1
|
Khosravi A, Nnamdi P, May A, Slattery K, Sammelson RE, Shi WQ. Structural Modifications Reveal Dual Functions of the C-4 Carbonyl Group in the Fatty Acid Chain of Ipomoeassin F. Molecules 2025; 30:400. [PMID: 39860269 PMCID: PMC11767275 DOI: 10.3390/molecules30020400] [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: 10/29/2024] [Revised: 12/18/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Ipomoeassin F (Ipom-F) is a plant-derived macrocyclic resin glycoside that potently inhibits cancer cell growth through blockage of Sec61-mediated protein translocation at the endoplasmic reticulum. Recently, detailed structural information on how Ipom-F binds to Sec61α was obtained using Cryo-EM, which discovered that polar interactions between asparagine-300 (N300) in Sec61α and four oxygens in Ipom-F are crucial. One of the four oxygens is from the carbonyl group at C-4 of the fatty acid chain. In contrast, our previous structure-activity relationship (SAR) studies suggest that the carbonyl group is not essential. To resolve this discrepancy, we designed and synthesized two new open-chain analogues (10 and 11); 10 without the C-4 carbonyl had a dramatic activity loss, whereas 11 with an amide functional group was even more potent than Ipom-F. These new SAR data, in conjunction with some previous SAR information, imply two functional roles of the C-4 carbonyl: (1) to form H-bonds with N300; and (2) to regulate interactions of the fatty acid chain with membrane lipids. Impacts of these dual functions on antiproliferation depend on the overall structure of an Ipom-F derivative. Moreover, 11 can serve as a lead compound for developing future amino acid/peptide-modified analogues of Ipom-F with improved therapeutic properties.
Collapse
Affiliation(s)
- Arman Khosravi
- Department of Chemistry, Ball State University, Muncie, IN 47306, USA; (A.K.); (P.N.); (K.S.); (R.E.S.)
| | - Precious Nnamdi
- Department of Chemistry, Ball State University, Muncie, IN 47306, USA; (A.K.); (P.N.); (K.S.); (R.E.S.)
| | - Alexa May
- Chemistry Department, Michigan State University, East Lansing, MI 48824, USA;
| | - Kelsey Slattery
- Department of Chemistry, Ball State University, Muncie, IN 47306, USA; (A.K.); (P.N.); (K.S.); (R.E.S.)
| | - Robert E. Sammelson
- Department of Chemistry, Ball State University, Muncie, IN 47306, USA; (A.K.); (P.N.); (K.S.); (R.E.S.)
| | - Wei Q. Shi
- Department of Chemistry, Ball State University, Muncie, IN 47306, USA; (A.K.); (P.N.); (K.S.); (R.E.S.)
| |
Collapse
|
2
|
Chang J, Pickard A, Herrera JA, O'Keefe S, Garva R, Hartshorn M, Hoyle A, Dingle L, Knox J, Jowitt TA, Coy M, Wong J, Reid A, Lu Y, Zeltz C, Venkateswaran RV, Caswell PT, High S, Gullberg D, Kadler KE. Endocytic recycling is central to circadian collagen fibrillogenesis and disrupted in fibrosis. eLife 2025; 13:RP95842. [PMID: 39812558 PMCID: PMC11735028 DOI: 10.7554/elife.95842] [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] [Indexed: 01/16/2025] Open
Abstract
Collagen-I fibrillogenesis is crucial to health and development, where dysregulation is a hallmark of fibroproliferative diseases. Here, we show that collagen-I fibril assembly required a functional endocytic system that recycles collagen-I to assemble new fibrils. Endogenous collagen production was not required for fibrillogenesis if exogenous collagen was available, but the circadian-regulated vacuolar protein sorting (VPS) 33b and collagen-binding integrin α11 subunit were crucial to fibrillogenesis. Cells lacking VPS33B secrete soluble collagen-I protomers but were deficient in fibril formation, thus secretion and assembly are separately controlled. Overexpression of VPS33B led to loss of fibril rhythmicity and overabundance of fibrils, which was mediated through integrin α11β1. Endocytic recycling of collagen-I was enhanced in human fibroblasts isolated from idiopathic pulmonary fibrosis, where VPS33B and integrin α11 subunit were overexpressed at the fibrogenic front; this correlation between VPS33B, integrin α11 subunit, and abnormal collagen deposition was also observed in samples from patients with chronic skin wounds. In conclusion, our study showed that circadian-regulated endocytic recycling is central to homeostatic assembly of collagen fibrils and is disrupted in diseases.
Collapse
Affiliation(s)
- Joan Chang
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Adam Pickard
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Jeremy A Herrera
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Sarah O'Keefe
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Richa Garva
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Matthew Hartshorn
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Anna Hoyle
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Lewis Dingle
- Blond McIndoe Laboratories, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - John Knox
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Thomas A Jowitt
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Madeleine Coy
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Jason Wong
- Blond McIndoe Laboratories, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Adam Reid
- Blond McIndoe Laboratories, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Yinhui Lu
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Cédric Zeltz
- Department of Biomedicine and Centre for Cancer Biomarkers, Norwegian Center of Excellence, University of BergenBergenNorway
| | - Rajamiyer V Venkateswaran
- Manchester University National Health Service Foundation Trust, Manchester Academic Health Science CentreManchesterUnited Kingdom
| | - Patrick T Caswell
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Stephen High
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Donald Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, Norwegian Center of Excellence, University of BergenBergenNorway
| | - Karl E Kadler
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| |
Collapse
|
3
|
Sorout N, Helms V. Toward Understanding the Mechanism of Client-Selective Small Molecule Inhibitors of the Sec61 Translocon. J Mol Recognit 2025; 38:e3108. [PMID: 39394908 DOI: 10.1002/jmr.3108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/28/2024] [Accepted: 09/27/2024] [Indexed: 10/14/2024]
Abstract
The Sec61 translocon mediates the translocation of numerous, newly synthesized precursor proteins into the lumen of the endoplasmic reticulum or their integration into its membrane. Recently, structural biology revealed conformations of idle or substrate-engaged Sec61, and likewise its interactions with the accessory membrane proteins Sec62, Sec63, and TRAP, respectively. Several natural and synthetic small molecules have been shown to block Sec61-mediated protein translocation. Since this is a key step in protein biogenesis, broad inhibition is generally cytotoxic, which may be problematic for a putative drug target. Interestingly, several compounds exhibit client-selective modes of action, such that only translocation of certain precursor proteins was affected. Here, we discuss recent advances of structural biology, molecular modelling, and molecular screening that aim to use Sec61 as feasible drug target.
Collapse
Affiliation(s)
- Nidhi Sorout
- Center for Bioinformatics, Saarland University, Saarbrücken, Saarland, Germany
| | - Volkhard Helms
- Center for Bioinformatics, Saarland University, Saarbrücken, Saarland, Germany
| |
Collapse
|
4
|
Akolgo GA, Asiedu KB, Amewu RK. Exploring Mycolactone-The Unique Causative Toxin of Buruli Ulcer: Biosynthetic, Synthetic Pathways, Biomarker for Diagnosis, and Therapeutic Potential. Toxins (Basel) 2024; 16:528. [PMID: 39728786 PMCID: PMC11678992 DOI: 10.3390/toxins16120528] [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: 09/19/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Mycolactone is a complex macrolide toxin produced by Mycobacterium ulcerans, the causative agent of Buruli ulcer. The aim of this paper is to review the chemistry, biosynthetic, and synthetic pathways of mycolactone A/B to help develop an understanding of the mode of action of these polyketides as well as their therapeutic potential. The synthetic work has largely been driven by the desire to afford researchers enough (≥100 mg) of the pure toxins for systematic biological studies toward understanding their very high biological activities. The review focuses on pioneering studies of Kishi which elaborate first-, second-, and third-generation approaches to the synthesis of mycolactones A/B. The three generations focused on the construction of the key intermediates required for the mycolactone synthesis. Synthesis of the first generation involves assignment of the relative and absolute stereochemistry of the mycolactones A and B. This was accomplished by employing a linear series of 17 chemical steps (1.3% overall yield) using the mycolactone core. The second generation significantly improved the first generation in three ways: (1) by optimizing the selection of protecting groups; (2) by removing needless protecting group adjustments; and (3) by enhancing the stereoselectivity and overall synthetic efficiency. Though the synthetic route to the mycolactone core was longer than the first generation, the overall yield was significantly higher (8.8%). The third-generation total synthesis was specifically aimed at an efficient, scalable, stereoselective, and shorter synthesis of mycolactone. The synthesis of the mycolactone core was achieved in 14 linear chemical steps with 19% overall yield. Furthermore, a modular synthetic approach where diverse analogues of mycolactone A/B were synthesized via a cascade of catalytic and/or asymmetric reactions as well as several Pd-catalyzed key steps coupled with hydroboration reactions were reviewed. In addition, the review discusses how mycolactone is employed in the diagnosis of Buruli ulcer with emphasis on detection methods of mass spectrometry, immunological assays, RNA aptamer techniques, and fluorescent-thin layer chromatography (f-TLC) methods as diagnostic tools. We examined studies of the structure-activity relationship (SAR) of various analogues of mycolactone. The paper highlights the multiple biological consequences associated with mycolactone such as skin ulceration, host immunomodulation, and analgesia. These effects are attributed to various proposed mechanisms of actions including Wiskott-Aldrich Syndrome protein (WASP)/neural Wiskott-Aldrich Syndrome protein (N-WASP) inhibition, Sec61 translocon inhibition, angiotensin II type 2 receptor (AT2R) inhibition, and inhibition of mTOR. The possible application of novel mycolactone analogues produced based on SAR investigations as therapeutic agents for the treatment of inflammatory disorders and inflammatory pain are discussed. Additionally, their therapeutic potential as anti-viral and anti-cancer agents have also been addressed.
Collapse
Affiliation(s)
| | - Kingsley Bampoe Asiedu
- Department of Neglected Tropical Diseases, World Health Organization, 1211 Geneva, Switzerland;
| | | |
Collapse
|
5
|
Wong HH, Crudgington DRK, Siu L, Sanyal S. Flaviviruses induce ER-specific remodelling of protein synthesis. PLoS Pathog 2024; 20:e1012766. [PMID: 39621795 PMCID: PMC11637433 DOI: 10.1371/journal.ppat.1012766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 12/12/2024] [Accepted: 11/20/2024] [Indexed: 12/14/2024] Open
Abstract
Flaviviruses orchestrate a unique remodelling of the endoplasmic reticulum (ER) to facilitate translation and processing of their polyprotein, giving rise to virus replication compartments. While the signal recognition particle (SRP)-dependent pathway is the canonical route for ER-targeting of nascent cellular membrane proteins, it is unknown whether flaviviruses rely on this mechanism. Here we show that Zika virus bypasses the SRP receptor via extensive interactions between the viral non-structural proteins and the host translational machinery. Remarkably, Zika virus appears to maintain ER-localised translation via NS3-SRP54 interaction instead, unlike other viruses such as influenza. Viral proteins engage SRP54 and the translocon, selectively enriching for factors supporting membrane expansion and lipid metabolism while excluding RNA binding and antiviral stress granule proteins. Our findings reveal a sophisticated viral strategy to rewire host protein synthesis pathways and create a replication-favourable subcellular niche, providing insights into viral adaptation.
Collapse
Affiliation(s)
- Ho Him Wong
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR
| | | | - Lewis Siu
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR
| | - Sumana Sanyal
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR
- Sir William Dunn School of Pathology, South Parks Road, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
6
|
Hsieh LTH, Hall BS, Newcombe J, Mendum TA, Santana-Varela S, Umrania Y, Deery MJ, Shi WQ, Diaz-Delgado J, Salguero FJ, Simmonds RE. Mycolactone causes destructive Sec61-dependent loss of the endothelial glycocalyx and vessel basement membrane: a new indirect mechanism driving tissue necrosis in Mycobacterium ulcerans infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.21.529382. [PMID: 36865118 PMCID: PMC9980099 DOI: 10.1101/2023.02.21.529382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
The drivers of tissue necrosis in Mycobacterium ulcerans infection (Buruli ulcer disease) have historically been ascribed solely to the directly cytotoxic action of the diffusible exotoxin, mycolactone. However, its role in the clinically-evident vascular component of disease aetiology remains poorly explained. We have now dissected mycolactone's effects on primary vascular endothelial cells in vitro and in vivo. We show that mycolactone-induced changes in endothelial morphology, adhesion, migration, and permeability are dependent on its action at the Sec61 translocon. Unbiased quantitative proteomics identified a profound effect on proteoglycans, driven by rapid loss of type II transmembrane proteins of the Golgi, including enzymes required for glycosaminoglycan (GAG) synthesis, combined with a reduction in the core proteins themselves. Loss of the glycocalyx is likely to be of particular mechanistic importance, since knockdown of galactosyltransferase II (beta-1,3-galactotransferase 6; B3GALT6), the GAG linker-building enzyme, phenocopied the permeability and phenotypic changes induced by mycolactone. Additionally, mycolactone depleted many secreted basement membrane components and microvascular basement membranes were disrupted in vivo. Remarkably, exogenous addition of laminin-511 reduced endothelial cell rounding, restored cell attachment and reversed the defective migration caused by mycolactone. Hence supplementing mycolactone-depleted extracellular matrix may be a future therapeutic avenue, to improve wound healing rates.
Collapse
Affiliation(s)
| | - Belinda S Hall
- Dept of Microbial Sciences, School of Bioscience and Medicine, University of Surrey
| | - Jane Newcombe
- Dept of Microbial Sciences, School of Bioscience and Medicine, University of Surrey
| | - Tom A Mendum
- Dept of Microbial Sciences, School of Bioscience and Medicine, University of Surrey
| | - Sonia Santana-Varela
- Dept of Microbial Sciences, School of Bioscience and Medicine, University of Surrey
| | - Yagnesh Umrania
- Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK
| | - Michael J Deery
- Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK
| | - Wei Q Shi
- Department of Chemistry, Ball State University, Muncie, IN 47306, USA
| | - Josué Diaz-Delgado
- Texas A&M Veterinary Medical Diagnostic Laboratory, College Station, Texas, USA
| | | | - Rachel E Simmonds
- Dept of Microbial Sciences, School of Bioscience and Medicine, University of Surrey
| |
Collapse
|
7
|
Robeson L, Casanova‐Morales N, Burgos‐Bravo F, Alfaro‐Valdés HM, Lesch R, Ramírez‐Álvarez C, Valdivia‐Delgado M, Vega M, Matute RA, Schekman R, Wilson CAM. Characterization of the interaction between the Sec61 translocon complex and ppαF using optical tweezers. Protein Sci 2024; 33:e4996. [PMID: 38747383 PMCID: PMC11094780 DOI: 10.1002/pro.4996] [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: 10/19/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 05/19/2024]
Abstract
The Sec61 translocon allows the translocation of secretory preproteins from the cytosol to the endoplasmic reticulum lumen during polypeptide biosynthesis. These proteins possess an N-terminal signal peptide (SP) which docks at the translocon. SP mutations can abolish translocation and cause diseases, suggesting an essential role for this SP/Sec61 interaction. However, a detailed biophysical characterization of this binding is still missing. Here, optical tweezers force spectroscopy was used to characterize the kinetic parameters of the dissociation process between Sec61 and the SP of prepro-alpha-factor. The unbinding parameters including off-rate constant and distance to the transition state were obtained by fitting rupture force data to Dudko-Hummer-Szabo models. Interestingly, the translocation inhibitor mycolactone increases the off-rate and accelerates the SP/Sec61 dissociation, while also weakening the interaction. Whereas the translocation deficient mutant containing a single point mutation in the SP abolished the specificity of the SP/Sec61 binding, resulting in an unstable interaction. In conclusion, we characterize quantitatively the dissociation process between the signal peptide and the translocon, and how the unbinding parameters are modified by a translocation inhibitor.
Collapse
Affiliation(s)
- Luka Robeson
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y FarmacéuticasUniversidad de ChileSantiagoChile
| | - Nathalie Casanova‐Morales
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y FarmacéuticasUniversidad de ChileSantiagoChile
- Facultad de Artes LiberalesUniversidad Adolfo IbáñezSantiagoChile
| | - Francesca Burgos‐Bravo
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y FarmacéuticasUniversidad de ChileSantiagoChile
- California Institute for Quantitative Biosciences, Howard Hughes Medical InstituteUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Hilda M. Alfaro‐Valdés
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y FarmacéuticasUniversidad de ChileSantiagoChile
| | - Robert Lesch
- Department of Molecular and Cellular Biology, Howard Hughes Medical InstituteUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Carolina Ramírez‐Álvarez
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y FarmacéuticasUniversidad de ChileSantiagoChile
| | - Mauricio Valdivia‐Delgado
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y FarmacéuticasUniversidad de ChileSantiagoChile
| | - Marcela Vega
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y FarmacéuticasUniversidad de ChileSantiagoChile
| | - Ricardo A. Matute
- Centro Integrativo de Biología y Química Aplicada (CIBQA)Universidad Bernardo O'HigginsSantiagoChile
- Division of Chemistry and Chemical EngineeringCalifornia Institute of TechnologyPasadenaCaliforniaUSA
| | - Randy Schekman
- Department of Molecular and Cellular Biology, Howard Hughes Medical InstituteUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Christian A. M. Wilson
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y FarmacéuticasUniversidad de ChileSantiagoChile
| |
Collapse
|
8
|
Lyu Z, Genereux JC. Quantitative Measurement of Transthyretin Mistargeting by Proximity Labeling and Parallel Reaction Monitoring. FRONTIERS IN CHEMICAL BIOLOGY 2023; 2:1288188. [PMID: 38173467 PMCID: PMC10764115 DOI: 10.3389/fchbi.2023.1288188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Proximity labeling is a powerful approach for characterizing subcellular proteomes. We recently demonstrated that proximity labeling can be used to identify mistrafficking of secretory proteins, such as occurs during pre-emptive quality control (pre-QC) following endoplasmic reticulum (ER) stress. This assay depends on protein quantification by immunoblotting and densitometry, which sometimes suffers from poor sensitivity. Here, we integrate parallel reaction monitoring (PRM) mass spectrometry to enable a more quantitative platform, and assess how chemical ER stressors impact pre-QC of the model secretory protein transthyretin in HEK293T cells. We find that some drug treatments affect labeling efficiency, which can be controlled for by normalizing to APEX2 auto-labeling. While some chemical ER stress inducers including Brefeldin A and thapsigargin induce pre-QC, tunicamycin and dithiothreitol do not, indicating ER stress alone is not sufficient. This finding contrasts with the canonical model of pre-QC induction, and establishes the utility of our platform.
Collapse
Affiliation(s)
- Ziqi Lyu
- Department of Chemistry, University of California, Riverside, Riverside, CA, United States
| | - Joseph C. Genereux
- Department of Chemistry, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
9
|
Itskanov S, Wang L, Junne T, Sherriff R, Xiao L, Blanchard N, Shi WQ, Forsyth C, Hoepfner D, Spiess M, Park E. A common mechanism of Sec61 translocon inhibition by small molecules. Nat Chem Biol 2023; 19:1063-1071. [PMID: 37169959 PMCID: PMC11458068 DOI: 10.1038/s41589-023-01337-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 04/12/2023] [Indexed: 05/13/2023]
Abstract
The Sec61 complex forms a protein-conducting channel in the endoplasmic reticulum membrane that is required for secretion of soluble proteins and production of many membrane proteins. Several natural and synthetic small molecules specifically inhibit Sec61, generating cellular effects that are useful for therapeutic purposes, but their inhibitory mechanisms remain unclear. Here we present near-atomic-resolution structures of human Sec61 inhibited by a comprehensive panel of structurally distinct small molecules-cotransin, decatransin, apratoxin, ipomoeassin, mycolactone, cyclotriazadisulfonamide and eeyarestatin. All inhibitors bind to a common lipid-exposed pocket formed by the partially open lateral gate and plug domain of Sec61. Mutations conferring resistance to the inhibitors are clustered at this binding pocket. The structures indicate that Sec61 inhibitors stabilize the plug domain in a closed state, thereby preventing the protein-translocation pore from opening. Our study provides the atomic details of Sec61-inhibitor interactions and the structural framework for further pharmacological studies and drug design.
Collapse
Affiliation(s)
- Samuel Itskanov
- Biophysics Graduate Program, University of California, Berkeley, Berkeley, CA, USA
| | - Laurie Wang
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Tina Junne
- Biozentrum, University of Basel, Basel, Switzerland
| | - Rumi Sherriff
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Li Xiao
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Nicolas Blanchard
- Université de Haute-Alsace, Université de Strasbourg, CNRS, LIMA, UMR 7042, Mulhouse, France
| | - Wei Q Shi
- Department of Chemistry, Ball State University, Muncie, IN, USA
| | - Craig Forsyth
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Dominic Hoepfner
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Forum 1 Novartis Campus, Basel, Switzerland
| | | | - Eunyong Park
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
10
|
Lyu Z, Genereux JC. Quantitative Measurement of Secretory Protein Mistargeting by Proximity Labeling and Parallel Reaction Monitoring. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.19.549095. [PMID: 37503147 PMCID: PMC10370094 DOI: 10.1101/2023.07.19.549095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Proximity labeling is a powerful approach for characterizing subcellular proteomes. We recently demonstrated that proximity labeling can be used to identify mistrafficking of secretory proteins, such as occurs during pre-emptive quality control (pre-QC) following endoplasmic reticulum (ER) stress. This assay depends on protein quantification by immunoblotting and densitometry, which is only semi-quantitative and suffers from poor sensitivity. Here, we integrate parallel reaction monitoring mass spectrometry to enable a more quantitative platform for ER import. PRM as opposed to densitometry improves quantification of transthyretin mistargeting while also achieving at least a ten-fold gain in sensitivity. The multiplexing of PRM also enabled us to evaluate a series of normalization approaches, revealing that normalization to auto-labeled APEX2 peroxidase is necessary to account for drug treatment-dependent changes in labeling efficiency. We apply this approach to systematically characterize the relationship between chemical ER stressors and ER pre-QC induction in HEK293T cells. Using dual-FLAG-tagged transthyretin (FLAGTTR) as a model secretory protein, we find that Brefeldin A treatment as well as ER calcium depletion cause pre-QC, while tunicamycin and dithiothreitol do not, indicating ER stress alone is not sufficient. This finding contrasts with the canonical model of pre-QC induction, and establishes the utility of our platform.
Collapse
Affiliation(s)
- Ziqi Lyu
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521
| | - Joseph C. Genereux
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521
| |
Collapse
|
11
|
Schäfer AB, Steenhuis M, Jim KK, Neef J, O’Keefe S, Whitehead RC, Swanton E, Wang B, Halbedel S, High S, van Dijl JM, Luirink J, Wenzel M. Dual Action of Eeyarestatin 24 on Sec-Dependent Protein Secretion and Bacterial DNA. ACS Infect Dis 2023; 9:253-269. [PMID: 36637435 PMCID: PMC9926488 DOI: 10.1021/acsinfecdis.2c00404] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Indexed: 01/14/2023]
Abstract
Eeyarestatin 24 (ES24) is a promising new antibiotic with broad-spectrum activity. It shares structural similarity with nitrofurantoin (NFT), yet appears to have a distinct and novel mechanism: ES24 was found to inhibit SecYEG-mediated protein transport and membrane insertion in Gram-negative bacteria. However, possible additional targets have not yet been explored. Moreover, its activity was notably better against Gram-positive bacteria, for which its mechanism of action had not yet been investigated. We have used transcriptomic stress response profiling, phenotypic assays, and protein secretion analyses to investigate the mode of action of ES24 in comparison with NFT using the Gram-positive model bacterium Bacillus subtilis and have compared our findings to Gram-negative Escherichia coli. Here, we show the inhibition of Sec-dependent protein secretion in B. subtilis and additionally provide evidence for DNA damage, probably caused by the generation of reactive derivatives of ES24. Interestingly, ES24 caused a gradual dissipation of the membrane potential, which led to delocalization of cytokinetic proteins and subsequent cell elongation in E. coli. However, none of those effects were observed in B. subtilis, thereby suggesting that ES24 displays distinct mechanistic differences with respect to Gram-positive and Gram-negative bacteria. Despite its structural similarity to NFT, ES24 profoundly differed in our phenotypic analysis, which implies that it does not share the NFT mechanism of generalized macromolecule and structural damage. Importantly, ES24 outperformed NFT in vivo in a zebrafish embryo pneumococcal infection model. Our results suggest that ES24 not only inhibits the Sec translocon, but also targets bacterial DNA and, in Gram-negative bacteria, the cell membrane.
Collapse
Affiliation(s)
- Ann-Britt Schäfer
- Division
of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Maurice Steenhuis
- Molecular
Microbiology, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Kin Ki Jim
- Department
of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers - Location Vrije Universiteit
Amsterdam, 1081 HZ Amsterdam, The Netherlands
- Amsterdam
Institute for Infection and Immunity, Amsterdam
University Medical Centers, 1081 HZ Amsterdam, The Netherlands
| | - Jolanda Neef
- Department
of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, 9700 RB Groningen, The Netherlands
| | - Sarah O’Keefe
- School
of
Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Roger C. Whitehead
- School
of Chemistry, Faculty of Science and Engineering, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Eileithyia Swanton
- School
of
Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Biwen Wang
- Bacterial
Cell Biology and Physiology, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Sven Halbedel
- FG11
Division of Enteropathogenic Bacteria and Legionella, Robert Koch Institute, 38855 Wernigerode, Germany
- Institute
for Medical Microbiology and Hospital Hygiene, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Stephen High
- School
of
Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Jan Maarten van Dijl
- Department
of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, 9700 RB Groningen, The Netherlands
| | - Joen Luirink
- Molecular
Microbiology, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Michaela Wenzel
- Division
of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| |
Collapse
|
12
|
Itskanov S, Park E. Mechanism of Protein Translocation by the Sec61 Translocon Complex. Cold Spring Harb Perspect Biol 2023; 15:a041250. [PMID: 35940906 PMCID: PMC9808579 DOI: 10.1101/cshperspect.a041250] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The endoplasmic reticulum (ER) is a major site for protein synthesis, folding, and maturation in eukaryotic cells, responsible for production of secretory proteins and most integral membrane proteins. The universally conserved protein-conducting channel Sec61 complex mediates core steps in these processes by translocating hydrophilic polypeptide segments of client proteins across the ER membrane and integrating hydrophobic transmembrane segments into the membrane. The Sec61 complex associates with several other molecular machines and enzymes to enable substrate engagement with the channel and coordination of protein translocation with translation, protein folding, and/or post-translational modifications. Recent cryo-electron microscopy and functional studies of these translocon complexes have greatly advanced our mechanistic understanding of Sec61-dependent protein biogenesis at the ER. Here, we will review the current models for how the Sec61 channel performs its functions in coordination with partner complexes.
Collapse
Affiliation(s)
- Samuel Itskanov
- Biophysics Graduate Program
- California Institute for Quantitative Biosciences
| | - Eunyong Park
- California Institute for Quantitative Biosciences
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, USA
| |
Collapse
|
13
|
da Hora GCA, Nguyen JDM, Swanson JMJ. Can membrane composition traffic toxins? Mycolactone and preferential membrane interactions. Biophys J 2022; 121:4260-4270. [PMID: 36258678 PMCID: PMC9703097 DOI: 10.1016/j.bpj.2022.10.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/31/2022] [Accepted: 10/13/2022] [Indexed: 12/14/2022] Open
Abstract
Mycolactone is a cytotoxic and immunosuppressive macrolide produced by Mycobacterium ulcerans and the sole causative agent of the neglected tropical skin disease Buruli ulcer. The toxin acts by invading host cells and interacting with intracellular targets to disrupt multiple fundamental cellular processes. Mycolactone's amphiphilic nature enables strong interactions with lipophilic environments, including cellular membranes; however, the specificity of these interactions and the role of membranes in the toxin's pathogenicity remain unknown. It is likely that preferential interactions with lipophilic carriers play a key role in the toxin's distribution in the host, which, if understood, could provide insights to aid in the development of needed diagnostics for Buruli ulcer disease. In this work, molecular dynamics simulations were combined with enhanced free-energy sampling to characterize mycolactone's association with and permeation through models of the mammalian endoplasmic reticulum (ER) and plasma membranes (PMs). We find that increased order in the PMs not only leads to a different permeation mechanism compared with that in the ER membrane but also an energetic driving force for ER localization. Increased hydration, membrane deformation, and preferential interactions with unsaturated lipid tails stabilize the toxin in the ER membrane, while disruption of lipid packing is a destabilizing force in the PMs.
Collapse
Affiliation(s)
| | - John D M Nguyen
- Department of Chemistry, University of Utah, Salt Lake City, Utah
| | | |
Collapse
|
14
|
O'Keefe S, Pool MR, High S. Membrane protein biogenesis at the ER: the highways and byways. FEBS J 2022; 289:6835-6862. [PMID: 33960686 DOI: 10.1111/febs.15905] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 01/13/2023]
Abstract
The Sec61 complex is the major protein translocation channel of the endoplasmic reticulum (ER), where it plays a central role in the biogenesis of membrane and secretory proteins. Whilst Sec61-mediated protein translocation is typically coupled to polypeptide synthesis, suggestive of significant complexity, an obvious characteristic of this core translocation machinery is its surprising simplicity. Over thirty years after its initial discovery, we now understand that the Sec61 complex is in fact the central piece of an elaborate jigsaw puzzle, which can be partly solved using new research findings. We propose that the Sec61 complex acts as a dynamic hub for co-translational protein translocation at the ER, proactively recruiting a range of accessory complexes that enhance and regulate its function in response to different protein clients. It is now clear that the Sec61 complex does not have a monopoly on co-translational insertion, with some transmembrane proteins preferentially utilising the ER membrane complex instead. We also have a better understanding of post-insertion events, where at least one membrane-embedded chaperone complex can capture the newly inserted transmembrane domains of multi-span proteins and co-ordinate their assembly into a native structure. Having discovered this array of Sec61-associated components and competitors, our next challenge is to understand how they act together in order to expand the range and complexity of the membrane proteins that can be synthesised at the ER. Furthermore, this diversity of components and pathways may open up new opportunities for targeted therapeutic interventions designed to selectively modulate protein biogenesis at the ER.
Collapse
Affiliation(s)
- Sarah O'Keefe
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Martin R Pool
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Stephen High
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| |
Collapse
|
15
|
Lyu Z, Sycks MM, Espinoza MF, Nguyen KK, Montoya MR, Galapate CM, Mei L, Genereux JC. Monitoring Protein Import into the Endoplasmic Reticulum in Living Cells with Proximity Labeling. ACS Chem Biol 2022; 17:1963-1977. [PMID: 35675579 DOI: 10.1021/acschembio.2c00405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The proper trafficking of eukaryotic proteins is essential to cellular function. Genetic, environmental, and other stresses can induce protein mistargeting and, in turn, threaten cellular protein homeostasis. Current methods for measuring protein mistargeting are difficult to translate to living cells, and thus the role of cellular signaling networks in stress-dependent protein mistargeting processes, such as ER pre-emptive quality control (ER pQC), is difficult to parse. Herein, we use genetically encoded peroxidases to characterize protein import into the endoplasmic reticulum (ER). We show that the ERHRP/cytAPEX pair provides good selectivity and sensitivity for both multiplexed protein labeling and for identifying protein mistargeting, using the known ER pQC substrate transthyretin (TTR). Although ERHRP labeling induces formation of detergent-resistant TTR aggregates, this is minimized by using low ERHRP expression, without loss of labeling efficiency. cytAPEX labeling recovers TTR that is mistargeted as a consequence of Sec61 inhibition or ER stress-induced ER pQC. Furthermore, we discover that stress-free activation of the ER stress-associated transcription factor ATF6 recapitulates the TTR import deficiency of ER pQC. Hence, proximity labeling is an effective strategy for characterizing factors that influence ER protein import in living cells.
Collapse
Affiliation(s)
- Ziqi Lyu
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Melody M Sycks
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Mateo F Espinoza
- Graduate Program of Microbiology, University of California, Riverside, California 92521, United States
| | - Khanh K Nguyen
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Maureen R Montoya
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Cheska M Galapate
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Liangyong Mei
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Joseph C Genereux
- Department of Chemistry, University of California, Riverside, California 92521, United States.,Graduate Program of Microbiology, University of California, Riverside, California 92521, United States
| |
Collapse
|
16
|
Synthesis, Biological Evaluation and Docking Studies of Ring-Opened Analogues of Ipomoeassin F. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27144419. [PMID: 35889292 PMCID: PMC9320607 DOI: 10.3390/molecules27144419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022]
Abstract
The plant-derived macrocyclic resin glycoside ipomoeassin F (Ipom-F) binds to Sec61α and significantly disrupts multiple aspects of Sec61-mediated protein biogenesis at the endoplasmic reticulum, ultimately leading to cell death. However, extensive assessment of Ipom-F as a molecular tool and a therapeutic lead is hampered by its limited production scale, largely caused by intramolecular assembly of the macrocyclic ring. Here, using in vitro and/or in cellula biological assays to explore the first series of ring-opened analogues for the ipomoeassins, and indeed all resin glycosides, we provide clear evidence that macrocyclic integrity is not required for the cytotoxic inhibition of Sec61-dependent protein translocation by Ipom-F. Furthermore, our modeling suggests that open-chain analogues of Ipom-F can interact with multiple sites on the Sec61α subunit, most likely located at a previously identified binding site for mycolactone and/or the so-called lateral gate. Subsequent in silico-aided design led to the discovery of the stereochemically simplified analogue 3 as a potent, alternative lead compound that could be synthesized much more efficiently than Ipom-F and will accelerate future ipomoeassin research in chemical biology and drug discovery. Our work may also inspire further exploration of ring-opened analogues of other resin glycosides.
Collapse
|
17
|
Leznicki P, Schneider HO, Harvey JV, Shi WQ, High S. Co-translational biogenesis of lipid droplet integral membrane proteins. J Cell Sci 2022; 135:272279. [PMID: 34558621 PMCID: PMC8627552 DOI: 10.1242/jcs.259220] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/16/2021] [Indexed: 12/18/2022] Open
Abstract
Membrane proteins destined for lipid droplets (LDs), a major intracellular storage site for neutral lipids, are inserted into the endoplasmic reticulum (ER) and then trafficked to LDs where they reside in a hairpin loop conformation. Here, we show that LD membrane proteins can be delivered to the ER either co- or post-translationally and that their membrane-embedded region specifies pathway selection. The co-translational route for LD membrane protein biogenesis is insensitive to a small molecule inhibitor of the Sec61 translocon, Ipomoeassin F, and instead relies on the ER membrane protein complex (EMC) for membrane insertion. This route may even result in a transient exposure of the short N termini of some LD membrane proteins to the ER lumen, followed by putative topological rearrangements that would enable their transmembrane segment to form a hairpin loop and N termini to face the cytosol. Our study reveals an unexpected complexity to LD membrane protein biogenesis and identifies a role for the EMC during their co-translational insertion into the ER. Summary: Insertion of many lipid droplet membrane proteins into the ER is co-translational, mediated by the ER membrane protein complex and may involve topology reorientation.
Collapse
Affiliation(s)
- Pawel Leznicki
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | | | - Jada V Harvey
- Department of Chemistry, Ball State University, Muncie, IN 47306, USA
| | - Wei Q Shi
- Department of Chemistry, Ball State University, Muncie, IN 47306, USA
| | - Stephen High
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| |
Collapse
|
18
|
Hall BS, Hsieh LTH, Sacre S, Simmonds RE. The One That Got Away: How Macrophage-Derived IL-1β Escapes the Mycolactone-Dependent Sec61 Blockade in Buruli Ulcer. Front Immunol 2022; 12:788146. [PMID: 35154073 PMCID: PMC8826060 DOI: 10.3389/fimmu.2021.788146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
Buruli ulcer (BU), caused by Mycobacterium ulcerans, is a devastating necrotizing skin disease. Key to its pathogenesis is mycolactone, the exotoxin virulence factor that is both immunosuppressive and cytotoxic. The discovery that the essential Sec61 translocon is the major cellular target of mycolactone explains much of the disease pathology, including the immune blockade. Sec61 inhibition leads to a loss in production of nearly all cytokines from monocytes, macrophages, dendritic cells and T cells, as well as antigen presentation pathway proteins and costimulatory molecules. However, there has long been evidence that the immune system is not completely incapable of responding to M. ulcerans infection. In particular, IL-1β was recently shown to be present in BU lesions, and to be induced from M. ulcerans-exposed macrophages in a mycolactone-dependent manner. This has important implications for our understanding of BU, showing that mycolactone can act as the "second signal" for IL-1β production without inhibiting the pathways of unconventional secretion it uses for cellular release. In this Perspective article, we validate and discuss this recent advance, which is entirely in-line with our understanding of mycolactone's inhibition of the Sec61 translocon. However, we also show that the IL-1 receptor, which uses the conventional secretory pathway, is sensitive to mycolactone blockade at Sec61. Hence, a more complete understanding of the mechanisms regulating IL-1β function in skin tissue, including the transient intra-macrophage stage of M. ulcerans infection, is urgently needed to uncover the double-edged sword of IL-1β in BU pathogenesis, treatment and wound healing.
Collapse
Affiliation(s)
- Belinda S Hall
- Department of Microbial Sciences, School of Bioscience and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Louise Tzung-Harn Hsieh
- Department of Microbial Sciences, School of Bioscience and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Sandra Sacre
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| | - Rachel E Simmonds
- Department of Microbial Sciences, School of Bioscience and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
19
|
Domenger A, Choisy C, Baron L, Mayau V, Perthame E, Deriano L, Arnulf B, Bories JC, Dadaglio G, Demangel C. The Sec61 translocon is a therapeutic vulnerability in multiple myeloma. EMBO Mol Med 2022; 14:e14740. [PMID: 35014767 PMCID: PMC8899908 DOI: 10.15252/emmm.202114740] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is an incurable malignancy characterized by the uncontrolled expansion of plasma cells in the bone marrow. While proteasome inhibitors like bortezomib efficiently halt MM progression, drug resistance inevitably develop, and novel therapeutic approaches are needed. Here, we used a recently discovered Sec61 inhibitor, mycolactone, to assess the interest of disrupting MM proteostasis via protein translocation blockade. In human MM cell lines, mycolactone caused rapid defects in secretion of immunoglobulins and expression of pro‐survival interleukin (IL)‐6 receptor and CD40, whose activation stimulates IL‐6 production. Mycolactone also triggered pro‐apoptotic endoplasmic reticulum stress responses synergizing with bortezomib for induction of MM cell death and overriding acquired resistance to the proteasome inhibitor. Notably, the mycolactone–bortezomib combination rapidly killed patient‐derived MM cells ex vivo, but not normal mononuclear cells. In immunodeficient mice engrafted with MM cells, it demonstrated superior therapeutic efficacy over single drug treatments, without inducing toxic side effects. Collectively, these findings establish Sec61 blockers as novel anti‐MM agents and reveal the interest of targeting both the translocon and the proteasome in proteostasis‐addicted tumors.
Collapse
Affiliation(s)
- Antoine Domenger
- Unité d'Immunobiologie de l'Infection, Institut Pasteur, INSERM U1224, Université de Paris, Paris, France.,Sorbonne Paris Cité, Université de Paris, Paris, France
| | - Caroline Choisy
- INSERM U976, Institut de Recherche Saint Louis, Université de Paris, Paris, France
| | - Ludivine Baron
- Unité d'Immunobiologie de l'Infection, Institut Pasteur, INSERM U1224, Université de Paris, Paris, France
| | - Véronique Mayau
- Unité d'Immunobiologie de l'Infection, Institut Pasteur, INSERM U1224, Université de Paris, Paris, France
| | - Emeline Perthame
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université de Paris, Paris, France
| | - Ludovic Deriano
- Unité d'Intégrité du Génome, Immunité et Cancer, Equipe Labellisée Ligue Contre Le Cancer, Institut Pasteur, INSERM U1223, Université de Paris, Paris, France
| | - Bertrand Arnulf
- INSERM U976, Institut de Recherche Saint Louis, Université de Paris, Paris, France.,APHP Department of Immuno-Hematology, Hôpital Saint Louis, Paris, France
| | | | - Gilles Dadaglio
- Unité d'Immunobiologie de l'Infection, Institut Pasteur, INSERM U1224, Université de Paris, Paris, France
| | - Caroline Demangel
- Unité d'Immunobiologie de l'Infection, Institut Pasteur, INSERM U1224, Université de Paris, Paris, France
| |
Collapse
|
20
|
Abstract
Mycobacterium ulcerans, the causative agent of Buruli ulcer disease, is unique among human pathogens in its capacity to produce mycolactone, a diffusible macrolide with immunosuppressive and cytotoxic properties. Recent studies have shown that mycolactone operates by inhibiting the host membrane translocation complex (Sec61), with an unprecedented potency compared to previously identified Sec61 blockers. Mycolactone binding to the pore-forming subunit of Sec61 inhibits its capacity to transport nascent secretory and membrane proteins into the endoplasmic reticulum, leading to their cytosolic degradation by the ubiquitin:proteasome system. In T lymphocytes, Sec61 blockade by mycolactone manifests as a sharp decrease in the cell's ability to express homing receptors and release cytokines following activation. Sustained exposure of human cells to mycolactone typically generates proteotoxic stress responses in their cytosol and endoplasmic reticulum (ER), ultimately inducing apoptosis. Here we describe cell-free systems for studying Sec61-mediated protein translocation that allow the impact of mycolactone on the biogenesis of secretory and membrane proteins to be probed. We also describe biological assays of mycolactone-driven inhibition of Sec61 providing rapid and sensitive means to quantitatively assess the presence of the toxin in biological samples.
Collapse
|
21
|
Aberrant stromal tissue factor localisation and mycolactone-driven vascular dysfunction, exacerbated by IL-1β, are linked to fibrin formation in Buruli ulcer lesions. PLoS Pathog 2022; 18:e1010280. [PMID: 35100311 PMCID: PMC8846541 DOI: 10.1371/journal.ppat.1010280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 02/15/2022] [Accepted: 01/13/2022] [Indexed: 12/23/2022] Open
Abstract
Buruli ulcer (BU) is a neglected tropical disease caused by subcutaneous infection with Mycobacterium ulcerans and its exotoxin mycolactone. BU displays coagulative necrosis and widespread fibrin deposition in affected skin tissues. Despite this, the role of the vasculature in BU pathogenesis remains almost completely unexplored. We hypothesise that fibrin-driven ischemia can be an ‘indirect’ route to mycolactone-dependent tissue necrosis by a mechanism involving vascular dysfunction. Here, we tracked >900 vessels within contiguous tissue sections from eight BU patient biopsies. Our aim was to evaluate their vascular and coagulation biomarker phenotype and explore potential links to fibrin deposition. We also integrated this with our understanding of mycolactone’s mechanism of action at Sec61 and its impact on proteins involved in maintaining normal vascular function. Our findings showed that endothelial cell dysfunction is common in skin tissue adjacent to necrotic regions. There was little evidence of primary haemostasis, perhaps due to mycolactone-dependent depletion of endothelial von Willebrand factor. Instead, fibrin staining appeared to be linked to the extrinsic pathway activator, tissue factor (TF). There was significantly greater than expected fibrin staining around vessels that had TF staining within the stroma, and this correlated with the distance it extended from the vessel basement membrane. TF-induced fibrin deposition in these locations would require plasma proteins outside of vessels, therefore we investigated whether mycolactone could increase vascular permeability in vitro. This was indeed the case, and leakage was further exacerbated by IL-1β. Mycolactone caused the loss of endothelial adherens and tight junctions by the depletion of VE-cadherin, TIE-1, TIE-2 and JAM-C; all Sec61-dependent proteins. Taken together, our findings suggest that both vascular and lymphatic vessels in BU lesions become “leaky” during infection, due to the unique action of mycolactone, allowing TF-containing structures and plasma proteins into skin tissue, ultimately leading to local coagulopathy and tissue ischemia. To date, the debilitating skin disease Buruli ulcer remains a public health concern and financial burden in low or middle-income countries, especially in tropical regions. Late diagnosis is frequent in remote areas, perhaps due to the painlessness of the disease. Hence patients often present with large, destructive opened ulcers leading to delayed wound closure or even lifelong disability. The infectious agent produces a toxin called mycolactone that drives the disease. We previously found evidence that the vascular system is disrupted by mycolactone in these lesions, and now we have further explored potential explanations for these findings by looking at the expression of vascular markers in BU. In a detailed analysis of patient skin punch biopsies, we identified distinct expression patterns of certain proteins and found that tissue factor, which initiates the so-called extrinsic pathway of blood clotting, is particularly important. Mycolactone is able to disrupt the barrier function of the endothelium, further aggravating the diseased phenotype, which may explain how clotting factors access the tissue. Altogether, such localised hypercoagulation in Buruli ulcer skin lesions may contribute to the development of the lesion.
Collapse
|
22
|
Mycolactone enhances the Ca2+ leak from endoplasmic reticulum by trapping Sec61 translocons in a Ca2+ permeable state. Biochem J 2021; 478:4005-4024. [PMID: 34726690 PMCID: PMC8650850 DOI: 10.1042/bcj20210345] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/19/2021] [Accepted: 11/02/2021] [Indexed: 01/17/2023]
Abstract
The Mycobacterium ulcerans exotoxin, mycolactone, is an inhibitor of co-translational translocation via the Sec61 complex. Mycolactone has previously been shown to bind to, and alter the structure of the major translocon subunit Sec61α, and change its interaction with ribosome nascent chain complexes. In addition to its function in protein translocation into the ER, Sec61 also plays a key role in cellular Ca2+ homeostasis, acting as a leak channel between the endoplasmic reticulum (ER) and cytosol. Here, we have analysed the effect of mycolactone on cytosolic and ER Ca2+ levels using compartment-specific sensors. We also used molecular docking analysis to explore potential interaction sites for mycolactone on translocons in various states. These results show that mycolactone enhances the leak of Ca2+ ions via the Sec61 translocon, resulting in a slow but substantial depletion of ER Ca2+. This leak was dependent on mycolactone binding to Sec61α because resistance mutations in this protein completely ablated the increase. Molecular docking supports the existence of a mycolactone-binding transient inhibited state preceding translocation and suggests mycolactone may also bind Sec61α in its idle state. We propose that delayed ribosomal release after translation termination and/or translocon ‘breathing' during rapid transitions between the idle and intermediate-inhibited states allow for transient Ca2+ leak, and mycolactone's stabilisation of the latter underpins the phenotype observed.
Collapse
|
23
|
Inhibitors of the Sec61 Complex and Novel High Throughput Screening Strategies to Target the Protein Translocation Pathway. Int J Mol Sci 2021; 22:ijms222112007. [PMID: 34769437 PMCID: PMC8585047 DOI: 10.3390/ijms222112007] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/25/2021] [Accepted: 10/29/2021] [Indexed: 02/08/2023] Open
Abstract
Proteins targeted to the secretory pathway start their intracellular journey by being transported across biological membranes such as the endoplasmic reticulum (ER). A central component in this protein translocation process across the ER is the Sec61 translocon complex, which is only intracellularly expressed and does not have any enzymatic activity. In addition, Sec61 translocon complexes are difficult to purify and to reconstitute. Screening for small molecule inhibitors impairing its function has thus been notoriously difficult. However, such translocation inhibitors may not only be valuable tools for cell biology, but may also represent novel anticancer drugs, given that cancer cells heavily depend on efficient protein translocation into the ER to support their fast growth. In this review, different inhibitors of protein translocation will be discussed, and their specific mode of action will be compared. In addition, recently published screening strategies for small molecule inhibitors targeting the whole SRP-Sec61 targeting/translocation pathway will be summarized. Of note, slightly modified assays may be used in the future to screen for substances affecting SecYEG, the bacterial ortholog of the Sec61 complex, in order to identify novel antibiotic drugs.
Collapse
|
24
|
Overview: Mycolactone , the Macrolide Toxin of Mycobacterium ulcerans. Methods Mol Biol 2021. [PMID: 34643906 DOI: 10.1007/978-1-0716-1779-3_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The acquisition by a Mycobacterium marinum-like progenitor of a plasmid encoding enzymes for the biosynthesis of the highly potent macrolide toxin mycolactone has set off the evolution of M. ulcerans toward a new mycobacterial species. While the selective advantage of producing mycolactone for survival in environmental niche(s) of the pathogen is unclear, there is no doubt that the cytotoxic, immunomodulatory, and analgesic properties of mycolactone are key for the establishment and progression of M. ulcerans infections in the host. Improved procedures for the isolation, handling, and detection of the amphiphilic and light-sensitive toxin have facilitated studies to unravel molecular mechanisms of mycolactone action on host cells in vitro and on cellular and immune responses in animal models. The pivotal role of mycolactone in the pathology of Buruli ulcer and the fact that the toxin has not been associated with other pathogens make it an ideal target for therapeutics/vaccines aiming at mycolactone neutralization and for the development of assays for the diagnosis of the disease.
Collapse
|
25
|
Tello Rubio B, Bugault F, Baudon B, Raynal B, Brûlé S, Morel JD, Saint-Auret S, Blanchard N, Demangel C, Guenin-Macé L. Molecular Mechanisms Underpinning the Circulation and Cellular Uptake of Mycobacterium ulcerans Toxin Mycolactone. Front Pharmacol 2021; 12:733496. [PMID: 34603049 PMCID: PMC8481864 DOI: 10.3389/fphar.2021.733496] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/06/2021] [Indexed: 11/13/2022] Open
Abstract
Mycolactone is a diffusible lipid toxin produced by Mycobacterium ulcerans, the causative agent of Buruli ulcer disease. Altough bacterially derived mycolactone has been shown to traffic from cutaneous foci of infection to the bloodstream, the mechanisms underpinning its access to systemic circulation and import by host cells remain largely unknown. Using biophysical and cell-based approaches, we demonstrate that mycolactone specific association to serum albumin and lipoproteins is necessary for its solubilization and is a major mechanism to regulate its bioavailability. We also demonstrate that Scavenger Receptor (SR)-B1 contributes to the cellular uptake of mycolactone. Overall, we suggest a new mechanism of transport and cell entry, challenging the dogma that the toxin enters host cells via passive diffusion.
Collapse
Affiliation(s)
- Bruno Tello Rubio
- Immunobiology of Infection Unit, INSERM U1221, Institut Pasteur, Paris, France
| | - Florence Bugault
- Immunobiology of Infection Unit, INSERM U1221, Institut Pasteur, Paris, France
| | - Blandine Baudon
- Immunobiology of Infection Unit, INSERM U1221, Institut Pasteur, Paris, France
| | - Bertrand Raynal
- Plateforme de Biophysique Moléculaire, UMR 3528 CNRS, Institut Pasteur, Paris, France
| | - Sébastien Brûlé
- Plateforme de Biophysique Moléculaire, UMR 3528 CNRS, Institut Pasteur, Paris, France
| | - Jean-David Morel
- Immunobiology of Infection Unit, INSERM U1221, Institut Pasteur, Paris, France
| | - Sarah Saint-Auret
- CNRS, LIMA, UMR 7042, Université de Haute-Alsace, Université de Strasbourg, Mulhouse, France
| | - Nicolas Blanchard
- CNRS, LIMA, UMR 7042, Université de Haute-Alsace, Université de Strasbourg, Mulhouse, France
| | - Caroline Demangel
- Immunobiology of Infection Unit, INSERM U1221, Institut Pasteur, Paris, France
| | - Laure Guenin-Macé
- Immunobiology of Infection Unit, INSERM U1221, Institut Pasteur, Paris, France
| |
Collapse
|
26
|
Llibre A, Dedicoat M, Burel JG, Demangel C, O’Shea MK, Mauro C. Host Immune-Metabolic Adaptations Upon Mycobacterial Infections and Associated Co-Morbidities. Front Immunol 2021; 12:747387. [PMID: 34630426 PMCID: PMC8495197 DOI: 10.3389/fimmu.2021.747387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022] Open
Abstract
Mycobacterial diseases are a major public health challenge. Their causative agents include, in order of impact, members of the Mycobacterium tuberculosis complex (causing tuberculosis), Mycobacterium leprae (causing leprosy), and non-tuberculous mycobacterial pathogens including Mycobacterium ulcerans. Macrophages are mycobacterial targets and they play an essential role in the host immune response to mycobacteria. This review aims to provide a comprehensive understanding of the immune-metabolic adaptations of the macrophage to mycobacterial infections. This metabolic rewiring involves changes in glycolysis and oxidative metabolism, as well as in the use of fatty acids and that of metals such as iron, zinc and copper. The macrophage metabolic adaptations result in changes in intracellular metabolites, which can post-translationally modify proteins including histones, with potential for shaping the epigenetic landscape. This review will also cover how critical tuberculosis co-morbidities such as smoking, diabetes and HIV infection shape host metabolic responses and impact disease outcome. Finally, we will explore how the immune-metabolic knowledge gained in the last decades can be harnessed towards the design of novel diagnostic and therapeutic tools, as well as vaccines.
Collapse
Affiliation(s)
- Alba Llibre
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Martin Dedicoat
- Department of Infectious Diseases, Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Julie G. Burel
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Caroline Demangel
- Immunobiology of Infection Unit, Institut Pasteur, INSERM U1224, Paris, France
| | - Matthew K. O’Shea
- Department of Infectious Diseases, Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Claudio Mauro
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
27
|
O’Keefe S, Zong G, Duah KB, Andrews LE, Shi WQ, High S. An alternative pathway for membrane protein biogenesis at the endoplasmic reticulum. Commun Biol 2021; 4:828. [PMID: 34211117 PMCID: PMC8249459 DOI: 10.1038/s42003-021-02363-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
The heterotrimeric Sec61 complex is a major site for the biogenesis of transmembrane proteins (TMPs), accepting nascent TMP precursors that are targeted to the endoplasmic reticulum (ER) by the signal recognition particle (SRP). Unlike most single-spanning membrane proteins, the integration of type III TMPs is completely resistant to small molecule inhibitors of the Sec61 translocon. Using siRNA-mediated depletion of specific ER components, in combination with the potent Sec61 inhibitor ipomoeassin F (Ipom-F), we show that type III TMPs utilise a distinct pathway for membrane integration at the ER. Hence, following SRP-mediated delivery to the ER, type III TMPs can uniquely access the membrane insertase activity of the ER membrane complex (EMC) via a mechanism that is facilitated by the Sec61 translocon. This alternative EMC-mediated insertion pathway allows type III TMPs to bypass the Ipom-F-mediated blockade of membrane integration that is seen with obligate Sec61 clients.
Collapse
Affiliation(s)
- Sarah O’Keefe
- grid.5379.80000000121662407School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Guanghui Zong
- grid.164295.d0000 0001 0941 7177Department of Chemistry and Biochemistry, University of Maryland, College Park, MD USA
| | - Kwabena B. Duah
- grid.252754.30000 0001 2111 9017Department of Chemistry, Ball State University, Muncie, IN USA
| | - Lauren E. Andrews
- grid.252754.30000 0001 2111 9017Department of Chemistry, Ball State University, Muncie, IN USA
| | - Wei Q. Shi
- grid.252754.30000 0001 2111 9017Department of Chemistry, Ball State University, Muncie, IN USA
| | - Stephen High
- grid.5379.80000000121662407School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
28
|
Roboti P, O'Keefe S, Duah KB, Shi WQ, High S. Ipomoeassin-F disrupts multiple aspects of secretory protein biogenesis. Sci Rep 2021; 11:11562. [PMID: 34079010 PMCID: PMC8173012 DOI: 10.1038/s41598-021-91107-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
The Sec61 complex translocates nascent polypeptides into and across the membrane of the endoplasmic reticulum (ER), providing access to the secretory pathway. In this study, we show that Ipomoeassin-F (Ipom-F), a selective inhibitor of protein entry into the ER lumen, blocks the in vitro translocation of certain secretory proteins and ER lumenal folding factors whilst barely affecting others such as albumin. The effects of Ipom-F on protein secretion from HepG2 cells are twofold: reduced ER translocation combined, in some cases, with defective ER lumenal folding. This latter issue is most likely a consequence of Ipom-F preventing the cell from replenishing its ER lumenal chaperones. Ipom-F treatment results in two cellular stress responses: firstly, an upregulation of stress-inducible cytosolic chaperones, Hsp70 and Hsp90; secondly, an atypical unfolded protein response (UPR) linked to the Ipom-F-mediated perturbation of ER function. Hence, although levels of spliced XBP1 and CHOP mRNA and ATF4 protein increase with Ipom-F, the accompanying increase in the levels of ER lumenal BiP and GRP94 seen with tunicamycin are not observed. In short, although Ipom-F reduces the biosynthetic load of newly synthesised secretory proteins entering the ER lumen, its effects on the UPR preclude the cell restoring ER homeostasis.
Collapse
Affiliation(s)
- Peristera Roboti
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK.
| | - Sarah O'Keefe
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Kwabena B Duah
- Department of Chemistry, Ball State University, Muncie, IN, 47306, USA
| | - Wei Q Shi
- Department of Chemistry, Ball State University, Muncie, IN, 47306, USA
| | - Stephen High
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
29
|
Lenz KD, Klosterman KE, Mukundan H, Kubicek-Sutherland JZ. Macrolides: From Toxins to Therapeutics. Toxins (Basel) 2021; 13:347. [PMID: 34065929 PMCID: PMC8150546 DOI: 10.3390/toxins13050347] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 12/17/2022] Open
Abstract
Macrolides are a diverse class of hydrophobic compounds characterized by a macrocyclic lactone ring and distinguished by variable side chains/groups. Some of the most well characterized macrolides are toxins produced by marine bacteria, sea sponges, and other species. Many marine macrolide toxins act as biomimetic molecules to natural actin-binding proteins, affecting actin polymerization, while other toxins act on different cytoskeletal components. The disruption of natural cytoskeletal processes affects cell motility and cytokinesis, and can result in cellular death. While many macrolides are toxic in nature, others have been shown to display therapeutic properties. Indeed, some of the most well known antibiotic compounds, including erythromycin, are macrolides. In addition to antibiotic properties, macrolides have been shown to display antiviral, antiparasitic, antifungal, and immunosuppressive actions. Here, we review each functional class of macrolides for their common structures, mechanisms of action, pharmacology, and human cellular targets.
Collapse
Affiliation(s)
| | | | | | - Jessica Z. Kubicek-Sutherland
- Physical Chemistry and Applied Spectroscopy, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA; (K.D.L.); (K.E.K.); (H.M.)
| |
Collapse
|
30
|
Amoako YA, Loglo AD, Frimpong M, Agbavor B, Abass MK, Amofa G, Ofori E, Ampadu E, Asiedu K, Stienstra Y, Wansbrough-Jones M, van der Werf T, Phillips RO. Co-infection of HIV in patients with Buruli ulcer disease in Central Ghana. BMC Infect Dis 2021; 21:331. [PMID: 33832460 PMCID: PMC8028811 DOI: 10.1186/s12879-021-06009-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/22/2021] [Indexed: 12/13/2022] Open
Abstract
Background Previous studies have reported that presence and severity of Buruli ulcer (BU) may reflect the underlying immunosuppression in HIV infected individuals by causing increased incidence of multiple, larger and ulcerated lesions. We report cases of BU-HIV coinfection and the accompanying programmatic challenges encountered in central Ghana. Methods Patients with PCR confirmed BU in central Ghana who were HIV positive were identified and their BU01 forms were retrieved and reviewed in further detail. A combined 16S rRNA reverse transcriptase / IS2404 qPCR assay was used to assess the Mycobacterium ulcerans load. The characteristics of coinfected patients (BU+HIV+) were compared with a group of matched controls. Results The prevalence of HIV in this BU cohort was 2.4% (compared to national HIV prevalence of 1.7%). Eight of 9 BU+HIV+ patients had a single lesion and ulcers were the most common lesion type. The lesions presented were predominantly category II (5/9) followed by category I lesions. The median (IQR) time to healing was 14 (8–28) weeks in the BU+HIV+ compared to 28 (12–33) weeks in the control BU+HIV− group (p = 0.360). Only one BU+HIV+ developed a paradoxical reaction at week 16 but the lesion healed completely at week 20. The median bacterial load (16SrRNA) of BU+HIV+ patients was 750 copies /ml (95% CI 0–398,000) versus 500 copies/ml (95% CI 0–126,855,500) in BU+HIV− group. Similarly, the median count using the IS2404 assay was 500 copies/ml (95% CI 0–500) for BU+HIV+ patients versus 500 copies/ml (95% CI 500–31,000) for BU+HIV− patients. BU+HIV− patients mounted a significantly higher interferon-γ response compared to the BU+HIV+ co-infected patients with respective median (range) responses of [1687(81.11–4399) pg/ml] versus [137.5(4.436–1406) pg/ml, p = 0.03]. There were challenges with the integration of HIV and BU care in this cohort. Conclusion The prevalence of HIV in the BU+ infected population was not significantly increased when compared to the prevalence of HIV in the general population. There was no clear relationship between BU lesion severity and HIV viral load or CD4 counts. Efforts should be made to encourage the integration of care of patients with BU-HIV coinfection.
Collapse
Affiliation(s)
- Yaw Ampem Amoako
- School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana. .,Department of Medicine, Komfo Anokye Teaching Hospital, Kumasi, Ghana. .,Skin NTD's Research Group, Kumasi Centre for Collaborative Research in Tropical Medicine, Kumasi, Ghana.
| | - Aloysius Dzigbordi Loglo
- School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.,Skin NTD's Research Group, Kumasi Centre for Collaborative Research in Tropical Medicine, Kumasi, Ghana
| | - Michael Frimpong
- School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.,Skin NTD's Research Group, Kumasi Centre for Collaborative Research in Tropical Medicine, Kumasi, Ghana
| | - Bernadette Agbavor
- School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.,Skin NTD's Research Group, Kumasi Centre for Collaborative Research in Tropical Medicine, Kumasi, Ghana
| | | | | | | | - Edwin Ampadu
- National Buruli ulcer Control Programme, Ghana Health Service, Accra, Ghana
| | - Kingsley Asiedu
- Department of Neglected Tropical Diseases, WHO, Geneva, Switzerland
| | - Ymkje Stienstra
- Department of Medicine/ Infectious Diseases, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | | | - Tjip van der Werf
- Department of Medicine/ Infectious Diseases, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Richard Odame Phillips
- School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.,Department of Medicine, Komfo Anokye Teaching Hospital, Kumasi, Ghana.,Skin NTD's Research Group, Kumasi Centre for Collaborative Research in Tropical Medicine, Kumasi, Ghana
| |
Collapse
|
31
|
Fevereiro J, Fraga AG, Pedrosa J. Genetics in the Host-Mycobacterium ulcerans interaction. Immunol Rev 2021; 301:222-241. [PMID: 33682158 DOI: 10.1111/imr.12958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 11/30/2022]
Abstract
Buruli ulcer is an emerging infectious disease associated with high morbidity and unpredictable outbreaks. It is caused by Mycobacterium ulcerans, a slow-growing pathogen evolutionarily shaped by the acquisition of a plasmid involved in the production of a potent macrolide-like cytotoxin and by genome rearrangements and downsizing. These events culminated in an uncommon infection pattern, whereby M. ulcerans is both able to induce the initiation of the inflammatory cascade and the cell death of its proponents, as well as to survive within the phagosome and in the extracellular milieu. In such extreme conditions, the host is sentenced to rely on a highly orchestrated genetic landscape to be able to control the infection. We here revisit the dynamics of M. ulcerans infection, drawing parallels from other mycobacterioses and integrating the most recent knowledge on its evolution and pathogenicity in its interaction with the host immune response.
Collapse
Affiliation(s)
- João Fevereiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Alexandra G Fraga
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Jorge Pedrosa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
32
|
O'Keefe S, Roboti P, Duah KB, Zong G, Schneider H, Shi WQ, High S. Ipomoeassin-F inhibits the in vitro biogenesis of the SARS-CoV-2 spike protein and its host cell membrane receptor. J Cell Sci 2021; 134:jcs257758. [PMID: 33468620 PMCID: PMC7904091 DOI: 10.1242/jcs.257758] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
In order to produce proteins essential for their propagation, many pathogenic human viruses, including SARS-CoV-2, the causative agent of COVID-19 respiratory disease, commandeer host biosynthetic machineries and mechanisms. Three major structural proteins, the spike, envelope and membrane proteins, are amongst several SARS-CoV-2 components synthesised at the endoplasmic reticulum (ER) of infected human cells prior to the assembly of new viral particles. Hence, the inhibition of membrane protein synthesis at the ER is an attractive strategy for reducing the pathogenicity of SARS-CoV-2 and other obligate viral pathogens. Using an in vitro system, we demonstrate that the small molecule inhibitor ipomoeassin F (Ipom-F) potently blocks the Sec61-mediated ER membrane translocation and/or insertion of three therapeutic protein targets for SARS-CoV-2 infection; the viral spike and ORF8 proteins together with angiotensin-converting enzyme 2, the host cell plasma membrane receptor. Our findings highlight the potential for using ER protein translocation inhibitors such as Ipom-F as host-targeting, broad-spectrum antiviral agents.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Sarah O'Keefe
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Peristera Roboti
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Kwabena B Duah
- Department of Chemistry, Ball State University, Muncie, IN 47306, USA
| | - Guanghui Zong
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA
| | - Hayden Schneider
- Department of Chemistry, Ball State University, Muncie, IN 47306, USA
| | - Wei Q Shi
- Department of Chemistry, Ball State University, Muncie, IN 47306, USA
| | - Stephen High
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
33
|
Demangel C. Immunity against Mycobacterium ulcerans: The subversive role of mycolactone. Immunol Rev 2021; 301:209-221. [PMID: 33607704 DOI: 10.1111/imr.12956] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/19/2021] [Accepted: 01/19/2021] [Indexed: 12/11/2022]
Abstract
Mycobacterium ulcerans causes Buruli ulcer, a neglected tropical skin disease manifesting as chronic wounds that can leave victims with major, life-long deformity and disability. Differently from other mycobacterial pathogens, M ulcerans produces mycolactone, a diffusible lipid factor with unique cytotoxic and immunomodulatory properties. Both traits result from mycolactone targeting Sec61, the entry point of the secretory pathway in eukaryotic cells. By inhibiting Sec61, mycolactone prevents the host cell's production of secreted proteins, and most of its transmembrane proteins. This molecular blockade dramatically alters the functions of immune cells, thereby the generation of protective immunity. Moreover, sustained inhibition of Sec61 triggers proteotoxic stress responses leading to apoptotic cell death, which can stimulate vigorous immune responses. The dynamics of bacterial production of mycolactone and elimination by infected hosts thus critically determine the balance between its immunostimulatory and immunosuppressive effects. Following an introduction summarizing the essential information on Buruli ulcer disease, this review focuses on the current state of knowledge regarding mycolactone's regulation and biodistribution. We then detail the consequences of mycolactone-mediated Sec61 blockade on initiation and maintenance of innate and adaptive immune responses. Finally, we discuss the key questions to address in order to improve immunity to M ulcerans, and how increased knowledge of mycolactone biology may pave the way to innovative therapeutics.
Collapse
Affiliation(s)
- Caroline Demangel
- Immunobiology of Infection Unit, INSERM U1221, Institut Pasteur, Paris, France
| |
Collapse
|
34
|
O’Keefe S, Roboti P, Duah KB, Zong G, Schneider H, Shi WQ, High S. Ipomoeassin-F inhibits the in vitro biogenesis of the SARS-CoV-2 spike protein and its host cell membrane receptor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2020.11.24.390039. [PMID: 33269350 PMCID: PMC7709170 DOI: 10.1101/2020.11.24.390039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In order to produce proteins essential for their propagation, many pathogenic human viruses, including SARS-CoV-2 the causative agent of COVID-19 respiratory disease, commandeer host biosynthetic machineries and mechanisms. Three major structural proteins, the spike, envelope and membrane proteins, are amongst several SARS-CoV-2 components synthesised at the endoplasmic reticulum (ER) of infected human cells prior to the assembly of new viral particles. Hence, the inhibition of membrane protein synthesis at the ER is an attractive strategy for reducing the pathogenicity of SARS-CoV-2 and other obligate viral pathogens. Using an in vitro system, we demonstrate that the small molecule inhibitor ipomoeassin F (Ipom-F) potently blocks the Sec61-mediated ER membrane translocation/insertion of three therapeutic protein targets for SARS-CoV-2 infection; the viral spike and ORF8 proteins together with angiotensin-converting enzyme 2, the host cell plasma membrane receptor. Our findings highlight the potential for using ER protein translocation inhibitors such as Ipom-F as host-targeting, broad-spectrum, antiviral agents.
Collapse
Affiliation(s)
- Sarah O’Keefe
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Peristera Roboti
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Kwabena B. Duah
- Department of Chemistry, Ball State University, Muncie, Indiana 47306, USA
| | - Guanghui Zong
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, USA
| | - Hayden Schneider
- Department of Chemistry, Ball State University, Muncie, Indiana 47306, USA
| | - Wei Q. Shi
- Department of Chemistry, Ball State University, Muncie, Indiana 47306, USA
| | - Stephen High
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| |
Collapse
|
35
|
Zong G, Hu Z, Duah KB, Andrews LE, Zhou J, O'Keefe S, Whisenhunt L, Shim JS, Du Y, High S, Shi WQ. Ring Expansion Leads to a More Potent Analogue of Ipomoeassin F. J Org Chem 2020; 85:16226-16235. [PMID: 33264019 PMCID: PMC7808706 DOI: 10.1021/acs.joc.0c01659] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
![]()
Two
new ring-size-varying analogues (2 and 3) of ipomoeassin F were synthesized and evaluated. Improved cytotoxicity
(IC50: from 1.8 nM) and in vitro protein translocation
inhibition (IC50: 35 nM) derived from ring expansion imply
that the binding pocket of Sec61α (isoform 1) can accommodate
further structural modifications, likely in the fatty acid portion.
Streamlined preparation of the key diol intermediate 5 enabled gram-scale production, allowing us to establish that ipomoeassin
F is biologically active in vivo (MTD: ∼3 mg/kg).
Collapse
Affiliation(s)
- Guanghui Zong
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Zhijian Hu
- Angion Biomedica Corp., 51 Charles Lindbergh Boulevard, Uniondale, New York 11553, United States
| | - Kwabena Baffour Duah
- Department of Chemistry, Ball State University, Muncie, Indiana 47306, United States
| | - Lauren E Andrews
- Department of Chemistry, Ball State University, Muncie, Indiana 47306, United States
| | - Jianhong Zhou
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Sarah O'Keefe
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Lucas Whisenhunt
- Thermo Fisher Scientific, 6173 E. Old Marion Highway, Florence, South Carolina 29501, United States
| | - Joong Sup Shim
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, 999078 Taipa, Macau SAR China
| | - Yuchun Du
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Stephen High
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Wei Q Shi
- Department of Chemistry, Ball State University, Muncie, Indiana 47306, United States
| |
Collapse
|
36
|
Colucci-Guyon E, Rifflet A, Saint-Auret S, da Costa A, Boucontet L, Laval T, Prehaud C, Blanchard N, Levraud JP, Boneca IG, Demangel C, Guenin-Macé L. Spatiotemporal analysis of mycolactone distribution in vivo reveals partial diffusion in the central nervous system. PLoS Negl Trop Dis 2020; 14:e0008878. [PMID: 33264290 PMCID: PMC7710047 DOI: 10.1371/journal.pntd.0008878] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/13/2020] [Indexed: 01/26/2023] Open
Abstract
Mycobacterium ulcerans, the causative agent of Buruli ulcer (BU) disease, is unique amongst human pathogens in its capacity to produce a lipid toxin called mycolactone. While previous studies have demonstrated that bacterially-released mycolactone diffuses beyond infection foci, the spatiotemporal distribution of mycolactone remained largely unknown. Here, we used the zebrafish model to provide the first global kinetic analysis of mycolactone's diffusion in vivo, and multicellular co-culture systems to address the critical question of the toxin's access to the brain. Zebrafish larvae were injected with a fluorescent-derivative of mycolactone to visualize the in vivo diffusion of the toxin from the peripheral circulation. A rapid, body-wide distribution of mycolactone was observed, with selective accumulation in tissues near the injection site and brain, together with an important excretion through the gastro-intestinal tract. Our conclusion that mycolactone reached the central nervous system was reinforced by an in cellulo model of human blood brain barrier and a mouse model of M. ulcerans-infection. Here we show that mycolactone has a broad but heterogenous profile of distribution in vivo. Our investigations in vitro and in vivo support the view that a fraction of bacterially-produced mycolactone gains access to the central nervous system. The relative persistence of mycolactone in the bloodstream suggests that assays of circulating mycolactone are relevant for BU disease monitoring and treatment optimization.
Collapse
Affiliation(s)
- Emma Colucci-Guyon
- Macrophages and Development of Immunity, Institut Pasteur, CNRS UMR 3738, Paris, France
| | - Aline Rifflet
- Institut Pasteur, Unité Biologie et génétique de la paroi bactérienne, Paris 75724, France; CNRS, UMR 2001 “Microbiologie intégrative et moléculaire”, Paris 75015, France; INSERM, groupe Avenir, Paris, France
| | - Sarah Saint-Auret
- Université de Haute-Alsace, Université de Strasbourg, CNRS, LIMA, UMR 7042, Mulhouse, France
| | | | - Laurent Boucontet
- Macrophages and Development of Immunity, Institut Pasteur, CNRS UMR 3738, Paris, France
| | - Thomas Laval
- Immunobiology of Infection Unit, Institut Pasteur, INSERM U1221, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | - Nicolas Blanchard
- Université de Haute-Alsace, Université de Strasbourg, CNRS, LIMA, UMR 7042, Mulhouse, France
| | - Jean-Pierre Levraud
- Macrophages and Development of Immunity, Institut Pasteur, CNRS UMR 3738, Paris, France
| | - Ivo G. Boneca
- Institut Pasteur, Unité Biologie et génétique de la paroi bactérienne, Paris 75724, France; CNRS, UMR 2001 “Microbiologie intégrative et moléculaire”, Paris 75015, France; INSERM, groupe Avenir, Paris, France
| | - Caroline Demangel
- Immunobiology of Infection Unit, Institut Pasteur, INSERM U1221, Paris, France
| | - Laure Guenin-Macé
- Immunobiology of Infection Unit, Institut Pasteur, INSERM U1221, Paris, France
- * E-mail:
| |
Collapse
|
37
|
Targeting of HER/ErbB family proteins using broad spectrum Sec61 inhibitors coibamide A and apratoxin A. Biochem Pharmacol 2020; 183:114317. [PMID: 33152346 DOI: 10.1016/j.bcp.2020.114317] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 01/17/2023]
Abstract
Coibamide A is a potent cancer cell toxin and one of a select group of natural products that inhibit protein entry into the secretory pathway via a direct inhibition of the Sec61 protein translocon. Many Sec61 client proteins are clinically relevant drug targets once trafficked to their final destination in or outside the cell, however the use of Sec61 inhibitors to block early biosynthesis of specific proteins is at a pre-clinical stage. In the present study we evaluated the action of coibamide A against human epidermal growth factor receptor (HER, ErbB) proteins in representative breast and lung cancer cell types. HERs were selected for this study as they represent a family of Sec61 clients that is frequently dysregulated in human cancers, including coibamide-sensitive cell types. Although coibamide A inhibits biogenesis of a broad range of Sec61 substrate proteins in a presumed substrate-nonselective manner, endogenous HER3 (ErbB-3) and EGFR (ErbB-1) proteins were more sensitive to coibamide A, and the related Sec61 inhibitor apratoxin A, than HER2 (ErbB-2). Despite this rank order of sensitivity (HER3 > EGFR > HER2), Sec61-dependent inhibition by coibamide A was sufficient to decrease cell surface expression of HER2. We report that coibamide A- or apratoxin A-mediated block of HER3 entry into the secretory pathway is unlikely to be mediated by the HER3 signal peptide alone. HER3 (G11L/S15L), that is fully resistant to the highly substrate-selective cotransin analogue CT8, was more resistant than wild-type HER3 but only at low coibamide A (3 nM) concentrations; HER3 (G11L/S15L) expression was inhibited by higher concentrations of either natural product. Time- and concentration-dependent decreases in HER protein expression induced a commensurate reduction in AKT/MAPK signaling in breast and lung cancer cell types and loss in cell viability. Coibamide A potentiated the cytotoxic efficacy of small molecule kinase inhibitors lapatinib and erlotinib in breast and lung cancer cell types, respectively. These data indicate that natural product modulators of Sec61 function have value as chemical probes to interrogate HER/ErbB signaling in treatment-resistant human cancers.
Collapse
|
38
|
Förster B, Demangel C, Thye T. Mycolactone induces cell death by SETD1B-dependent degradation of glutathione. PLoS Negl Trop Dis 2020; 14:e0008709. [PMID: 33006969 PMCID: PMC7556509 DOI: 10.1371/journal.pntd.0008709] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 10/14/2020] [Accepted: 08/13/2020] [Indexed: 12/21/2022] Open
Abstract
Mycobacterium ulcerans is a human pathogen that causes a necrotizing skin disease known as Buruli ulcer. Necrosis of infected skin is driven by bacterial production of mycolactone, a diffusible exotoxin targeting the host translocon (Sec61). By blocking Sec61, mycolactone prevents the transport of nascent secretory proteins into the endoplasmic reticulum of host cells. This triggers pro-apoptotic stress responses partially depending on activation of the ATF4 transcription factor. To gain further insight into the molecular pathways mediating the cytotoxic effects of mycolactone we conducted the first haploid genetic screen with the M. ulcerans toxin in KBM-7 cells. This approach allowed us to identify the histone methyltransferase SETD1B as a novel mediator of mycolactone-induced cell death. CRISPR/Cas9-based inactivation of SETD1B rendered cells resistant to lethal doses of the toxin, highlighting the critical importance of this gene’s expression. To understand how SETD1B contributes to mycolactone cytotoxicity, we compared the transcriptomes of wild-type (WT) and SETD1B knockout KBM-7 cells upon exposure to the toxin. While ATF4 effectors were upregulated by mycolactone in both WT and SETD1B knockout cells, mycolactone selectively induced the expression of pro-apoptotic genes in WT cells. Among those genes we identified CHAC1, which codes for a major glutathione (GSH)-degrading enzyme, and whose strong upregulation in mycolactone-treated WT cells correlated with a marked reduction in GSH protein level. Moreover, GSH supplementation conferred cells with substantial protection against the toxic effects of mycolactone. Our data thus identify SETD1B/CHAC1/GSH as a novel, epigenetic mechanism connecting Sec61 blockade with apoptotic cell death. They suggest that GSH-based treatments might have the capacity to limit skin necrosis in Buruli ulcer. The human pathogen Mycobacterium ulcerans causes a necrotizing skin disease known as Buruli ulcer. The major toxin of the mycobacteria, mycolactone, prevents the transport of secretory proteins into the endoplasmic reticulum, and thereby triggers a deadly stress response. We conducted the first haploid genetic screen to identify host factors with impact on mycolactone toxicity. This enabled us to identify the histone methyltransferase SETD1B as a novel mediator of mycolactone-induced cell death. RNA analyses of wild-type cells and resistant SETD1B knockout cells treated with mycolactone then showed a selective induction of genes implicated in programmed cell-death only in wild-type cells. This was accompanied by a marked reduction of the antioxidant glutathione, which might cause the mycolactone induced cell death.
Collapse
Affiliation(s)
- Birgit Förster
- Bernhard Nocht Institute for Tropical Medicine, Dept. Infectious Disease Epidemiology, Hamburg, Germany
| | - Caroline Demangel
- Immunobiology Infection Unit, Institut Pasteur, INSERM U1221, Paris, France
| | - Thorsten Thye
- Bernhard Nocht Institute for Tropical Medicine, Dept. Infectious Disease Epidemiology, Hamburg, Germany
- * E-mail:
| |
Collapse
|
39
|
Steenhuis M, Koningstein GM, Oswald J, Pick T, O'Keefe S, Koch HG, Cavalié A, Whitehead RC, Swanton E, High S, Luirink J. Eeyarestatin 24 impairs SecYEG-dependent protein trafficking and inhibits growth of clinically relevant pathogens. Mol Microbiol 2020; 115:28-40. [PMID: 32798330 PMCID: PMC8273874 DOI: 10.1111/mmi.14589] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/29/2020] [Indexed: 02/06/2023]
Abstract
Eeyarestatin 1 (ES1) is an inhibitor of endoplasmic reticulum (ER) associated protein degradation, Sec61‐dependent Ca2+ homeostasis and protein translocation into the ER. Recently, evidence was presented showing that a smaller analog of ES1, ES24, targets the Sec61‐translocon, and captures it in an open conformation that is translocation‐incompetent. We now show that ES24 impairs protein secretion and membrane protein insertion in Escherichia coli via the homologous SecYEG‐translocon. Transcriptomic analysis suggested that ES24 has a complex mode of action, probably involving multiple targets. Interestingly, ES24 shows antibacterial activity toward clinically relevant strains. Furthermore, the antibacterial activity of ES24 is equivalent to or better than that of nitrofurantoin, a known antibiotic that, although structurally similar to ES24, does not interfere with SecYEG‐dependent protein trafficking. Like nitrofurantoin, we find that ES24 requires activation by the NfsA and NfsB nitroreductases, suggesting that the formation of highly reactive nitroso intermediates is essential for target inactivation in vivo.
Collapse
Affiliation(s)
- Maurice Steenhuis
- Department of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit, Amsterdam, the Netherlands
| | - Gregory M Koningstein
- Department of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit, Amsterdam, the Netherlands
| | - Julia Oswald
- Institute of Biochemistry and Molecular Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tillman Pick
- Experimental and Clinical Pharmacology and Toxicology, Saarland University, Homburg, Germany
| | - Sarah O'Keefe
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Hans-Georg Koch
- Institute of Biochemistry and Molecular Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Adolfo Cavalié
- Experimental and Clinical Pharmacology and Toxicology, Saarland University, Homburg, Germany
| | - Roger C Whitehead
- School of Chemistry, Faculty of Science and Engineering, University of Manchester, Manchester, UK
| | - Eileithyia Swanton
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Stephen High
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Joen Luirink
- Department of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit, Amsterdam, the Netherlands
| |
Collapse
|
40
|
Structure of the Inhibited State of the Sec Translocon. Mol Cell 2020; 79:406-415.e7. [PMID: 32692975 PMCID: PMC7427319 DOI: 10.1016/j.molcel.2020.06.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/30/2020] [Accepted: 06/05/2020] [Indexed: 02/06/2023]
Abstract
Protein secretion in eukaryotes and prokaryotes involves a universally conserved protein translocation channel formed by the Sec61 complex. Unrelated small-molecule natural products and synthetic compounds inhibit Sec61 with differential effects for different substrates or for Sec61 from different organisms, making this a promising target for therapeutic intervention. To understand the mode of inhibition and provide insight into the molecular mechanism of this dynamic translocon, we determined the structure of mammalian Sec61 inhibited by the Mycobacterium ulcerans exotoxin mycolactone via electron cryo-microscopy. Unexpectedly, the conformation of inhibited Sec61 is optimal for substrate engagement, with mycolactone wedging open the cytosolic side of the lateral gate. The inability of mycolactone-inhibited Sec61 to effectively transport substrate proteins implies that signal peptides and transmembrane domains pass through the site occupied by mycolactone. This provides a foundation for understanding the molecular mechanism of Sec61 inhibitors and reveals novel features of translocon function and dynamics. The inhibited Sec translocon adopts a conformation optimal for substrate engagement The inhibitor mycolactone wedges open the lateral gate of Sec61α Mycolactone blocks the path taken by the signal peptide during engagement Resistance mutations are likely to operate by modulating translocon dynamics
Collapse
|
41
|
Lumangtad LA, Bell TW. The signal peptide as a new target for drug design. Bioorg Med Chem Lett 2020; 30:127115. [PMID: 32209293 PMCID: PMC7138182 DOI: 10.1016/j.bmcl.2020.127115] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/06/2020] [Accepted: 03/15/2020] [Indexed: 01/16/2023]
Abstract
Many current and potential drug targets are membrane-bound or secreted proteins that are expressed and transported via the Sec61 secretory pathway. They are targeted to translocon channels across the membrane of the endoplasmic reticulum (ER) by signal peptides (SPs), which are temporary structures on the N-termini of their nascent chains. During translation, such proteins enter the lumen and membrane of the ER by a process known as co-translational translocation. Small molecules have been found that interfere with this process, decreasing protein expression by recognizing the unique structures of the SPs of particular proteins. The SP may thus become a validated target for designing drugs for numerous disorders, including certain hereditary diseases.
Collapse
Affiliation(s)
| | - Thomas W Bell
- Department of Chemistry, University of Nevada, Reno, NV 89557-0216, USA.
| |
Collapse
|
42
|
Luesch H, Paavilainen VO. Natural products as modulators of eukaryotic protein secretion. Nat Prod Rep 2020; 37:717-736. [PMID: 32067014 DOI: 10.1039/c9np00066f] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Covering: up to the end of 2019Diverse natural product small molecules have allowed critical insights into processes that govern eukaryotic cells' ability to secrete cytosolically synthesized secretory proteins into their surroundings or to insert newly synthesized integral membrane proteins into the lipid bilayer of the endoplasmic reticulum. In addition, many components of the endoplasmic reticulum, required for protein homeostasis or other processes such as lipid metabolism or maintenance of calcium homeostasis, are being investigated for their potential in modulating human disease conditions such as cancer, neurodegenerative conditions and diabetes. In this review, we cover recent findings up to the end of 2019 on natural products that influence protein secretion or impact ER protein homeostasis, and serve as powerful chemical tools to understand protein flux through the mammalian secretory pathway and as leads for the discovery of new therapeutics.
Collapse
Affiliation(s)
- Hendrik Luesch
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, P.O. Box 100485, Gainesville, Florida 32610, USA.
| | | |
Collapse
|
43
|
Abstract
Natural products from microorganisms are important small molecules that play roles in various biological processes like cellular growth, motility, nutrient acquisition, stress response, biofilm formation, and defense. It is hypothesized that pathogens exploit these molecules to regulate virulence and persistence during infections. Here, we present selected examples of signaling natural products from human pathogenic bacteria that use these metabolites to gain a competitive advantage. Targeting these signaling systems provides novel strategies to antimicrobial treatments.
Collapse
Affiliation(s)
- Zhijuan Hu
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, 201 Gilman Hall, Berkeley, California 94720, United States
| | - Wenjun Zhang
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, 201 Gilman Hall, Berkeley, California 94720, United States
- Chan Zuckerberg Biohub, San Francisco, California 94158, United States
| |
Collapse
|
44
|
Reynaert ML, Dupoiron D, Yeramian E, Marsollier L, Brodin P. Could Mycolactone Inspire New Potent Analgesics? Perspectives and Pitfalls. Toxins (Basel) 2019; 11:toxins11090516. [PMID: 31487908 PMCID: PMC6783859 DOI: 10.3390/toxins11090516] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/26/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022] Open
Abstract
Pain currently represents the most common symptom for which medical attention is sought by patients. The available treatments have limited effectiveness and significant side-effects. In addition, most often, the duration of analgesia is short. Today, the handling of pain remains a major challenge. One promising alternative for the discovery of novel potent analgesics is to take inspiration from Mother Nature; in this context, the detailed investigation of the intriguing analgesia implemented in Buruli ulcer, an infectious disease caused by the bacterium Mycobacterium ulcerans and characterized by painless ulcerative lesions, seems particularly promising. More precisely, in this disease, the painless skin ulcers are caused by mycolactone, a polyketide lactone exotoxin. In fact, mycolactone exerts a wide range of effects on the host, besides being responsible for analgesia, as it has been shown notably to modulate the immune response or to provoke apoptosis. Several cellular mechanisms and different targets have been proposed to account for the analgesic effect of the toxin, such as nerve degeneration, the inhibition of inflammatory mediators and the activation of angiotensin II receptor 2. In this review, we discuss the current knowledge in the field, highlighting possible controversies. We first discuss the different pain-mimicking experimental models that were used to study the effect of mycolactone. We then detail the different variants of mycolactone that were used in such models. Overall, based on the results and the discussions, we conclude that the development of mycolactone-derived molecules can represent very promising perspectives for new analgesic drugs, which could be effective for specific pain indications.
Collapse
Affiliation(s)
- Marie-Line Reynaert
- France Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Denis Dupoiron
- Institut de Cancérologie de l'Ouest Paul Papin, 15 rue André Boquel-49055 Angers, France
| | - Edouard Yeramian
- Unité de Microbiologie Structurale, Institut Pasteur, CNRS, Univ. Paris, F-75015 Paris, France
| | - Laurent Marsollier
- Equipe ATIP AVENIR, CRCINA, INSERM, Univ. Nantes, Univ. Angers, 4 rue Larrey, F-49933 Angers, France.
| | - Priscille Brodin
- France Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France.
| |
Collapse
|
45
|
Gehringer M, Mäder P, Gersbach P, Pfeiffer B, Scherr N, Dangy JP, Pluschke G, Altmann KH. Configurationally Stabilized Analogs of M. ulcerans Exotoxins Mycolactones A and B Reveal the Importance of Side Chain Geometry for Mycolactone Virulence. Org Lett 2019; 21:5853-5857. [PMID: 31295000 DOI: 10.1021/acs.orglett.9b01947] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Mycolactones A/B (1a/b) are exotoxins of Mycobacterium ulcerans that are the molecular cause of Buruli ulcer. 1a/b represent a rapidly equilibrating mixture of Z/E isomers about the C4'═C5' double bond of the C5-side chain. Here, we describe the syntheses of mycolactone analogs with configurationally stable C5-side chains (2a, E mimetic; 2b/c, Z mimetics). Based on the cytotoxicity of 2a-c, the Δ4',5'-trans isomer of mycolactones A/B (1b) appears to be the major virulence factor.
Collapse
Affiliation(s)
- Matthias Gehringer
- Swiss Federal Institute of Technology (ETH) Zürich , Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences , Vladimir-Prelog-Weg 4 , 8093 Zürich , Switzerland
| | - Patrick Mäder
- Swiss Federal Institute of Technology (ETH) Zürich , Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences , Vladimir-Prelog-Weg 4 , 8093 Zürich , Switzerland
| | - Philipp Gersbach
- Swiss Federal Institute of Technology (ETH) Zürich , Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences , Vladimir-Prelog-Weg 4 , 8093 Zürich , Switzerland
| | - Bernhard Pfeiffer
- Swiss Federal Institute of Technology (ETH) Zürich , Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences , Vladimir-Prelog-Weg 4 , 8093 Zürich , Switzerland
| | - Nicole Scherr
- Swiss Tropical and Public Health Institute , Socinstrasse 57 , 4002 Basel , Switzerland
| | - Jean-Pierre Dangy
- Swiss Tropical and Public Health Institute , Socinstrasse 57 , 4002 Basel , Switzerland
| | - Gerd Pluschke
- Swiss Tropical and Public Health Institute , Socinstrasse 57 , 4002 Basel , Switzerland
| | - Karl-Heinz Altmann
- Swiss Federal Institute of Technology (ETH) Zürich , Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences , Vladimir-Prelog-Weg 4 , 8093 Zürich , Switzerland
| |
Collapse
|
46
|
Aydin F, Sun R, Swanson JMJ. Mycolactone Toxin Membrane Permeation: Atomistic versus Coarse-Grained MARTINI Simulations. Biophys J 2019; 117:87-98. [PMID: 31174850 PMCID: PMC6626831 DOI: 10.1016/j.bpj.2019.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 05/02/2019] [Accepted: 05/07/2019] [Indexed: 02/06/2023] Open
Abstract
Mycolactone, a cytotoxic and immunosuppressive macrolide produced by Mycobacterium ulcerans, is the central virulent factor in the skin disease Buruli ulcer. This multifunctional cytotoxin affects fundamental cellular processes such as cell adhesion, immune response, and cell death by targeting various cellular structures. Developing effective diagnostics that target mycolactone has been challenging, potentially because of suspected interactions with lipophilic architectures, including membranes. To better understand the pathogenesis of Buruli ulcer disease, aid in the development of diagnostics, and learn how amphiphiles in general use lipid trafficking to navigate the host environment, we seek to understand the nature of mycolactone-membrane interactions. Herein, we characterize how the two dominant isomers of mycolactone (A and B) interact with and permeate DPPC membranes with all-atom molecular dynamics simulations employing transition-tempered metadynamics and compare these results to those obtained by MARTINI coarse-grained simulations. Our all-atom simulations reveal that both isomers have a strong preference to associate with the membrane, although their mechanisms and energetics of membrane permeation differ slightly. Water molecules are found to play an important role in the permeation process. Although the MARTINI coarse-grained simulations give the correct free energy of membrane association, they fail to capture the mechanism of permeation and role of water during permeation as seen in all-atom simulations.
Collapse
Affiliation(s)
- Fikret Aydin
- Department of Chemistry and Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois
| | - Rui Sun
- Department of Chemistry and Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois
| | - Jessica M J Swanson
- Department of Chemistry and Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois.
| |
Collapse
|
47
|
Zong G, Hu Z, O’Keefe S, Tranter D, Iannotti MJ, Baron L, Hall B, Corfield K, Paatero AO, Henderson MJ, Roboti P, Zhou J, Sun X, Govindarajan M, Rohde JM, Blanchard N, Simmonds R, Inglese J, Du Y, Demangel C, High S, Paavilainen VO, Shi WQ. Ipomoeassin F Binds Sec61α to Inhibit Protein Translocation. J Am Chem Soc 2019; 141:8450-8461. [PMID: 31059257 PMCID: PMC6627486 DOI: 10.1021/jacs.8b13506] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Ipomoeassin F is a potent natural cytotoxin that inhibits growth of many tumor cell lines with single-digit nanomolar potency. However, its biological and pharmacological properties have remained largely unexplored. Building upon our earlier achievements in total synthesis and medicinal chemistry, we used chemical proteomics to identify Sec61α (protein transport protein Sec61 subunit alpha isoform 1), the pore-forming subunit of the Sec61 protein translocon, as a direct binding partner of ipomoeassin F in living cells. The interaction is specific and strong enough to survive lysis conditions, enabling a biotin analogue of ipomoeassin F to pull down Sec61α from live cells, yet it is also reversible, as judged by several experiments including fluorescent streptavidin staining, delayed competition in affinity pulldown, and inhibition of TNF biogenesis after washout. Sec61α forms the central subunit of the ER protein translocation complex, and the binding of ipomoeassin F results in a substantial, yet selective, inhibition of protein translocation in vitro and a broad ranging inhibition of protein secretion in live cells. Lastly, the unique resistance profile demonstrated by specific amino acid single-point mutations in Sec61α provides compelling evidence that Sec61α is the primary molecular target of ipomoeassin F and strongly suggests that the binding of this natural product to Sec61α is distinctive. Therefore, ipomoeassin F represents the first plant-derived, carbohydrate-based member of a novel structural class that offers new opportunities to explore Sec61α function and to further investigate its potential as a therapeutic target for drug discovery.
Collapse
Affiliation(s)
- Guanghui Zong
- †Department
of Chemistry and Biochemistry and ⬡Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701, United States,Department
of Chemistry and Biochemistry, University
of Maryland, College Park, Maryland 20742, United States
| | - Zhijian Hu
- †Department
of Chemistry and Biochemistry and ⬡Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Sarah O’Keefe
- School
of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Dale Tranter
- University
of Helsinki, HiLIFE, Helsinki, Finland,Institute
of Biotechnology, Helsinki, Finland
| | - Michael J. Iannotti
- National
Center for Advancing Translational Sciences, National
Institutes of Health, Rockville, Maryland 20850, United States
| | - Ludivine Baron
- Immunobiology
of Infection Unit, Institut Pasteur, 75015 Paris, France,INSERM, U1221, 75005 Paris, France
| | - Belinda Hall
- Department
of Microbial Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, Surrey GU2 7XH, United Kingdom
| | - Katherine Corfield
- Department
of Microbial Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, Surrey GU2 7XH, United Kingdom
| | - Anja O. Paatero
- University
of Helsinki, HiLIFE, Helsinki, Finland,Institute
of Biotechnology, Helsinki, Finland
| | - Mark J. Henderson
- National
Center for Advancing Translational Sciences, National
Institutes of Health, Rockville, Maryland 20850, United States
| | - Peristera Roboti
- School
of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Jianhong Zhou
- †Department
of Chemistry and Biochemistry and ⬡Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Xianwei Sun
- †Department
of Chemistry and Biochemistry and ⬡Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701, United States,Department
of Radiology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Mugunthan Govindarajan
- †Department
of Chemistry and Biochemistry and ⬡Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701, United States,Emory
Institute for Drug Development, Emory University, 954 Gatewood Road, Atlanta, Georgia 30329, United States
| | - Jason M. Rohde
- National
Center for Advancing Translational Sciences, National
Institutes of Health, Rockville, Maryland 20850, United States
| | - Nicolas Blanchard
- Université
de Haute-Alsace, Université de Strasbourg, CNRS, LIMA, UMR 7042, 68000 Mulhouse, France
| | - Rachel Simmonds
- Department
of Microbial Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, Surrey GU2 7XH, United Kingdom,
| | - James Inglese
- National
Center for Advancing Translational Sciences, National
Institutes of Health, Rockville, Maryland 20850, United States,
| | - Yuchun Du
- †Department
of Chemistry and Biochemistry and ⬡Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701, United States,
| | - Caroline Demangel
- Immunobiology
of Infection Unit, Institut Pasteur, 75015 Paris, France,INSERM, U1221, 75005 Paris, France,
| | - Stephen High
- School
of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom,
| | - Ville O. Paavilainen
- University
of Helsinki, HiLIFE, Helsinki, Finland,Institute
of Biotechnology, Helsinki, Finland,
| | - Wei Q. Shi
- †Department
of Chemistry and Biochemistry and ⬡Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701, United States,Department
of Chemistry, Ball State University, Muncie, Indiana 47306, United States,;
| |
Collapse
|
48
|
Klein W, Rutz C, Eckhard J, Provinciael B, Specker E, Neuenschwander M, Kleinau G, Scheerer P, von Kries JP, Nazaré M, Vermeire K, Schülein R. Use of a sequential high throughput screening assay to identify novel inhibitors of the eukaryotic SRP-Sec61 targeting/translocation pathway. PLoS One 2018; 13:e0208641. [PMID: 30543669 PMCID: PMC6292634 DOI: 10.1371/journal.pone.0208641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 11/20/2018] [Indexed: 11/22/2022] Open
Abstract
The SRP-Sec61 targeting/translocation pathway of eukaryotic cells targets nascent protein chains to the membrane of the endoplasmic reticulum. Using this machinery, secretory proteins are translocated across this membrane whereas membrane proteins are integrated into the lipid bilayer. One of the key players of the pathway is the protein-conducting Sec61 (translocon) complex of the endoplasmic reticulum. The Sec61 complex has no enzymatic activity, is expressed only intracellularly and is difficult to purify and to reconstitute. Screening for small molecule inhibitors impairing its functions is thus notoriously difficult. Such inhibitors may not only be valuable tools for cell biology, they may also represent novel anti-tumor drugs. Here we have developed a two-step, sequential screening assay for inhibitors of the whole SRP-Sec61 targeting/translocation pathway which might include molecules affecting Sec61 complex functions. The resulting hit compounds were analyzed using a whole cell biosynthesis assay and a cell free transcription/translation/translocation assay. Using this methodology, we identified novel compounds inhibiting this pathway. Following structure-based back screening, one of these substances was analyzed in more detail and we could show that it indeed impairs translocation at the level of the Sec61 complex. A slightly modified methodology may be used in the future to screen for substances affecting SecYEG, the bacterial ortholog of the Sec61 complex in order to derive novel antibiotic drugs.
Collapse
Affiliation(s)
- Wolfgang Klein
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Claudia Rutz
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Jamina Eckhard
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Becky Provinciael
- KU Leuven, Department of Microbiology and Immunology, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Edgar Specker
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | | | - Gunnar Kleinau
- Charité—Universitätsmedizin Berlin, Campus Charité Mitte, Charité Centrum Grundlagenmedizin CC2, Institut für Medizinische Physik und Biophysik, Group Protein X-ray Crystallography & Signal Transduction, Berlin, Germany
| | - Patrick Scheerer
- Charité—Universitätsmedizin Berlin, Campus Charité Mitte, Charité Centrum Grundlagenmedizin CC2, Institut für Medizinische Physik und Biophysik, Group Protein X-ray Crystallography & Signal Transduction, Berlin, Germany
| | | | - Marc Nazaré
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Kurt Vermeire
- KU Leuven, Department of Microbiology and Immunology, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Ralf Schülein
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
- * E-mail:
| |
Collapse
|
49
|
Demangel C, High S. Sec61 blockade by mycolactone: A central mechanism in Buruli ulcer disease. Biol Cell 2018; 110:237-248. [PMID: 30055020 DOI: 10.1111/boc.201800030] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/19/2018] [Indexed: 12/14/2022]
Abstract
Infection with Mycobacterium ulcerans results in a necrotising skin disease known as a Buruli ulcer, the pathology of which is directly linked to the bacterial production of the toxin mycolactone. Recent studies have identified the protein translocation machinery of the endoplasmic reticulum (ER) membrane as the primary cellular target of mycolactone, and shown that the toxin binds to the core subunit of the Sec61 complex. Mycolactone binding strongly inhibits the capacity of the Sec61 translocon to transport newly synthesised membrane and secretory proteins into and across the ER membrane. Since the ER acts as the entry point for the mammalian secretory pathway, and hence regulates initial access to the entire endomembrane system, mycolactone-treated cells have a reduced ability to produce a range of proteins including secretory cytokines and plasma membrane receptors. The global effect of this molecular blockade of protein translocation at the ER is that the host is unable to mount an effective immune response to the underlying mycobacterial infection. Prolonged exposure to mycolactone is normally cytotoxic, since it triggers stress responses activating the transcription factor ATF4 and ultimately inducing apoptosis.
Collapse
Affiliation(s)
- Caroline Demangel
- Immunobiology of Infection Unit, Institut Pasteur, Paris, France.,INSERM, U1221, Paris, France
| | - Stephen High
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| |
Collapse
|
50
|
Embgenbroich M, Burgdorf S. Current Concepts of Antigen Cross-Presentation. Front Immunol 2018; 9:1643. [PMID: 30061897 PMCID: PMC6054923 DOI: 10.3389/fimmu.2018.01643] [Citation(s) in RCA: 262] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/04/2018] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells have the ability to efficiently present internalized antigens on major histocompatibility complex (MHC) I molecules. This process is termed cross-presentation and is important role in the generation of an immune response against viruses and tumors, after vaccinations or in the induction of immune tolerance. The molecular mechanisms enabling cross-presentation have been topic of intense debate since many years. However, a clear view on these mechanisms remains difficult, partially due to important remaining questions, controversial results and discussions. Here, we give an overview of the current concepts of antigen cross-presentation and focus on a description of the major cross-presentation pathways, the role of retarded antigen degradation for efficient cross-presentation, the dislocation of antigens from endosomal compartment into the cytosol, the reverse transport of proteasome-derived peptides for loading on MHC I and the translocation of the cross-presentation machinery from the ER to endosomes. We try to highlight recent advances, discuss some of the controversial data and point out some of the major open questions in the field.
Collapse
Affiliation(s)
- Maria Embgenbroich
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Sven Burgdorf
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|