Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
J Org Chem ; 88(18): 12914-12923, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37656942

ABSTRACT

Herein we report a modular synthetic method for the preparation of diaryl-substituted cyclohexenone acids starting from phenyl pyruvate and suitable enones. When the reaction is carried out in alkaline tert-butanol or toluene solutions in microwave-assisted conditions mainly anti configuration products are obtained with up to 86% isolated yield. However, when the reaction is carried out in alkaline water, a mixture of products with anti and syn conformations is obtained with up to 98% overall isolated yield. Mechanistically the product with anti conformation forms by a hemiketal-oxy-Cope type [3,3]-sigmatropic rearrangement-intramolecular aldol condensation route and syn product by an intermolecular aldol condensation-electrocyclization (disrotatory type) route.

2.
EMBO Rep ; 24(7): e56467, 2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37155564

ABSTRACT

The APOE4 variant of apolipoprotein E (apoE) is the most prevalent genetic risk allele associated with late-onset Alzheimer's disease (AD). ApoE interacts with complement regulator factor H (FH), but the role of this interaction in AD pathogenesis is unknown. Here we elucidate the mechanism by which isoform-specific binding of apoE to FH alters Aß1-42-mediated neurotoxicity and clearance. Flow cytometry and transcriptomic analysis reveal that apoE and FH reduce binding of Aß1-42 to complement receptor 3 (CR3) and subsequent phagocytosis by microglia which alters expression of genes involved in AD. Moreover, FH forms complement-resistant oligomers with apoE/Aß1-42 complexes and the formation of these complexes is isoform specific with apoE2 and apoE3 showing higher affinity to FH than apoE4. These FH/apoE complexes reduce Aß1-42 oligomerization and toxicity, and colocalize with complement activator C1q deposited on Aß plaques in the brain. These findings provide an important mechanistic insight into AD pathogenesis and explain how the strongest genetic risk factor for AD predisposes for neuroinflammation in the early stages of the disease pathology.


Subject(s)
Alzheimer Disease , Apolipoprotein E4 , Humans , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Complement Factor H/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Neuroinflammatory Diseases , Apolipoproteins E/chemistry , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Amyloid beta-Peptides/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism
3.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35074912

ABSTRACT

Balanced synaptic inhibition, controlled by multiple synaptic adhesion proteins, is critical for proper brain function. MDGA1 (meprin, A-5 protein, and receptor protein-tyrosine phosphatase mu [MAM] domain-containing glycosylphosphatidylinositol anchor protein 1) suppresses synaptic inhibition in mammalian neurons, yet the molecular mechanisms underlying MDGA1-mediated negative regulation of GABAergic synapses remain unresolved. Here, we show that the MDGA1 MAM domain directly interacts with the extension domain of amyloid precursor protein (APP). Strikingly, MDGA1-mediated synaptic disinhibition requires the MDGA1 MAM domain and is prominent at distal dendrites of hippocampal CA1 pyramidal neurons. Down-regulation of APP in presynaptic GABAergic interneurons specifically suppressed GABAergic, but not glutamatergic, synaptic transmission strength and inputs onto both the somatic and dendritic compartments of hippocampal CA1 pyramidal neurons. Moreover, APP deletion manifested differential effects in somatostatin- and parvalbumin-positive interneurons in the hippocampal CA1, resulting in distinct alterations in inhibitory synapse numbers, transmission, and excitability. The infusion of MDGA1 MAM protein mimicked postsynaptic MDGA1 gain-of-function phenotypes that involve the presence of presynaptic APP. The overexpression of MDGA1 wild type or MAM, but not MAM-deleted MDGA1, in the hippocampal CA1 impaired novel object-recognition memory in mice. Thus, our results establish unique roles of APP-MDGA1 complexes in hippocampal neural circuits, providing unprecedented insight into trans-synaptic mechanisms underlying differential tuning of neuronal compartment-specific synaptic inhibition.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Hippocampus/metabolism , Hippocampus/physiopathology , Neural Cell Adhesion Molecules/genetics , Neural Inhibition , Synapses/metabolism , Amyloid beta-Protein Precursor/genetics , CA1 Region, Hippocampal , Carrier Proteins , Dendrites/metabolism , GABAergic Neurons/metabolism , Interneurons , Models, Biological , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/metabolism , Neural Inhibition/genetics , Protein Binding , Protein Interaction Domains and Motifs , Pyramidal Cells/metabolism , Receptors, GABA-B/metabolism , Synaptic Transmission
4.
Acta Crystallogr F Struct Biol Commun ; 78(Pt 1): 39-44, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34981774

ABSTRACT

Synaptic adhesion molecules are major organizers of the neuronal network and play a crucial role in the regulation of synapse development and maintenance in the brain. Synaptic adhesion-like molecules (SALMs) and leukocyte common antigen-related receptor protein tyrosine phosphatases (LAR-PTPs) are adhesion protein families with established synaptic function. Dysfunction of several synaptic adhesion molecules has been linked to cognitive disorders such as autism spectrum disorders and schizophrenia. A recent study of the binding and complex structure of SALM3 and PTPσ using small-angle X-ray scattering revealed a 2:2 complex similar to that observed for the interaction of human SALM5 and PTPδ. However, the molecular structure of the SALM3-PTPσ complex remains to be determined beyond the small-angle X-ray scattering model. Here, the expression, purification, crystallization and initial 6.5 Šresolution structure of the mouse SALM3-PTPσ complex are reported, which further verifies the formation of a 2:2 trans-heterotetrameric complex similar to the crystal structure of human SALM5-PTPδ and validates the architecture of the previously reported small-angle scattering-based solution structure of the SALM3-PTPσ complex. Details of the protein expression and purification, crystal optimization trials, and the initial structure solution and data analysis are provided.


Subject(s)
Receptor-Like Protein Tyrosine Phosphatases, Class 2 , Synapses , Animals , Crystallization , Crystallography, X-Ray , Mice , Receptor-Like Protein Tyrosine Phosphatases, Class 2/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Synapses/metabolism
5.
Sci Rep ; 10(1): 11557, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32665594

ABSTRACT

Synaptic adhesion molecules play an important role in the formation, maintenance and refinement of neuronal connectivity. Recently, several leucine rich repeat (LRR) domain containing neuronal adhesion molecules have been characterized including netrin G-ligands, SLITRKs and the synaptic adhesion-like molecules (SALMs). Dysregulation of these adhesion molecules have been genetically and functionally linked to various neurological disorders. Here we investigated the molecular structure and mechanism of ligand interactions for the postsynaptic SALM3 adhesion protein with its presynaptic ligand, receptor protein tyrosine phosphatase σ (PTPσ). We solved the crystal structure of the dimerized LRR domain of SALM3, revealing the conserved structural features and mechanism of dimerization. Furthermore, we determined the complex structure of SALM3 with PTPσ using small angle X-ray scattering, revealing a 2:2 complex similar to that observed for SALM5. Solution studies unraveled additional flexibility for the complex structure, but validated the uniform mode of action for SALM3 and SALM5 to promote synapse formation. The relevance of the key interface residues was further confirmed by mutational analysis with cellular binding assays and artificial synapse formation assays. Collectively, our results suggest that SALM3 dimerization is a pre-requisite for the SALM3-PTPσ complex to exert synaptogenic activity.


Subject(s)
Membrane Glycoproteins/chemistry , Membrane Glycoproteins/physiology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/chemistry , Synapses/physiology , Animals , Cell Adhesion Molecules, Neuronal/chemistry , Cell Differentiation , Crystallography, X-Ray , DNA Mutational Analysis , Drosophila , Fibronectins/chemistry , Glycosylation , HEK293 Cells , Humans , Ligands , Mice , Mice, Transgenic , Phosphoric Monoester Hydrolases/chemistry , Protein Domains , Protein Multimerization , Scattering, Radiation
6.
Anal Biochem ; 587: 113463, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31574254

ABSTRACT

Synaptic adhesion molecules, including presynaptic neurexins (NRXNs) and post-synaptic leucine-rich repeat transmembrane (LRRTM) proteins are important for development and maintenance of brain neuronal networks. NRXNs are probably the best characterized synaptic adhesion molecules, and one of the major presynaptic organizer proteins. The LRRTMs were found as ligands for NRXNs. Many of the synaptic adhesion proteins have been linked to neurological cognitive disorders, such as schizophrenia and autism spectrum disorders, making them targets of interest for both biological studies, and towards drug development. Therefore, we decided to develop a screening method to target the adhesion proteins, here the LRRTM-NRXN interaction, to find small molecule probes for further studies in cellular settings. To our knowledge, no potent small molecule compounds against the neuronal synaptic adhesion proteins are available. We utilized the AlphaScreen technology, and developed an assay targeting the NRXN-LRRTM2 interaction. We carried out screening of 2000 compounds and identified hits with moderate IC50-values. We also established an orthogonal in-cell Western blot assay to validate hits. This paves way for future development of specific high affinity compounds by further high throughput screening of larger compound libraries using the methods established here. The method could also be applied to screening other NRXN-ligand interactions.


Subject(s)
Calcium-Binding Proteins/antagonists & inhibitors , Nervous System Diseases/metabolism , Neural Cell Adhesion Molecules/antagonists & inhibitors , Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Drosophila , Leucine-Rich Repeat Proteins , Mice , Models, Molecular , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/metabolism , Proteins/chemistry , Proteins/metabolism
7.
ChemMedChem ; 13(22): 2400-2407, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30178912

ABSTRACT

In the era of increased antibiotic resistance, targeting enzymes involved in bacterial communication (quorum sensing) represents a new strategy to fight bacterial infections. LsrK is a kinase responsible for the phosphorylation of autoinducer-2, a signaling molecule involved in quorum sensing. Inhibiting LsrK would lead to quorum sensing inactivation and interfere with the pathogenesis. In this study, we built the first LsrK 3D model and performed virtual screening of a locally available database. Selected compounds were tested against LsrK, and the analogue search conducted based on the positive hits led to the identification of low-micromolar LsrK inhibitors. These results prove the utility of the model and provide the first class of LsrK inhibitors to be further optimized as antivirulence agents.


Subject(s)
Organic Chemicals/chemistry , Protein Kinase Inhibitors/chemistry , Quorum Sensing/drug effects , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Catalytic Domain , Chromobacterium/enzymology , Databases, Chemical , Drug Evaluation, Preclinical , Enzyme Assays , Escherichia coli/enzymology , Escherichia coli/genetics , Molecular Structure , Protein Conformation , Protein Kinases/chemistry , Protein Kinases/genetics , Salmonella typhimurium/enzymology , Structure-Activity Relationship
8.
Protein Eng Des Sel ; 31(5): 147-157, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29897575

ABSTRACT

Synaptic adhesion molecules play a crucial role in the regulation of synapse development and maintenance. Recently, several families of leucine-rich repeat (LRR) domain-containing neuronal adhesion molecules have been characterised, including netrin-G ligands, LRRTMs and the synaptic adhesion-like molecule (SALM) family proteins. Most of these are expressed at the excitatory glutamatergic synapses, and dysfunctions of these genes are genetically linked with cognitive disorders, such as autism spectrum disorders and schizophrenia. The SALM family proteins SALM3 and SALM5, similar to SLITRKs, have been shown to bind to the presynaptic receptor protein tyrosine phosphatase (RPTP) family ligands. Here, we present the 3.1 Å crystal structure of the SALM5 LRR-Ig-domain construct and biophysical studies that verify the crystallographic results. We show that SALM1, SALM3 and SALM5 form similar dimeric structures, in which the LRR domains form the dimer interface. Both SALM3 and SALM5 bind to RPTP immunoglobulin domains with micromolar affinity. SALM3 shows a clear preference for the RPTP ligands with the meB splice insert. Our structural studies and sequence conservation analysis suggests a ligand-binding site and mechanism for RPTP binding via the dimeric LRR domain region.


Subject(s)
Cell Adhesion Molecules, Neuronal/chemistry , Cell Adhesion Molecules, Neuronal/metabolism , Protein Multimerization , Receptor-Like Protein Tyrosine Phosphatases/metabolism , Animals , Extracellular Space/metabolism , Ligands , Models, Molecular , Protein Binding , Protein Domains , Protein Structure, Quaternary , Synapses/metabolism
9.
Nat Commun ; 7: 13596, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27922000

ABSTRACT

Membrane-bound pyrophosphatases (M-PPases), which couple proton/sodium ion transport to pyrophosphate synthesis/hydrolysis, are important in abiotic stress resistance and in the infectivity of protozoan parasites. Here, three M-PPase structures in different catalytic states show that closure of the substrate-binding pocket by helices 5-6 affects helix 13 in the dimer interface and causes helix 12 to move down. This springs a 'molecular mousetrap', repositioning a conserved aspartate and activating the nucleophilic water. Corkscrew motion at helices 6 and 16 rearranges the key ionic gate residues and leads to ion pumping. The pumped ion is above the ion gate in one of the ion-bound structures, but below it in the other. Electrometric measurements show a single-turnover event with a non-hydrolysable inhibitor, supporting our model that ion pumping precedes hydrolysis. We propose a complete catalytic cycle for both proton and sodium-pumping M-PPases, and one that also explains the basis for ion specificity.


Subject(s)
Cell Membrane/enzymology , Pyrophosphatases/metabolism , Thermotoga maritima/enzymology , Vigna/enzymology , Biocatalysis , Crystallography, X-Ray , Hydrogen Bonding , Hydrolysis , Ions , Kinetics , Models, Molecular , Protein Structure, Secondary , Proton Pumps/metabolism , Pyrophosphatases/chemistry , Sodium/metabolism
10.
Sci Rep ; 6: 33916, 2016 Sep 27.
Article in English | MEDLINE | ID: mdl-27671118

ABSTRACT

Chondroitin sulfate (CS) glycosaminoglycans inhibit regeneration in the adult central nervous system (CNS). We report here that HB-GAM (heparin-binding growth-associated molecule; also known as pleiotrophin), a CS-binding protein expressed at high levels in the developing CNS, reverses the role of the CS chains in neurite growth of CNS neurons in vitro from inhibition to activation. The CS-bound HB-GAM promotes neurite growth through binding to the cell surface proteoglycan glypican-2; furthermore, HB-GAM abrogates the CS ligand binding to the inhibitory receptor PTPσ (protein tyrosine phosphatase sigma). Our in vivo studies using two-photon imaging of CNS injuries support the in vitro studies and show that HB-GAM increases dendrite regeneration in the adult cerebral cortex and axonal regeneration in the adult spinal cord. Our findings may enable the development of novel therapies for CNS injuries.

11.
Biochemistry ; 55(6): 914-26, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26785044

ABSTRACT

Synaptic adhesion molecules are key components in development of the brain, and in the formation of neuronal circuits, as they are central in the assembly and maturation of chemical synapses. Several families of neuronal adhesion molecules have been identified such as the neuronal cell adhesion molecules, neurexins and neuroligins, and in particular recently several leucine-rich repeat proteins, e.g., Netrin G-ligands, SLITRKs, and LRRTMs. The LRRTMs form a family of four proteins. They have been implicated in excitatory glutamatergic synapse function and were specifically characterized as ligands for neurexins in excitatory synapse formation and maintenance. In addition, LRRTM3 and LRRTM4 have been found to be ligands for heparan sulfate proteoglycans, including glypican. We report here the crystal structure of a thermostabilized mouse LRRTM2, with a Tm 30 °C higher than that of the wild-type protein. We localized the neurexin binding site to the concave surface based on protein engineering, sequence conservation, and prior information about the interaction of the ligand with neurexins, which allowed us to propose a tentative model for the LRRTM-neurexin interaction complex. We also determined affinities of the thermostabilized LRRTM2 and wild-type LRRTM1 and LRRTM2 for neurexin-ß1 with and without Ca(2+). Cell culture studies and binding experiments show that the engineered protein is functional and capable of forming synapselike contacts. The structural and functional data presented here provide the first structure of an LRRTM protein and allow us to propose a model for the molecular mechanism of LRRTM function in the synaptic adhesion.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Models, Molecular , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Synapses/metabolism , Amino Acid Sequence , Animals , Calcium-Binding Proteins , Cell Adhesion Molecules, Neuronal/chemistry , Cells, Cultured , Crystallography, X-Ray , Drosophila , HEK293 Cells , Humans , Insecta , Membrane Proteins , Mice , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Neural Cell Adhesion Molecules/chemistry , Neurons/metabolism , Protein Binding/physiology , Protein Structure, Secondary , Protein Structure, Tertiary , Rats
12.
BMC Biochem ; 16: 8, 2015 Feb 28.
Article in English | MEDLINE | ID: mdl-25888394

ABSTRACT

BACKGROUND: The protein growth arrest specific-1 (GAS1) was discovered based on its ability to stop the cell cycle. During development it is involved in embryonic patterning, inhibits cell proliferation and mediates cell death, and has therefore been considered as a tumor suppressor. GAS1 is known to signal through two different cell membrane receptors: Rearranged during transformation (RET), and the sonic hedgehog receptor Patched-1. Sonic Hedgehog signalling is important in stem cell renewal and RET mediated signalling in neuronal survival. Disorders in both sonic hedgehog and RET signalling are connected to cancer progression. The neuroprotective effect of RET is controlled by glial cell-derived neurotrophic factor family ligands and glial cell-derived neurotrophic factor receptor alphas (GFRαs). Human Growth arrest specific-1 is a distant homolog of the GFRαs. RESULTS: We have produced and purified recombinant human GAS1 protein, and confirmed that GAS1 is a monomer in solution by static light scattering and small angle X-ray scattering analysis. The low resolution solution structure reveals that GAS1 is more elongated and flexible than the GFRαs, and the homology modelling of the individual domains show that they differ from GFRαs by lacking the amino acids for neurotrophic factor binding. In addition, GAS1 has an extended loop in the N-terminal domain that is conserved in vertebrates after the divergence of fishes and amphibians. CONCLUSIONS: We conclude that GAS1 most likely differs from GFRαs functionally, based on comparative structural analysis, while it is able to bind the extracellular part of RET in a neurotrophic factor independent manner, although with low affinity in solution. Our structural characterization indicates that GAS1 differs from GFRα's significantly also in its conformation, which probably reflects the functional differences between GAS1 and the GFRαs.


Subject(s)
Biophysical Phenomena , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Evolution, Molecular , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Protein Stability , Protein Structure, Tertiary , Sequence Analysis , Sequence Homology, Amino Acid , Solutions , Temperature
13.
J Virol ; 90(6): 2849-57, 2015 Dec 30.
Article in English | MEDLINE | ID: mdl-26719278

ABSTRACT

UNLABELLED: The enveloped negative-stranded RNA virus measles virus (MeV) is an important human pathogen. The nucleoprotein (N(0)) assembles with the viral RNA into helical ribonucleocapsids (NC) which are, in turn, coated by a helical layer of the matrix protein. The viral polymerase complex uses the NC as its template. The N(0) assembly onto the NC and the activity of the polymerase are regulated by the viral phosphoprotein (P). In this study, we pulled down an N(0)1₋408 fragment lacking most of its C-terminal tail domain by several affinity-tagged, N-terminal P fragments to map the N(0)-binding region of P to the first 48 amino acids. We showed biochemically and using P mutants the importance of the hydrophobic interactions for the binding. We fused an N(0) binding peptide, P1₋48, to the C terminus of an N(0)21₋408 fragment lacking both the N-terminal peptide and the C-terminal tail of N protein to reconstitute and crystallize the N(0)-P complex. We solved the X-ray structure of the resulting N(0)-P chimeric protein at a resolution of 2.7 Å. The structure reveals the molecular details of the conserved N(0)-P interface and explains how P chaperones N(0), preventing both self-assembly of N(0) and its binding to RNA. Finally, we propose a model for a preinitiation complex for RNA polymerization. IMPORTANCE: Measles virus is an important, highly contagious human pathogen. The nucleoprotein N binds only to viral genomic RNA and forms the helical ribonucleocapsid that serves as a template for viral replication. We address how N is regulated by another protein, the phosphoprotein (P), to prevent newly synthesized N from binding to cellular RNA. We describe the atomic model of an N-P complex and compare it to helical ribonucleocapsid. We thus provide insight into how P chaperones N and helps to start viral RNA synthesis. Our results provide a new insight into mechanisms of paramyxovirus replication. New data on the mechanisms of phosphoprotein chaperone action allows better understanding of virus genome replication and nucleocapsid assembly. We describe a conserved structural interface for the N-P interaction which could be a target for drug development to treat not only measles but also potentially other paramyxovirus diseases.


Subject(s)
Measles virus/chemistry , Nucleoproteins/chemistry , Nucleoproteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Centrifugation , Crystallography, X-Ray , DNA Mutational Analysis , Measles virus/genetics , Models, Molecular , Nucleocapsid Proteins , Nucleoproteins/genetics , Phosphoproteins/genetics , Protein Binding , Protein Conformation , Protein Interaction Mapping , Viral Proteins/genetics
14.
J Synchrotron Radiat ; 21(Pt 3): 638-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24763656

ABSTRACT

Transnational access (TNA) to national radiation sources is presently provided via programmes of the European Commission by BIOSTRUCT-X and CALIPSO with a major benefit for scientists from European countries. Entirely based on scientific merit, TNA allows all European scientists to realise synchrotron radiation experiments for addressing the Societal Challenges promoted in HORIZON2020. In addition, by TNA all European users directly take part in the development of the research infrastructure of facilities. The mutual interconnection of users and facilities is a strong prerequisite for future development of the research infrastructure of photon science. Taking into account the present programme structure of HORIZON2020, the European Synchrotron User Organization (ESUO) sees considerable dangers for the continuation of this successful collaboration in the future.

15.
PLoS One ; 9(2): e89910, 2014.
Article in English | MEDLINE | ID: mdl-24587117

ABSTRACT

Leucine-rich repeat transmembrane neuronal proteins (LRRTMs) form in mammals a family of four postsynaptic adhesion proteins, which have been shown to bind neurexins and heparan sulphate proteoglycan (HSPG) glypican on the presynaptic side. Mutations in the genes encoding LRRTMs and neurexins are implicated in human cognitive disorders such as schizophrenia and autism. Our analysis shows that in most jawed vertebrates, lrrtm1, lrrtm2, and lrrtm3 genes are nested on opposite strands of large conserved intron of α-catenin genes ctnna2, ctnna1, and ctnna3, respectively. No lrrtm genes could be found in tunicates or lancelets, while two lrrtm genes are found in the lamprey genome, one of which is adjacent to a single ctnna homolog. Based on similar highly positive net charge of lamprey LRRTMs and the HSPG-binding LRRTM3 and LRRTM4 proteins, we speculate that the ancestral LRRTM might have bound HSPG before acquiring neurexins as binding partners. Our model suggests that lrrtm gene translocated into the large ctnna intron in early vertebrates, and that subsequent duplications resulted in three lrrtm/ctnna gene pairs present in most jawed vertebrates. However, we detected three prominent exceptions: (1) the lrrtm3/ctnna3 gene structure is absent in the ray-finned fish genomes, (2) the genomes of clawed frogs contain ctnna1 but lack the corresponding nested (lrrtm2) gene, and (3) contain lrrtm3 gene in the syntenic position but lack the corresponding host (ctnna3) gene. We identified several other protein-coding nested gene structures of which either the host or the nested gene has presumably been lost in the frog or chicken lineages. Interestingly, majority of these nested genes comprise LRR domains.


Subject(s)
Evolution, Molecular , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Phylogeny , Vertebrates/genetics , alpha Catenin/genetics , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Chromosome Mapping , DNA Primers/genetics , Humans , Likelihood Functions , Models, Genetic , Mutation/genetics , Polymerase Chain Reaction , Sequence Alignment , Species Specificity , Synteny/genetics
16.
Mol Biosyst ; 10(5): 1023-34, 2014 May.
Article in English | MEDLINE | ID: mdl-24574026

ABSTRACT

Protein splicing is an autocatalytic process involving self-excision of an internal protein domain, the intein, and concomitant ligation of the two flanking sequences, the exteins, with a peptide bond. Protein splicing can also take place in trans by naturally split inteins or artificially split inteins, ligating the exteins on two different polypeptide chains into one polypeptide chain. Protein trans-splicing could work in foreign contexts by replacing the native extein sequences with other protein sequences. Protein ligation using protein trans-splicing increasingly becomes a useful tool for biotechnological applications such as semi-synthesis of proteins, segmental isotopic labeling, and in vivo protein engineering. However, only a few split inteins have been successfully applied for protein ligation. Naturally split inteins have been widely used, but they are cross-reactive to each other, limiting their applications to multiple-fragment ligation. Based on the three-dimensional structures including two newly determined intein structures, we derived 21 new split inteins from four highly efficient cis-splicing inteins, in order to develop novel split inteins suitable for protein ligation. We systematically compared trans-splicing of 24 split inteins and tested the cross-activities among them to identify orthogonal split intein fragments that could be used in chemical biology and biotechnological applications.


Subject(s)
Inteins , Protein Engineering/methods , Protein Splicing , Crystallography, X-Ray , Kinetics , Models, Biological , Models, Molecular , Terminology as Topic , Trans-Splicing
17.
Nat Chem Biol ; 9(10): 616-22, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23974115

ABSTRACT

Protein sequences are diversified on the DNA level by recombination and mutation and can be further increased on the RNA level by alternative RNA splicing, involving introns that have important roles in many biological processes. The protein version of introns (inteins), which catalyze protein splicing, were first reported in the 1990s. The biological roles of protein splicing still remain elusive because inteins neither provide any clear benefits nor have an essential role in their host organisms. We now report protein alternative splicing, in which new protein sequences can be produced by protein recombination by intermolecular domain swapping of inteins, as elucidated by NMR spectroscopy and crystal structures. We demonstrate that intein-mediated protein alternative splicing could be a new strategy to increase protein diversity (that is, functions) without any modification in genetic backgrounds. We also exploited it as a post-translational protein conformation-driven switch of protein functions (for example, as highly specific protein interference).


Subject(s)
Inteins , Protein Splicing , Proteins/chemistry , Proteins/metabolism , Crystallography, X-Ray , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular
18.
J Virol ; 87(15): 8388-98, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23698307

ABSTRACT

Biochemical reactions powered by ATP hydrolysis are fundamental for the movement of molecules and cellular structures. One such reaction is the encapsidation of the double-stranded DNA (dsDNA) genome of an icosahedrally symmetric virus into a preformed procapsid with the help of a genome-translocating NTPase. Such NTPases have been characterized in detail from both RNA and tailed DNA viruses. We present four crystal structures and the biochemical activity of a thermophilic NTPase, B204, from the nontailed, membrane-containing, hyperthermoacidophilic archaeal dsDNA virus Sulfolobus turreted icosahedral virus 2. These are the first structures of a genome-packaging NTPase from a nontailed, dsDNA virus with an archaeal host. The four structures highlight the catalytic cycle of B204, pinpointing the molecular movement between substrate-bound (open) and empty (closed) active sites. The protein is shown to bind both single-stranded and double-stranded nucleic acids and to have an optimum activity at 80°C and pH 4.5. The overall fold of B204 places it in the FtsK-HerA superfamily of P-loop ATPases, whose cellular and viral members have been suggested to share a DNA-translocating mechanism.


Subject(s)
Archaeal Viruses/enzymology , Archaeal Viruses/physiology , DNA Packaging , Nucleoside-Triphosphatase/chemistry , Nucleoside-Triphosphatase/metabolism , Sulfolobus/virology , Adenosine Triphosphate/metabolism , Catalytic Domain , Crystallography, X-Ray , DNA, Viral/metabolism , Enzyme Stability , Hydrogen-Ion Concentration , Hydrolysis , Models, Molecular , Protein Conformation , Temperature
19.
FEBS Lett ; 587(13): 1863-9, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23684653

ABSTRACT

Soluble inorganic pyrophosphatases (PPases) catalyse an essential reaction, the hydrolysis of pyrophosphate to inorganic phosphate. In addition, an evolutionarily ancient family of membrane-integral pyrophosphatases couple this hydrolysis to Na(+) and/or H(+) pumping, and so recycle some of the free energy from the pyrophosphate. The structures of the H(+)-pumping mung bean PPase and the Na(+)-pumping Thermotoga maritima PPase solved last year revealed an entirely novel membrane protein containing 16 transmembrane helices. The hydrolytic centre, well above the membrane, is linked by a charged "coupling funnel" to the ionic gate about 20Å away. By comparing the active sites, fluoride inhibition data and the various models for ion transport, we conclude that membrane-integral PPases probably use binding of pyrophosphate to drive pumping.


Subject(s)
Diphosphates/chemistry , Pyrophosphatases/chemistry , Animals , Bacterial Proteins/chemistry , Catalytic Domain , Humans , Hydrogen Bonding , Hydrolysis , Models, Molecular , Protein Structure, Secondary
20.
J Biol Chem ; 288(26): 18685-95, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23658013

ABSTRACT

Borrelia burgdorferi spirochetes that cause Lyme borreliosis survive for a long time in human serum because they successfully evade the complement system, an important arm of innate immunity. The outer surface protein E (OspE) of B. burgdorferi is needed for this because it recruits complement regulator factor H (FH) onto the bacterial surface to evade complement-mediated cell lysis. To understand this process at the molecular level, we used a structural approach. First, we solved the solution structure of OspE by NMR, revealing a fold that has not been seen before in proteins involved in complement regulation. Next, we solved the x-ray structure of the complex between OspE and the FH C-terminal domains 19 and 20 (FH19-20) at 2.83 Å resolution. The structure shows that OspE binds FH19-20 in a way similar to, but not identical with, that used by endothelial cells to bind FH via glycosaminoglycans. The observed interaction of OspE with FH19-20 allows the full function of FH in down-regulation of complement activation on the bacteria. This reveals the molecular basis for how B. burgdorferi evades innate immunity and suggests how OspE could be used as a potential vaccine antigen.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Borrelia burgdorferi/immunology , Complement Factor H/immunology , Lipoproteins/immunology , Lyme Disease/microbiology , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Endothelial Cells/metabolism , Glycosaminoglycans/metabolism , Humans , Hydrogen Bonding , Immunity, Innate , Lyme Disease/immunology , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL