vitro kinetic assays with recombinant proteins show that ezrin also is important for the activity of SOS itself. Ezrin interacts with GDP-Ras and with the Dbl homology (DH)/pleckstrin homology (PH) domains of SOS, bringing GDP-Ras to the proximity of the allosteric site of SOS. These actions of ezrin are antagonized by the neurofibromatosis type 2 tumor-suppressor protein merlin. We propose an additional essential step in SOS/Ras control that is relevant for human cancer as well as all physiological processes involving Ras.


Subject(s)
Cytoskeletal Proteins/metabolism , Guanosine Diphosphate/metabolism , MAP Kinase Signaling System , Neurofibromin 2/metabolism , Oncogene Protein p21(ras)/metabolism , Son of Sevenless Proteins/metabolism , Animals , Cytoskeletal Proteins/genetics , Guanosine Diphosphate/genetics , Humans , Mice , NIH 3T3 Cells , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neurofibromin 2/genetics , Oncogene Protein p21(ras)/genetics , Son of Sevenless Proteins/genetics
6.
J Immunol ; 191(7): 3594-604, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24014878

ABSTRACT

Autoantibodies targeting conformationally intact myelin oligodendrocyte glycoprotein (MOG) are found in different inflammatory diseases of the CNS, but their antigenic epitopes have not been mapped. We expressed mutants of MOG on human HeLa cells and analyzed sera from 111 patients (104 children, 7 adults) who recognized cell-bound human MOG, but had different diseases, including acute disseminated encephalomyelitis (ADEM), one episode of transverse myelitis or optic neuritis, multiple sclerosis (MS), anti-aquaporin-4 (AQP4)-negative neuromyelitis optica (NMO), and chronic relapsing inflammatory optic neuritis (CRION). We obtained insight into the recognition of epitopes in 98 patients. All epitopes identified were located at loops connecting the ß strands of MOG. The most frequently recognized MOG epitope was revealed by the P42S mutation positioned in the CC'-loop. Overall, we distinguished seven epitope patterns, including the one mainly recognized by mouse mAbs. In half of the patients, the anti-MOG response was directed to a single epitope. The epitope specificity was not linked to certain disease entities. Longitudinal analysis of 11 patients for up to 5 y indicated constant epitope recognition without evidence for intramolecular epitope spreading. Patients who rapidly lost their anti-MOG IgG still generated a long-lasting IgG response to vaccines, indicating that their loss of anti-MOG reactivity did not reflect a general lack of capacity for long-standing IgG responses. The majority of human anti-MOG Abs did not recognize rodent MOG, which has implications for animal studies. Our findings might assist in future detection of potential mimotopes and pave the way to Ag-specific depletion.


Subject(s)
Central Nervous System Diseases/immunology , Epitopes/chemistry , Epitopes/immunology , Inflammation/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Adolescent , Adult , Amino Acid Sequence , Amino Acid Substitution , Animals , Autoantibodies/immunology , Autoantibodies/metabolism , Central Nervous System Diseases/genetics , Child , Child, Preschool , Epitopes/metabolism , Female , Glycosylation , Humans , Immunoglobulin G/immunology , Infant , Inflammation/genetics , Male , Mice , Middle Aged , Models, Molecular , Mutation , Myelin-Oligodendrocyte Glycoprotein/genetics , Myelin-Oligodendrocyte Glycoprotein/metabolism , Protein Binding/immunology , Protein Conformation , Protein Stability , Sequence Alignment , Transfection , Young Adult
7.
Biol Chem ; 394(8): 1091-6, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23729564

ABSTRACT

Drosophila Toll receptors are involved in embryonic development and in the immune response of adult flies. In both processes, the Toll receptor ligand is the NGF-like cystine knot protein Spätzle. Here we present the expression of Toll receptor ectodomain in Schneider cells at high yields and demonstrate a high affinity interaction with the refolded and trypsin-processed Spätzle cystine knot domain dimer. Poorly and anisotropically diffracting crystals of the complex could be improved by deglycosylation and dehydration, paving the way for structural analyses of the Toll-Spätzle interaction.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Toll-Like Receptors/metabolism , Amino Acid Sequence , Animals , Cell Line , Cloning, Molecular , Crystallization , Drosophila/chemistry , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Gene Expression , Molecular Sequence Data , Protein Multimerization , Protein Refolding , Protein Structure, Tertiary , Toll-Like Receptors/chemistry , Toll-Like Receptors/genetics
9.
J Mol Biol ; 392(5): 1266-77, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19660473

ABSTRACT

12-Oxophytodienoate reductase 3 (OPR3) is a FMN-dependent oxidoreductase that catalyzes the reduction of the cyclopentenone (9S,13S)-12-oxophytodienoate [(9S,13S)-OPDA] to the corresponding cyclopentanone in the biosynthesis of the plant hormone jasmonic acid. In vitro, however, OPR3 reduces the jasmonic acid precursor (9S,13S)-OPDA as well as the enantiomeric (9R,13R)-OPDA, while its isozyme OPR1 is highly selective, accepting only (9R,13R)-OPDA as a substrate. To uncover the molecular determinants of this remarkable enantioselectivity, we determined the crystal structures of OPR1 and OPR3 in complex with the ligand p-hydroxybenzaldehyde. Structural comparison with the OPR1:(9R,13R)-OPDA complex and further biochemical and mutational analyses revealed that two active-site residues, Tyr78 and Tyr246 in OPR1 and Phe74 and His244 in OPR3, are critical for substrate filtering. The relatively smaller OPR3 residues allow formation of a wider substrate binding pocket that is less enantio-restrictive. Substitution of Phe74 and His244 by the corresponding OPR1 tyrosines resulted in an OPR3 mutant showing enhanced, OPR1-like substrate selectivity. Moreover, sequence analysis of the OPR family supports the filtering function of Tyr78 and Tyr246 and allows predictions with respect to substrate specificity and biological function of thus far uncharacterized OPR isozymes. The discovered structural features may also be relevant for other stereoselective proteins and guide the rational design of stereospecific enzymes for biotechnological applications.


Subject(s)
Oxidoreductases Acting on CH-CH Group Donors/chemistry , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Solanum lycopersicum/enzymology , Amino Acid Sequence , Amino Acid Substitution/genetics , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Oxidoreductases Acting on CH-CH Group Donors/genetics , Protein Structure, Tertiary , Sequence Alignment , Substrate Specificity
10.
J Mol Biol ; 385(4): 1015-31, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-19084535

ABSTRACT

Ezrin, radixin and moesin are a family of proteins that provide a link between the plasma membrane and the cortical actin cytoskeleton. The regulated targeting of ezrin to the plasma membrane and its association with cortical F-actin are more than likely functions necessary for a number of cellular processes, such as cell adhesion, motility, morphogenesis and cell signalling. The interaction with F-actin was originally mapped to the last 34 residues of ezrin, which correspond to the last three helices (alphaB, alphaC and alphaD) of the C-terminal tail. We set out to identify and mutate the ezrin/F-actin binding site in order to pinpoint the role of F-actin interaction in morphological processes as well as signal transduction. We report here the generation of an ezrin mutant defective in F-actin binding. We identified four actin-binding residues, T576, K577, R579 and I580, that form a contiguous patch on the surface of the last helix, alphaD. Interestingly, mutagenesis of R579 also eliminated the interaction of band four-point one, ezrin, radixin, moesin homology domains (FERM) and the C-terminal tail domain, identifying a hotspot of the FERM/tail interaction. In vivo expression of the ezrin mutant defective in F-actin binding and FERM/tail interaction (R579A) altered the normal cell surface structure dramatically and inhibited cell migration. Further, we showed that ezrin/F-actin binding is required for the receptor tyrosine kinase signal transfer to the Ras/MAP kinase signalling pathway. Taken together, these observations highlight the importance of ezrin/F-actin function in the development of dynamic membrane/actin structures critical for cell shape and motility, as well as signal transduction.


Subject(s)
Actins/metabolism , Cytoskeletal Proteins/metabolism , Mutant Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Amino Acids/metabolism , Animals , Cell Membrane/metabolism , Cytoskeletal Proteins/chemistry , Humans , Membrane Proteins/chemistry , Mice , Microfilament Proteins/chemistry , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , NIH 3T3 Cells , Phosphorylation , Phosphothreonine/metabolism , Point Mutation/genetics , Protein Binding , Protein Structure, Secondary , Protein Transport , Sus scrofa
11.
J Immunol ; 181(2): 1255-63, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606679

ABSTRACT

Conformational epitopes of myelin oligodendrocyte glycoprotein (MOG) provide a major target for demyelinating autoantibodies in experimental autoimmune encephalomyelitis and recent studies indicate that a similar situation may exist in multiple sclerosis. We recently solved the crystal structure of the extracellular domain of MOG (MOG(ex)) in complex with a Fab derived from the demyelinating mAb 8-18C5 and identified the conformational 8-18C5 epitope on MOG that is dominated by the surface exposed FG loop of MOG. To determine the importance of this epitope with regard to the polyclonal Ab response to MOG(ex) we investigated the effects of mutating His(103) and Ser(104), the two central amino acids of the FG loop, on Ab binding. Mutation of these two residues reduced binding of a panel of eight demyelinating conformation-dependent mAbs to <20% compared with binding to wild-type MOG(ex), whereas substitution of amino acids that do not contribute to the 8-18C5 epitope had only a minor effect on Ab binding. The same restriction was observed for the polyclonal MOG-specific Ab response of MOG DNA-vaccinated BALB/c and SJL/J mice. Our data demonstrate that the pathogenic anti-MOG Ab response primarily targets one immunodominant region centered at the FG loop of MOG. Comparison of the structure of MOG(ex) with the structures of related IgV-like domains yields a possible explanation for the focused Ab response.


Subject(s)
Autoantibodies/immunology , Epitopes/immunology , Myelin-Associated Glycoprotein/chemistry , Myelin-Associated Glycoprotein/immunology , Amino Acid Sequence , Animals , Antibody Specificity , Demyelinating Autoimmune Diseases, CNS/immunology , Epitopes/chemistry , Female , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/immunology , Myelin Proteins , Myelin-Associated Glycoprotein/metabolism , Myelin-Oligodendrocyte Glycoprotein , Point Mutation , Protein Conformation , Rats , Sequence Alignment
12.
J Biol Chem ; 282(17): 13003-10, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17303565

ABSTRACT

Uridine diphosphate-glucose pyrophosphorylase (UGPase) represents a ubiquitous enzyme, which catalyzes the formation of UDP-glucose, a key metabolite of the carbohydrate pathways of all organisms. In the protozoan parasite Leishmania major, which causes a broad spectrum of diseases and is transmitted to humans by sand fly vectors, UGPase represents a virulence factor because of its requirement for the synthesis of cell surface glycoconjugates. Here we present the crystal structures of the L. major UGPase in its uncomplexed apo form (open conformation) and in complex with UDP-glucose (closed conformation). The UGPase consists of three distinct domains. The N-terminal domain exhibits species-specific differences in length, which might permit distinct regulation mechanisms. The central catalytic domain resembles a Rossmann-fold and contains key residues that are conserved in many nucleotidyltransferases. The C-terminal domain forms a left-handed parallel beta-helix (LbetaH), which represents a rarely observed structural element. The presented structures together with mutagenesis analyses provide a basis for a detailed analysis of the catalytic mechanism and for the design of species-specific UGPase inhibitors.


Subject(s)
Leishmania major/enzymology , Protein Folding , Protozoan Proteins/chemistry , UTP-Glucose-1-Phosphate Uridylyltransferase/chemistry , Virulence Factors/chemistry , Animals , Apoenzymes/chemistry , Apoenzymes/metabolism , Catalysis , Crystallography, X-Ray , Glycoconjugates/biosynthesis , Glycoconjugates/chemistry , Leishmania major/pathogenicity , Protein Structure, Secondary , Protein Structure, Tertiary , Protozoan Proteins/metabolism , UTP-Glucose-1-Phosphate Uridylyltransferase/metabolism , Virulence Factors/metabolism
13.
Proc Natl Acad Sci U S A ; 103(39): 14337-42, 2006 Sep 26.
Article in English | MEDLINE | ID: mdl-16983071

ABSTRACT

12-Oxophytodienoate reductase (OPR) 3, a homologue of old yellow enzyme (OYE), catalyzes the reduction of 9S,13S-12-oxophytodienoate to the corresponding cyclopentanone, which is subsequently converted to the plant hormone jasmonic acid (JA). JA and JA derivatives, as well as 12-oxophytodienoate and related cyclopentenones, are known to regulate gene expression in plant development and defense. Together with other oxygenated fatty acid derivatives, they form the oxylipin signature in plants, which resembles the pool of prostaglandins in animals. Here, we report the crystal structure of OPR3 from tomato and of two OPR3 mutants. Although the catalytic residues of OPR3 and related OYEs are highly conserved, several characteristic differences can be discerned in the substrate-binding regions, explaining the remarkable substrate stereoselectivity of OPR isozymes. Interestingly, OPR3 crystallized as an extraordinary self-inhibited dimer. Mutagenesis studies and biochemical analysis confirmed a weak dimerization of OPR3 in vitro, which correlated with a loss of enzymatic activity. Based on structural data of OPR3, a putative mechanism for a strong and reversible dimerization of OPR3 in vivo that involves phosphorylation of OPR3 is suggested. This mechanism could contribute to the shaping of the oxylipin signature, which is critical for fine-tuning gene expression in plants.


Subject(s)
Oxidoreductases Acting on CH-CH Group Donors/chemistry , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Solanum lycopersicum/enzymology , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Dimerization , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Structure-Activity Relationship
14.
Eur J Immunol ; 34(8): 2065-71, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15259003

ABSTRACT

Myelin oligodendrocyte glycoprotein (MOG) is the only myelin protein known to initiate a demyelinating autoantibody response in EAE, an animal model for multiple sclerosis (MS). The pathophysiological significance of MOG-specific autoantibodies in MS is, however, controversial, as high titer antibody responses to MOG are also found in many patients with non-demyelinating neurological diseases. In this issue of the European Journal of Immunology, von Büdingen et al. demonstrate that demyelination in a primate model of MOG-induced EAE is mediated by MOG-specific antibodies directed against discontinuous, rather than linear, MOG epitopes. This functional segregation of pathogenic vs. non-pathogenic autoantibodies in terms of epitope specificity may be crucial to understand the relevance of MOG-specific responses in human disease. This commentary discusses these findings in the context of the structure and immunobiology of MOG, and their implications with respect to antibody-mediated demyelination in MS.


Subject(s)
Autoantibodies/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin Sheath/metabolism , Myelin-Associated Glycoprotein/immunology , Animals , Antibody Formation/immunology , Autoantibodies/chemistry , Callithrix , Disease Models, Animal , Epitopes/immunology , Multiple Sclerosis/immunology , Myelin Proteins , Myelin Sheath/immunology , Myelin-Associated Glycoprotein/chemistry , Myelin-Oligodendrocyte Glycoprotein , Protein Structure, Tertiary
15.
Eur J Biochem ; 271(15): 3208-14, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15265040

ABSTRACT

The amino acid residue tryptophan 27 of 6,7-dimethyl-8-ribityllumazine synthase of the yeast Schizosaccharomyces pombe was replaced by tyrosine. The structures of the W27Y mutant protein in complex with riboflavin, the substrate analogue 5-nitroso-6-ribitylamino-2,4(1H,3H)-pyrimidinedione, and the product analogue 6-carboxyethyl-7-oxo-8-ribityllumazine, were determined by X-ray crystallography at resolutions of 2.7-2.8 A. Whereas the indole system of W27 forms a coplanar pi-complex with riboflavin, the corresponding phenyl ring in the W27Y mutant establishes only peripheral contact with the heterocyclic ring system of the bound riboflavin. These findings provide an explanation for the absence of the long wavelength shift in optical absorption spectra of riboflavin bound to the mutant enzyme. The structures of the mutants are important tools for the interpretation of the unusual physical properties of riboflavin in complex with lumazine synthase.


Subject(s)
Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Riboflavin/metabolism , Schizosaccharomyces/enzymology , Binding Sites , Crystallography, X-Ray , Models, Molecular , Molecular Structure , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/genetics , Mutation/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Riboflavin/chemistry , Schizosaccharomyces/genetics , Structure-Activity Relationship , Tryptophan/genetics , Tryptophan/metabolism
16.
EMBO J ; 23(8): 1720-8, 2004 Apr 21.
Article in English | MEDLINE | ID: mdl-15057273

ABSTRACT

Protoporphyrinogen IX oxidase (PPO), the last common enzyme of haem and chlorophyll biosynthesis, catalyses the oxidation of protoporphyrinogen IX to protoporphyrin IX. The membrane-embedded flavoprotein is the target of a large class of herbicides. In humans, a defect in PPO is responsible for the dominantly inherited disease variegate porphyria. Here we present the crystal structure of mitochondrial PPO from tobacco complexed with a phenyl-pyrazol inhibitor. PPO forms a loosely associated dimer and folds into an FAD-binding domain of the p-hydroxybenzoate-hydrolase fold and a substrate-binding domain that enclose a narrow active site cavity beneath the FAD and an alpha-helical membrane-binding domain. The active site architecture suggests a specific substrate-binding mode compatible with the unusual six-electron oxidation. The membrane-binding domains can be docked onto the dimeric structure of human ferrochelatase, the next enzyme in haem biosynthesis, embedded in the opposite side of the membrane. This modelled transmembrane complex provides a structural explanation for the uncoupling of haem biosynthesis observed in variegate porphyria patients and in plants after inhibiting PPO.


Subject(s)
Chlorophyll/biosynthesis , Heme/biosynthesis , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Amino Acid Sequence , Binding Sites , Catalysis , Cell Membrane/enzymology , Crystallography, X-Ray , Dimerization , Ferrochelatase/metabolism , Flavoproteins , Humans , Mitochondria/enzymology , Mitochondrial Proteins , Models, Molecular , Molecular Sequence Data , Porphyria, Variegate/enzymology , Protein Structure, Quaternary , Protein Subunits/chemistry , Protein Subunits/metabolism , Protoporphyrinogen Oxidase , Sequence Alignment , Nicotiana/enzymology
17.
Proc Natl Acad Sci U S A ; 100(16): 9446-51, 2003 Aug 05.
Article in English | MEDLINE | ID: mdl-12874380

ABSTRACT

Multiple sclerosis is a chronic disease of the central nervous system (CNS) characterized by inflammation, demyelination, and axonal loss. The immunopathogenesis of demyelination in multiple sclerosis involves an autoantibody response to myelin oligodendrocyte glycoprotein (MOG), a type I transmembrane protein located at the surface of CNS myelin. Here we present the crystal structures of the extracellular domain of MOG (MOGIgd) at 1.45-A resolution and the complex of MOGIgd with the antigen-binding fragment (Fab) of the MOG-specific demyelinating monoclonal antibody 8-18C5 at 3.0-A resolution. MOGIgd adopts an IgV like fold with the A'GFCC'C" sheet harboring a cavity similar to the one used by the costimulatory molecule B7-2 to bind its ligand CTLA4. The antibody 8-18C5 binds to three loops located at the membrane-distal side of MOG with a surprisingly dominant contribution made by MOG residues 101-108 containing a strained loop that forms the upper edge of the putative ligand binding site. The sequence R101DHSYQEE108 is unique for MOG, whereas large parts of the remaining sequence are conserved in potentially tolerogenic MOG homologues expressed outside the immuno-privileged environment of the CNS. Strikingly, the only sequence identical to DHSYQEE was found in a Chlamydia trachomatis protein of unknown function, raising the possibility that Chlamydia infections may play a role in the MOG-specific autoimmune response in man. Our data provide the structural basis for the development of diagnostic and therapeutic strategies targeting the pathogenic autoantibody response to MOG.


Subject(s)
Antigens/chemistry , Myelin-Associated Glycoprotein/chemistry , Myelin-Associated Glycoprotein/immunology , Amino Acid Sequence , Animals , Binding Sites , Cell Membrane/metabolism , Chlamydia trachomatis/metabolism , Crystallography, X-Ray , Escherichia coli/metabolism , Ligands , Models, Molecular , Molecular Sequence Data , Myelin Proteins , Myelin-Oligodendrocyte Glycoprotein , Protein Conformation , Protein Folding , Protein Structure, Tertiary , Rats , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...