Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
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
2.
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
3.
ACS Comb Sci ; 20(4): 197-202, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29553252

ABSTRACT

We present a screening protocol utilizing small-angle X-ray scattering (SAXS) to obtain structural information on biomolecular interactions independent of prior knowledge, so as to complement affinity-based screening and provide leads for further exploration. This protocol categorizes ligand titrations by computing pairwise agreement between curves, and separately estimates affinities by quantifying complex formation as a departure from the linear sum properties of solution SAXS. The protocol is validated by sparse sequence search around the native poly uridine RNA motifs of the two-RRM domain Sex-lethal protein (Sxl). The screening of 35 RNA motifs between 4 to 10 nucleotides reveals a strong variation of resulting complexes, revealed to be preference-switching between 1:1 and 2:2 binding stoichiometries upon addition of structural modeling. Validation of select sequences in isothermal calorimetry and NMR titration retrieves domain-specific roles and function of a guanine anchor. These findings reinforce the suitability of SAXS as a complement in lead identification.


Subject(s)
RNA-Binding Proteins/chemistry , RNA/chemistry , Scattering, Small Angle , Binding Sites , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Conformation , Thermodynamics , X-Rays
4.
G3 (Bethesda) ; 8(1): 79-89, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29118030

ABSTRACT

Efficient preparation of high-quality sequencing libraries that well represent the biological sample is a key step for using next-generation sequencing in research. Tn5 enables fast, robust, and highly efficient processing of limited input material while scaling to the parallel processing of hundreds of samples. Here, we present a robust Tn5 transposase purification strategy based on an N-terminal His6-Sumo3 tag. We demonstrate that libraries prepared with our in-house Tn5 are of the same quality as those processed with a commercially available kit (Nextera XT), while they dramatically reduce the cost of large-scale experiments. We introduce improved purification strategies for two versions of the Tn5 enzyme. The first version carries the previously reported point mutations E54K and L372P, and stably produces libraries of constant fragment size distribution, even if the Tn5-to-input molecule ratio varies. The second Tn5 construct carries an additional point mutation (R27S) in the DNA-binding domain. This construct allows for adjustment of the fragment size distribution based on enzyme concentration during tagmentation, a feature that opens new opportunities for use of Tn5 in customized experimental designs. We demonstrate the versatility of our Tn5 enzymes in different experimental settings, including a novel single-cell polyadenylation site mapping protocol as well as ultralow input DNA sequencing.


Subject(s)
Gene Library , High-Throughput Nucleotide Sequencing/methods , Point Mutation , Recombinant Fusion Proteins/genetics , Transposases/genetics , Base Sequence , Cloning, Molecular/methods , DNA/genetics , DNA/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HeLa Cells , High-Throughput Nucleotide Sequencing/economics , Humans , Polyadenylation , Protein Binding , Recombinant Fusion Proteins/metabolism , Transposases/metabolism
5.
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
6.
Sci Rep ; 7(1): 9903, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28852099

ABSTRACT

The RNA-chaperone Hfq catalyses the annealing of bacterial small RNAs (sRNAs) with target mRNAs to regulate gene expression in response to environmental stimuli. Hfq acts on a diverse set of sRNA-mRNA pairs using a variety of different molecular mechanisms. Here, we present an unusual crystal structure showing two Hfq-RNA complexes interacting via their bound RNA molecules. The structure contains two Hfq6:A18 RNA assemblies positioned face-to-face, with the RNA molecules turned towards each other and connected via interdigitating base stacking interactions at the center. Biochemical data further confirm the observed interaction, and indicate that RNA-mediated contacts occur between Hfq-RNA complexes with various (ARN)X motif containing RNA sequences in vitro, including the stress response regulator OxyS and its target, fhlA. A systematic computational survey also shows that phylogenetically conserved (ARN)X motifs are present in a subset of sRNAs, some of which share similar modular architectures. We hypothesise that Hfq can co-opt RNA-RNA base stacking, an unanticipated structural trick, to promote the interaction of (ARN)X motif containing sRNAs with target mRNAs on a "speed-dating" fashion, thereby supporting their regulatory function.


Subject(s)
Escherichia coli Proteins/chemistry , Host Factor 1 Protein/chemistry , Nucleic Acid Conformation , RNA/chemistry , Amino Acid Motifs , Base Sequence , Binding Sites , Escherichia coli Proteins/metabolism , Host Factor 1 Protein/metabolism , Models, Molecular , Molecular Structure , Protein Binding , RNA/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Solutions/chemistry , Structure-Activity Relationship
7.
Nat Commun ; 7: 13855, 2016 12 19.
Article in English | MEDLINE | ID: mdl-27991587

ABSTRACT

Bromodomains are critical components of many chromatin modifying/remodelling proteins and are emerging therapeutic targets, yet how they interact with nucleosomes, rather than acetylated peptides, remains unclear. Using BRDT as a model, we characterized how the BET family of bromodomains interacts with site-specifically acetylated nucleosomes. Here we report that BRDT interacts with nucleosomes through its first (BD1), but not second (BD2) bromodomain, and that acetylated histone recognition by BD1 is complemented by a bromodomain-DNA interaction. Simultaneous DNA and histone recognition enhances BRDT's nucleosome binding affinity and specificity, and its ability to localize to acetylated chromatin in cells. Conservation of DNA binding in bromodomains of BRD2, BRD3 and BRD4, indicates that bivalent nucleosome recognition is a key feature of these bromodomains and possibly others. Our results elucidate the molecular mechanism of BRDT association with nucleosomes and identify structural features of the BET bromodomains that may be targeted for therapeutic inhibition.


Subject(s)
Nuclear Proteins/chemistry , Nucleosomes/chemistry , Acetylation , Amino Acid Sequence , Histones , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Nucleosomes/metabolism , Protein Binding , Protein Domains , Protein Processing, Post-Translational , Protein Structure, Tertiary
8.
Cell Rep ; 12(9): 1519-30, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26299969

ABSTRACT

Many cellular processes involve the recruitment of proteins to specific membranes, which are decorated with distinctive lipids that act as docking sites. The phosphoinositides form signaling hubs, and we examine mechanisms underlying recruitment. We applied a physiological, quantitative, liposome microarray-based assay to measure the membrane-binding properties of 91 pleckstrin homology (PH) domains, the most common phosphoinositide-binding target. 10,514 experiments quantified the role of phosphoinositides in membrane recruitment. For most domains examined, the observed binding specificity implied cooperativity with additional signaling lipids. Analyses of PH domains with similar lipid-binding profiles identified a conserved motif, mutations in which-including some found in human cancers-induced discrete changes in binding affinities in vitro and protein mislocalization in vivo. The data set reveals cooperativity as a key mechanism for membrane recruitment and, by enabling the interpretation of disease-associated mutations, suggests avenues for the design of small molecules targeting PH domains.


Subject(s)
Cell Membrane/metabolism , Fungal Proteins/metabolism , Phosphatidylinositols/metabolism , Chaetomium/metabolism , Fungal Proteins/chemistry , Protein Binding , Protein Structure, Tertiary , Protein Transport , Saccharomyces cerevisiae/metabolism
9.
Cell Rep ; 12(4): 587-98, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26190108

ABSTRACT

In many animals, the germ plasm segregates germline from soma during early development. Oskar protein is known for its ability to induce germ plasm formation and germ cells in Drosophila. However, the molecular basis of germ plasm formation remains unclear. Here, we show that Oskar is an RNA-binding protein in vivo, crosslinking to nanos, polar granule component, and germ cell-less mRNAs, each of which has a role in germline formation. Furthermore, we present high-resolution crystal structures of the two Oskar domains. RNA-binding maps in vitro to the C-terminal domain, which shows structural similarity to SGNH hydrolases. The highly conserved N-terminal LOTUS domain forms dimers and mediates Oskar interaction with the germline-specific RNA helicase Vasa in vitro. Our findings suggest a dual function of Oskar in RNA and Vasa binding, providing molecular clues to its germ plasm function.


Subject(s)
DEAD-box RNA Helicases/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila/metabolism , Amino Acid Sequence , Animals , Binding Sites , Drosophila/chemistry , Molecular Sequence Data , Protein Binding , RNA, Messenger/metabolism
10.
Dev Cell ; 33(2): 150-62, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25898165

ABSTRACT

Clathrin-mediated endocytosis, the main trafficking route from the plasma membrane to the cytoplasm, is critical to many fundamental cellular processes. Clathrin, coupled to the membrane by adaptor proteins, is thought to play a major structural role in endocytosis by self-assembling into a cage-like lattice around the forming vesicle. Although clathrin adaptors are essential for endocytosis, little is known about their structural role in this process. Here we show that the membrane-binding domains of two conserved clathrin adaptors, Sla2 and Ent1, co-assemble in a PI(4,5)P2-dependent manner to form organized lattices on membranes. We determined the structure of the co-assembled lattice by electron cryo-microscopy and designed mutations that specifically impair the lattice formation in vitro. We show that these mutations block endocytosis in vivo. We suggest that clathrin adaptors not only link the polymerized clathrin to the membrane but also form an oligomeric structure, which is essential for membrane remodeling during endocytosis.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Dictyostelium/metabolism , Endocytosis/physiology , Equilibrative Nucleoside Transporter 1/metabolism , Yeasts/metabolism , Biological Transport , Cell Membrane/metabolism , Cytoskeletal Proteins , Phosphorylation , Protein Structure, Tertiary , Transport Vesicles
11.
Structure ; 23(1): 149-160, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25543256

ABSTRACT

The small, highly conserved Kti11 alias Dph3 protein encoded by the Kluyveromyces lactis killer toxin insensitive gene KTI11/DPH3 is involved in the diphthamide modification of eukaryotic elongation factor 2 and, together with Kti13, in Elongator-dependent tRNA wobble base modifications, thereby affecting the speed and accuracy of protein biosynthesis through two distinct mechanisms. We have solved the crystal structures of Saccharomyces cerevisiae Kti13 and the Kti11/Kti13 heterodimer at 2.4 and 2.9 Å resolution, respectively, and validated interacting residues through mutational analysis in vitro and in vivo. We show that metal coordination by Kti11 and its heterodimerization with Kti13 are essential for both translational control mechanisms. Our structural and functional analyses identify Kti13 as an additional component of the diphthamide modification pathway and provide insight into the molecular mechanisms that allow the Kti11/Kti13 heterodimer to coregulate two consecutive steps in ribosomal protein synthesis.


Subject(s)
Peptide Elongation Factor 2/metabolism , RNA, Transfer/metabolism , Repressor Proteins/chemistry , Repressor Proteins/physiology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/physiology , Models, Molecular , Organisms, Genetically Modified , Peptide Elongation Factor 2/chemistry , Protein Biosynthesis/genetics , Protein Multimerization , Protein Structure, Quaternary , RNA Processing, Post-Transcriptional/genetics , RNA, Transfer/chemistry , Repressor Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
12.
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
13.
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
14.
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
15.
Nat Struct Mol Biol ; 20(4): 508-14, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23474712

ABSTRACT

ADP-ribosylation is a reversible post-translational modification with wide-ranging biological functions in all kingdoms of life. A variety of enzymes use NAD(+) to transfer either single or multiple ADP-ribose (ADPr) moieties onto distinct amino acid substrates, often in response to DNA damage or other stresses. Poly-ADPr-glycohydrolase readily reverses poly-ADP-ribosylation induced by the DNA-damage sensor PARP1 and other enzymes, but it does not remove the most proximal ADPr linked to the target amino acid. Searches for enzymes capable of fully reversing cellular mono-ADP-ribosylation back to the unmodified state have proved elusive, which leaves a gap in the understanding of this modification. Here, we identify a family of macrodomain enzymes present in viruses, yeast and animals that reverse cellular ADP-ribosylation by acting on mono-ADP-ribosylated substrates. Our discoveries establish the complete reversibility of PARP-catalyzed cellular ADP-ribosylation as a regulatory modification.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , Proteins/metabolism , Amino Acid Sequence , Biocatalysis , Models, Molecular , Molecular Sequence Data , N-Glycosyl Hydrolases/chemistry , N-Glycosyl Hydrolases/metabolism , Protein Binding , Protein Processing, Post-Translational , Sequence Homology, Amino Acid
16.
Structure ; 20(4): 604-17, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22483108

ABSTRACT

The viral oncoprotein E6 is an essential factor for cervical cancers induced by "high-risk" mucosal HPV. Among other oncogenic activities, E6 recruits the ubiquitin ligase E6AP to promote the ubiquitination and subsequent proteasomal degradation of p53. E6 is prone to self-association, which long precluded its structural analysis. Here we found that E6 specifically dimerizes through its N-terminal domain and that disruption of the dimer interface strongly increases E6 solubility. This allowed us to raise structural data covering the entire HPV16 E6 protein, including the high-resolution NMR structures of the two zinc-binding domains of E6 and a robust data-driven model structure of the N-terminal domain homodimer. Interestingly, homodimer interface mutations that disrupt E6 self-association also inactivate E6-mediated p53 degradation. These data suggest that E6 needs to self-associate via its N-terminal domain to promote the polyubiquitination of p53 by E6AP.


Subject(s)
Oncogene Proteins, Viral/chemistry , Repressor Proteins/chemistry , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Binding Sites , Escherichia coli , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Nuclear Magnetic Resonance, Biomolecular , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...