Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Mol Cell ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38908370

ABSTRACT

Protein folding is assisted by molecular chaperones that bind nascent polypeptides during mRNA translation. Several structurally distinct classes of chaperones promote de novo folding, suggesting that their activities are coordinated at the ribosome. We used biochemical reconstitution and structural proteomics to explore the molecular basis for cotranslational chaperone action in bacteria. We found that chaperone binding is disfavored close to the ribosome, allowing folding to precede chaperone recruitment. Trigger factor recognizes compact folding intermediates that expose an extensive unfolded surface, and dictates DnaJ access to nascent chains. DnaJ uses a large surface to bind structurally diverse intermediates and recruits DnaK to sequence-diverse solvent-accessible sites. Neither Trigger factor, DnaJ, nor DnaK destabilize cotranslational folding intermediates. Instead, the chaperones collaborate to protect incipient structure in the nascent polypeptide well beyond the ribosome exit tunnel. Our findings show how the chaperone network selects and modulates cotranslational folding intermediates.

2.
Wellcome Open Res ; 8: 327, 2023.
Article in English | MEDLINE | ID: mdl-37766847

ABSTRACT

Background: In eukaryotes, replication stress activates a checkpoint response, which facilitates genome duplication by stabilising the replisome. How the checkpoint kinases regulate the replisome remains poorly understood. The aim of this study is to identify new targets of checkpoint kinases within the replisome during replication stress. Methods: Here we use an unbiased biotin proximity-ligation approach in Saccharomyces cerevisiae to identify new interactors and substrates of the checkpoint kinase Rad53 in vivo. Results: From this screen, we identified the replication initiation factor Sld7 as a Rad53 substrate, and Pol1, the catalytic subunit of polymerase a, as a Rad53-interactor. We showed that CDK phosphorylation of Pol1 mediates its interaction with Rad53. Combined with other interactions between Rad53 and the replisome, this Rad53-Pol1 interaction is important for viability and replisome progression during replication stress. Conclusions: Together, we explain how the interactions of Rad53 with the replisome are controlled by both replication stress and the cell cycle, and why these interactions might be important for coordinating the stabilisation of both the leading and lagging strand machineries.

3.
Mol Cell ; 83(13): 2332-2346.e8, 2023 Jul 06.
Article in English | MEDLINE | ID: mdl-37339624

ABSTRACT

Modular SCF (SKP1-CUL1-Fbox) ubiquitin E3 ligases orchestrate multiple cellular pathways in eukaryotes. Their variable SKP1-Fbox substrate receptor (SR) modules enable regulated substrate recruitment and subsequent proteasomal degradation. CAND proteins are essential for the efficient and timely exchange of SRs. To gain structural understanding of the underlying molecular mechanism, we reconstituted a human CAND1-driven exchange reaction of substrate-bound SCF alongside its co-E3 ligase DCNL1 and visualized it by cryo-EM. We describe high-resolution structural intermediates, including a ternary CAND1-SCF complex, as well as conformational and compositional intermediates representing SR- or CAND1-dissociation. We describe in molecular detail how CAND1-induced conformational changes in CUL1/RBX1 provide an optimized DCNL1-binding site and reveal an unexpected dual role for DCNL1 in CAND1-SCF dynamics. Moreover, a partially dissociated CAND1-SCF conformation accommodates cullin neddylation, leading to CAND1 displacement. Our structural findings, together with functional biochemical assays, help formulate a detailed model for CAND-SCF regulation.


Subject(s)
Cullin Proteins , SKP Cullin F-Box Protein Ligases , Humans , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Cullin Proteins/metabolism , Transcription Factors/metabolism , Carrier Proteins/metabolism
4.
Mol Cell ; 82(22): 4324-4339.e8, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36347259

ABSTRACT

ATG9A and ATG2A are essential core members of the autophagy machinery. ATG9A is a lipid scramblase that allows equilibration of lipids across a membrane bilayer, whereas ATG2A facilitates lipid flow between tethered membranes. Although both have been functionally linked during the formation of autophagosomes, the molecular details and consequences of their interaction remain unclear. By combining data from peptide arrays, crosslinking, and hydrogen-deuterium exchange mass spectrometry together with cryoelectron microscopy, we propose a molecular model of the ATG9A-2A complex. Using this integrative structure modeling approach, we identify several interfaces mediating ATG9A-2A interaction that would allow a direct transfer of lipids from ATG2A into the lipid-binding perpendicular branch of ATG9A. Mutational analyses combined with functional activity assays demonstrate their importance for autophagy, thereby shedding light on this protein complex at the heart of autophagy.


Subject(s)
Autophagosomes , Autophagy , Cryoelectron Microscopy , Biological Assay , Lipids
5.
Nat Microbiol ; 7(10): 1686-1701, 2022 10.
Article in English | MEDLINE | ID: mdl-36123441

ABSTRACT

During bacterial cell division, filaments of tubulin-like FtsZ form the Z-ring, which is the cytoplasmic scaffold for divisome assembly. In Escherichia coli, the actin homologue FtsA anchors the Z-ring to the membrane and recruits divisome components, including bitopic FtsN. FtsN regulates the periplasmic peptidoglycan synthase FtsWI. To characterize how FtsA regulates FtsN, we applied electron microscopy to show that E. coli FtsA forms antiparallel double filaments on lipid monolayers when bound to the cytoplasmic tail of FtsN. Using X-ray crystallography, we demonstrate that Vibrio maritimus FtsA crystallizes as an equivalent double filament. We identified an FtsA-FtsN interaction site in the IA-IC interdomain cleft of FtsA using X-ray crystallography and confirmed that FtsA forms double filaments in vivo by site-specific cysteine cross-linking. FtsA-FtsN double filaments reconstituted in or on liposomes prefer negative Gaussian curvature, like those of MreB, the actin-like protein of the elongasome. We propose that curved antiparallel FtsA double filaments together with treadmilling FtsZ filaments organize septal peptidoglycan synthesis in the division plane.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Actins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cysteine/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Lipids , Liposomes , Membrane Proteins/metabolism , Peptidoglycan/metabolism , Tubulin/metabolism
6.
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
7.
Mol Cell Proteomics ; 21(2): 100192, 2022 02.
Article in English | MEDLINE | ID: mdl-34979241

ABSTRACT

The amount of any given protein in the brain is determined by the rates of its synthesis and destruction, which are regulated by different cellular mechanisms. Here, we combine metabolic labeling in live mice with global proteomic profiling to simultaneously quantify both the flux and amount of proteins in mouse models of neurodegeneration. In multiple models, protein turnover increases were associated with increasing pathology. This method distinguishes changes in protein expression mediated by synthesis from those mediated by degradation. In the AppNL-F knockin mouse model of Alzheimer's disease, increased turnover resulted from imbalances in both synthesis and degradation, converging on proteins associated with synaptic vesicle recycling (Dnm1, Cltc, Rims1) and mitochondria (Fis1, Ndufv1). In contrast to disease models, aging in wild-type mice caused a widespread decrease in protein recycling associated with a decrease in autophagic flux. Overall, this simple multidimensional approach enables a comprehensive mapping of proteome dynamics and identifies affected proteins in mouse models of disease and other live animal test settings.


Subject(s)
Alzheimer Disease , Proteome , Aging , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Mammals/metabolism , Mice , Mice, Transgenic , Proteome/metabolism , Proteomics/methods
8.
Nucleic Acids Res ; 50(2): 1128-1146, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34951455

ABSTRACT

Biogenesis of the U5 small nuclear ribonucleoprotein (snRNP) is an essential and highly regulated process. In particular, PRPF8, one of U5 snRNP main components, requires HSP90 working in concert with R2TP, a cochaperone complex containing RUVBL1 and RUVBL2 AAA-ATPases, and additional factors that are still poorly characterized. Here, we use biochemistry, interaction mapping, mass spectrometry and cryoEM to study the role of ZNHIT2 in the regulation of the R2TP chaperone during the biogenesis of PRPF8. ZNHIT2 forms a complex with R2TP which depends exclusively on the direct interaction of ZNHIT2 with the RUVBL1-RUVBL2 ATPases. The cryoEM analysis of this complex reveals that ZNHIT2 alters the conformation and nucleotide state of RUVBL1-RUVBL2, affecting its ATPase activity. We characterized the interactions between R2TP, PRPF8, ZNHIT2, ECD and AAR2 proteins. Interestingly, PRPF8 makes a direct interaction with R2TP and this complex can incorporate ZNHIT2 and other proteins involved in the biogenesis of PRPF8 such as ECD and AAR2. Together, these results show that ZNHIT2 participates in the assembly of the U5 snRNP as part of a network of contacts between assembly factors required for PRPF8 biogenesis and the R2TP-HSP90 chaperone, while concomitantly regulating the structure and nucleotide state of R2TP.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Carrier Proteins/metabolism , DNA Helicases/metabolism , Phosphoproteins/metabolism , RNA Precursors/metabolism , RNA, Messenger/metabolism , HEK293 Cells , Humans , Protein Binding , RNA Splicing , RNA-Binding Proteins
9.
Commun Biol ; 4(1): 1273, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34754068

ABSTRACT

Bromodomain-containing protein 4 (BRD4) is an epigenetic reader and oncology drug target that regulates gene transcription through binding to acetylated chromatin via bromodomains. Phosphorylation by casein kinase II (CK2) regulates BRD4 function, is necessary for active transcription and is involved in resistance to BRD4 drug inhibition in triple-negative breast cancer. Here, we provide the first biophysical analysis of BRD4 phospho-regulation. Using integrative structural biology, we show that phosphorylation by CK2 modulates the dimerization of human BRD4. We identify two conserved regions, a coiled-coil motif and the Basic-residue enriched Interaction Domain (BID), essential for the BRD4 structural rearrangement, which we term the phosphorylation-dependent dimerization domain (PDD). Finally, we demonstrate that bivalent inhibitors induce a conformational change within BRD4 dimers in vitro and in cancer cells. Our results enable the proposal of a model for BRD4 activation critical for the characterization of its protein-protein interaction network and for the development of more specific therapeutics.


Subject(s)
Cell Cycle Proteins/genetics , Gene Expression Regulation , Transcription Factors/genetics , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Cycle Proteins/metabolism , Humans , Phosphorylation , Transcription Factors/metabolism
10.
Nat Commun ; 12(1): 6313, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34728620

ABSTRACT

Cross-talk between distinct protein post-translational modifications is critical for an effective DNA damage response. Arginine methylation plays an important role in maintaining genome stability, but how this modification integrates with other enzymatic activities is largely unknown. Here, we identify the deubiquitylating enzyme USP11 as a previously uncharacterised PRMT1 substrate, and demonstrate that the methylation of USP11 promotes DNA end-resection and the repair of DNA double strand breaks (DSB) by homologous recombination (HR), an event that is independent from another USP11-HR activity, the deubiquitylation of PALB2. We also show that PRMT1 is a ubiquitylated protein that it is targeted for deubiquitylation by USP11, which regulates the ability of PRMT1 to bind to and methylate MRE11. Taken together, our findings reveal a specific role for USP11 during the early stages of DSB repair, which is mediated through its ability to regulate the activity of the PRMT1-MRE11 pathway.


Subject(s)
Arginine/metabolism , MRE11 Homologue Protein/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Rad51 Recombinase/metabolism , Recombinational DNA Repair , Repressor Proteins/metabolism , Thiolester Hydrolases/metabolism , Ubiquitination , Arginine/chemistry , Cell Line , DNA Damage , Genomic Instability , Humans , Methylation
11.
Cell Rep ; 36(1): 109317, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34233195

ABSTRACT

The R2TP (RUVBL1-RUVBL2-RPAP3-PIH1D1) complex, in collaboration with heat shock protein 90 (HSP90), functions as a chaperone for the assembly and stability of protein complexes, including RNA polymerases, small nuclear ribonucleoprotein particles (snRNPs), and phosphatidylinositol 3-kinase (PI3K)-like kinases (PIKKs) such as TOR and SMG1. PIKK stabilization depends on an additional complex of TELO2, TTI1, and TTI2 (TTT), whose structure and function are poorly understood. The cryoelectron microscopy (cryo-EM) structure of the human R2TP-TTT complex, together with biochemical experiments, reveals the mechanism of TOR recruitment to the R2TP-TTT chaperone. The HEAT-repeat TTT complex binds the kinase domain of TOR, without blocking its activity, and delivers TOR to the R2TP chaperone. In addition, TTT regulates the R2TP chaperone by inhibiting RUVBL1-RUVBL2 ATPase activity and by modulating the conformation and interactions of the PIH1D1 and RPAP3 components of R2TP. Taken together, our results show how TTT couples the recruitment of TOR to R2TP with the regulation of this chaperone system.


Subject(s)
Molecular Chaperones/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphatases/metabolism , Cryoelectron Microscopy , Humans , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Protein Domains , Protein Interaction Mapping , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/ultrastructure , Structure-Activity Relationship
12.
Sci Transl Med ; 13(602)2021 07 14.
Article in English | MEDLINE | ID: mdl-34261798

ABSTRACT

Lung and bladder cancers are mostly incurable because of the early development of drug resistance and metastatic dissemination. Hence, improved therapies that tackle these two processes are urgently needed to improve clinical outcome. We have identified RSK4 as a promoter of drug resistance and metastasis in lung and bladder cancer cells. Silencing this kinase, through either RNA interference or CRISPR, sensitized tumor cells to chemotherapy and hindered metastasis in vitro and in vivo in a tail vein injection model. Drug screening revealed several floxacin antibiotics as potent RSK4 activation inhibitors, and trovafloxacin reproduced all effects of RSK4 silencing in vitro and in/ex vivo using lung cancer xenograft and genetically engineered mouse models and bladder tumor explants. Through x-ray structure determination and Markov transient and Deuterium exchange analyses, we identified the allosteric binding site and revealed how this compound blocks RSK4 kinase activation through binding to an allosteric site and mimicking a kinase autoinhibitory mechanism involving the RSK4's hydrophobic motif. Last, we show that patients undergoing chemotherapy and adhering to prophylactic levofloxacin in the large placebo-controlled randomized phase 3 SIGNIFICANT trial had significantly increased (P = 0.048) long-term overall survival times. Hence, we suggest that RSK4 inhibition may represent an effective therapeutic strategy for treating lung and bladder cancer.


Subject(s)
Lung Neoplasms , Urinary Bladder Neoplasms , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Humans , Lung/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics
13.
Angew Chem Int Ed Engl ; 60(19): 10919-10927, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33616271

ABSTRACT

Many natural metalloenzymes assemble from proteins and biosynthesised complexes, generating potent catalysts by changing metal coordination. Here we adopt the same strategy to generate artificial metalloenzymes (ArMs) using ligand exchange to unmask catalytic activity. By systematically testing RuII (η6 -arene)(bipyridine) complexes designed to facilitate the displacement of functionalised bipyridines, we develop a fast and robust procedure for generating new enzymes via ligand exchange in a protein that has not evolved to bind such a complex. The resulting metal cofactors form peptidic coordination bonds but also retain a non-biological ligand. Tandem mass spectrometry and 19 F NMR spectroscopy were used to characterise the organometallic cofactors and identify the protein-derived ligands. By introduction of ruthenium cofactors into a 4-helical bundle, transfer hydrogenation catalysts were generated that displayed a 35-fold rate increase when compared to the respective small molecule reaction in solution.


Subject(s)
Metalloproteins/metabolism , Organometallic Compounds/chemistry , Ruthenium/chemistry , Catalysis , Fluorine , Hydrogenation , Ligands , Magnetic Resonance Spectroscopy , Metalloproteins/chemistry , Molecular Structure , Organometallic Compounds/metabolism , Ruthenium/metabolism
14.
Cell Rep ; 33(12): 108529, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33357431

ABSTRACT

Upon DNA damage, the ALC1/CHD1L nucleosome remodeling enzyme (remodeler) is activated by binding to poly(ADP-ribose). How activated ALC1 recognizes the nucleosome, as well as how this recognition is coupled to remodeling, is unknown. Here, we show that remodeling by ALC1 requires a wild-type acidic patch on the entry side of the nucleosome. The cryo-electron microscopy structure of a nucleosome-ALC1 linker complex reveals a regulatory linker segment that binds to the acidic patch. Mutations within this interface alter the dynamics of ALC1 recruitment to DNA damage and impede the ATPase and remodeling activities of ALC1. Full activation requires acidic patch-linker segment interactions that tether the remodeler to the nucleosome and couple ATP hydrolysis to nucleosome mobilization. Upon DNA damage, such a requirement may be used to modulate ALC1 activity via changes in the nucleosome acidic patches.


Subject(s)
DNA Damage , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Nucleosomes/metabolism , Animals , Histones/metabolism , Humans , Poly (ADP-Ribose) Polymerase-1/metabolism , Xenopus laevis
15.
Elife ; 92020 11 04.
Article in English | MEDLINE | ID: mdl-33146611

ABSTRACT

The outer-membrane of Gram-negative bacteria is critical for surface adhesion, pathogenicity, antibiotic resistance and survival. The major constituent - hydrophobic ß-barrel Outer-Membrane Proteins (OMPs) - are first secreted across the inner-membrane through the Sec-translocon for delivery to periplasmic chaperones, for example SurA, which prevent aggregation. OMPs are then offloaded to the ß-Barrel Assembly Machinery (BAM) in the outer-membrane for insertion and folding. We show the Holo-TransLocon (HTL) - an assembly of the protein-channel core-complex SecYEG, the ancillary sub-complex SecDF, and the membrane 'insertase' YidC - contacts BAM through periplasmic domains of SecDF and YidC, ensuring efficient OMP maturation. Furthermore, the proton-motive force (PMF) across the inner-membrane acts at distinct stages of protein secretion: (1) SecA-driven translocation through SecYEG and (2) communication of conformational changes via SecDF across the periplasm to BAM. The latter presumably drives efficient passage of OMPs. These interactions provide insights of inter-membrane organisation and communication, the importance of which is becoming increasingly apparent.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bacterial Secretion Systems/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Bacterial Outer Membrane Proteins/genetics , Bacterial Secretion Systems/genetics , Models, Molecular , Protein Conformation , Protein Transport
16.
Science ; 369(6508): 1220-1227, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32883864

ABSTRACT

A key step in translational initiation is the recruitment of the 43S preinitiation complex by the cap-binding complex [eukaryotic initiation factor 4F (eIF4F)] at the 5' end of messenger RNA (mRNA) to form the 48S initiation complex (i.e., the 48S). The 48S then scans along the mRNA to locate a start codon. To understand the mechanisms involved, we used cryo-electron microscopy to determine the structure of a reconstituted human 48S The structure reveals insights into early events of translation initiation complex assembly, as well as how eIF4F interacts with subunits of eIF3 near the mRNA exit channel in the 43S The location of eIF4F is consistent with a slotting model of mRNA recruitment and suggests that downstream mRNA is unwound at least in part by being "pulled" through the 40S subunit during scanning.


Subject(s)
Eukaryotic Initiation Factor-3/chemistry , Eukaryotic Initiation Factor-4F/chemistry , Peptide Chain Initiation, Translational , Adenosine Triphosphate/chemistry , Codon, Initiator , Cryoelectron Microscopy , Humans , Hydrolysis , RNA, Messenger/chemistry
17.
Nat Struct Mol Biol ; 27(1): 78-83, 2020 01.
Article in English | MEDLINE | ID: mdl-31907454

ABSTRACT

The human integral membrane protein SERINC5 potently restricts HIV-1 infectivity and sensitizes the virus to antibody-mediated neutralization. Here, using cryo-EM, we determine the structures of human SERINC5 and its orthologue from Drosophila melanogaster at subnanometer and near-atomic resolution, respectively. The structures reveal a novel fold comprised of ten transmembrane helices organized into two subdomains and bisected by a long diagonal helix. A lipid binding groove and clusters of conserved residues highlight potential functional sites. A structure-based mutagenesis scan identified surface-exposed regions and the interface between the subdomains of SERINC5 as critical for HIV-1-restriction activity. The same regions are also important for viral sensitization to neutralizing antibodies, directly linking the antiviral activity of SERINC5 with remodeling of the HIV-1 envelope glycoprotein.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , Membrane Proteins/chemistry , Membrane Proteins/immunology , Animals , Drosophila Proteins/chemistry , Drosophila Proteins/ultrastructure , Drosophila melanogaster/chemistry , Humans , Membrane Proteins/ultrastructure , Models, Molecular , Protein Conformation , Protein Domains , Protein Multimerization
18.
Nature ; 575(7781): 234-237, 2019 11.
Article in English | MEDLINE | ID: mdl-31666700

ABSTRACT

The Fanconi anaemia (FA) pathway repairs DNA damage caused by endogenous and chemotherapy-induced DNA crosslinks, and responds to replication stress1,2. Genetic inactivation of this pathway by mutation of genes encoding FA complementation group (FANC) proteins impairs development, prevents blood production and promotes cancer1,3. The key molecular step in the FA pathway is the monoubiquitination of a pseudosymmetric heterodimer of FANCD2-FANCI4,5 by the FA core complex-a megadalton multiprotein E3 ubiquitin ligase6,7. Monoubiquitinated FANCD2 then recruits additional protein factors to remove the DNA crosslink or to stabilize the stalled replication fork. A molecular structure of the FA core complex would explain how it acts to maintain genome stability. Here we reconstituted an active, recombinant FA core complex, and used cryo-electron microscopy and mass spectrometry to determine its structure. The FA core complex comprises two central dimers of the FANCB and FA-associated protein of 100 kDa (FAAP100) subunits, flanked by two copies of the RING finger subunit, FANCL. These two heterotrimers act as a scaffold to assemble the remaining five subunits, resulting in an extended asymmetric structure. Destabilization of the scaffold would disrupt the entire complex, resulting in a non-functional FA pathway. Thus, the structure provides a mechanistic basis for the low numbers of patients with mutations in FANCB, FANCL and FAAP100. Despite a lack of sequence homology, FANCB and FAAP100 adopt similar structures. The two FANCL subunits are in different conformations at opposite ends of the complex, suggesting that each FANCL has a distinct role. This structural and functional asymmetry of dimeric RING finger domains may be a general feature of E3 ligases. The cryo-electron microscopy structure of the FA core complex provides a foundation for a detailed understanding of its E3 ubiquitin ligase activity and DNA interstrand crosslink repair.


Subject(s)
Cryoelectron Microscopy , Fanconi Anemia Complementation Group Proteins/chemistry , Fanconi Anemia Complementation Group Proteins/ultrastructure , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Protein Subunits/chemistry , Animals , Chickens , Fanconi Anemia/enzymology , Fanconi Anemia Complementation Group L Protein/chemistry , Fanconi Anemia Complementation Group L Protein/ultrastructure , Mass Spectrometry , Models, Molecular , Protein Domains , Protein Multimerization , Structure-Activity Relationship , Ubiquitination
19.
Nature ; 576(7786): 321-325, 2019 12.
Article in English | MEDLINE | ID: mdl-31597161

ABSTRACT

Host infection by pathogenic mycobacteria, such as Mycobacterium tuberculosis, is facilitated by virulence factors that are secreted by type VII secretion systems1. A molecular understanding of the type VII secretion mechanism has been hampered owing to a lack of three-dimensional structures of the fully assembled secretion apparatus. Here we report the cryo-electron microscopy structure of a membrane-embedded core complex of the ESX-3/type VII secretion system from Mycobacterium smegmatis. The core of the ESX-3 secretion machine consists of four protein components-EccB3, EccC3, EccD3 and EccE3, in a 1:1:2:1 stoichiometry-which form two identical protomers. The EccC3 coupling protein comprises a flexible array of four ATPase domains, which are linked to the membrane through a stalk domain. The domain of unknown function (DUF) adjacent to the stalk is identified as an ATPase domain that is essential for secretion. EccB3 is predominantly periplasmatic, but a small segment crosses the membrane and contacts the stalk domain. This suggests that conformational changes in the stalk domain-triggered by substrate binding at the distal end of EccC3 and subsequent ATP hydrolysis in the DUF-could be coupled to substrate secretion to the periplasm. Our results reveal that the architecture of type VII secretion systems differs markedly from that of other known secretion machines2, and provide a structural understanding of these systems that will be useful for the design of antimicrobial strategies that target bacterial virulence.


Subject(s)
Cryoelectron Microscopy , Mycobacterium smegmatis/chemistry , Type VII Secretion Systems/chemistry , Type VII Secretion Systems/ultrastructure , Actinobacteria/chemistry , Actinobacteria/enzymology , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/isolation & purification , Adenosine Triphosphatases/ultrastructure , Adenosine Triphosphate/metabolism , Models, Molecular , Mycobacterium smegmatis/enzymology , Mycobacterium smegmatis/ultrastructure , Protein Domains , Protein Structure, Quaternary , Protein Subunits/chemistry , Protein Subunits/isolation & purification , Structure-Activity Relationship , Thermomonospora , Type VII Secretion Systems/isolation & purification
20.
Cell Host Microbe ; 24(6): 761-775.e6, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30503508

ABSTRACT

TRIM5 is a RING domain E3 ubiquitin ligase with potent antiretroviral function. TRIM5 assembles into a hexagonal lattice on retroviral capsids, causing envelopment of the infectious core. Concomitantly, TRIM5 initiates innate immune signaling and orchestrates disassembly of the viral particle, yet how these antiviral responses are regulated by capsid recognition is unclear. We show that hexagonal assembly triggers N-terminal polyubiquitination of TRIM5 that collectively drives antiviral responses. In uninfected cells, N-terminal monoubiquitination triggers non-productive TRIM5 turnover. Upon TRIM5 assembly on virus, a trivalent RING arrangement allows elongation of N-terminally anchored K63-linked ubiquitin chains (N-K63-Ub). N-K63-Ub drives TRIM5 innate immune stimulation and proteasomal degradation. Inducing ubiquitination before TRIM5 assembly triggers premature degradation and ablates antiviral restriction. Conversely, driving N-K63 ubiquitination after TRIM5 assembly enhances innate immune signaling. Thus, the hexagonal geometry of TRIM5's antiviral lattice converts a capsid-binding protein into a multifunctional antiviral platform.


Subject(s)
Carrier Proteins/metabolism , Immunity, Innate/immunology , Retroviridae Infections/immunology , Ubiquitin-Protein Ligases/metabolism , Animals , Antiviral Restriction Factors , Capsid/chemistry , Capsid/metabolism , Carrier Proteins/genetics , HEK293 Cells , Humans , Leukemia Virus, Murine/enzymology , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/immunology , Mice , Mice, Inbred C57BL , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/genetics , Polysaccharides, Bacterial/metabolism , Retroviridae Infections/metabolism , Retroviridae Infections/virology , THP-1 Cells , Tripartite Motif Proteins , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...