1
|
Wen ZT, Ellepola K, Wu H. MecA: A Multifunctional ClpP-Dependent and Independent Regulator in Gram-Positive Bacteria. Mol Microbiol 2025; 123:433-438. [PMID: 40070161 DOI: 10.1111/mmi.15356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/19/2025]
Abstract
MecA is a broadly conserved adaptor protein in Gram-positive bacteria, mediating the recognition and degradation of specific target proteins by ClpCP protease complexes. MecA binds target proteins, often through recognition of degradation tags or motifs, and delivers them to the ClpC ATPase, which unfolds and translocates the substrates into the ClpP protease barrel for degradation. MecA activity is tightly regulated through interactions with ClpC ATPase and other factors, ensuring precise control over protein degradation and cellular homeostasis. Beyond proteolysis, emerging evidence highlights a ClpP-independent role of MecA in modulating the function of its targets, including key enzymes and transcriptional factors involved in biosynthetic and metabolic pathways. However, the full scope and mechanisms of ClpP-independent MecA regulation remain unclear, warranting further investigation.
Collapse
Affiliation(s)
- Zezhang T Wen
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Department of Microbiology, Immunology and Parasitology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Kassapa Ellepola
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, Illinois, USA
| | - Hui Wu
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
2
|
Massoni SC, Evans NJ, Hantke I, Fenton C, Torpey JH, Collins KM, Krysztofinska EM, Muench JH, Thapaliya A, Martínez-Lumbreras S, Hart Ferrell S, Slater C, Wang X, Fekade R, Obwar S, Yin S, Vazquez A, Prior CB, Turgay K, Isaacson RL, Camp AH. MdfA is a novel ClpC adaptor protein that functions in the developing Bacillus subtilis spore. Genes Dev 2025; 39:gad.352498.124. [PMID: 40086879 PMCID: PMC11960690 DOI: 10.1101/gad.352498.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/13/2025] [Indexed: 03/16/2025]
Abstract
Bacterial protein degradation machinery consists of chaperone-protease complexes that play vital roles in bacterial growth and development and have sparked interest as novel antimicrobial targets. ClpC-ClpP (ClpCP) is one such chaperone-protease complex, recruited by adaptors to specific functions in the model bacterium Bacillus subtilis and other Gram-positive bacteria, including the pathogens Staphylococcus aureus and Mycobacterium tuberculosis Here we have identified a new ClpCP adaptor protein, MdfA (metabolic differentiation factor A; formerly YjbA), in a genetic screen for factors that help drive B. subtilis toward metabolic dormancy during spore formation. A knockout of mdfA stimulates gene expression in the developing spore, while aberrant expression of mdfA during vegetative growth is toxic. MdfA binds directly to ClpC to induce its oligomerization and ATPase activity, and this interaction is required for the in vivo effects of mdfA Finally, a cocrystal structure reveals that MdfA binds to the ClpC N-terminal domain at a location analogous to that on the M. tuberculosis ClpC1 protein where bactericidal cyclic peptides bind. Altogether, our data and that of an accompanying study by Riley and colleagues support a model in which MdfA induces ClpCP-mediated degradation of metabolic enzymes in the developing spore, helping drive it toward metabolic dormancy.
Collapse
Affiliation(s)
- Shawn C Massoni
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts 01075, USA
| | - Nicola J Evans
- Department of Chemistry, King's College London, London SE1 1DB, United Kingdom
| | - Ingo Hantke
- Institute for Microbiology, Leibniz Universität Hannover, Hannover 30419, Germany
| | - Colleen Fenton
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts 01075, USA
| | - James H Torpey
- Department of Chemistry, King's College London, London SE1 1DB, United Kingdom
| | - Katherine M Collins
- Department of Chemistry, King's College London, London SE1 1DB, United Kingdom
| | | | - Janina H Muench
- Department of Chemistry, King's College London, London SE1 1DB, United Kingdom
| | - Arjun Thapaliya
- Department of Chemistry, King's College London, London SE1 1DB, United Kingdom
| | | | - Sé Hart Ferrell
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts 01075, USA
| | - Celia Slater
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts 01075, USA
| | - Xinyue Wang
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts 01075, USA
| | - Ruth Fekade
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts 01075, USA
| | - Sandra Obwar
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts 01075, USA
| | - Siyu Yin
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts 01075, USA
| | - Alishba Vazquez
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts 01075, USA
| | - Christopher B Prior
- Department of Mathematical Sciences, Durham University, Durham DH1 3LE, United Kingdom
| | - Kürşad Turgay
- Institute for Microbiology, Leibniz Universität Hannover, Hannover 30419, Germany
- Max Planck Unit for the Science of Pathogens, Berlin 10117, Germany
| | - Rivka L Isaacson
- Department of Chemistry, King's College London, London SE1 1DB, United Kingdom;
| | - Amy H Camp
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts 01075, USA;
| |
Collapse
|
3
|
Annis MY, Ravenburg CM, van Wijk KJ. Uvr motifs regulate the chloroplast Clp chaperone-protease system. TRENDS IN PLANT SCIENCE 2025; 30:269-282. [PMID: 39448301 DOI: 10.1016/j.tplants.2024.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/12/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024]
Abstract
Chloroplast proteostasis relies on diverse proteases, including the essential Clp chaperone-protease system. Two chloroplast ClpC AAA+ chaperones and the plant-specific adaptor ClpF contain an Uvr motif with predicted coiled-coiled structures implicated in protein-protein interactions. Head-to-head contacts between Uvr motifs in middle (M)-domains regulate the oligomerization and activation of several bacterial Clp chaperones. Interestingly, in arabidopsis (Arabidopsis thaliana), this Uvr motif is found in six additional chloroplast proteins (Executer1, Executer2, and Uvr1-4). Here, we first summarize evidence that Uvr motifs regulate proteostasis in bacteria. Based on this evidence and recent results in arabidopsis, we postulate that arabidopsis Uvr motif proteins regulate chloroplast Clp proteolysis. We propose specific working hypotheses to test the function of the Uvr motif in chloroplast proteostasis.
Collapse
Affiliation(s)
- Marissa Y Annis
- Section of Plant Biology, School of Integrative Plant Sciences (SIPS), Cornell University, Ithaca, NY 14853, USA
| | - Claire M Ravenburg
- Section of Plant Biology, School of Integrative Plant Sciences (SIPS), Cornell University, Ithaca, NY 14853, USA
| | - Klaas J van Wijk
- Section of Plant Biology, School of Integrative Plant Sciences (SIPS), Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
4
|
Ellepola K, Shields RC, Kajfasz JK, Zhang H, Lemos JA, Wu H, Wen ZT. MecA in Streptococcus mutans is a multi-functional protein. mSphere 2024; 9:e0030824. [PMID: 39530674 PMCID: PMC11656736 DOI: 10.1128/msphere.00308-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024] Open
Abstract
Our recent studies have shown that deficiency of MecA in Streptococcus mutans significantly affects cell division, growth, and biofilm formation. In this study, an in vitro mixed-species model, proteomics, and affinity pull-down assays were used to further characterize the MecA-mediated regulation in S. mutans. The results showed that compared with the wild type, UA159, the mecA mutant significantly reduced its production of glucans and weakened its ability to facilitate mixed-species biofilm formation. Relative to the wild type, the mecA mutant also displayed unique characteristics, including colony morphology, growth rate, and biofilm formation that did not fully resemble any of the clpP, clpX, clpE, clpCE, and clpC individual or combinational mutants. Deletion of mecA was shown to result in alteration of >337 proteins, including down expression of GtfBC&D and adhesin P1. More than 277 proteins were differentially expressed in response to clpP deletion, including increased expression of GtfB. By cross-referencing the two proteomes, a distinctive set of proteins was found to be altered in the mecA mutant, indicating a ClpP-independent role of MecA in the regulation of S. mutans. When analyzed using affinity pull-down, ClpC, ClpX, ClpE, and CcpA were among the members identified in the MecA-associated complex. Further analysis using a bacterial two-hybrid system confirmed CcpA, ClpX, and ClpE as members of the MecA interactome. These results further suggest that MecA in S. mutans is more than an adapter of the Clp-proteolytic machinery, although the mechanism that underlies the Clp-independent regulation and its impact on S. mutans pathophysiology await further investigation. IMPORTANCE MecA is known as an adaptor protein that works in concerto with ATPase ClpC and protease ClpP in the regulated proteolysis machinery. The results presented here provide further evidence that MecA in S. mutans, a keystone cariogenic bacterium, plays a significant role in its ability to facilitate mixed-species biofilm formation, a trait critical to its cariogenicity. Proteomics analysis, along with affinity pull-down and bacterial two-hybrid system, further confirm that MecA can also regulate S. mutans physiology and biofilm formation through pathways independent of the Clp proteolytic machinery, although how it functions independently of Clp awaits further investigation.
Collapse
Affiliation(s)
- Kassapa Ellepola
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Robert C. Shields
- Department of Biological Sciences, Arkansas State University, Jonesboro, Arkansas, USA
| | - Jessica K. Kajfasz
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Hua Zhang
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Jose A. Lemos
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Hui Wu
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Zezhang T. Wen
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Department of Microbiology, Immunology and Parasitology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| |
Collapse
|
5
|
Morreale FE, Kleine S, Leodolter J, Junker S, Hoi DM, Ovchinnikov S, Okun A, Kley J, Kurzbauer R, Junk L, Guha S, Podlesainski D, Kazmaier U, Boehmelt G, Weinstabl H, Rumpel K, Schmiedel VM, Hartl M, Haselbach D, Meinhart A, Kaiser M, Clausen T. BacPROTACs mediate targeted protein degradation in bacteria. Cell 2022; 185:2338-2353.e18. [PMID: 35662409 PMCID: PMC9240326 DOI: 10.1016/j.cell.2022.05.009] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/15/2022] [Accepted: 05/10/2022] [Indexed: 12/14/2022]
Abstract
Hijacking the cellular protein degradation system offers unique opportunities for drug discovery, as exemplified by proteolysis-targeting chimeras. Despite their great promise for medical chemistry, so far, it has not been possible to reprogram the bacterial degradation machinery to interfere with microbial infections. Here, we develop small-molecule degraders, so-called BacPROTACs, that bind to the substrate receptor of the ClpC:ClpP protease, priming neo-substrates for degradation. In addition to their targeting function, BacPROTACs activate ClpC, transforming the resting unfoldase into its functional state. The induced higher-order oligomer was visualized by cryo-EM analysis, providing a structural snapshot of activated ClpC unfolding a protein substrate. Finally, drug susceptibility and degradation assays performed in mycobacteria demonstrate in vivo activity of BacPROTACs, allowing selective targeting of endogenous proteins via fusion to an established degron. In addition to guiding antibiotic discovery, the BacPROTAC technology presents a versatile research tool enabling the inducible degradation of bacterial proteins.
Collapse
Affiliation(s)
- Francesca E Morreale
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Stefan Kleine
- University of Duisburg-Essen, Center of Medical Biotechnology, Faculty of Biology, 45141 Essen, Germany
| | - Julia Leodolter
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Sabryna Junker
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - David M Hoi
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Stepan Ovchinnikov
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Anastasia Okun
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Juliane Kley
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Robert Kurzbauer
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Lukas Junk
- Saarland University, Organic Chemistry I, 66123 Saarbrücken, Germany
| | - Somraj Guha
- Saarland University, Organic Chemistry I, 66123 Saarbrücken, Germany
| | - David Podlesainski
- University of Duisburg-Essen, Center of Medical Biotechnology, Faculty of Biology, 45141 Essen, Germany
| | - Uli Kazmaier
- Saarland University, Organic Chemistry I, 66123 Saarbrücken, Germany
| | - Guido Boehmelt
- Boehringer Ingelheim RCV GmbH & Co KG, 1120 Vienna, Austria
| | | | - Klaus Rumpel
- Boehringer Ingelheim RCV GmbH & Co KG, 1120 Vienna, Austria
| | | | - Markus Hartl
- Max Perutz Laboratories, Vienna Biocenter, 1030 Vienna, Austria
| | - David Haselbach
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Anton Meinhart
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Markus Kaiser
- University of Duisburg-Essen, Center of Medical Biotechnology, Faculty of Biology, 45141 Essen, Germany.
| | - Tim Clausen
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria; Medical University of Vienna, 1030 Vienna, Austria.
| |
Collapse
|
6
|
Hawkins PME, Hoi DM, Cheung CY, Wang T, Quan D, Sasi VM, Liu DY, Linington RG, Jackson CJ, Oehlers SH, Cook GM, Britton WJ, Clausen T, Payne RJ. Potent Bactericidal Antimycobacterials Targeting the Chaperone ClpC1 Based on the Depsipeptide Natural Products Ecumicin and Ohmyungsamycin A. J Med Chem 2022; 65:4893-4908. [PMID: 35293761 DOI: 10.1021/acs.jmedchem.1c02122] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ohmyungsamycin A and ecumicin are structurally related cyclic depsipeptide natural products that possess activity against Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB). Herein, we describe the design and synthesis of a library of analogues of these two natural products using an efficient solid-phase synthesis and late-stage macrolactamization strategy. Lead analogues possessed potent activity against Mtb in vitro (minimum inhibitory concentration 125-500 nM) and were shown to inhibit protein degradation by the mycobacterial ClpC1-ClpP1P2 protease with an associated enhancement of ClpC1 ATPase activity. The most promising analogue from the series exhibited rapid bactericidal killing activity against Mtb, capable of sterilizing cultures after 7 days, and retained bactericidal activity against hypoxic non-replicating Mtb. This natural product analogue was also active in an in vivo zebrafish model of infection.
Collapse
Affiliation(s)
- Paige M E Hawkins
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - David M Hoi
- Research Institute of Molecular Pathology (IMP), Dr-Bohr-Gasse 7, Vienna 1030, Austria
| | - Chen-Yi Cheung
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin 9016, New Zealand
| | - Trixie Wang
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Diana Quan
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Vishnu Mini Sasi
- Research School of Chemistry, Australian National University, Acton, 2601 Australian Capital Territory, Australia
| | - Dennis Y Liu
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Roger G Linington
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Colin J Jackson
- Research School of Chemistry, Australian National University, Acton, 2601 Australian Capital Territory, Australia
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Gregory M Cook
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin 9016, New Zealand
| | - Warwick J Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Sydney, New South Wales 2006, Australia.,Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Tim Clausen
- Research Institute of Molecular Pathology (IMP), Dr-Bohr-Gasse 7, Vienna 1030, Austria
| | - Richard J Payne
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
7
|
Division of labor between the pore-1 loops of the D1 and D2 AAA+ rings coordinates substrate selectivity of the ClpAP protease. J Biol Chem 2021; 297:101407. [PMID: 34780718 PMCID: PMC8666677 DOI: 10.1016/j.jbc.2021.101407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 11/24/2022] Open
Abstract
ClpAP, an ATP-dependent protease consisting of ClpA, a double-ring hexameric unfoldase of the ATPases associated with diverse cellular activities superfamily, and the ClpP peptidase, degrades damaged and unneeded proteins to support cellular proteostasis. ClpA recognizes many protein substrates directly, but it can also be regulated by an adapter, ClpS, that modifies ClpA’s substrate profile toward N-degron substrates. Conserved tyrosines in the 12 pore-1 loops lining the central channel of the stacked D1 and D2 rings of ClpA are critical for degradation, but the roles of these residues in individual steps during direct or adapter-mediated degradation are poorly understood. Using engineered ClpA hexamers with zero, three, or six pore-1 loop mutations in each ATPases associated with diverse cellular activities superfamily ring, we found that active D1 pore loops initiate productive engagement of substrates, whereas active D2 pore loops are most important for mediating the robust unfolding of stable native substrates. In complex with ClpS, active D1 pore loops are required to form a high affinity ClpA•ClpS•substrate complex, but D2 pore loops are needed to “tug on” and remodel ClpS to transfer the N-degron substrate to ClpA. Overall, we find that the pore-1 loop tyrosines in D1 are critical for direct substrate engagement, whereas ClpS-mediated substrate delivery requires unique contributions from both the D1 and D2 pore loops. In conclusion, our study illustrates how pore loop engagement, substrate capture, and powering of the unfolding/translocation steps are distributed between the two rings of ClpA, illuminating new mechanistic features that may be common to double-ring protein unfolding machines.
Collapse
|
8
|
Abstract
The Borrelia spp. are tick-borne pathogenic spirochetes that include the agents of Lyme disease and relapsing fever. As part of their life cycle, the spirochetes traffic between the tick vector and the vertebrate host, which requires significant physiological changes and remodeling of their outer membranes and proteome. This crucial proteome resculpting is carried out by a diverse set of proteases, adaptor proteins, and related chaperones. Despite its small genome, Borrelia burgdorferi has dedicated a large percentage of its genome to proteolysis, including a full complement of ATP-dependent proteases. Energy-driven proteolysis appears to be an important physiological feature of this dual-life-cycle bacterium. The proteolytic arsenal of Borrelia is strategically deployed for disposal of proteins no longer required as they move from one stage to another or are transferred from one host to another. Likewise, the Borrelia spp. are systemic organisms that need to break down and move through host tissues and barriers, and so their unique proteolytic resources, both endogenous and borrowed, make movement more feasible. Both the Lyme disease and relapsing fever Borrelia spp. bind plasminogen as well as numerous components of the mammalian plasminogen-activating system. This recruitment capacity endows the spirochetes with a borrowed proteolytic competency that can lead to increased invasiveness.
Collapse
|
9
|
Izert MA, Klimecka MM, Górna MW. Applications of Bacterial Degrons and Degraders - Toward Targeted Protein Degradation in Bacteria. Front Mol Biosci 2021; 8:669762. [PMID: 34026843 PMCID: PMC8138137 DOI: 10.3389/fmolb.2021.669762] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/15/2021] [Indexed: 12/28/2022] Open
Abstract
A repertoire of proteolysis-targeting signals known as degrons is a necessary component of protein homeostasis in every living cell. In bacteria, degrons can be used in place of chemical genetics approaches to interrogate and control protein function. Here, we provide a comprehensive review of synthetic applications of degrons in targeted proteolysis in bacteria. We describe recent advances ranging from large screens employing tunable degradation systems and orthogonal degrons, to sophisticated tools and sensors for imaging. Based on the success of proteolysis-targeting chimeras as an emerging paradigm in cancer drug discovery, we discuss perspectives on using bacterial degraders for studying protein function and as novel antimicrobials.
Collapse
Affiliation(s)
| | | | - Maria Wiktoria Górna
- Structural Biology Group, Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, Warsaw, Poland
| |
Collapse
|
10
|
Kim G, Lee SG, Han S, Jung J, Jeong HS, Hyun JK, Rhee DK, Kim HM, Lee S. ClpL is a functionally active tetradecameric AAA+ chaperone, distinct from hexameric/dodecameric ones. FASEB J 2020; 34:14353-14370. [PMID: 32910525 DOI: 10.1096/fj.202000843r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/23/2020] [Accepted: 08/11/2020] [Indexed: 01/17/2023]
Abstract
AAA+ (ATPases associated with diverse cellular activities) chaperones are involved in a plethora of cellular activities to ensure protein homeostasis. The function of AAA+ chaperones is mostly modulated by their hexameric/dodecameric quaternary structures. Here we report the structural and biochemical characterizations of a tetradecameric AAA+ chaperone, ClpL from Streptococcus pneumoniae. ClpL exists as a tetradecamer in solution in the presence of ATP. The cryo-EM structure of ClpL at 4.5 Å resolution reveals a striking tetradecameric arrangement. Solution structures of ClpL derived from small-angle X-ray scattering data suggest that the tetradecameric ClpL could assume a spiral conformation found in active hexameric/dodecameric AAA+ chaperone structures. Vertical positioning of the middle domain accounts for the head-to-head arrangement of two heptameric rings. Biochemical activity assays with site-directed mutagenesis confirmed the critical roles of residues both in the integrity of the tetradecameric arrangement and activities of ClpL. Non-conserved Q321 and R670 are crucial in the heptameric ring assembly of ClpL. These results establish that ClpL is a functionally active tetradecamer, clearly distinct from hexameric/dodecameric AAA+ chaperones.
Collapse
Affiliation(s)
- Gyuhee Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Seong-Gyu Lee
- Center for Biomolecular and Cellular Structure, Institute for Basic Science (IBS), Daejeon, Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Seungsu Han
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Jaeeun Jung
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | | | - Jae-Kyung Hyun
- Korea Basic Science Institute, Cheongju, Korea.,Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Dong-Kwon Rhee
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Ho Min Kim
- Center for Biomolecular and Cellular Structure, Institute for Basic Science (IBS), Daejeon, Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Sangho Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea.,Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
11
|
Ziemski M, Leodolter J, Taylor G, Kerschenmeyer A, Weber-Ban E. Genome-wide interaction screen for Mycobacterium tuberculosis ClpCP protease reveals toxin-antitoxin systems as a major substrate class. FEBS J 2020; 288:111-126. [PMID: 32301575 DOI: 10.1111/febs.15335] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/12/2020] [Accepted: 04/14/2020] [Indexed: 12/30/2022]
Abstract
In Mycobacterium tuberculosis (Mtb), the Clp protease degradation pathway, mediated by the modular ClpCP and ClpXP protease complexes, is essential for growth and presents an attractive drug target. Employing a bacterial adenylate cyclase two-hybrid (BACTH) screening approach that we adapted to screen the proteome of an Mtb ORF library, we identify protein interaction partners of the ClpC1 chaperone on a genome-wide level. Our results demonstrate that bipartite type II toxin-antitoxin (TA) systems represent a major substrate class. Out of the 67 type II TA systems known in Mtb, 25 appear as ClpC1 interaction partners in the BACTH screen, including members of the VapBC, MazEF, and ParDE families, as well as a RelBE member that was identified biochemically. We show that antitoxins of the Vap and Rel families are degraded by ClpCP in vitro. We also demonstrate that ClpCP is responsible for mediating the N-end rule pathway, since the adaptor protein ClpS supports ClpC-dependent degradation of an N-end rule model substrate in vitro.
Collapse
Affiliation(s)
- Michal Ziemski
- Institute of Molecular Biology & Biophysics, ETH Zurich, Switzerland
| | - Julia Leodolter
- Institute of Molecular Biology & Biophysics, ETH Zurich, Switzerland
| | - Gabrielle Taylor
- Institute of Molecular Biology & Biophysics, ETH Zurich, Switzerland
| | | | - Eilika Weber-Ban
- Institute of Molecular Biology & Biophysics, ETH Zurich, Switzerland
| |
Collapse
|
12
|
Wolf NM, Lee H, Zagal D, Nam JW, Oh DC, Lee H, Suh JW, Pauli GF, Cho S, Abad-Zapatero C. Structure of the N-terminal domain of ClpC1 in complex with the antituberculosis natural product ecumicin reveals unique binding interactions. ACTA CRYSTALLOGRAPHICA SECTION D-STRUCTURAL BIOLOGY 2020; 76:458-471. [PMID: 32355042 PMCID: PMC7193532 DOI: 10.1107/s2059798320004027] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 03/22/2020] [Indexed: 11/10/2022]
Abstract
The biological processes related to protein homeostasis in Mycobacterium tuberculosis, the etiologic agent of tuberculosis, have recently been established as critical pathways for therapeutic intervention. Proteins of particular interest are ClpC1 and the ClpC1-ClpP1-ClpP2 proteasome complex. The structure of the potent antituberculosis macrocyclic depsipeptide ecumicin complexed with the N-terminal domain of ClpC1 (ClpC1-NTD) is presented here. Crystals of the ClpC1-NTD-ecumicin complex were monoclinic (unit-cell parameters a = 80.0, b = 130.0, c = 112.0 Å, β = 90.07°; space group P21; 12 complexes per asymmetric unit) and diffracted to 2.5 Å resolution. The structure was solved by molecular replacement using the self-rotation function to resolve space-group ambiguities. The new structure of the ecumicin complex showed a unique 1:2 (target:ligand) stoichiometry exploiting the intramolecular dyad in the α-helical fold of the target N-terminal domain. The structure of the ecumicin complex unveiled extensive interactions in the uniquely extended N-terminus, a critical binding site for the known cyclopeptide complexes. This structure, in comparison with the previously reported rufomycin I complex, revealed unique features that could be relevant for understanding the mechanism of action of these potential antituberculosis drug leads. Comparison of the ecumicin complex and the ClpC1-NTD-L92S/L96P double-mutant structure with the available structures of rufomycin I and cyclomarin A complexes revealed a range of conformational changes available to this small N-terminal helical domain and the minor helical alterations involved in the antibiotic-resistance mechanism. The different modes of binding and structural alterations could be related to distinct modes of action.
Collapse
Affiliation(s)
- Nina M Wolf
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Hyun Lee
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Daniel Zagal
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Joo Won Nam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Dong Chan Oh
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Hanki Lee
- Center for Nutraceutical and Pharmaceutical Materials, Myongji University, Cheoin-gu, Yongin-si, Gyeonggi-do 17058, Republic of Korea
| | - Joo Won Suh
- Center for Nutraceutical and Pharmaceutical Materials, Myongji University, Cheoin-gu, Yongin-si, Gyeonggi-do 17058, Republic of Korea
| | - Guido F Pauli
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sanghyun Cho
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Celerino Abad-Zapatero
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
13
|
Wolf NM, Lee H, Choules MP, Pauli GF, Phansalkar R, Anderson JR, Gao W, Ren J, Santarsiero BD, Lee H, Cheng J, Jin YY, Ho NA, Duc NM, Suh JW, Abad-Zapatero C, Cho S. High-Resolution Structure of ClpC1-Rufomycin and Ligand Binding Studies Provide a Framework to Design and Optimize Anti-Tuberculosis Leads. ACS Infect Dis 2019; 5:829-840. [PMID: 30990022 DOI: 10.1021/acsinfecdis.8b00276] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Addressing the urgent need to develop novel drugs against drug-resistant Mycobacterium tuberculosis ( M. tb) strains, ecumicin (ECU) and rufomycin I (RUFI) are being explored as promising new leads targeting cellular proteostasis via the caseinolytic protein ClpC1. Details of the binding topology and chemical mode of (inter)action of these cyclopeptides help drive further development of novel potency-optimized entities as tuberculosis drugs. ClpC1 M. tb protein constructs with mutations driving resistance to ECU and RUFI show reduced binding affinity by surface plasmon resonance (SPR). Despite certain structural similarities, ECU and RUFI resistant mutation sites did not overlap in their SPR binding patterns. SPR competition experiments show ECU prevents RUFI binding, whereas RUFI partially inhibits ECU binding. The X-ray structure of the ClpC1-NTD-RUFI complex reveals distinct differences compared to the previously reported ClpC1-NTD-cyclomarin A structure. Surprisingly, the complex structure revealed that the epoxide moiety of RUFI opened and covalently bound to ClpC1-NTD via the sulfur atom of Met1. Furthermore, RUFI analogues indicate that the epoxy group of RUFI is critical for binding and bactericidal activity. The outcomes demonstrate the significance of ClpC1 as a novel target and the importance of SAR analysis of identified macrocyclic peptides for drug discovery.
Collapse
Affiliation(s)
- Nina M. Wolf
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Hyun Lee
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Biophysics Core at the Research Resource Center, University of Illinois at Chicago, 1100 S. Ashland Street, Chicago, Illinois 60612, United States
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Mary P. Choules
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Guido F. Pauli
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Rasika Phansalkar
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Jeffrey R. Anderson
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Wei Gao
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Jinhong Ren
- Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, 900 S. Ashland Street, Chicago, Illinois 60612, United States
| | - Bernard D. Santarsiero
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, 900 S. Ashland Street, Chicago, Illinois 60612, United States
| | - Hanki Lee
- Center for Nutraceutical and Pharmaceutical Materials, College of Natural Sciences, Myongji University, 116 Myongji-ro, Cheoin-gu, Yongin-si, Gyeonggi-do 17058, Republic of Korea
| | - Jinhua Cheng
- Center for Nutraceutical and Pharmaceutical Materials, College of Natural Sciences, Myongji University, 116 Myongji-ro, Cheoin-gu, Yongin-si, Gyeonggi-do 17058, Republic of Korea
- Division of Bioscience and Bioinformatics, College of Natural Sciences, Myongji University, 116 Myongji-ro, Cheoin-gu, Yongin-si, Gyeonggi-do 17058, Republic of Korea
| | - Ying-Yu Jin
- Center for Nutraceutical and Pharmaceutical Materials, College of Natural Sciences, Myongji University, 116 Myongji-ro, Cheoin-gu, Yongin-si, Gyeonggi-do 17058, Republic of Korea
- Division of Bioscience and Bioinformatics, College of Natural Sciences, Myongji University, 116 Myongji-ro, Cheoin-gu, Yongin-si, Gyeonggi-do 17058, Republic of Korea
| | - Ngoc Anh Ho
- Center for Nutraceutical and Pharmaceutical Materials, College of Natural Sciences, Myongji University, 116 Myongji-ro, Cheoin-gu, Yongin-si, Gyeonggi-do 17058, Republic of Korea
| | - Nguyen Minh Duc
- Center for Nutraceutical and Pharmaceutical Materials, College of Natural Sciences, Myongji University, 116 Myongji-ro, Cheoin-gu, Yongin-si, Gyeonggi-do 17058, Republic of Korea
| | - Joo-Won Suh
- Center for Nutraceutical and Pharmaceutical Materials, College of Natural Sciences, Myongji University, 116 Myongji-ro, Cheoin-gu, Yongin-si, Gyeonggi-do 17058, Republic of Korea
- Division of Bioscience and Bioinformatics, College of Natural Sciences, Myongji University, 116 Myongji-ro, Cheoin-gu, Yongin-si, Gyeonggi-do 17058, Republic of Korea
| | - Celerino Abad-Zapatero
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, 900 S. Ashland Street, Chicago, Illinois 60612, United States
| | - Sanghyun Cho
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| |
Collapse
|
14
|
De A, Jorgensen AN, Beatty WL, Lemos J, Wen ZT. Deficiency of MecA in Streptococcus mutans Causes Major Defects in Cell Envelope Biogenesis, Cell Division, and Biofilm Formation. Front Microbiol 2018; 9:2130. [PMID: 30254619 PMCID: PMC6141683 DOI: 10.3389/fmicb.2018.02130] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/20/2018] [Indexed: 12/28/2022] Open
Abstract
MecA is an adaptor protein that guides the ClpC/P-mediated proteolysis. A S. mutans MecA-deficient mutant was constructed by double-crossover allelic exchange and analyzed for the effects of such a deficiency on cell biology and biofilm formation. Unlike the wild-type, UA159, the mecA mutant, TW416, formed mucoid and smooth colonies, severely clumped in broth and had a reduced growth rate. Transmission electron microscopy analysis revealed that TW416 grows primarily in chains of giant “swollen” cells with multiple asymmetric septa, unlike the coccoid form of UA159. As compared to UA159, biofilm formation by TW416 was significantly reduced regardless of the carbohydrate sources used for growth (P < 0.001). Western blot analysis of TW416 whole cell lysates showed a reduced expression of the glucosyltransferase GtfC and GtfB, as well as the P1 and WapA adhesins providing an explanation for the defective biofilm formation of TW416. When analyzed by a colorimetric assay, the cell wall phosphate of the mutant murein sacculi was almost 20-fold lower than the parent strain (P < 0.001). Interestingly, however, when analyzed using immunoblotting of the murein sacculi preps with UA159 whole cell antiserum as a probe, TW416 was shown to possess significantly higher signal intensity as compared to the wild-type. There is also evidence that MecA in S. mutans is more than an adaptor protein, although how it modulates the bacterial pathophysiology, including cell envelope biogenesis, cell division, and biofilm formation awaits further investigation.
Collapse
Affiliation(s)
- Arpan De
- Department of Comprehensive Dentistry and Biomaterials, University of Florida, Gainesville, FL, United States
| | - Ashton N Jorgensen
- Department of Comprehensive Dentistry and Biomaterials, University of Florida, Gainesville, FL, United States
| | - Wandy L Beatty
- Center of Oral and Craniofacial Biology, University of Florida, Gainesville, FL, United States
| | - Jose Lemos
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Zezhang T Wen
- Department of Comprehensive Dentistry and Biomaterials, University of Florida, Gainesville, FL, United States.,Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO, United States.,Department of Oral Biology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
15
|
Son M, Kaspar J, Ahn SJ, Burne RA, Hagen SJ. Threshold regulation and stochasticity from the MecA/ClpCP proteolytic system in Streptococcus mutans competence. Mol Microbiol 2018; 110:914-930. [PMID: 29873131 PMCID: PMC6281771 DOI: 10.1111/mmi.13992] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2018] [Indexed: 12/28/2022]
Abstract
Many bacterial species use the MecA/ClpCP proteolytic system to block entry into genetic competence. In Streptococcus mutans, MecA/ClpCP degrades ComX (also called SigX), an alternative sigma factor for the comY operon and other late competence genes. Although the mechanism of MecA/ClpCP has been studied in multiple Streptococcus species, its role within noisy competence pathways is poorly understood. S. mutans competence can be triggered by two different peptides, CSP and XIP, but it is not known whether MecA/ClpCP acts similarly for both stimuli, how it affects competence heterogeneity, and how its regulation is overcome. We have studied the effect of MecA/ClpCP on the activation of comY in individual S. mutans cells. Our data show that MecA/ClpCP is active under both XIP and CSP stimulation, that it provides threshold control of comY, and that it adds noise in comY expression. Our data agree quantitatively with a model in which MecA/ClpCP prevents adventitious entry into competence by sequestering or intercepting low levels of ComX. Competence is permitted when ComX levels exceed a threshold, but cell‐to‐cell heterogeneity in MecA levels creates variability in that threshold. Therefore, MecA/ClpCP provides a stochastic switch, located downstream of the already noisy comX, that enhances phenotypic diversity.
Collapse
Affiliation(s)
- M Son
- Department of Physics, University of Florida, Gainesville, FL 32611, USA
| | - J Kaspar
- Department of Oral Biology, University of Florida, Gainesville, FL 32610, USA
| | - S J Ahn
- Department of Oral Biology, University of Florida, Gainesville, FL 32610, USA
| | - R A Burne
- Department of Oral Biology, University of Florida, Gainesville, FL 32610, USA
| | - S J Hagen
- Department of Physics, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
16
|
Tanner AW, Carabetta VJ, Dubnau D. ClpC and MecA, components of a proteolytic machine, prevent Spo0A-P-dependent transcription without degradation. Mol Microbiol 2018; 108:178-186. [PMID: 29446505 PMCID: PMC5897911 DOI: 10.1111/mmi.13928] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2018] [Indexed: 01/23/2023]
Abstract
In Bacillus subtilis, a proteolytic machine composed of MecA, ClpC and ClpP degrades the transcription factor ComK, controlling its accumulation during growth. MecA also inhibits sporulation and biofilm formation by down-regulating spoIIG and sinI, genes that are dependent for their transcription on the phosphorylated protein Spo0A-P. Additionally, MecA has been shown to interact in vitro with Spo0A. Although the inhibitory effect on transcription requires MecA's binding partner ClpC, inhibition is not accompanied by the degradation of Spo0A, pointing to a previously unsuspected regulatory mechanism involving these proteins. Here, we further investigate the MecA and ClpC effects on Spo0A-P-dependent transcription. We show that MecA inhibits the transcription of several Spo0A-P activated genes, but fails to de-repress several Spo0A-P repressed promoters. This demonstrates that MecA and ClpC do not act by preventing the binding of Spo0A-P to its target promoters. Consistent with this, MecA by itself has no effect in vitro on the transcription from PspoIIG while the addition of both MecA and ClpC has a strong inhibitory effect. A complex of MecA and ClpC likely binds to Spo0A-P on its target promoters, preventing the activation of transcription. Thus, components of a degradative machine have been harnessed to directly repress transcription.
Collapse
Affiliation(s)
- Andrew W. Tanner
- Public Health Research Institute Center, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Valerie J. Carabetta
- Public Health Research Institute Center, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - David Dubnau
- Public Health Research Institute Center, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| |
Collapse
|
17
|
Carroni M, Franke KB, Maurer M, Jäger J, Hantke I, Gloge F, Linder D, Gremer S, Turgay K, Bukau B, Mogk A. Regulatory coiled-coil domains promote head-to-head assemblies of AAA+ chaperones essential for tunable activity control. eLife 2017; 6. [PMID: 29165246 PMCID: PMC5699869 DOI: 10.7554/elife.30120] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/22/2017] [Indexed: 12/20/2022] Open
Abstract
Ring-forming AAA+ chaperones exert ATP-fueled substrate unfolding by threading through a central pore. This activity is potentially harmful requiring mechanisms for tight repression and substrate-specific activation. The AAA+ chaperone ClpC with the peptidase ClpP forms a bacterial protease essential to virulence and stress resistance. The adaptor MecA activates ClpC by targeting substrates and stimulating ClpC ATPase activity. We show how ClpC is repressed in its ground state by determining ClpC cryo-EM structures with and without MecA. ClpC forms large two-helical assemblies that associate via head-to-head contacts between coiled-coil middle domains (MDs). MecA converts this resting state to an active planar ring structure by binding to MD interaction sites. Loss of ClpC repression in MD mutants causes constitutive activation and severe cellular toxicity. These findings unravel an unexpected regulatory concept executed by coiled-coil MDs to tightly control AAA+ chaperone activity.
Collapse
Affiliation(s)
- Marta Carroni
- Swedish Cryo-EM Facility, Science for Life Laboratory Stockholm University, Solna, Sweden
| | - Kamila B Franke
- DKFZ-ZMBH Alliance, Center for Molecular Biology of the University of Heidelberg (ZMBH) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Maurer
- DKFZ-ZMBH Alliance, Center for Molecular Biology of the University of Heidelberg (ZMBH) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jasmin Jäger
- DKFZ-ZMBH Alliance, Center for Molecular Biology of the University of Heidelberg (ZMBH) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ingo Hantke
- Institute for Microbiology, Leibniz Universität Hannover, Hannover, Germany
| | | | - Daniela Linder
- DKFZ-ZMBH Alliance, Center for Molecular Biology of the University of Heidelberg (ZMBH) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Gremer
- DKFZ-ZMBH Alliance, Center for Molecular Biology of the University of Heidelberg (ZMBH) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kürşad Turgay
- Institute for Microbiology, Leibniz Universität Hannover, Hannover, Germany
| | - Bernd Bukau
- DKFZ-ZMBH Alliance, Center for Molecular Biology of the University of Heidelberg (ZMBH) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Axel Mogk
- DKFZ-ZMBH Alliance, Center for Molecular Biology of the University of Heidelberg (ZMBH) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
18
|
Miller JM, Chaudhary H, Marsee JD. Phylogenetic analysis predicts structural divergence for proteobacterial ClpC proteins. J Struct Biol 2017; 201:52-62. [PMID: 29129755 DOI: 10.1016/j.jsb.2017.11.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/06/2017] [Accepted: 11/08/2017] [Indexed: 12/29/2022]
Abstract
Regulated proteolysis is required in all organisms for the removal of misfolded or degradation-tagged protein substrates in cellular quality control pathways. The molecular machines that catalyze this process are known as ATP-dependent proteases with examples that include ClpAP and ClpCP. Clp/Hsp100 subunits form ring-structures that couple the energy of ATP binding and hydrolysis to protein unfolding and subsequent translocation of denatured protein into the compartmentalized ClpP protease for degradation. Copies of the clpA, clpC, clpE, clpK, and clpL genes are present in all characterized bacteria and their gene products are highly conserved in structure and function. However, the evolutionary relationship between these proteins remains unclear. Here we report a comprehensive phylogenetic analysis that suggests divergent evolution yielded ClpA from an ancestral ClpC protein and that ClpE/ClpL represent intermediates between ClpA/ClpC. This analysis also identifies a group of proteobacterial ClpC proteins that are likely not functional in regulated proteolysis. Our results strongly suggest that bacterial ClpC proteins should not be assumed to all function identically due to the structural differences identified here.
Collapse
Affiliation(s)
- Justin M Miller
- Middle Tennessee State University, Department of Chemistry, 1301 East Main Street, Murfreesboro, TN 37132, United States.
| | - Hamza Chaudhary
- Middle Tennessee State University, Department of Chemistry, 1301 East Main Street, Murfreesboro, TN 37132, United States
| | - Justin D Marsee
- Middle Tennessee State University, Department of Chemistry, 1301 East Main Street, Murfreesboro, TN 37132, United States
| |
Collapse
|
19
|
Variability of Protein Structure Models from Electron Microscopy. Structure 2017; 25:592-602.e2. [PMID: 28262392 DOI: 10.1016/j.str.2017.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 01/10/2017] [Accepted: 02/11/2017] [Indexed: 11/23/2022]
Abstract
An increasing number of biomolecular structures are solved by electron microscopy (EM). However, the quality of structure models determined from EM maps vary substantially. To understand to what extent structure models are supported by information embedded in EM maps, we used two computational structure refinement methods to examine how much structures can be refined using a dataset of 49 maps with accompanying structure models. The extent of structure modification as well as the disagreement between refinement models produced by the two computational methods scaled inversely with the global and the local map resolutions. A general quantitative estimation of deviations of structures for particular map resolutions are provided. Our results indicate that the observed discrepancy between the deposited map and the refined models is due to the lack of structural information present in EM maps and thus these annotations must be used with caution for further applications.
Collapse
|
20
|
Olivares AO, Nager AR, Iosefson O, Sauer RT, Baker TA. Mechanochemical basis of protein degradation by a double-ring AAA+ machine. Nat Struct Mol Biol 2014; 21:871-5. [PMID: 25195048 PMCID: PMC4190165 DOI: 10.1038/nsmb.2885] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 08/06/2014] [Indexed: 02/08/2023]
Abstract
Molecular machines containing double or single AAA+ rings power energy-dependent protein degradation and other critical cellular processes, including disaggregation and remodeling of macromolecular complexes. How the mechanical activities of double-ring and single-ring AAA+ enzymes differ is unknown. Using single-molecule optical trapping, we determine how the double-ring ClpA enzyme from Escherichia coli mechanically degrades proteins in complex with the ClpP peptidase. We demonstrate that ClpA unfolds some protein substrates substantially faster than the single-ring ClpX enzyme, which also degrades substrates in collaboration with ClpP. We find that ClpA is a slower polypeptide translocase and moves in physical steps that are smaller and more regular than steps taken by ClpX. These direct measurements of protein unfolding and translocation define the core mechanochemical behavior of a double-ring AAA+ machine and provide insight into the degradation of proteins that unfold via metastable intermediates.
Collapse
Affiliation(s)
- Adrian O Olivares
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Andrew R Nager
- 1] Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [2]
| | - Ohad Iosefson
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Robert T Sauer
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Tania A Baker
- 1] Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [2] Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
21
|
Zeymer C, Barends TRM, Werbeck ND, Schlichting I, Reinstein J. Elements in nucleotide sensing and hydrolysis of the AAA+ disaggregation machine ClpB: a structure-based mechanistic dissection of a molecular motor. ACTA ACUST UNITED AC 2014; 70:582-95. [PMID: 24531492 PMCID: PMC3940203 DOI: 10.1107/s1399004713030629] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 11/07/2013] [Indexed: 12/12/2022]
Abstract
ATPases of the AAA+ superfamily are large oligomeric molecular machines that remodel their substrates by converting the energy from ATP hydrolysis into mechanical force. This study focuses on the molecular chaperone ClpB, the bacterial homologue of Hsp104, which reactivates aggregated proteins under cellular stress conditions. Based on high-resolution crystal structures in different nucleotide states, mutational analysis and nucleotide-binding kinetics experiments, the ATPase cycle of the C-terminal nucleotide-binding domain (NBD2), one of the motor subunits of this AAA+ disaggregation machine, is dissected mechanistically. The results provide insights into nucleotide sensing, explaining how the conserved sensor 2 motif contributes to the discrimination between ADP and ATP binding. Furthermore, the role of a conserved active-site arginine (Arg621), which controls binding of the essential Mg2+ ion, is described. Finally, a hypothesis is presented as to how the ATPase activity is regulated by a conformational switch that involves the essential Walker A lysine. In the proposed model, an unusual side-chain conformation of this highly conserved residue stabilizes a catalytically inactive state, thereby avoiding unnecessary ATP hydrolysis.
Collapse
Affiliation(s)
- Cathleen Zeymer
- Department of Biomolecular Mechanisms, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Thomas R M Barends
- Department of Biomolecular Mechanisms, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Nicolas D Werbeck
- Department of Biomolecular Mechanisms, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Ilme Schlichting
- Department of Biomolecular Mechanisms, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Jochen Reinstein
- Department of Biomolecular Mechanisms, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| |
Collapse
|
22
|
Doyle SM, Genest O, Wickner S. Protein rescue from aggregates by powerful molecular chaperone machines. Nat Rev Mol Cell Biol 2013; 14:617-29. [PMID: 24061228 DOI: 10.1038/nrm3660] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Protein quality control within the cell requires the interplay of many molecular chaperones and proteases. When this quality control system is disrupted, polypeptides follow pathways leading to misfolding, inactivity and aggregation. Among the repertoire of molecular chaperones are remarkable proteins that forcibly untangle protein aggregates, called disaggregases. Structural and biochemical studies have led to new insights into how these proteins collaborate with co-chaperones and utilize ATP to power protein disaggregation. Understanding how energy-dependent protein disaggregating machines function is universally important and clinically relevant, as protein aggregation is linked to medical conditions such as Alzheimer's disease, Parkinson's disease, amyloidosis and prion diseases.
Collapse
Affiliation(s)
- Shannon M Doyle
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bldg. 37, Room 5144, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|