Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Cell ; 171(3): 588-600.e24, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-28988770

ABSTRACT

Condensin protein complexes coordinate the formation of mitotic chromosomes and thereby ensure the successful segregation of replicated genomes. Insights into how condensin complexes bind to chromosomes and alter their topology are essential for understanding the molecular principles behind the large-scale chromatin rearrangements that take place during cell divisions. Here, we identify a direct DNA-binding site in the eukaryotic condensin complex, which is formed by its Ycg1Cnd3 HEAT-repeat and Brn1Cnd2 kleisin subunits. DNA co-crystal structures reveal a conserved, positively charged groove that accommodates the DNA double helix. A peptide loop of the kleisin subunit encircles the bound DNA and, like a safety belt, prevents its dissociation. Firm closure of the kleisin loop around DNA is essential for the association of condensin complexes with chromosomes and their DNA-stimulated ATPase activity. Our data suggest a sophisticated molecular basis for anchoring condensin complexes to chromosomes that enables the formation of large-sized chromatin loops.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromosomes/metabolism , DNA-Binding Proteins/metabolism , Eukaryota/metabolism , Fungal Proteins/metabolism , Multiprotein Complexes/metabolism , Adenosine Triphosphatases/chemistry , Amino Acid Sequence , Chaetomium/metabolism , Chromosomes/chemistry , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/chemistry , Eukaryota/chemistry , Fungal Proteins/chemistry , HeLa Cells , Humans , Models, Molecular , Multiprotein Complexes/chemistry , Saccharomyces cerevisiae/metabolism , Sequence Alignment
2.
Nature ; 499(7456): 111-4, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23698368

ABSTRACT

Facilitates chromatin transcription (FACT) is a conserved histone chaperone that reorganizes nucleosomes and ensures chromatin integrity during DNA transcription, replication and repair. Key to the broad functions of FACT is its recognition of histones H2A-H2B (ref. 2). However, the structural basis for how histones H2A-H2B are recognized and how this integrates with the other functions of FACT, including the recognition of histones H3-H4 and other nuclear factors, is unknown. Here we reveal the crystal structure of the evolutionarily conserved FACT chaperone domain Spt16M from Chaetomium thermophilum, in complex with the H2A-H2B heterodimer. A novel 'U-turn' motif scaffolded onto a Rtt106-like module embraces the α1 helix of H2B. Biochemical and in vivo assays validate the structure and dissect the contribution of histone tails and H3-H4 towards Spt16M binding. Furthermore, we report the structure of the FACT heterodimerization domain that connects FACT to replicative polymerases. Our results show that Spt16M makes several interactions with histones, which we suggest allow the module to invade the nucleosome gradually and block the strongest interaction of H2B with DNA. FACT would thus enhance 'nucleosome breathing' by re-organizing the first 30 base pairs of nucleosomal histone-DNA contacts. Our snapshot of the engagement of the chaperone with H2A-H2B and the structures of all globular FACT domains enable the high-resolution analysis of the vital chaperoning functions of FACT, shedding light on how the complex promotes the activity of enzymes that require nucleosome reorganization.


Subject(s)
Chaetomium/chemistry , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Histones/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Amino Acid Motifs , Conserved Sequence , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , DNA Replication , Histones/chemistry , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Nucleosomes/chemistry , Nucleosomes/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Substrate Specificity
3.
Nature ; 475(7356): 408-11, 2011 Jul 13.
Article in English | MEDLINE | ID: mdl-21753750

ABSTRACT

Many cellular functions involve multi-domain proteins, which are composed of structurally independent modules connected by flexible linkers. Although it is often well understood how a given domain recognizes a cognate oligonucleotide or peptide motif, the dynamic interaction of multiple domains in the recognition of these ligands remains to be characterized. Here we have studied the molecular mechanisms of the recognition of the 3'-splice-site-associated polypyrimidine tract RNA by the large subunit of the human U2 snRNP auxiliary factor (U2AF65) as a key early step in pre-mRNA splicing. We show that the tandem RNA recognition motif domains of U2AF65 adopt two remarkably distinct domain arrangements in the absence or presence of a strong (that is, high affinity) polypyrimidine tract. Recognition of sequence variations in the polypyrimidine tract RNA involves a population shift between these closed and open conformations. The equilibrium between the two conformations functions as a molecular rheostat that quantitatively correlates the natural variations in polypyrimidine tract nucleotide composition, length and functional strength to the efficiency to recruit U2 snRNP to the intron during spliceosome assembly. Mutations that shift the conformational equilibrium without directly affecting RNA binding modulate splicing activity accordingly. Similar mechanisms of cooperative multi-domain conformational selection may operate more generally in the recognition of degenerate nucleotide or amino acid motifs by multi-domain proteins.


Subject(s)
Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing/physiology , RNA, Messenger/metabolism , Ribonucleoproteins/chemistry , Ribonucleoproteins/metabolism , Amino Acid Motifs , Base Sequence , Humans , Introns/genetics , Ligands , Models, Molecular , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Tertiary , Pyrimidines/metabolism , RNA Splice Sites/genetics , RNA, Messenger/genetics , Spliceosomes/chemistry , Spliceosomes/metabolism , Splicing Factor U2AF , Substrate Specificity
4.
Mol Cell ; 30(4): 507-18, 2008 May 23.
Article in English | MEDLINE | ID: mdl-18498752

ABSTRACT

Pygo and BCL9/Legless transduce the Wnt signal by promoting the transcriptional activity of beta-catenin/Armadillo in normal and malignant cells. We show that human and Drosophila Pygo PHD fingers associate with their cognate HD1 domains from BCL9/Legless to bind specifically to the histone H3 tail methylated at lysine 4 (H3K4me). The crystal structures of ternary complexes between PHD, HD1, and two different H3K4me peptides reveal a unique mode of histone tail recognition: efficient histone binding requires HD1 association, and the PHD-HD1 complex binds preferentially to H3K4me2 while displaying insensitivity to methylation of H3R2. Therefore, this is a prime example of histone tail binding by a PHD finger (of Pygo) being modulated by a cofactor (BCL9/Legless). Rescue experiments in Drosophila indicate that Wnt signaling outputs depend on histone decoding. The specificity of this process provided by the Pygo-BCL9/Legless complex suggests that this complex facilitates an early step in the transition from gene silence to Wnt-induced transcription.


Subject(s)
Drosophila Proteins/metabolism , Histones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Signal Transduction/physiology , Wnt Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster , Histones/chemistry , Histones/genetics , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Lysine/metabolism , Methylation , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Binding , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Transcription Factors
5.
Plant J ; 78(1): 31-43, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24461462

ABSTRACT

Brassinosteroids, which control plant growth and development, are sensed by the membrane receptor kinase BRASSINOSTEROID INSENSITIVE 1 (BRI1). Brassinosteroid binding to the BRI1 leucine-rich repeat (LRR) domain induces heteromerisation with a SOMATIC EMBRYOGENESIS RECEPTOR KINASE (SERK)-family co-receptor. This process allows the cytoplasmic kinase domains of BRI1 and SERK to interact, trans-phosphorylate and activate each other. Here we report crystal structures of the BRI1 kinase domain in its activated form and in complex with nucleotides. BRI1 has structural features reminiscent of both serine/threonine and tyrosine kinases, providing insight into the evolution of dual-specificity kinases in plants. Phosphorylation of Thr1039, Ser1042 and Ser1044 causes formation of a catalytically competent activation loop. Mapping previously identified serine/threonine and tyrosine phosphorylation sites onto the structure, we analyse their contribution to brassinosteroid signaling. The location of known genetic missense alleles provide detailed insight into the BRI1 kinase mechanism, while our analyses are inconsistent with a previously reported guanylate cyclase activity. We identify a protein interaction surface on the C-terminal lobe of the kinase and demonstrate that the isolated BRI1, SERK2 and SERK3 cytoplasmic segments form homodimers in solution and have a weak tendency to heteromerise. We propose a model in which heterodimerisation of the BRI1 and SERK ectodomains brings their cytoplasmic kinase domains in a catalytically competent arrangement, an interaction that can be modulated by the BRI1 inhibitor protein BKI1.


Subject(s)
Arabidopsis/metabolism , Brassinosteroids/metabolism , Serine/metabolism , Signal Transduction/physiology , Arabidopsis/genetics , Arabidopsis Proteins/metabolism , Mutation , Phosphorylation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Threonine/metabolism
6.
J Biol Chem ; 288(21): 15110-20, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23569204

ABSTRACT

Saccharomyces cerevisiae τ55, a subunit of the RNA polymerase III-specific general transcription factor TFIIIC, comprises an N-terminal histidine phosphatase domain (τ55-HPD) whose catalytic activity and cellular function is poorly understood. We solved the crystal structures of τ55-HPD and its closely related paralogue Huf and used in silico docking methods to identify phosphoserine- and phosphotyrosine-containing peptides as possible substrates that were subsequently validated using in vitro phosphatase assays. A comparative phosphoproteomic study identified additional phosphopeptides as possible targets that show the involvement of these two phosphatases in the regulation of a variety of cellular functions. Our results identify τ55-HPD and Huf as bona fide protein phosphatases, characterize their substrate specificities, and provide a small set of regulated phosphosite targets in vivo.


Subject(s)
Phosphoric Monoester Hydrolases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Transcription Factors, TFIII/chemistry , Crystallography, X-Ray , Molecular Docking Simulation , Phosphoric Monoester Hydrolases/genetics , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors, TFIII/genetics
7.
EMBO J ; 28(13): 1965-77, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19494831

ABSTRACT

Polycomb group (PcG) proteins repress transcription by modifying chromatin structure in target genes. dSfmbt is a subunit of the Drosophila melanogaster PcG protein complex PhoRC and contains four malignant brain tumour (MBT) repeats involved in the recognition of various mono- and dimethylated histone peptides. Here, we present the crystal structure of the four-MBT-repeat domain of dSfmbt in complex with a mono-methylated histone H4 peptide. Only a single histone peptide binds to the four-MBT-repeat domain. Mutational analyses show high-affinity binding with low peptide sequence selectivity through combinatorial interaction of the methyl-lysine with an aromatic cage and positively charged flanking residues with the surrounding negatively charged surface of the fourth MBT repeat. dSfmbt directly interacts with the PcG protein Scm, a related MBT-repeat protein with similar methyl-lysine binding activity. dSfmbt and Scm co-occupy Polycomb response elements of target genes in Drosophila and they strongly synergize in the repression of these target genes, suggesting that the combined action of these two MBT proteins is crucial for Polycomb silencing.


Subject(s)
Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Histones/chemistry , Histones/metabolism , Lysine/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Histones/genetics , Humans , Lysine/chemistry , Methylation , Models, Molecular , Molecular Sequence Data , Point Mutation , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Protein Binding , Protein Structure, Tertiary , Repressor Proteins/genetics , Sequence Alignment
8.
RNA ; 17(11): 1973-82, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21947263

ABSTRACT

X-chromosome inactivation (XCI) in female mammals depends on the noncoding RNA X inactivation specific transcript (Xist). The mechanism of chromosome-wide silencing by Xist is poorly understood. While it is established that the 5' region of Xist RNA, comprising the A-repeats and holding 7.5-8.5 copies of a conserved 26-mer sequence, is essential for Xist-mediated silencing, high-resolution structural information for the A-repeats is not available. Here, we report the three-dimensional solution structure of a 14-mer hairpin in the 5' region of a human A-repeat. This hairpin is remarkably stable and adopts a novel AUCG tetraloop fold, the integrity of which is required for silencing. We show that, contrary to previous predictions, the 3' region of single or tandem A-repeats mediates duplex formation in vitro. Significantly, mutations in this region disrupt the inter-repeat duplex formation in vitro and abrogate the silencing function of Xist A-repeats in vivo. Our data suggest that the complete A-repeat region may be stabilized by inter-repeat duplex formation and, as such, may provide a platform for multimerization and specific recognition of the AUCG tetraloops by trans-acting factors.


Subject(s)
RNA Folding , RNA, Untranslated/chemistry , Repetitive Sequences, Nucleic Acid , Animals , Dimerization , Humans , Models, Molecular , Nucleic Acid Conformation , RNA, Long Noncoding , RNA, Untranslated/metabolism
9.
Mol Syst Biol ; 8: 571, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22373819

ABSTRACT

Protein post-translational modifications (PTMs) represent important regulatory states that when combined have been hypothesized to act as molecular codes and to generate a functional diversity beyond genome and transcriptome. We systematically investigate the interplay of protein phosphorylation with other post-transcriptional regulatory mechanisms in the genome-reduced bacterium Mycoplasma pneumoniae. Systematic perturbations by deletion of its only two protein kinases and its unique protein phosphatase identified not only the protein-specific effect on the phosphorylation network, but also a modulation of proteome abundance and lysine acetylation patterns, mostly in the absence of transcriptional changes. Reciprocally, deletion of the two putative N-acetyltransferases affects protein phosphorylation, confirming cross-talk between the two PTMs. The measured M. pneumoniae phosphoproteome and lysine acetylome revealed that both PTMs are very common, that (as in Eukaryotes) they often co-occur within the same protein and that they are frequently observed at interaction interfaces and in multifunctional proteins. The results imply previously unreported hidden layers of post-transcriptional regulation intertwining phosphorylation with lysine acetylation and other mechanisms that define the functional state of a cell.


Subject(s)
Acetylesterase/metabolism , Genome Size/genetics , Lysine/metabolism , Metabolic Networks and Pathways/genetics , Pneumonia, Mycoplasma/genetics , Protein Kinases/metabolism , Acetylation , Catalytic Domain/genetics , Evolution, Molecular , Gene Regulatory Networks/genetics , Gene Regulatory Networks/physiology , Genome, Bacterial/genetics , Metabolic Networks and Pathways/physiology , Models, Biological , Organisms, Genetically Modified , Phosphorylation/physiology , Pneumonia, Mycoplasma/metabolism , Protein Processing, Post-Translational/genetics , Proteome/genetics , Proteome/metabolism
10.
Proc Natl Acad Sci U S A ; 107(40): 17427-32, 2010 Oct 05.
Article in English | MEDLINE | ID: mdl-20858733

ABSTRACT

Invertases are highly regulated enzymes with essential functions in carbohydrate partitioning, sugar signaling, and plant development. Here we present the 2.6 Å crystal structure of Arabidopsis cell-wall invertase 1 (INV1) in complex with a protein inhibitor (CIF, or cell-wall inhibitor of ß-fructosidase) from tobacco. The structure identifies a small amino acid motif in CIF that directly targets the invertase active site. The activity of INV1 and its interaction with CIF are strictly pH-dependent with a maximum at about pH 4.5. At this pH, isothermal titration calorimetry reveals that CIF tightly binds its target with nanomolar affinity. CIF competes with sucrose (Suc) for the same binding site, suggesting that both the extracellular Suc concentration and the pH changes regulate association of the complex. A conserved glutamate residue in the complex interface was previously identified as an important quantitative trait locus affecting fruit quality, which implicates the invertase-inhibitor complex as a main regulator of carbon partitioning in plants. Comparison of the CIF/INV1 structure with the complex between the structurally CIF-related pectin methylesterase inhibitor (PMEI) and pectin methylesterase indicates a common targeting mechanism in PMEI and CIF. However, CIF and PMEI use distinct surface areas to selectively inhibit very different enzymatic scaffolds.


Subject(s)
Arabidopsis Proteins/antagonists & inhibitors , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Cell Wall/enzymology , Hydrogen-Ion Concentration , Protein Conformation , beta-Fructofuranosidase/antagonists & inhibitors , beta-Fructofuranosidase/chemistry , beta-Fructofuranosidase/metabolism , Amino Acid Sequence , Arabidopsis/anatomy & histology , Arabidopsis Proteins/genetics , Binding Sites , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Sequence Alignment , Nicotiana , beta-Fructofuranosidase/genetics
11.
J Biol Chem ; 286(26): 23388-96, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21550984

ABSTRACT

Drosophila Nurf55 is a component of different chromatin-modifying complexes, including the PRC2 (Polycomb repressive complex 2). Based on the 1.75-Å crystal structure of Nurf55 bound to histone H4 helix 1, we analyzed interactions of Nurf55 (Nurf55 or p55 in fly and RbAp48/46 in human) with the N-terminal tail of histone H3, the first helix of histone H4, and an N-terminal fragment of the PRC2 subunit Su(z)12 using isothermal calorimetry and pulldown experiments. Site-directed mutagenesis identified the binding site of histone H3 at the top of the Nurf55 WD40 propeller. Unmodified or K9me3- or K27me3-containing H3 peptides were bound with similar affinities, whereas the affinity for K4me3-containing H3 peptides was reduced. Helix 1 of histone H4 and Su(z)12 bound to the edge of the ß-propeller using overlapping binding sites. Our results show similarities in the recognition of histone H4 and Su(z)12 and identify Nurf55 as a versatile interactor that simultaneously contacts multiple partners.


Subject(s)
Drosophila Proteins/chemistry , Histone-Lysine N-Methyltransferase/chemistry , Histones/chemistry , Repressor Proteins/chemistry , Retinoblastoma-Binding Protein 4/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Protein Structure, Secondary , Repressor Proteins/genetics , Repressor Proteins/metabolism , Retinoblastoma-Binding Protein 4/genetics , Retinoblastoma-Binding Protein 4/metabolism
12.
STAR Protoc ; 3(4): 101894, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36595894

ABSTRACT

Apolipoprotein E (ApoE) particles are responsible for packing and transporting lipids throughout aqueous environments. We detail steps to assess in vitro particles forming from artificial membranes using right-angle light scattering and to measure their size using dynamic light scattering. We further describe how to generate in cellulo ApoE particles containing triacylglycerol under fatty-acid-induced stress. We also detail steps to isolate them from cell secretome by immunoprecipitation and analyze their lipid cargo by thin-layer chromatography. For complete details on the use and execution of this protocol, please refer to Lindner et al. (2022).1.


Subject(s)
Apolipoproteins E , Fatty Acids , Apolipoproteins E/chemistry
13.
Cell Rep ; 38(9): 110435, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35235798

ABSTRACT

Apolipoprotein E transports lipids and couples metabolism between astrocytes and neurons. The E4 variant (APOE4) affects these functions and represents a genetic predisposition for Alzheimer's disease, but the molecular mechanisms remain elusive. We show that ApoE produces different types of lipoproteins via distinct lipidation pathways. ApoE forms high-density lipoprotein (HDL)-like, cholesterol-rich particles via the ATP-binding cassette transporter 1 (ABCA1), a mechanism largely unaffected by ApoE polymorphism. Alternatively, ectopic accumulation of fat in astrocytes, a stress-associated condition, redirects ApoE toward the assembly and secretion of triacylglycerol-rich lipoproteins, a process boosted by the APOE4 variant. We demonstrate in vitro that ApoE can detect triacylglycerol in membranes and spontaneously assemble lipoprotein particles (10-20 nm) rich in unsaturated triacylglycerol, and that APOE4 has remarkable properties behaving as a strong triacylglycerol binder. We propose that fatty APOE4 astrocytes have reduced ability to clear toxic fatty acids from the extracellular milieu, because APOE4 reroutes them back to secretion.


Subject(s)
Apolipoprotein E4 , Astrocytes , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Apolipoproteins E/metabolism , Astrocytes/metabolism , Protein Isoforms/metabolism , Triglycerides/metabolism
14.
J Biol Chem ; 285(43): 32810-32817, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20720017

ABSTRACT

Cysteine synthesis in bacteria and plants is catalyzed by serine acetyltransferase (SAT) and O-acetylserine (thiol)-lyase (OAS-TL), which form the hetero-oligomeric cysteine synthase complex (CSC). In plants, but not in bacteria, the CSC is assumed to control cellular sulfur homeostasis by reversible association of the subunits. Application of size exclusion chromatography, analytical ultracentrifugation, and isothermal titration calorimetry revealed a hexameric structure of mitochondrial SAT from Arabidopsis thaliana (AtSATm) and a 2:1 ratio of the OAS-TL dimer to the SAT hexamer in the CSC. Comparable results were obtained for the composition of the cytosolic SAT from A. thaliana (AtSATc) and the cytosolic SAT from Glycine max (Glyma16g03080, GmSATc) and their corresponding CSCs. The hexameric SAT structure is also supported by the calculated binding energies between SAT trimers. The interaction sites of dimers of AtSATm trimers are identified using peptide arrays. A negative Gibbs free energy (ΔG = -33 kcal mol(-1)) explains the spontaneous formation of the AtCSCs, whereas the measured SAT:OAS-TL affinity (K(D) = 30 nm) is 10 times weaker than that of bacterial CSCs. Free SAT from bacteria is >100-fold more sensitive to feedback inhibition by cysteine than AtSATm/c. The sensitivity of plant SATs to cysteine is further decreased by CSC formation, whereas the feedback inhibition of bacterial SAT by cysteine is not affected by CSC formation. The data demonstrate highly similar quaternary structures of the CSCs from bacteria and plants but emphasize differences with respect to the affinity of CSC formation (K(D)) and the regulation of cysteine sensitivity of SAT within the CSC.


Subject(s)
Arabidopsis/enzymology , Cysteine Synthase/chemistry , Mitochondria/enzymology , Plant Proteins/chemistry , Bacteria/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cysteine/chemistry , Cysteine/metabolism , Cysteine Synthase/metabolism , Plant Proteins/metabolism , Protein Structure, Quaternary , Glycine max/enzymology
15.
Mol Syst Biol ; 6: 430, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21119626

ABSTRACT

Protein-metabolite networks are central to biological systems, but are incompletely understood. Here, we report a screen to catalog protein-lipid interactions in yeast. We used arrays of 56 metabolites to measure lipid-binding fingerprints of 172 proteins, including 91 with predicted lipid-binding domains. We identified 530 protein-lipid associations, the majority of which are novel. To show the data set's biological value, we studied further several novel interactions with sphingolipids, a class of conserved bioactive lipids with an elusive mode of action. Integration of live-cell imaging suggests new cellular targets for these molecules, including several with pleckstrin homology (PH) domains. Validated interactions with Slm1, a regulator of actin polarization, show that PH domains can have unexpected lipid-binding specificities and can act as coincidence sensors for both phosphatidylinositol phosphates and phosphorylated sphingolipids.


Subject(s)
High-Throughput Screening Assays/methods , Lipid Metabolism , Saccharomyces cerevisiae Proteins/analysis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/chemistry , Algorithms , Fatty Acid-Binding Proteins/analysis , Fatty Acid-Binding Proteins/chemistry , Fatty Acid-Binding Proteins/metabolism , Lipid Metabolism/physiology , Lipid-Linked Proteins/analysis , Lipid-Linked Proteins/chemistry , Lipid-Linked Proteins/metabolism , Lipids/analysis , Metabolome , Models, Biological , Protein Array Analysis/methods , Protein Binding , Protein Interaction Domains and Motifs/physiology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Validation Studies as Topic
16.
Proc Natl Acad Sci U S A ; 105(26): 8884-9, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18579787

ABSTRACT

The FACT complex is a conserved cofactor for RNA polymerase II elongation through nucleosomes. FACT bears histone chaperone activity and contributes to chromatin integrity. However, the molecular mechanisms behind FACT function remain elusive. Here we report biochemical, structural, and mutational analyses that identify the peptidase homology domain of the Schizosaccharomyces pombe FACT large subunit Spt16 (Spt16-N) as a binding module for histones H3 and H4. The 2.1-A crystal structure of Spt16-N reveals an aminopeptidase P fold whose enzymatic activity has been lost. Instead, the highly conserved fold directly binds histones H3-H4 through a tight interaction with their globular core domains, as well as with their N-terminal tails. Mutations within a conserved surface pocket in Spt16-N or posttranslational modification of the histone H4 tail reduce interaction in vitro, whereas the globular domains of H3-H4 and the H3 tail bind distinct Spt16-N surfaces. Our analysis suggests that the N-terminal domain of Spt16 may add to the known H2A-H2B chaperone activity of FACT by including a H3-H4 tail and H3-H4 core binding function mediated by the N terminus of Spt16. We suggest that these interactions may aid FACT-mediated nucleosome reorganization events.


Subject(s)
Aminopeptidases/chemistry , Histones/metabolism , Peptide Hydrolases/chemistry , Peptide Hydrolases/metabolism , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces/metabolism , Aminopeptidases/metabolism , Catalysis , Enzyme Activation , Histones/chemistry , Models, Molecular , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Schizosaccharomyces/enzymology , Schizosaccharomyces pombe Proteins/metabolism
17.
Glycobiology ; 20(8): 1010-20, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20466650

ABSTRACT

The endoplasmic reticulum malectin is a highly conserved protein in the animal kingdom that has no counterpart so far in lower organisms. We recently determined the structure of its conserved domain and found a highly selective binding to Glc(2)Man(9)GlcNAc(2), an intermediate of N-glycosylation. In our quest for putative ligands during the initial characterization of the protein, we noticed that the malectin domain is highly specific for diglucosides but quite tolerant towards the linkage of the glucosidic bond. To understand the molecular requirements for the observed promiscuity of the malectin domain, here we analyze the binding to a range of diglucosides through comparison of the protein chemical shift perturbation patterns and the saturation transfer difference spectra of the ligands including two maltose-mimicking drugs. A comparison of the maltose-bound structure of the malectin domain with the complex of the native ligand nigerose reveals why malectin is able to tolerate such a diversity of ligands.


Subject(s)
Glucosides/chemistry , Lectins/chemistry , Membrane Proteins/chemistry , Animals , Endoplasmic Reticulum/chemistry , Lectins/isolation & purification , Magnetic Resonance Spectroscopy/standards , Membrane Proteins/isolation & purification , Models, Molecular , Protein Structure, Tertiary , Reference Standards , Xenopus laevis
18.
J Virol ; 83(17): 8759-70, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19553340

ABSTRACT

Papillomaviruses (PVs) are a large family of small DNA viruses infecting mammals, reptiles, and birds. PV infection induces cell proliferation that may lead to the formation of orogenital or skin tumors. PV-induced cell proliferation has been related mainly to the expression of two small oncoproteins, E6 and E7. In mammalian PVs, E6 contains two 70-residue zinc-binding repeats, whereas E7 consists of a natively unfolded N-terminal region followed by a zinc-binding domain which folds as an obligate homodimer. Here, we show that both the novel francolin bird PV Francolinus leucoscepus PV type 1 (FlPV-1) and the chaffinch bird PV Fringilla coelebs PV contain unusual E6 and E7 proteins. The avian E7 proteins contain an extended unfolded N terminus and a zinc-binding domain of reduced size, whereas the avian E6 proteins consist of a single zinc-binding domain. A comparable single-domain E6 protein may have existed in a common ancestor of mammalian and avian PVs. Mammalian E6 C-terminal domains are phylogenetically related to those of single-domain avian E6, whereas mammalian E6 N-terminal domains seem to have emerged by duplication and subsequently diverged from the original ancestral domain. In avian and mammalian cells, both FlPV-1 E6 and FlPV-1 E7 were evenly expressed in the cytoplasm and the nucleus. Finally, samples of full-length FlPV-1 E6 and the FlPV-1 E7 C-terminal zinc-binding domain were prepared for biophysical analysis. Both constructs were highly soluble and well folded, according to nuclear magnetic resonance spectroscopy measurements.


Subject(s)
Bird Diseases/virology , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomavirus Infections/veterinary , Amino Acid Sequence , Animals , Birds , Cell Nucleus/chemistry , Cluster Analysis , Cytoplasm/chemistry , DNA, Viral/chemistry , DNA, Viral/genetics , Evolution, Molecular , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Oncogene Proteins, Viral/analysis , Papillomaviridae/isolation & purification , Phylogeny , Protein Folding , Protein Structure, Tertiary , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology
19.
Eur J Nucl Med Mol Imaging ; 37(7): 1397-407, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20157706

ABSTRACT

PURPOSE: Recombinant antibodies isolated from human antibody libraries have excellent affinities and high target specificity. As full-length IgGs are cleared inadequately slowly from the circulation, the aim of this work was to figure out which kind of recombinant antibody fragment proves to be appropriate for imaging epithelial cell adhesion molecule (EpCAM)-expressing tumours with the short-living radioisotope (68)Ga. METHODS: In order to combine the promising tumour targeting properties of antibodies with (68)Ga, four antibody variants with the same specificity and origin only differing in molecular weight were constructed for comparison. Therefore, the binding domains of a single-chain fragment variable (scFv) isolated from a human naïve antibody library were modified genetically to construct the respective full-length IgG, the tria- and diabody variants. These molecules were conjugated with the bifunctional chelating agent N,N'-bis[2-hydroxy-5-(carboxyethyl)benzyl]ethylenediamine-N,N'-diacetic acid (HBED-CC) to enable (68)Ga labelling at ambient temperature and compared in biodistribution and immuno-PET imaging experiments. RESULTS: The antibody variants with identical specificity proved to have the correct molecular weight, high binding affinity and specificity to their antigen, EpCAM. Radiometal complexation was efficiently performed at room temperature leading to (68)Ga-labelled antibodies with unchanged binding properties compared to the original antibody variants. The best targeting properties were obtained with the scFv and especially with the diabody. The triabody showed higher absolute tumour uptake but only moderate clearance from circulation. CONCLUSION: The antibody variants differed considerably in normal organ uptake, clearance from circulation and tumour accumulation. The data demonstrate the feasibility of imaging solid tumours with the (68)Ga-labelled diabody format. This type of recombinant protein might be a promising carrier even for the short-lived radiometal (68)Ga to support e.g. the management of immunotherapy which may provide important information regarding receptor expression of solid tumours.


Subject(s)
Immunoglobulin Fragments , Neoplasms/diagnostic imaging , Neoplasms/immunology , Positron-Emission Tomography/methods , Recombinant Proteins , Animals , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Epithelial Cells/metabolism , Female , Gallium Radioisotopes , Gene Expression Regulation, Neoplastic , Humans , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/metabolism , Mice , Neoplasms/genetics , Neoplasms/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics
20.
Plant J ; 54(6): 1063-75, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18346196

ABSTRACT

In plants, the first committed enzyme for glutathione biosynthesis, gamma-glutamylcysteine ligase (GCL), is under multiple controls. The recent elucidation of GCL structure from Brassica juncea (BjGCL) has revealed the presence of two intramolecular disulfide bridges (CC1, CC2), which both strongly impact on GCL activity in vitro. Here we demonstrate that cysteines of CC1 are confined to plant species from the Rosids clade, and are absent in other plant families. Conversely, cysteines of CC2 involved in the monomer-dimer transition in BjGCL are not only conserved in the plant kingdom, but are also conserved in the evolutionarily related alpha- (and some gamma-) proteobacterial GCLs. Focusing on the role of CC2 for GCL redox regulation, we have extended our analysis to all available plant (31; including moss and algal) and related proteobacterial GCL (46) protein sequences. Amino acids contributing to the homodimer interface in BjGCL are highly conserved among plant GCLs, but are not conserved in related proteobacterial GCLs. To probe the significance of this distinction, recombinant GCLs from Nicotiana tabacum (NtGCL), Agrobacterium tumefaciens (AtuGCL, alpha-proteobacteria) and Xanthomonas campestris (XcaGCL, gamma-proteobacteria) were analyzed for their redox response. As expected, NtGCL forms a homodimer under oxidizing conditions, and is activated more than threefold. Conversely, proteobacterial GCLs remain monomeric under oxidizing and reducing conditions, and their activities are not inhibited by DTT, despite the presence of CC2. We conclude that although plant GCLs are evolutionarily related to proteobacterial GCLs, redox regulation of their GCLs via CC2-dependent dimerization has been acquired later in evolution, possibly as a consequence of compartmentation in the redox-modulated plastid environment.


Subject(s)
Glutamate-Cysteine Ligase/metabolism , Nicotiana/enzymology , Protein Multimerization , Agrobacterium tumefaciens/enzymology , Amino Acid Sequence , Bacterial Proteins/metabolism , Cloning, Molecular , DNA, Bacterial/genetics , DNA, Plant/genetics , Evolution, Molecular , Models, Molecular , Molecular Sequence Data , Oxidation-Reduction , Plant Proteins/metabolism , Protein Structure, Quaternary , Recombinant Proteins/metabolism , Sequence Alignment , Xanthomonas campestris/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL