Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 30
1.
Nat Commun ; 15(1): 3301, 2024 Apr 26.
Article En | MEDLINE | ID: mdl-38671004

Diphthamide is a modified histidine residue unique for eukaryotic translation elongation factor 2 (eEF2), a key ribosomal protein. Loss of this evolutionarily conserved modification causes developmental defects through unknown mechanisms. In a patient with compound heterozygous mutations in Diphthamide Biosynthesis 1 (DPH1) and impaired eEF2 diphthamide modification, we observe multiple defects in neural crest (NC)-derived tissues. Knockin mice harboring the patient's mutations and Xenopus embryos with Dph1 depleted also display NC defects, which can be attributed to reduced proliferation in the neuroepithelium. DPH1 depletion facilitates dissociation of eEF2 from ribosomes and association with p53 to promote transcription of the cell cycle inhibitor p21, resulting in inhibited proliferation. Knockout of one p21 allele rescues the NC phenotypes in the knockin mice carrying the patient's mutations. These findings uncover an unexpected role for eEF2 as a transcriptional coactivator for p53 to induce p21 expression and NC defects, which is regulated by diphthamide modification.


Cyclin-Dependent Kinase Inhibitor p21 , Histidine , Histidine/analogs & derivatives , Minor Histocompatibility Antigens , Neural Crest , Peptide Elongation Factor 2 , Tumor Suppressor Protein p53 , Tumor Suppressor Proteins , Animals , Neural Crest/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Humans , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Mice , Peptide Elongation Factor 2/metabolism , Peptide Elongation Factor 2/genetics , Histidine/metabolism , Ribosomes/metabolism , Mutation , Cell Proliferation , Xenopus laevis , Female , Gene Knock-In Techniques , Xenopus , Male , Mice, Knockout
2.
bioRxiv ; 2024 Apr 12.
Article En | MEDLINE | ID: mdl-38352471

All bacteria possess ATP-dependent proteases that destroy cytosolic proteins. These enzymes help cells mitigate proteotoxic stress, adapt to changing nutrient availability, regulate virulence phenotypes, and transition to pathogenic lifestyles. Moreover, ATP-dependent proteases have emerged as promising antibacterial and antivirulence targets in a variety of pathogens. The physiological roles of these proteases are largely defined by the complement of proteins that they degrade. Substrates are typically recognized in a highly selective manner, often via short unstructured sequences termed degrons. While a few degrons have been identified and rigorously characterized, we lack a systematic understanding of how proteases select valid degrons from the vast complexity of protein sequence space. Here, we describe a novel high-throughput screening approach in Escherichia coli that couples proteolysis of a protein toxin to cell survival. We used this method to screen a combinatorial library of C-terminal pentapeptide sequences for functionality as proteolytic degrons in wild type E. coli, and in strains lacking components of the ClpXP and ClpAP proteases. By examining the competitive enrichment of sequences over time, we found that about one percent of pentapeptide tags lead to toxin proteolysis. Interestingly, the most enriched degrons were ClpXP-dependent and highly similar to the ssrA tag, one of the most extensively characterized degrons in bacteria. Among ssrA-like sequences, we observed that specific upstream residues correlate with successful recognition. The lack of diversity among strongly enriched sequences suggests that ssrA-like tags comprise a uniquely potent class of short C-terminal degron in E. coli. Efficient proteolysis of substrates lacking such degrons likely requires adaptors or multivalent interactions. These findings broaden our understanding of the constraints that shape the bacterial proteolytic landscape. Our screening approach may be broadly applicable to probing aspects of proteolytic substrate selection in other bacterial systems.

3.
Microbiol Spectr ; 11(4): e0454822, 2023 08 17.
Article En | MEDLINE | ID: mdl-37341639

The prevalence of drug-resistant Mycobacterium tuberculosis infections has prompted extensive efforts to exploit new drug targets in this globally important pathogen. ClpC1, the unfoldase component of the essential ClpC1P1P2 protease, has emerged as one particularly promising antibacterial target. However, efforts to identify and characterize compounds that impinge on ClpC1 activity are constrained by our limited knowledge of Clp protease function and regulation. To expand our understanding of ClpC1 physiology, we employed a coimmunoprecipitation and mass spectrometry workflow to identify proteins that interact with ClpC1 in Mycolicibacterium smegmatis, a surrogate for M. tuberculosis. We identify a diverse panel of interaction partners, many of which coimmunoprecipitate with both the regulatory N-terminal domain and the ATPase core of ClpC1. Notably, our interactome analysis establishes MSMEI_3879, a truncated gene product unique to M. smegmatis, as a novel proteolytic substrate. Degradation of MSMEI_3879 by ClpC1P1P2 in vitro requires exposure of its N-terminal sequence, reinforcing the idea that ClpC1 selectively recognizes disordered motifs on substrates. Fluorescent substrates incorporating MSMEI_3879 may be useful in screening for novel ClpC1-targeting antibiotics to help address the challenge of M. tuberculosis drug resistance. IMPORTANCE Drug-resistant tuberculosis infections are a major challenge to global public health. Much effort has been invested in identifying new drug targets in the causative pathogen, Mycobacterium tuberculosis. One such target is the ClpC1 unfoldase. Compounds have been identified that kill M. tuberculosis by disrupting ClpC1 activity, yet the physiological function of ClpC1 in cells has remained poorly defined. Here, we identify interaction partners of ClpC1 in a model mycobacterium. By building a broader understanding of the role of this prospective drug target, we can more effectively develop compounds that inhibit its essential cellular activities.


Mycobacterium tuberculosis , Tuberculosis , Humans , Heat-Shock Proteins/genetics , Bacterial Proteins/metabolism , Molecular Chaperones/metabolism , Anti-Bacterial Agents/pharmacology , Tuberculosis/microbiology , Mycobacterium smegmatis/metabolism
4.
Front Microbiol ; 14: 1161764, 2023.
Article En | MEDLINE | ID: mdl-37138635

All clades of bacteria possess Hsp100/Clp family unfoldase enzymes that contribute to aspects of protein quality control. In Actinomycetota, these include ClpB, which functions as an independent chaperone and disaggregase, and ClpC, which cooperates with the ClpP1P2 peptidase to carry out regulated proteolysis of client proteins. We initially sought to algorithmically catalog Clp unfoldase orthologs from Actinomycetota into ClpB and ClpC categories. In the process, we uncovered a phylogenetically distinct third group of double-ringed Clp enzymes, which we term ClpI. ClpI enzymes are architecturally similar to ClpB and ClpC, with intact ATPase modules and motifs associated with substrate unfolding and translation. While ClpI possess an M-domain similar in length to that of ClpC, its N-terminal domain is more variable than the strongly conserved N-terminal domain of ClpC. Surprisingly, ClpI sequences are divisible into sub-classes that either possess or lack the LGF-motifs required for stable assembly with ClpP1P2, suggesting distinct cellular roles. The presence of ClpI enzymes likely provides bacteria with expanded complexity and regulatory control over protein quality control programs, supplementing the conserved roles of ClpB and ClpC.

5.
ACS Chem Biol ; 18(4): 724-733, 2023 04 21.
Article En | MEDLINE | ID: mdl-32083462

Proteolytic complexes in Mycobacterium tuberculosis (Mtb), the deadliest bacterial pathogen, are major foci in tuberculosis drug development programs. The Clp proteases, which are essential for Mtb viability, are high-priority targets. These proteases function through the collaboration of ClpP1P2, a barrel-shaped heteromeric peptidase, with associated ATP-dependent chaperones like ClpX and ClpC1 that recognize and unfold specific substrates in an ATP-dependent fashion. The critical interaction of the peptidase and its unfoldase partners is blocked by the competitive binding of acyldepsipeptide antibiotics (ADEPs) to the interfaces of the ClpP2 subunits. The resulting inhibition of Clp protease activity is lethal to Mtb. Here, we report the surprising discovery that a fragment of the ADEPs retains anti-Mtb activity yet stimulates rather than inhibits the ClpXP1P2-catalyzed degradation of proteins. Our data further suggest that the fragment stabilizes the ClpXP1P2 complex and binds ClpP1P2 in a fashion distinct from that of the intact ADEPs. A structure-activity relationship study of the bioactive fragment defines the pharmacophore and points the way toward the development of new drug leads for the treatment of tuberculosis.


Anti-Bacterial Agents , Mycobacterium tuberculosis , Tuberculosis , Humans , Adenosine Triphosphate/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Endopeptidase Clp/chemistry , Molecular Chaperones/metabolism , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Peptide Hydrolases/drug effects , Peptide Hydrolases/metabolism , Tuberculosis/drug therapy
6.
Microbiol Spectr ; 10(5): e0204222, 2022 10 26.
Article En | MEDLINE | ID: mdl-36214676

Tuberculosis is a leading cause of worldwide infectious mortality. The prevalence of multidrug-resistant Mycobacterium tuberculosis infections drives an urgent need to exploit new drug targets. One such target is the ATP-dependent protease ClpC1P1P2, which is strictly essential for viability. However, few proteolytic substrates of mycobacterial ClpC1P1P2 have been identified to date. Recent studies in Bacillus subtilis have shown that the orthologous ClpCP protease recognizes proteolytic substrates bearing posttranslational arginine phosphorylation. While several lines of evidence suggest that ClpC1P1P2 is similarly capable of recognizing phosphoarginine-bearing proteins, the existence of phosphoarginine modifications in mycobacteria has remained in question. Here, we confirm the presence of posttranslational phosphoarginine modifications in Mycolicibacterium smegmatis, a nonpathogenic surrogate of M. tuberculosis. Using a phosphopeptide enrichment workflow coupled with shotgun phosphoproteomics, we identified arginine phosphosites on several functionally diverse targets within the M. smegmatis proteome. Interestingly, phosphoarginine modifications are not upregulated by heat stress, suggesting divergent roles in mycobacteria and Bacillus. Our findings provide new evidence supporting the existence of phosphoarginine-mediated proteolysis by ClpC1P1P2 in mycobacteria and other actinobacterial species. IMPORTANCE Mycobacteria that cause tuberculosis infections employ proteolytic pathways that modulate cellular behavior by destroying specific proteins in a highly regulated manner. Some proteolytic enzymes have emerged as novel antibacterial targets against drug-resistant tuberculosis infections. However, we have only a limited understanding of how these enzymes function in the cell and how they select proteins for destruction. Some proteolytic enzymes are capable of recognizing proteins that carry an unusual chemical modification, arginine phosphorylation. Here, we confirm the existence of arginine phosphorylation in mycobacterial proteins. Our work expands our understanding of a promising drug target in an important global pathogen.


Mycobacterium tuberculosis , Tuberculosis , Humans , Phosphorylation , Proteome/metabolism , Phosphopeptides/metabolism , Bacterial Proteins/metabolism , Mycobacterium smegmatis/metabolism , Arginine/metabolism , Mycobacterium tuberculosis/metabolism , ATP-Dependent Proteases/metabolism , Anti-Bacterial Agents
7.
Protein Sci ; 28(4): 756-765, 2019 04.
Article En | MEDLINE | ID: mdl-30767302

IGF-motif loops project from the hexameric ring of ClpX and are required for docking with the self-compartmentalized ClpP peptidase, which consists of heptameric rings stacked back-to-back. Here, we show that ATP or ATPγS support assembly by changing the conformation of the ClpX ring, bringing the IGF loops closer to each other and allowing efficient multivalent contacts with docking clefts on ClpP. In single-chain ClpX pseudohexamers, deletion of one or two IGF loops modestly slows association with ClpP but strongly accelerates dissociation of ClpXP complexes. We probe how changes in the sequence and length of the IGF loops affect ClpX-ClpP interactions and show that deletion of one or two IGF loops slows ATP-dependent proteolysis by ClpXP. We also find that ClpXP degradation is less processive when two IGF loops are deleted.


ATPases Associated with Diverse Cellular Activities/metabolism , Endopeptidase Clp/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Molecular Chaperones/metabolism , ATPases Associated with Diverse Cellular Activities/chemistry , Adenosine Triphosphate/metabolism , Amino Acid Motifs , Amino Acid Sequence , Endopeptidase Clp/chemistry , Escherichia coli/chemistry , Escherichia coli Proteins/chemistry , Molecular Chaperones/chemistry , Protein Conformation , Protein Multimerization , Proteolysis , Substrate Specificity
8.
Int J Sports Med ; 40(1): 31-37, 2019 Jan.
Article En | MEDLINE | ID: mdl-30458551

Circulating venous bubbles after dives are associated with symptoms of decompression sickness in adults. Up to now it is not known to what extent children and adolescents are subjected to a bubble formation during their shallow dives and if there are possible indications for that. The aim of this pilot study is to investigate whether bubbles and/or symptoms occur after standardised repeated dives performed by young divers. 28 children and adolescents (13.5±1.1 years) carried out two 25 min dives to a depth of 10 m with a 90 min surface interval. Before and after, echocardiographic data were recorded and evaluated with regard to circulating bubbles with an extended Eftedal-Brubakk-Scale by 2 different examiners. Bubbles were observed for a total of 6 subjects, Grade I (n=5) and Grade III (n=1). None of them showed any symptoms of decompression sickness. No differences were established regarding potential influencing factors on bubble formation between the groups with and without bubbles. The results indicate that even relatively shallow and short dives can generate venous bubbles in children and adolescents. To what extent this relates to the decompression sickness or clinical symptoms cannot be validated at this point.


Diving/physiology , Embolism, Air/diagnosis , Adolescent , Child , Decompression Sickness/physiopathology , Echocardiography , Female , Humans , Male , Pilot Projects
9.
ACS Chem Biol ; 11(6): 1552-1560, 2016 06 17.
Article En | MEDLINE | ID: mdl-27003103

The ClpXP protease assembles in a reaction in which an ATP-bound ring hexamer of ClpX binds to one or both heptameric rings of the ClpP peptidase. Contacts between ClpX IGF-loops and clefts on a ClpP ring stabilize the complex. How ClpXP stability is maintained during the ATP-hydrolysis cycle that powers mechanical unfolding and translocation of protein substrates is poorly understood. Here, we use a real-time kinetic assay to monitor the effects of nucleotides on the assembly and disassembly of ClpXP. When ATP is present, complexes containing single-chain ClpX assemble via an intermediate and remain intact until transferred into buffers containing ADP or no nucleotides. ATP binding to high-affinity subunits of the ClpX hexamer prevents rapid dissociation, but additional subunits must be occupied to promote assembly. Small-molecule acyldepsipeptides, which compete with the IGF loops of ClpX for ClpP-cleft binding, cause exceptionally rapid dissociation of otherwise stable ClpXP complexes, suggesting that the IGF-loop interactions with ClpP must be highly dynamic. Our results indicate that the ClpX hexamer spends almost no time in an ATP-free state during the ATPase cycle, allowing highly processive degradation of protein substrates.


Adenosine Triphosphatases/chemistry , Adenosine Triphosphate/chemistry , Endopeptidase Clp/chemistry , Escherichia coli Proteins/chemistry , Molecular Chaperones/chemistry , ATPases Associated with Diverse Cellular Activities , Adenosine Diphosphate/chemistry , Adenosine Triphosphate/analogs & derivatives , Biosensing Techniques , Depsipeptides/chemistry , Hydrolysis , Kinetics , Models, Chemical , Protein Multimerization , Protein Stability , Protein Structure, Quaternary , Streptavidin/chemistry
10.
Chembiochem ; 16(13): 1875-1879, 2015 Sep 07.
Article En | MEDLINE | ID: mdl-26147653

The cyclic acyldepsipeptide (ADEP) antibiotics act by binding the ClpP peptidase and dysregulating its activity. Their exocyclic N-acylphenylalanine is thought to structurally mimic the ClpP-binding, (I/L)GF tripeptide loop of the peptidase's accessory ATPases. We found that ADEP analogues with exocyclic N-acyl tripeptides or dipeptides resembling the (I/L)GF motif were weak ClpP activators and had no bioactivity. In contrast, ADEP analogues possessing difluorophenylalanine N-capped with methyl-branched acyl groups-like the side chains of residues in the (I/L)GF motifs-were superior to the parent ADEP with respect to both ClpP activation and bioactivity. We contend that the ADEP's N-acylphenylalanine moiety is not simply a stand-in for the ATPases' (I/L)GF motif; it likely has physicochemical properties that are better suited for ClpP binding. Further, our finding that the methyl-branching on the acyl group of the ADEPs improves activity opens new avenues for optimization.

11.
Nat Struct Mol Biol ; 22(5): 411-6, 2015 May.
Article En | MEDLINE | ID: mdl-25866879

The hexameric AAA+ ring of Escherichia coli ClpX, an ATP-dependent machine for protein unfolding and translocation, functions with the ClpP peptidase to degrade target substrates. For efficient function, ClpX subunits must switch between nucleotide-loadable (L) and nucleotide-unloadable (U) conformations, but the roles of switching are uncertain. Moreover, it is controversial whether working AAA+-ring enzymes assume symmetric or asymmetric conformations. Here, we show that a covalent ClpX ring with one subunit locked in the U conformation catalyzes robust ATP hydrolysis, with each unlocked subunit able to bind and hydrolyze ATP, albeit with highly asymmetric position-specific affinities. Preventing U↔L interconversion in one subunit alters the cooperativity of ATP hydrolysis and reduces the efficiency of substrate binding, unfolding and degradation, showing that conformational switching enhances multiple aspects of wild-type ClpX function. These results support an asymmetric and probabilistic model of AAA+-ring activity.


Adenosine Triphosphatases/metabolism , Endopeptidase Clp/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Molecular Chaperones/metabolism , Protein Conformation , Protein Unfolding , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphate/metabolism , Binding Sites , Models, Molecular , Nucleotides/metabolism , Protein Binding , Protein Subunits/metabolism
12.
Structure ; 23(2): 352-63, 2015 Feb 03.
Article En | MEDLINE | ID: mdl-25620000

F-BAR domains control membrane interactions in endocytosis, cytokinesis, and cell signaling. Although they are generally thought to bind curved membranes containing negatively charged phospholipids, numerous functional studies argue that differences in lipid-binding selectivities of F-BAR domains are functionally important. Here, we compare membrane-binding properties of the Saccharomyces cerevisiae F-BAR domains in vitro and in vivo. Whereas some F-BAR domains (such as Bzz1p and Hof1p F-BARs) bind equally well to all phospholipids, the F-BAR domain from the RhoGAP Rgd1p preferentially binds phosphoinositides. We determined X-ray crystal structures of F-BAR domains from Hof1p and Rgd1p, the latter bound to an inositol phosphate. The structures explain phospholipid-binding selectivity differences and reveal an F-BAR phosphoinositide binding site that is fully conserved in a mammalian RhoGAP called Gmip and is partly retained in certain other F-BAR domains. Our findings reveal previously unappreciated determinants of F-BAR domain lipid-binding specificity and provide a basis for its prediction from sequence.


Binding Sites/genetics , GTPase-Activating Proteins/chemistry , Inositol Phosphates/metabolism , Models, Molecular , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Amino Acid Sequence , Crystallography, X-Ray , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Inositol Phosphates/genetics , Molecular Sequence Data , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Species Specificity
13.
Proc Natl Acad Sci U S A ; 111(43): E4587-95, 2014 Oct 28.
Article En | MEDLINE | ID: mdl-25267638

Caseinolytic peptidase P (ClpP), a double-ring peptidase with 14 subunits, collaborates with ATPases associated with diverse activities (AAA+) partners to execute ATP-dependent protein degradation. Although many ClpP enzymes self-assemble into catalytically active homo-tetradecamers able to cleave small peptides, the Mycobacterium tuberculosis enzyme consists of discrete ClpP1 and ClpP2 heptamers that require a AAA+ partner and protein-substrate delivery or a peptide agonist to stabilize assembly of the active tetradecamer. Here, we show that cyclic acyldepsipeptides (ADEPs) and agonist peptides synergistically activate ClpP1P2 by mimicking AAA+ partners and substrates, respectively, and determine the structure of the activated complex. Our studies establish the basis of heteromeric ClpP1P2 assembly and function, reveal tight coupling between the conformations of each ring, show that ADEPs bind only to one ring but appear to open the axial pores of both rings, provide a foundation for rational drug development, and suggest strategies for studying the roles of individual ClpP1 and ClpP2 rings in Clp-family proteolysis.


Adenosine Triphosphatases/metabolism , Bacterial Proteins/chemistry , Models, Biological , Mycobacterium tuberculosis/enzymology , Peptide Hydrolases/metabolism , Protein Subunits/chemistry , Bacterial Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , Enzyme Activation , Enzyme Stability , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Protein Binding , Protein Multimerization , Protein Subunits/metabolism , Substrate Specificity
14.
Chembiochem ; 15(15): 2216-20, 2014 Oct 13.
Article En | MEDLINE | ID: mdl-25212124

The development of new antibacterial agents, particularly those with unique biological targets, is essential to keep pace with the inevitable emergence of drug resistance in pathogenic bacteria. We identified the minimal structural component of the cyclic acyldepsipeptide (ADEP) antibiotics that exhibits antibacterial activity. We found that N-acyldifluorophenylalanine fragments function via the same mechanism of action as ADEPs, as evidenced by the requirement of ClpP for the fragments' antibacterial activity, the ability of fragments to activate Bacillus subtilis ClpP in vitro, and the capacity of an N-acyldifluorophenylalanine affinity matrix to capture ClpP from B. subtilis cell lysates. N-acyldifluorophenylalanine fragments are much simpler in structure than the full ADEPs and are also highly amenable to structural diversification. Thus, the stage has been set for the development of non-peptide activators of ClpP that can be used as antibacterial agents.


Anti-Bacterial Agents/pharmacology , Bacillus subtilis/drug effects , Depsipeptides/pharmacology , Endopeptidase Clp/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Bacillus subtilis/enzymology , Depsipeptides/chemistry , Dose-Response Relationship, Drug , Endopeptidase Clp/chemistry , Endopeptidase Clp/metabolism , Enzyme Activation/drug effects , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship
15.
Cell ; 158(3): 647-58, 2014 Jul 31.
Article En | MEDLINE | ID: mdl-25083874

ClpXP and other AAA+ proteases recognize, mechanically unfold, and translocate target proteins into a chamber for proteolysis. It is not known whether these remarkable molecular machines operate by a stochastic or sequential mechanism or how power strokes relate to the ATP-hydrolysis cycle. Single-molecule optical trapping allows ClpXP unfolding to be directly visualized and reveals translocation steps of ∼1-4 nm in length, but how these activities relate to solution degradation and the physical properties of substrate proteins remains unclear. By studying single-molecule degradation using different multidomain substrates and ClpXP variants, we answer many of these questions and provide evidence for stochastic unfolding and translocation. We also present a mechanochemical model that accounts for single-molecule, biochemical, and structural results for our observation of enzymatic memory in translocation stepping, for the kinetics of translocation steps of different sizes, and for probabilistic but highly coordinated subunit activity within the ClpX ring.


Endopeptidase Clp/chemistry , Endopeptidase Clp/metabolism , Adenosine Triphosphate/metabolism , Escherichia coli Proteins/chemistry , Models, Molecular , Protein Structure, Tertiary , Protein Unfolding , Proteolysis
16.
Mol Microbiol ; 93(4): 617-28, 2014 Aug.
Article En | MEDLINE | ID: mdl-24976069

Mycobacterial Clp-family proteases function via collaboration of the heteromeric ClpP1P2 peptidase with a AAA+ partner, ClpX or ClpC1. These enzymes are essential for M. tuberculosis viability and are validated antibacterial drug targets, but the requirements for assembly and regulation of functional proteolytic complexes are poorly understood. Here, we report the reconstitution of protein degradation by mycobacterial Clp proteases in vitro and describe novel features of these enzymes that distinguish them from orthologues in other bacteria. Both ClpX and ClpC1 catalyse ATP-dependent unfolding and degradation of native protein substrates in conjunction with ClpP1P2, but neither mediates protein degradation with just ClpP1 or ClpP2. ClpP1P2 alone has negligible peptidase activity, but is strongly stimulated by translocation of protein substrates into ClpP1P2 by either AAA+ partner. Interestingly, our results support a model in which both binding of a AAA+ partner and protein-substrate delivery are required to stabilize active ClpP1P2. Our model has implications for therapeutically targeting ClpP1P2 in dormant M. tuberculosis, and our reconstituted systems should facilitate identification of novel Clp protease inhibitors and activators.


Bacterial Proteins/metabolism , Endopeptidase Clp/metabolism , Heat-Shock Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Models, Biological , Protein Folding , Proteolysis
17.
J Am Chem Soc ; 136(5): 1922-9, 2014 Feb 05.
Article En | MEDLINE | ID: mdl-24422534

The cyclic acyldepsipeptide (ADEP) antibiotics are a new class of antibacterial agents that kill bacteria via a mechanism that is distinct from all clinically used drugs. These molecules bind and dysregulate the activity of the ClpP peptidase. The potential of these antibiotics as antibacterial drugs has been enhanced by the elimination of pharmacological liabilities through medicinal chemistry efforts. Here, we demonstrate that the ADEP conformation observed in the ADEP-ClpP crystal structure is fortified by transannular hydrogen bonding and can be further stabilized by judicious replacement of constituent amino acids within the peptidolactone core structure with more conformationally constrained counterparts. Evidence supporting constraint of the molecule into the bioactive conformer was obtained by measurements of deuterium-exchange kinetics of hydrogens that were proposed to be engaged in transannular hydrogen bonds. We show that the rigidified ADEP analogs bind and activate ClpP at lower concentrations in vitro. Remarkably, these compounds have up to 1200-fold enhanced antibacterial activity when compared to those with the peptidolactone core structure common to two ADEP natural products. This study compellingly demonstrates how rational modulation of conformational dynamics may be used to improve the bioactivities of natural products.


Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Depsipeptides/chemistry , Depsipeptides/pharmacology , Anti-Bacterial Agents/chemical synthesis , Crystallography, X-Ray , Depsipeptides/chemical synthesis , Enterococcus faecalis/drug effects , Hydrogen Bonding , Microbial Sensitivity Tests , Protein Conformation , Staphylococcus aureus/drug effects , Streptococcus pneumoniae/drug effects , Structure-Activity Relationship
18.
ACS Chem Biol ; 8(12): 2669-77, 2013 Dec 20.
Article En | MEDLINE | ID: mdl-24047344

There is rapidly mounting evidence that intracellular proteases in bacteria are compelling targets for antibacterial drugs. Multiple reports suggest that the human pathogen Mycobacterium tuberculosis and other actinobacteria may be particularly sensitive to small molecules that perturb the activities of self-compartmentalized peptidases, which catalyze intracellular protein turnover as components of ATP-dependent proteolytic machines. Here, we report chemical syntheses and evaluations of structurally diverse ß-lactones, which have a privileged structure for selective, suicide inhibition of the self-compartmentalized ClpP peptidase. ß-Lactones with certain substituents on the α- and ß-carbons were found to be toxic to M. tuberculosis. Using an affinity-labeled analogue of a bioactive ß-lactone in a series of chemical proteomic experiments, we selectively captured the ClpP1P2 peptidase from live cultures of two different actinobacteria that are related to M. tuberculosis. Importantly, we found that the growth inhibitory ß-lactones also inactivate the M. tuberculosis ClpP1P2 peptidase in vitro via formation of a covalent adduct at the ClpP2 catalytic serine. Given the potent antibacterial activity of these compounds and their medicinal potential, we sought to identify innate mechanisms of resistance. Using a genome mining strategy, we identified a genetic determinant of ß-lactone resistance in Streptomyces coelicolor, a non-pathogenic relative of M. tuberculosis. Collectively, these findings validate the potential of ClpP inhibition as a strategy in antibacterial drug development and define a mechanism by which bacteria could resist the toxic effects of ClpP inhibitors.


Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Drug Resistance, Bacterial/drug effects , Lactones/chemistry , Mycobacterium tuberculosis/drug effects , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Chromatography, Liquid , Drug Resistance, Bacterial/genetics , Genetic Complementation Test , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Lactones/pharmacology , Microbial Sensitivity Tests , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Proteolysis , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Streptomyces coelicolor/chemistry , Streptomyces coelicolor/enzymology , Streptomyces coelicolor/genetics , Structure-Activity Relationship , Tandem Mass Spectrometry
19.
Structure ; 21(7): 1214-24, 2013 Jul 02.
Article En | MEDLINE | ID: mdl-23791944

The epidermal growth factor receptor (EGFR) is implicated in human cancers and is the target of several classes of therapeutic agents, including antibody-based drugs. Here, we describe X-ray crystal structures of the extracellular region of EGFR in complex with three inhibitory nanobodies, the variable domains of heavy chain only antibodies (VHH). VHH domains, the smallest natural antigen-binding modules, are readily engineered for diagnostic and therapeutic applications. All three VHH domains prevent ligand-induced EGFR activation, but use two distinct mechanisms. 7D12 sterically blocks ligand binding to EGFR in a manner similar to that of cetuximab. EgA1 and 9G8 bind an epitope near the EGFR domain II/III junction, preventing receptor conformational changes required for high-affinity ligand binding and dimerization. This epitope is accessible to the convex VHH paratope but inaccessible to the flatter paratope of monoclonal antibodies. Appreciating the modes of binding and inhibition of these VHH domains will aid in developing them for tumor imaging and/or cancer therapy.


ErbB Receptors/chemistry , Single-Domain Antibodies/chemistry , Antibodies, Monoclonal, Humanized/chemistry , Antineoplastic Agents/chemistry , Binding Sites , Binding, Competitive , Cetuximab , Crystallography, X-Ray , Cystine/chemistry , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Single-Domain Antibodies/metabolism
20.
Cell ; 153(3): 628-39, 2013 Apr 25.
Article En | MEDLINE | ID: mdl-23622246

ClpX, a AAA+ ring homohexamer, uses the energy of ATP binding and hydrolysis to power conformational changes that unfold and translocate target proteins into the ClpP peptidase for degradation. In multiple crystal structures, some ClpX subunits adopt nucleotide-loadable conformations, others adopt unloadable conformations, and each conformational class exhibits substantial variability. Using mutagenesis of individual subunits in covalently tethered hexamers together with fluorescence methods to assay the conformations and nucleotide-binding properties of these subunits, we demonstrate that dynamic interconversion between loadable and unloadable conformations is required to couple ATP hydrolysis by ClpX to mechanical work. ATP binding to different classes of subunits initially drives staged allosteric changes, which set the conformation of the ring to allow hydrolysis and linked mechanical steps. Subunit switching between loadable and unloadable conformations subsequently isomerizes or resets the configuration of the nucleotide-loaded ring and is required for mechanical function.


Adenosine Triphosphatases/chemistry , Adenosine Triphosphate/metabolism , Endopeptidase Clp/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , Molecular Chaperones/chemistry , Nucleotides/metabolism , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Endopeptidase Clp/genetics , Endopeptidase Clp/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Models, Molecular , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Protein Conformation , Protein Folding , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Proteolysis
...