Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Mol Cell ; 82(13): 2490-2504.e12, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35584695

ABSTRACT

Most eukaryotic messenger RNAs (mRNAs) are processed at their 3' end by the cleavage and polyadenylation specificity factor (CPF/CPSF). CPF mediates the endonucleolytic cleavage of the pre-mRNA and addition of a polyadenosine (poly(A)) tail, which together define the 3' end of the mature transcript. The activation of CPF is highly regulated to maintain the fidelity of RNA processing. Here, using cryo-EM of yeast CPF, we show that the Mpe1 subunit directly contacts the polyadenylation signal sequence in nascent pre-mRNA. The region of Mpe1 that contacts RNA also promotes the activation of CPF endonuclease activity and controls polyadenylation. The Cft2 subunit of CPF antagonizes the RNA-stabilized configuration of Mpe1. In vivo, the depletion or mutation of Mpe1 leads to widespread defects in transcription termination by RNA polymerase II, resulting in transcription interference on neighboring genes. Together, our data suggest that Mpe1 plays a major role in accurate 3' end processing, activating CPF, and ensuring timely transcription termination.


Subject(s)
RNA Precursors , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , mRNA Cleavage and Polyadenylation Factors , Amino Acid Sequence , Cryoelectron Microscopy , Polyadenylation , Protein Binding , Protein Structure, Tertiary , RNA Precursors/genetics , RNA, Messenger/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
2.
Nature ; 616(7958): 828-835, 2023 04.
Article in English | MEDLINE | ID: mdl-37020021

ABSTRACT

Newly made mRNAs are processed and packaged into mature ribonucleoprotein complexes (mRNPs) and are recognized by the essential transcription-export complex (TREX) for nuclear export1,2. However, the mechanisms of mRNP recognition and three-dimensional mRNP organization are poorly understood3. Here we report cryo-electron microscopy and tomography structures of reconstituted and endogenous human mRNPs bound to the 2-MDa TREX complex. We show that mRNPs are recognized through multivalent interactions between the TREX subunit ALYREF and mRNP-bound exon junction complexes. Exon junction complexes can multimerize through ALYREF, which suggests a mechanism for mRNP organization. Endogenous mRNPs form compact globules that are coated by multiple TREX complexes. These results reveal how TREX may simultaneously recognize, compact and protect mRNAs to promote their packaging for nuclear export. The organization of mRNP globules provides a framework to understand how mRNP architecture facilitates mRNA biogenesis and export.


Subject(s)
Active Transport, Cell Nucleus , Cell Nucleus , RNA, Messenger , Transcription, Genetic , Humans , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cryoelectron Microscopy , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Exons
3.
Mol Cell ; 80(6): 1039-1054.e6, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33301732

ABSTRACT

Eukaryotic SMC complexes, cohesin, condensin, and Smc5/6, use ATP hydrolysis to power a plethora of functions requiring organization and restructuring of eukaryotic chromosomes in interphase and during mitosis. The Smc5/6 mechanism of action and its activity on DNA are largely unknown. Here we purified the budding yeast Smc5/6 holocomplex and characterized its core biochemical and biophysical activities. Purified Smc5/6 exhibits DNA-dependent ATP hydrolysis and SUMO E3 ligase activity. We show that Smc5/6 binds DNA topologically with affinity for supercoiled and catenated DNA templates. Employing single-molecule assays to analyze the functional and dynamic characteristics of Smc5/6 bound to DNA, we show that Smc5/6 locks DNA plectonemes and can compact DNA in an ATP-dependent manner. These results demonstrate that the Smc5/6 complex recognizes DNA tertiary structures involving juxtaposed helices and might modulate DNA topology by plectoneme stabilization and local compaction.


Subject(s)
Cell Cycle Proteins/genetics , Multiprotein Complexes/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Adenosine Triphosphatases/genetics , Biophysical Phenomena , Cell Cycle Proteins/ultrastructure , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/ultrastructure , DNA-Binding Proteins/genetics , Humans , Interphase/genetics , Mitosis/genetics , Multiprotein Complexes/ultrastructure , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/ultrastructure , Sumoylation/genetics , Cohesins
4.
Mol Cell ; 79(6): 917-933.e9, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32755595

ABSTRACT

Despite key roles in sister chromatid cohesion and chromosome organization, the mechanism by which cohesin rings are loaded onto DNA is still unknown. Here we combine biochemical approaches and cryoelectron microscopy (cryo-EM) to visualize a cohesin loading intermediate in which DNA is locked between two gates that lead into the cohesin ring. Building on this structural framework, we design experiments to establish the order of events during cohesin loading. In an initial step, DNA traverses an N-terminal kleisin gate that is first opened upon ATP binding and then closed as the cohesin loader locks the DNA against the ATPase gate. ATP hydrolysis will lead to ATPase gate opening to complete DNA entry. Whether DNA loading is successful or results in loop extrusion might be dictated by a conserved kleisin N-terminal tail that guides the DNA through the kleisin gate. Our results establish the molecular basis for cohesin loading onto DNA.


Subject(s)
Cell Cycle Proteins/ultrastructure , Chromatids/ultrastructure , Chromosomal Proteins, Non-Histone/ultrastructure , DNA/ultrastructure , Sister Chromatid Exchange/genetics , Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , Chromatids/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosome Segregation/genetics , Cryoelectron Microscopy , DNA/genetics , Nucleic Acid Conformation , Protein Conformation , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/ultrastructure , Cohesins
5.
Nature ; 578(7796): 627-630, 2020 02.
Article in English | MEDLINE | ID: mdl-32025030

ABSTRACT

Thyroglobulin (TG) is the protein precursor of thyroid hormones, which are essential for growth, development and the control of metabolism in vertebrates1,2. Hormone synthesis from TG occurs in the thyroid gland via the iodination and coupling of pairs of tyrosines, and is completed by TG proteolysis3. Tyrosine proximity within TG is thought to enable the coupling reaction but hormonogenic tyrosines have not been clearly identified, and the lack of a three-dimensional structure of TG has prevented mechanistic understanding4. Here we present the structure of full-length human thyroglobulin at a resolution of approximately 3.5 Å, determined by cryo-electron microscopy. We identified all of the hormonogenic tyrosine pairs in the structure, and verified them using site-directed mutagenesis and in vitro hormone-production assays using human TG expressed in HEK293T cells. Our analysis revealed that the proximity, flexibility and solvent exposure of the tyrosines are the key characteristics of hormonogenic sites. We transferred the reaction sites from TG to an engineered tyrosine donor-acceptor pair in the unrelated bacterial maltose-binding protein (MBP), which yielded hormone production with an efficiency comparable to that of TG. Our study provides a framework to further understand the production and regulation of thyroid hormones.


Subject(s)
Cryoelectron Microscopy , Thyroglobulin/chemistry , Thyroglobulin/ultrastructure , Bacterial Proteins/chemistry , HEK293 Cells , Humans , Maltose-Binding Proteins/chemistry , Models, Molecular , Mutation , Reproducibility of Results , Solvents/chemistry , Thyroglobulin/genetics , Thyroid Hormones/biosynthesis , Thyroid Hormones/metabolism , Tyrosine/chemistry , Tyrosine/genetics , Tyrosine/metabolism
6.
EMBO J ; 40(8): e106164, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33734450

ABSTRACT

Dynactin is a 1.1 MDa complex that activates the molecular motor dynein for ultra-processive transport along microtubules. In order to do this, it forms a tripartite complex with dynein and a coiled-coil adaptor. Dynactin consists of an actin-related filament whose length is defined by its flexible shoulder domain. Despite previous cryo-EM structures, the molecular architecture of the shoulder and pointed end of the filament is still poorly understood due to the lack of high-resolution information in these regions. Here we combine multiple cryo-EM datasets and define precise masking strategies for particle signal subtraction and 3D classification. This overcomes domain flexibility and results in high-resolution maps into which we can build the shoulder and pointed end. The unique architecture of the shoulder securely houses the p150 subunit and positions the four identical p50 subunits in different conformations to bind dynactin's filament. The pointed end map allows us to build the first structure of p62 and reveals the molecular basis for cargo adaptor binding to different sites at the pointed end.


Subject(s)
Dynactin Complex/chemistry , Cryoelectron Microscopy , Dynactin Complex/metabolism , Humans , Molecular Dynamics Simulation , Protein Domains , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/metabolism
7.
Mol Syst Biol ; 19(4): e11544, 2023 04 12.
Article in English | MEDLINE | ID: mdl-36815589

ABSTRACT

Accurately modeling the structures of proteins and their complexes using artificial intelligence is revolutionizing molecular biology. Experimental data enable a candidate-based approach to systematically model novel protein assemblies. Here, we use a combination of in-cell crosslinking mass spectrometry and co-fractionation mass spectrometry (CoFrac-MS) to identify protein-protein interactions in the model Gram-positive bacterium Bacillus subtilis. We show that crosslinking interactions prior to cell lysis reveals protein interactions that are often lost upon cell lysis. We predict the structures of these protein interactions and others in the SubtiWiki database with AlphaFold-Multimer and, after controlling for the false-positive rate of the predictions, we propose novel structural models of 153 dimeric and 14 trimeric protein assemblies. Crosslinking MS data independently validates the AlphaFold predictions and scoring. We report and validate novel interactors of central cellular machineries that include the ribosome, RNA polymerase, and pyruvate dehydrogenase, assigning function to several uncharacterized proteins. Our approach uncovers protein-protein interactions inside intact cells, provides structural insight into their interaction interfaces, and is applicable to genetically intractable organisms, including pathogenic bacteria.


Subject(s)
Artificial Intelligence , Proteomics , Proteomics/methods , Proteins/chemistry , Mass Spectrometry/methods , Molecular Biology
8.
PLoS Pathog ; 17(8): e1009775, 2021 08.
Article in English | MEDLINE | ID: mdl-34339457

ABSTRACT

Viruses have evolved means to manipulate the host's ubiquitin-proteasome system, in order to down-regulate antiviral host factors. The Vpx/Vpr family of lentiviral accessory proteins usurp the substrate receptor DCAF1 of host Cullin4-RING ligases (CRL4), a family of modular ubiquitin ligases involved in DNA replication, DNA repair and cell cycle regulation. CRL4DCAF1 specificity modulation by Vpx and Vpr from certain simian immunodeficiency viruses (SIV) leads to recruitment, poly-ubiquitylation and subsequent proteasomal degradation of the host restriction factor SAMHD1, resulting in enhanced virus replication in differentiated cells. To unravel the mechanism of SIV Vpr-induced SAMHD1 ubiquitylation, we conducted integrative biochemical and structural analyses of the Vpr protein from SIVs infecting Cercopithecus cephus (SIVmus). X-ray crystallography reveals commonalities between SIVmus Vpr and other members of the Vpx/Vpr family with regard to DCAF1 interaction, while cryo-electron microscopy and cross-linking mass spectrometry highlight a divergent molecular mechanism of SAMHD1 recruitment. In addition, these studies demonstrate how SIVmus Vpr exploits the dynamic architecture of the multi-subunit CRL4DCAF1 assembly to optimise SAMHD1 ubiquitylation. Together, the present work provides detailed molecular insight into variability and species-specificity of the evolutionary arms race between host SAMHD1 restriction and lentiviral counteraction through Vpx/Vpr proteins.


Subject(s)
Cullin Proteins/chemistry , Gene Products, vpr/metabolism , Proteasome Endopeptidase Complex/chemistry , SAM Domain and HD Domain-Containing Protein 1/chemistry , Ubiquitination , Virus Replication , Amino Acid Sequence , Animals , Cryoelectron Microscopy , Cullin Proteins/metabolism , Gene Products, vpr/genetics , NEDD8 Protein/chemistry , NEDD8 Protein/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , SAM Domain and HD Domain-Containing Protein 1/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
9.
Anal Chem ; 94(22): 7779-7786, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35613060

ABSTRACT

Proteome-wide crosslinking mass spectrometry studies have coincided with the advent of mass spectrometry (MS)-cleavable crosslinkers that can reveal the individual masses of the two crosslinked peptides. However, recently, such studies have also been published with noncleavable crosslinkers, suggesting that MS-cleavability is not essential. We therefore examined in detail the advantages and disadvantages of using the commonly used MS-cleavable crosslinker, disuccinimidyl sulfoxide (DSSO). Indeed, DSSO gave rise to signature peptide fragments with a distinct mass difference (doublet) for nearly all identified crosslinked peptides. Surprisingly, we could show that it was not these peptide masses that proved the main advantage of MS cleavability of the crosslinker, but improved peptide backbone fragmentation which reduces the ambiguity of peptide identifications. This also holds true for another commonly used MS-cleavable crosslinker, DSBU. We show furthermore that the more intricate MS3-based data acquisition approaches lack sensitivity and specificity, causing them to be outperformed by the simpler and faster stepped higher-energy collisional dissociation (HCD) method. This understanding will guide future developments and applications of proteome-wide crosslinking mass spectrometry.


Subject(s)
Peptides , Proteome , Cross-Linking Reagents/chemistry , Mass Spectrometry/methods , Peptides/chemistry
10.
J Proteome Res ; 19(1): 327-336, 2020 01 03.
Article in English | MEDLINE | ID: mdl-31746214

ABSTRACT

The field of structural biology is increasingly focusing on studying proteins in situ, i.e., in their greater biological context. Cross-linking mass spectrometry (CLMS) is contributing to this effort, typically through the use of mass spectrometry (MS)-cleavable cross-linkers. Here, we apply the popular noncleavable cross-linker disuccinimidyl suberate (DSS) to human mitochondria and identify 5518 distance restraints between protein residues. Each distance restraint on proteins or their interactions provides structural information within mitochondria. Comparing these restraints to protein data bank (PDB)-deposited structures and comparative models reveals novel protein conformations. Our data suggest, among others, substrates and protein flexibility of mitochondrial heat shock proteins. Through this study, we bring forward two central points for the progression of CLMS towards large-scale in situ structural biology: First, clustered conflicts of cross-link data reveal in situ protein conformation states in contrast to error-rich individual conflicts. Second, noncleavable cross-linkers are compatible with proteome-wide studies.


Subject(s)
Cross-Linking Reagents/chemistry , Mass Spectrometry/methods , Mitochondria/chemistry , Mitochondrial Proteins/chemistry , Chromatography, Gel , Humans , K562 Cells , Mitochondrial Proteins/analysis , Protein Conformation , Protein Interaction Maps , Succinimides/chemistry , Workflow
11.
Mol Syst Biol ; 15(9): e8994, 2019 09.
Article in English | MEDLINE | ID: mdl-31556486

ABSTRACT

We present a concise workflow to enhance the mass spectrometric detection of crosslinked peptides by introducing sequential digestion and the crosslink identification software xiSEARCH. Sequential digestion enhances peptide detection by selective shortening of long tryptic peptides. We demonstrate our simple 12-fraction protocol for crosslinked multi-protein complexes and cell lysates, quantitative analysis, and high-density crosslinking, without requiring specific crosslinker features. This overall approach reveals dynamic protein-protein interaction sites, which are accessible, have fundamental functional relevance and are therefore ideally suited for the development of small molecule inhibitors.


Subject(s)
Mass Spectrometry/methods , Protein Interaction Mapping/methods , Proteins/chemistry , Proteomics/methods , Cytosol/chemistry , Humans , K562 Cells , Models, Molecular , Peptide Fragments/chemistry , Protein Conformation , Software
12.
Mol Cell Proteomics ; 17(12): 2387-2401, 2018 12.
Article in English | MEDLINE | ID: mdl-30181345

ABSTRACT

Protein post-translational modifications (PTMs) have an indispensable role in living cells as they expand chemical diversity of the proteome, providing a fine regulatory layer that can govern protein-protein interactions in changing environmental conditions. Here we investigated the effects of acetylation and phosphorylation on the stability of subunit interactions in purified Saccharomyces cerevisiae complexes, namely exosome, RNA polymerase II and proteasome. We propose a computational framework that consists of conformational sampling of the complexes by molecular dynamics simulations, followed by Gibbs energy calculation by MM/GBSA. After benchmarking against published tools such as FoldX and Mechismo, we could apply the framework for the first time on large protein assemblies with the aim of predicting the effects of PTMs located on interfaces of subunits on binding stability. We discovered that acetylation predominantly contributes to subunits' interactions in a locally stabilizing manner, while phosphorylation shows the opposite effect. Even though the local binding contributions of PTMs may be predictable to an extent, the long range effects and overall impact on subunits' binding were only captured because of our dynamical approach. Employing the developed, widely applicable workflow on other large systems will shed more light on the roles of PTMs in protein complex formation.


Subject(s)
Exosome Multienzyme Ribonuclease Complex/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , RNA Polymerase II/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Acetylation , Benchmarking , Computational Biology/methods , Exosome Multienzyme Ribonuclease Complex/chemistry , Mass Spectrometry , Molecular Dynamics Simulation , Phosphorylation , Proteasome Endopeptidase Complex/chemistry , Protein Binding , Protein Structure, Secondary , Proteome/metabolism , RNA Polymerase II/chemistry , Tandem Affinity Purification
13.
Anal Chem ; 91(11): 6953-6961, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31045356

ABSTRACT

The number of publications in the field of chemical cross-linking combined with mass spectrometry (XL-MS) to derive constraints for protein three-dimensional structure modeling and to probe protein-protein interactions has increased during the last years. As the technique is now becoming routine for in vitro and in vivo applications in proteomics and structural biology there is a pressing need to define protocols as well as data analysis and reporting formats. Such consensus formats should become accepted in the field and be shown to lead to reproducible results. This first, community-based harmonization study on XL-MS is based on the results of 32 groups participating worldwide. The aim of this paper is to summarize the status quo of XL-MS and to compare and evaluate existing cross-linking strategies. Our study therefore builds the framework for establishing best practice guidelines to conduct cross-linking experiments, perform data analysis, and define reporting formats with the ultimate goal of assisting scientists to generate accurate and reproducible XL-MS results.


Subject(s)
Cross-Linking Reagents/chemistry , Mass Spectrometry/methods , Serum Albumin, Bovine/analysis , Serum Albumin, Bovine/chemistry , Laboratories , Mass Spectrometry/instrumentation , Reproducibility of Results
14.
Mol Syst Biol ; 13(7): 936, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28743795

ABSTRACT

The arrangement of proteins into complexes is a key organizational principle for many cellular functions. Although the topology of many complexes has been systematically analyzed in isolation, their molecular sociology in situ remains elusive. Here, we show that crude cellular extracts of a eukaryotic thermophile, Chaetomium thermophilum, retain basic principles of cellular organization. Using a structural proteomics approach, we simultaneously characterized the abundance, interactions, and structure of a third of the C. thermophilum proteome within these extracts. We identified 27 distinct protein communities that include 108 interconnected complexes, which dynamically associate with each other and functionally benefit from being in close proximity in the cell. Furthermore, we investigated the structure of fatty acid synthase within these extracts by cryoEM and this revealed multiple, flexible states of the enzyme in adaptation to its association with other complexes, thus exemplifying the need for in situ studies. As the components of the captured protein communities are known-at both the protein and complex levels-this study constitutes another step forward toward a molecular understanding of subcellular organization.


Subject(s)
Chaetomium/metabolism , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Cellular Microenvironment , Cross-Linking Reagents , Cryoelectron Microscopy , Fatty Acid Synthase, Type II/chemistry , Fatty Acid Synthase, Type II/metabolism , Fatty Acid Synthase, Type II/ultrastructure , Fungal Proteins/ultrastructure , Mass Spectrometry , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Protein Interaction Mapping , Protein Interaction Maps , Proteomics , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism , Systems Biology
15.
Mol Syst Biol ; 11(1): 780, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25609650

ABSTRACT

Identifying all essential genomic components is critical for the assembly of minimal artificial life. In the genome-reduced bacterium Mycoplasma pneumoniae, we found that small ORFs (smORFs; < 100 residues), accounting for 10% of all ORFs, are the most frequently essential genomic components (53%), followed by conventional ORFs (49%). Essentiality of smORFs may be explained by their function as members of protein and/or DNA/RNA complexes. In larger proteins, essentiality applied to individual domains and not entire proteins, a notion we could confirm by expression of truncated domains. The fraction of essential non-coding RNAs (ncRNAs) non-overlapping with essential genes is 5% higher than of non-transcribed regions (0.9%), pointing to the important functions of the former. We found that the minimal essential genome is comprised of 33% (269,410 bp) of the M. pneumoniae genome. Our data highlight an unexpected hidden layer of smORFs with essential functions, as well as non-coding regions, thus changing the focus when aiming to define the minimal essential genome.


Subject(s)
DNA, Bacterial/genetics , Genome, Bacterial , Mycoplasma pneumoniae/genetics , Open Reading Frames , RNA, Untranslated/genetics , Genes, Essential , Protein Conformation , Sequence Analysis, DNA , Transcription, Genetic
16.
Proc Natl Acad Sci U S A ; 110(15): 5881-6, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23530218

ABSTRACT

We show that the M2 isoform of pyruvate kinase (M2PYK) exists in equilibrium between monomers and tetramers regulated by allosteric binding of naturally occurring small-molecule metabolites. Phenylalanine stabilizes an inactive T-state tetrameric conformer and inhibits M2PYK with an IC50 value of 0.24 mM, whereas thyroid hormone (triiodo-L-thyronine, T3) stabilizes an inactive monomeric form of M2PYK with an IC50 of 78 nM. The allosteric activator fructose-1,6-bisphosphate [F16BP, AC50 (concentration that gives 50% activation) of 7 µM] shifts the equilibrium to the tetrameric active R-state, which has a similar activity to that of the constitutively fully active isoform M1PYK. Proliferation assays using HCT-116 cells showed that addition of inhibitors phenylalanine and T3 both increased cell proliferation, whereas addition of the activator F16BP reduced proliferation. F16BP abrogates the inhibitory effect of both phenylalanine and T3, highlighting a dominant role of M2PYK allosteric activation in the regulation of cancer proliferation. X-ray structures show constitutively fully active M1PYK and F16BP-bound M2PYK in an R-state conformation with a lysine at the dimer-interface acting as a peg in a hole, locking the active tetramer conformation. Binding of phenylalanine in an allosteric pocket induces a 13° rotation of the protomers, destroying the peg-in-hole R-state interface. This distinct T-state tetramer is stabilized by flipped out Trp/Arg side chains that stack across the dimer interface. X-ray structures and biophysical binding data of M2PYK complexes explain how, at a molecular level, fluctuations in concentrations of amino acids, thyroid hormone, and glucose metabolites switch M2PYK on and off to provide the cell with a nutrient sensing and growth signaling mechanism.


Subject(s)
Cell Proliferation , Pyruvate Kinase/metabolism , Allosteric Site , Amino Acid Sequence , Catalytic Domain , Cell Line, Tumor , Crystallography, X-Ray , Dimerization , Humans , Inhibitory Concentration 50 , Molecular Sequence Data , Phenylalanine/chemistry , Protein Conformation , Protein Structure, Tertiary , Triiodothyronine/chemistry
17.
Nat Commun ; 15(1): 7692, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227404

ABSTRACT

The inhibitor of κB (IκB) kinase (IKK) is a central regulator of NF-κB signaling. All IKK complexes contain hetero- or homodimers of the catalytic IKKß and/or IKKα subunits. Here, we identify a YDDΦxΦ motif, which is conserved in substrates of canonical (IκBα, IκBß) and alternative (p100) NF-κB pathways, and which mediates docking to catalytic IKK dimers. We demonstrate a quantitative correlation between docking affinity and IKK activity related to IκBα phosphorylation/degradation. Furthermore, we show that phosphorylation of the motif's conserved tyrosine, an event previously reported to promote IκBα accumulation and inhibition of NF-κB gene expression, suppresses the docking interaction. Results from integrated structural analyzes indicate that the motif binds to a groove at the IKK dimer interface. Consistently, suppression of IKK dimerization also abolishes IκBα substrate binding. Finally, we show that an optimized bivalent motif peptide inhibits NF-κB signaling. This work unveils a function for IKKα/ß dimerization in substrate motif recognition.


Subject(s)
Amino Acid Motifs , I-kappa B Kinase , NF-kappa B , Protein Multimerization , I-kappa B Kinase/metabolism , I-kappa B Kinase/chemistry , I-kappa B Kinase/genetics , Humans , NF-kappa B/metabolism , Phosphorylation , Protein Binding , Signal Transduction , NF-KappaB Inhibitor alpha/metabolism , NF-KappaB Inhibitor alpha/genetics , Molecular Docking Simulation , HEK293 Cells , Substrate Specificity
18.
Essays Biochem ; 67(2): 215-228, 2023 03 29.
Article in English | MEDLINE | ID: mdl-36734207

ABSTRACT

Cross-linking mass spectrometry has become an established technology to provide structural information on the topology and dynamics of protein complexes. Readily accessible workflows can provide detailed data on simplified systems, such as purified complexes. However, using this technology to study the structure of protein complexes in situ, such as in organelles, cells, and even tissues, is still a technological frontier. The complexity of these systems remains a considerable challenge, but there have been dramatic improvements in sample handling, data acquisition, and data processing. Here, we summarise these developments and describe the paths towards comprehensive and comparative structural interactomes by cross-linking mass spectrometry.


Subject(s)
Proteomics , Proteomics/methods , Mass Spectrometry/methods , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/metabolism
19.
Nat Struct Mol Biol ; 30(9): 1314-1322, 2023 09.
Article in English | MEDLINE | ID: mdl-37653243

ABSTRACT

Translation affects messenger RNA stability and, in yeast, this is mediated by the Ccr4-Not deadenylation complex. The details of this process in mammals remain unclear. Here, we use cryogenic electron microscopy (cryo-EM) and crosslinking mass spectrometry to show that mammalian CCR4-NOT specifically recognizes ribosomes that are stalled during translation elongation in an in vitro reconstituted system with rabbit and human components. Similar to yeast, mammalian CCR4-NOT inserts a helical bundle of its CNOT3 subunit into the empty E site of the ribosome. Our cryo-EM structure shows that CNOT3 also locks the L1 stalk in an open conformation to inhibit further translation. CCR4-NOT is required for stable association of the nonconstitutive subunit CNOT4, which ubiquitinates the ribosome, likely to signal stalled translation elongation. Overall, our work shows that human CCR4-NOT not only detects but also enforces ribosomal stalling to couple translation and mRNA decay.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Humans , Animals , Rabbits , Mammals , Ribosomes , Ubiquitination , Mass Spectrometry , Transcription Factors , Receptors, CCR4 , Ribonucleases
20.
Commun Biol ; 6(1): 552, 2023 05 22.
Article in English | MEDLINE | ID: mdl-37217784

ABSTRACT

The oxoglutarate dehydrogenase complex (OGDHc) participates in the tricarboxylic acid cycle and, in a multi-step reaction, decarboxylates α-ketoglutarate, transfers succinyl to CoA, and reduces NAD+. Due to its pivotal role in metabolism, OGDHc enzymatic components have been studied in isolation; however, their interactions within the endogenous OGDHc remain elusive. Here, we discern the organization of a thermophilic, eukaryotic, native OGDHc in its active state. By combining biochemical, biophysical, and bioinformatic methods, we resolve its composition, 3D architecture, and molecular function at 3.35 Å resolution. We further report the high-resolution cryo-EM structure of the OGDHc core (E2o), which displays various structural adaptations. These include hydrogen bonding patterns confining interactions of OGDHc participating enzymes (E1o-E2o-E3), electrostatic tunneling that drives inter-subunit communication, and the presence of a flexible subunit (E3BPo), connecting E2o and E3. This multi-scale analysis of a succinyl-CoA-producing native cell extract provides a blueprint for structure-function studies of complex mixtures of medical and biotechnological value.


Subject(s)
Citric Acid Cycle , Ketoglutarate Dehydrogenase Complex , Ketoglutarate Dehydrogenase Complex/chemistry , Ketoglutarate Dehydrogenase Complex/metabolism , Acyl Coenzyme A/metabolism , Cytoplasm
SELECTION OF CITATIONS
SEARCH DETAIL