1
|
Binoy A, Nanjan P, Chellamuthu K, Liu H, Zhu S. A click chemistry-based biorthogonal approach for the detection and identification of protein lysine malonylation for osteoarthritis research. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.12.628274. [PMID: 39713453 PMCID: PMC11661220 DOI: 10.1101/2024.12.12.628274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Lysine malonylation is a post-translational modification where a malonyl group, characterized by a negatively charged carboxylate, is covalently attached to the Ɛ-amino side chain of lysine, influencing protein structure and function. Our laboratory identified Mak upregulation in cartilage under aging and obesity, contributing to osteoarthritis (OA). Current antibody-based detection methods face limitations in identifying Mak targets. Here, we introduce an alkyne-functionalized probe, MA-diyne, which metabolically incorporates into proteins, enabling copper(I) ion-catalyzed click reactions to conjugate labeled proteins with azide-based fluorescent dyes or affinity purification tags. In-gel fluorescence confirms MA-diyne incorporation into proteins across various cell types and species, including mouse chondrocytes, adipocytes, Hek293T cells, and C. elegans. Pull-down experiments identified known Mak proteins such as GAPDH and Aldolase. The extent of MA-diyne modification was higher in Sirtuin 5-deficient cells suggesting these modified proteins are Sirtuin 5 substrates. Pulse-chase experiments confirmed the dynamic nature of protein malonylation. Quantitative proteomics identified 1136 proteins corresponding to 8903 peptides with 429 proteins showing 1-fold increase in labeled group. Sirtuin 5 regulated 374 of these proteins. Pull down of newly identified proteins such as β-actin and Stat3 was also done. This study highlights MA-diyne as a powerful chemical tool to investigate the molecular targets and functions of lysine malonylation in OA conditions.
Collapse
Affiliation(s)
- Anupama Binoy
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine (HCOM), Ohio University, Athens, OH, 45701, USA
- Ohio Musculoskeletal and Neurological Institute (OMNI), Heritage College of Osteopathic Medicine (HCOM), Ohio University, Athens, OH, 45701, USA
| | - Pandurangan Nanjan
- Department of Chemistry, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore Campus, Tamilnadu, 641112, India
| | - Kavya Chellamuthu
- Department of Chemistry, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore Campus, Tamilnadu, 641112, India
| | - Huanhuan Liu
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine (HCOM), Ohio University, Athens, OH, 45701, USA
- Ohio Musculoskeletal and Neurological Institute (OMNI), Heritage College of Osteopathic Medicine (HCOM), Ohio University, Athens, OH, 45701, USA
| | - Shouan Zhu
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine (HCOM), Ohio University, Athens, OH, 45701, USA
- Ohio Musculoskeletal and Neurological Institute (OMNI), Heritage College of Osteopathic Medicine (HCOM), Ohio University, Athens, OH, 45701, USA
- Diabetes Institute (DI), Heritage College of Osteopathic Medicine (HCOM), Ohio University, Athens, OH, 45701, USA
| |
Collapse
|
2
|
Amiri A, Abedanzadeh S, Davaeil B, Shaabani A, Moosavi-Movahedi AA. Protein click chemistry and its potential for medical applications. Q Rev Biophys 2024; 57:e6. [PMID: 38619322 DOI: 10.1017/s0033583524000027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
A revolution in chemical biology occurred with the introduction of click chemistry. Click chemistry plays an important role in protein chemistry modifications, providing specific, sensitive, rapid, and easy-to-handle methods. Under physiological conditions, click chemistry often overlaps with bioorthogonal chemistry, defined as reactions that occur rapidly and selectively without interfering with biological processes. Click chemistry is used for the posttranslational modification of proteins based on covalent bond formations. With the contribution of click reactions, selective modification of proteins would be developed, representing an alternative to other technologies in preparing new proteins or enzymes for studying specific protein functions in different biological processes. Click-modified proteins have potential in diverse applications such as imaging, labeling, sensing, drug design, and enzyme technology. Due to the promising role of proteins in disease diagnosis and therapy, this review aims to highlight the growing applications of click strategies in protein chemistry over the last two decades, with a special emphasis on medicinal applications.
Collapse
Affiliation(s)
- Ahmad Amiri
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | | | - Bagher Davaeil
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ahmad Shaabani
- Department of Chemistry, Shahid Beheshti University, Tehran, Iran
| | | |
Collapse
|
3
|
Weigert Muñoz A, Zhao W, Sieber SA. Monitoring host-pathogen interactions using chemical proteomics. RSC Chem Biol 2024; 5:73-89. [PMID: 38333198 PMCID: PMC10849124 DOI: 10.1039/d3cb00135k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/09/2023] [Indexed: 02/10/2024] Open
Abstract
With the rapid emergence and the dissemination of microbial resistance to conventional chemotherapy, the shortage of novel antimicrobial drugs has raised a global health threat. As molecular interactions between microbial pathogens and their mammalian hosts are crucial to establish virulence, pathogenicity, and infectivity, a detailed understanding of these interactions has the potential to reveal novel therapeutic targets and treatment strategies. Bidirectional molecular communication between microbes and eukaryotes is essential for both pathogenic and commensal organisms to colonise their host. In particular, several devastating pathogens exploit host signalling to adjust the expression of energetically costly virulent behaviours. Chemical proteomics has emerged as a powerful tool to interrogate the protein interaction partners of small molecules and has been successfully applied to advance host-pathogen communication studies. Here, we present recent significant progress made by this approach and provide a perspective for future studies.
Collapse
Affiliation(s)
- Angela Weigert Muñoz
- Center for Functional Protein Assemblies, Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich Ernst-Otto-Fischer-Straße 8 D-85748 Garching Germany
| | - Weining Zhao
- College of Pharmacy, Shenzhen Technology University Shenzhen 518118 China
| | - Stephan A Sieber
- Center for Functional Protein Assemblies, Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich Ernst-Otto-Fischer-Straße 8 D-85748 Garching Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Germany
| |
Collapse
|
4
|
Saini G, Parasa MK, Clayton KN, Fraseur JG, Bolton SC, Lin KP, Wereley ST, Kinzer-Ursem TL. Immobilization of azide-functionalized proteins to micro- and nanoparticles directly from cell lysate. Mikrochim Acta 2023; 191:46. [PMID: 38129631 PMCID: PMC10739308 DOI: 10.1007/s00604-023-06068-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/23/2023] [Indexed: 12/23/2023]
Abstract
Immobilization of proteins and enzymes on solid supports has been utilized in a variety of applications, from improved protein stability on supported catalysts in industrial processes to fabrication of biosensors, biochips, and microdevices. A critical requirement for these applications is facile yet stable covalent conjugation between the immobilized and fully active protein and the solid support to produce stable, highly bio-active conjugates. Here, we report functionalization of solid surfaces (gold nanoparticles and magnetic beads) with bio-active proteins using site-specific and biorthogonal labeling and azide-alkyne cycloaddition, a click chemistry. Specifically, we recombinantly express and selectively label calcium-dependent proteins, calmodulin and calcineurin, and cAMP-dependent protein kinase A (PKA) with N-terminal azide-tags for efficient conjugation to nanoparticles and magnetic beads. We successfully immobilized the proteins on to the solid supports directly from the cell lysate with click chemistry, forgoing the step of purification. This approach is optimized to yield low particle aggregation and high levels of protein activity post-conjugation. The entire process enables streamlined workflows for bioconjugation and highly active conjugated proteins.
Collapse
Affiliation(s)
- Gunjan Saini
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47906, USA
| | - Mrugesh Krishna Parasa
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47906, USA
| | - Katherine N Clayton
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47906, USA
| | - Julia G Fraseur
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47906, USA
| | - Scott C Bolton
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47906, USA
| | - Kevin P Lin
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47906, USA
| | - Steven T Wereley
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47906, USA
| | - Tamara L Kinzer-Ursem
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47906, USA.
| |
Collapse
|
5
|
Chan AW, Broncel M, Yifrach E, Haseley NR, Chakladar S, Andree E, Herneisen AL, Shortt E, Treeck M, Lourido S. Analysis of CDPK1 targets identifies a trafficking adaptor complex that regulates microneme exocytosis in Toxoplasma. eLife 2023; 12:RP85654. [PMID: 37933960 PMCID: PMC10629828 DOI: 10.7554/elife.85654] [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: 11/08/2023] Open
Abstract
Apicomplexan parasites use Ca2+-regulated exocytosis to secrete essential virulence factors from specialized organelles called micronemes. Ca2+-dependent protein kinases (CDPKs) are required for microneme exocytosis; however, the molecular events that regulate trafficking and fusion of micronemes with the plasma membrane remain unresolved. Here, we combine sub-minute resolution phosphoproteomics and bio-orthogonal labeling of kinase substrates in Toxoplasma gondii to identify 163 proteins phosphorylated in a CDPK1-dependent manner. In addition to known regulators of secretion, we identify uncharacterized targets with predicted functions across signaling, gene expression, trafficking, metabolism, and ion homeostasis. One of the CDPK1 targets is a putative HOOK activating adaptor. In other eukaryotes, HOOK homologs form the FHF complex with FTS and FHIP to activate dynein-mediated trafficking of endosomes along microtubules. We show the FHF complex is partially conserved in T. gondii, consisting of HOOK, an FTS homolog, and two parasite-specific proteins (TGGT1_306920 and TGGT1_316650). CDPK1 kinase activity and HOOK are required for the rapid apical trafficking of micronemes as parasites initiate motility. Moreover, parasites lacking HOOK or FTS display impaired microneme protein secretion, leading to a block in the invasion of host cells. Taken together, our work provides a comprehensive catalog of CDPK1 targets and reveals how vesicular trafficking has been tuned to support a parasitic lifestyle.
Collapse
Affiliation(s)
- Alex W Chan
- Whitehead Institute for Biomedical ResearchCambridgeUnited States
- Biology Department, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Malgorzata Broncel
- Signaling in Apicomplexan Parasites Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Eden Yifrach
- Whitehead Institute for Biomedical ResearchCambridgeUnited States
| | - Nicole R Haseley
- Whitehead Institute for Biomedical ResearchCambridgeUnited States
| | | | - Elena Andree
- Whitehead Institute for Biomedical ResearchCambridgeUnited States
| | - Alice L Herneisen
- Whitehead Institute for Biomedical ResearchCambridgeUnited States
- Biology Department, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Emily Shortt
- Whitehead Institute for Biomedical ResearchCambridgeUnited States
| | - Moritz Treeck
- Signaling in Apicomplexan Parasites Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Sebastian Lourido
- Whitehead Institute for Biomedical ResearchCambridgeUnited States
- Biology Department, Massachusetts Institute of TechnologyCambridgeUnited States
| |
Collapse
|
6
|
Alexander AK, Elshahawi SI. Promiscuous Enzymes for Residue-Specific Peptide and Protein Late-Stage Functionalization. Chembiochem 2023; 24:e202300372. [PMID: 37338668 PMCID: PMC10496146 DOI: 10.1002/cbic.202300372] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 06/21/2023]
Abstract
The late-stage functionalization of peptides and proteins holds significant promise for drug discovery and facilitates bioorthogonal chemistry. This selective functionalization leads to innovative advances in in vitro and in vivo biological research. However, it is a challenging endeavor to selectively target a certain amino acid or position in the presence of other residues containing reactive groups. Biocatalysis has emerged as a powerful tool for selective, efficient, and economical modifications of molecules. Enzymes that have the ability to modify multiple complex substrates or selectively install nonnative handles have wide applications. Herein, we highlight enzymes with broad substrate tolerance that have been demonstrated to modify a specific amino acid residue in simple or complex peptides and/or proteins at late-stage. The different substrates accepted by these enzymes are mentioned together with the reported downstream bioorthogonal reactions that have benefited from the enzymatic selective modifications.
Collapse
Affiliation(s)
- Ashley K Alexander
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Rinker Health Science Campus, Irvine, CA 92618, USA
| | - Sherif I Elshahawi
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Rinker Health Science Campus, Irvine, CA 92618, USA
| |
Collapse
|
7
|
Chan AW, Broncel M, Yifrach E, Haseley N, Chakladar S, Andree E, Herneisen AL, Shortt E, Treeck M, Lourido S. Analysis of CDPK1 targets identifies a trafficking adaptor complex that regulates microneme exocytosis in Toxoplasma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.11.523553. [PMID: 36712004 PMCID: PMC9882037 DOI: 10.1101/2023.01.11.523553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Apicomplexan parasites use Ca2+-regulated exocytosis to secrete essential virulence factors from specialized organelles called micronemes. Ca2+-dependent protein kinases (CDPKs) are required for microneme exocytosis; however, the molecular events that regulate trafficking and fusion of micronemes with the plasma membrane remain unresolved. Here, we combine sub-minute resolution phosphoproteomics and bio-orthogonal labeling of kinase substrates in Toxoplasma gondii to identify 163 proteins phosphorylated in a CDPK1-dependent manner. In addition to known regulators of secretion, we identify uncharacterized targets with predicted functions across signaling, gene expression, trafficking, metabolism, and ion homeostasis. One of the CDPK1 targets is a putative HOOK activating adaptor. In other eukaryotes, HOOK homologs form the FHF complex with FTS and FHIP to activate dynein-mediated trafficking of endosomes along microtubules. We show the FHF complex is partially conserved in T. gondii, consisting of HOOK, an FTS homolog, and two parasite-specific proteins (TGGT1_306920 and TGGT1_316650). CDPK1 kinase activity and HOOK are required for the rapid apical trafficking of micronemes as parasites initiate motility. Moreover, parasites lacking HOOK or FTS display impaired microneme protein secretion, leading to a block in the invasion of host cells. Taken together, our work provides a comprehensive catalog of CDPK1 targets and reveals how vesicular trafficking has been tuned to support a parasitic lifestyle.
Collapse
Affiliation(s)
- Alex W Chan
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Biology Department, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Malgorzata Broncel
- Signaling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, London, UK
| | - Eden Yifrach
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Nicole Haseley
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | | | - Elena Andree
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Alice L Herneisen
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Biology Department, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emily Shortt
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Moritz Treeck
- Signaling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, London, UK
| | - Sebastian Lourido
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Biology Department, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
8
|
Michaluk P, Rusakov DA. Monitoring cell membrane recycling dynamics of proteins using whole-cell fluorescence recovery after photobleaching of pH-sensitive genetic tags. Nat Protoc 2022; 17:3056-3079. [PMID: 36064755 DOI: 10.1038/s41596-022-00732-4] [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: 11/21/2021] [Accepted: 06/07/2022] [Indexed: 11/08/2022]
Abstract
Population behavior of signaling molecules on the cell surface is key to their adaptive function. Live imaging of proteins tagged with fluorescent molecules has been an essential tool in understanding this behavior. Typically, genetic or chemical tags are used to target molecules present throughout the cell, whereas antibody-based tags label the externally exposed molecular domains only. Both approaches could potentially overlook the intricate process of in-out membrane recycling in which target molecules appear or disappear on the cell surface. This limitation is overcome by using a pH-sensitive fluorescent tag, such as Super-Ecliptic pHluorin (SEP), because its emission depends on whether it resides inside or outside the cell. Here we focus on the main glial glutamate transporter GLT1 and describe a genetic design that equips GLT1 molecules with SEP without interfering with the transporter's main function. Expressing GLT1-SEP in astroglia in cultures or in hippocampal slices enables monitoring of the real-time dynamics of the cell-surface and cytosolic fractions of the transporter in living cells. Whole-cell fluorescence recovery after photobleaching and quantitative image-kinetic analysis of the resulting time-lapse images enables assessment of the rate of GLT1-SEP recycling on the cell surface, a fundamental trafficking parameter unattainable previously. The present protocol takes 15-20 d to set up cell preparations, and 2-3 d to carry out live cell experiments and data analyses. The protocol can be adapted to study different membrane molecules of interest, particularly those proteins whose lifetime on the cell surface is critical to their adaptive function.
Collapse
Affiliation(s)
- Piotr Michaluk
- UCL Queen Square Institute of Neurology, University College London, London, UK.
- BRAINCITY, Laboratory of Neurobiology, Nencki Institute of Experimental Biology PAS, Warsaw, Poland.
| | - Dmitri A Rusakov
- UCL Queen Square Institute of Neurology, University College London, London, UK.
| |
Collapse
|
9
|
Wang Z, Zhang D, Hu S, Bi X, Lescar J, Tam JP, Liu CF. PAL-Mediated Ligation for Protein and Cell-Surface Modification. Methods Mol Biol 2022; 2530:177-193. [PMID: 35761050 DOI: 10.1007/978-1-0716-2489-0_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Peptidyl Asx-specific ligases (PALs) effect peptide ligation by catalyzing transpeptidation reactions at Asn/Asp-peptide bonds. Owing to their high efficiency and mild aqueous reaction conditions, these ligases have emerged as powerful biotechnological tools for protein manipulation in recent years. PALs are enzymes of the asparaginyl endopeptidase (AEP) superfamily but have predominant transpeptidase activity as opposed to typical AEPs which are predominantly hydrolases. Butelase-1 and VyPAL2, two PALs discovered by our teams, have been used successfully in a wide range of applications, including macrocyclization of synthetic peptides and recombinant proteins, protein N- or C-terminal modification, and cell-surface labeling. As shown in numerous reports, PAL-mediated ligation is highly efficient at Asn junctions. Although considerably less efficient, Asp-specific ligation has also been shown to be practically useful under suitable conditions. Herein, we describe the methods of using VyPAL2 for protein macrocyclization and labeling at an Asp residue as well as for protein dual labeling through orthogonal Asp- and Asn-directed ligations. We also describe a method for cell-surface protein modification using butelase-1, demonstrating its advantageous features over previous methods.
Collapse
Affiliation(s)
- Zhen Wang
- School of Biological Science, Nanyang Technological University, Singapore, Singapore
| | - Dingpeng Zhang
- School of Biological Science, Nanyang Technological University, Singapore, Singapore
| | - Side Hu
- School of Biological Science, Nanyang Technological University, Singapore, Singapore
| | - Xiaobao Bi
- School of Biological Science, Nanyang Technological University, Singapore, Singapore
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Zhejiang, China
| | - Julien Lescar
- School of Biological Science, Nanyang Technological University, Singapore, Singapore
| | - James P Tam
- School of Biological Science, Nanyang Technological University, Singapore, Singapore
| | - Chuan-Fa Liu
- School of Biological Science, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
10
|
Kallemeijn WW, Lanyon-Hogg T, Panyain N, Goya Grocin A, Ciepla P, Morales-Sanfrutos J, Tate EW. Proteome-wide analysis of protein lipidation using chemical probes: in-gel fluorescence visualization, identification and quantification of N-myristoylation, N- and S-acylation, O-cholesterylation, S-farnesylation and S-geranylgeranylation. Nat Protoc 2021; 16:5083-5122. [PMID: 34707257 DOI: 10.1038/s41596-021-00601-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 07/05/2021] [Indexed: 02/08/2023]
Abstract
Protein lipidation is one of the most widespread post-translational modifications (PTMs) found in nature, regulating protein function, structure and subcellular localization. Lipid transferases and their substrate proteins are also attracting increasing interest as drug targets because of their dysregulation in many disease states. However, the inherent hydrophobicity and potential dynamic nature of lipid modifications makes them notoriously challenging to detect by many analytical methods. Chemical proteomics provides a powerful approach to identify and quantify these diverse protein modifications by combining bespoke chemical tools for lipidated protein enrichment with quantitative mass spectrometry-based proteomics. Here, we report a robust and proteome-wide approach for the exploration of five major classes of protein lipidation in living cells, through the use of specific chemical probes for each lipid PTM. In-cell labeling of lipidated proteins is achieved by the metabolic incorporation of a lipid probe that mimics the specific natural lipid, concomitantly wielding an alkyne as a bio-orthogonal labeling tag. After incorporation, the chemically tagged proteins can be coupled to multifunctional 'capture reagents' by using click chemistry, allowing in-gel fluorescence visualization or enrichment via affinity handles for quantitative chemical proteomics based on label-free quantification (LFQ) or tandem mass-tag (TMT) approaches. In this protocol, we describe the application of lipid probes for N-myristoylation, N- and S-acylation, O-cholesterylation, S-farnesylation and S-geranylgeranylation in multiple cell lines to illustrate both the workflow and data obtained in these experiments. We provide detailed workflows for method optimization, sample preparation for chemical proteomics and data processing. A properly trained researcher (e.g., technician, graduate student or postdoc) can complete all steps from optimizing metabolic labeling to data processing within 3 weeks. This protocol enables sensitive and quantitative analysis of lipidated proteins at a proteome-wide scale at native expression levels, which is critical to understanding the role of lipid PTMs in health and disease.
Collapse
Affiliation(s)
- Wouter W Kallemeijn
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK
- The Francis Crick Institute, London, UK
| | - Thomas Lanyon-Hogg
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Nattawadee Panyain
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK
- Global Health Institute, Faculty of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Andrea Goya Grocin
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK
- The Francis Crick Institute, London, UK
| | - Paulina Ciepla
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK
- Institute of Chemical Sciences and Engineering (ISIC), Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Julia Morales-Sanfrutos
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK
- Proteomics Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Edward W Tate
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK.
- The Francis Crick Institute, London, UK.
| |
Collapse
|
11
|
Suazo KF, Park KY, Distefano MD. A Not-So-Ancient Grease History: Click Chemistry and Protein Lipid Modifications. Chem Rev 2021; 121:7178-7248. [PMID: 33821625 PMCID: PMC8820976 DOI: 10.1021/acs.chemrev.0c01108] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Protein lipid modification involves the attachment of hydrophobic groups to proteins via ester, thioester, amide, or thioether linkages. In this review, the specific click chemical reactions that have been employed to study protein lipid modification and their use for specific labeling applications are first described. This is followed by an introduction to the different types of protein lipid modifications that occur in biology. Next, the roles of click chemistry in elucidating specific biological features including the identification of lipid-modified proteins, studies of their regulation, and their role in diseases are presented. A description of the use of protein-lipid modifying enzymes for specific labeling applications including protein immobilization, fluorescent labeling, nanostructure assembly, and the construction of protein-drug conjugates is presented next. Concluding remarks and future directions are presented in the final section.
Collapse
Affiliation(s)
- Kiall F. Suazo
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455 USA
| | - Keun-Young Park
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455 USA
| | - Mark D. Distefano
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455 USA
| |
Collapse
|
12
|
De Rosa L, Di Stasi R, Romanelli A, D’Andrea LD. Exploiting Protein N-Terminus for Site-Specific Bioconjugation. Molecules 2021; 26:3521. [PMID: 34207845 PMCID: PMC8228110 DOI: 10.3390/molecules26123521] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/07/2021] [Accepted: 06/07/2021] [Indexed: 11/29/2022] Open
Abstract
Although a plethora of chemistries have been developed to selectively decorate protein molecules, novel strategies continue to be reported with the final aim of improving selectivity and mildness of the reaction conditions, preserve protein integrity, and fulfill all the increasing requirements of the modern applications of protein conjugates. The targeting of the protein N-terminal alpha-amine group appears a convenient solution to the issue, emerging as a useful and unique reactive site universally present in each protein molecule. Herein, we provide an updated overview of the methodologies developed until today to afford the selective modification of proteins through the targeting of the N-terminal alpha-amine. Chemical and enzymatic strategies enabling the selective labeling of the protein N-terminal alpha-amine group are described.
Collapse
Affiliation(s)
- Lucia De Rosa
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy; (L.D.R.); (R.D.S.)
| | - Rossella Di Stasi
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy; (L.D.R.); (R.D.S.)
| | - Alessandra Romanelli
- Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Via Venezian 21, 20133 Milano, Italy;
| | - Luca Domenico D’Andrea
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, CNR Via M. Bianco 9, 20131 Milano, Italy
| |
Collapse
|
13
|
Chamberlain LH, Shipston MJ, Gould GW. Regulatory effects of protein S-acylation on insulin secretion and insulin action. Open Biol 2021; 11:210017. [PMID: 33784857 PMCID: PMC8061761 DOI: 10.1098/rsob.210017] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/02/2021] [Indexed: 12/23/2022] Open
Abstract
Post-translational modifications (PTMs) such as phosphorylation and ubiquitination are well-studied events with a recognized importance in all aspects of cellular function. By contrast, protein S-acylation, although a widespread PTM with important functions in most physiological systems, has received far less attention. Perturbations in S-acylation are linked to various disorders, including intellectual disability, cancer and diabetes, suggesting that this less-studied modification is likely to be of considerable biological importance. As an exemplar, in this review, we focus on the newly emerging links between S-acylation and the hormone insulin. Specifically, we examine how S-acylation regulates key components of the insulin secretion and insulin response pathways. The proteins discussed highlight the diverse array of proteins that are modified by S-acylation, including channels, transporters, receptors and trafficking proteins and also illustrate the diverse effects that S-acylation has on these proteins, from membrane binding and micro-localization to regulation of protein sorting and protein interactions.
Collapse
Affiliation(s)
- Luke H. Chamberlain
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Michael J. Shipston
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Gwyn W. Gould
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
14
|
Bretherton RC, DeForest CA. The Art of Engineering Biomimetic Cellular Microenvironments. ACS Biomater Sci Eng 2021; 7:3997-4008. [PMID: 33523625 DOI: 10.1021/acsbiomaterials.0c01549] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cells and their surrounding microenvironment exist in dynamic reciprocity, where bidirectional feedback and feedforward crosstalk drives essential processes in development, homeostasis, and disease. With the ongoing explosion of customizable biomaterial innovation for dynamic cell culture, an ever-expanding suite of user-programmable scaffolds now exists to probe cell fate in response to spatiotemporally controlled biophysical and biochemical cues. Here, we highlight emerging trends in these efforts, emphasizing strategies that offer tunability over complex network mechanics, present biomolecular cues anisotropically, and harness cells as physiochemical actuators of the pericellular niche. Altogether, these material advances will lead to breakthroughs in our basic understanding of how cells interact with, integrate signals from, and influence their surrounding microenvironment.
Collapse
Affiliation(s)
- Ross C Bretherton
- Department of Bioengineering, University of Washington, Seattle, Washington 98105, United States.,Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States
| | - Cole A DeForest
- Department of Bioengineering, University of Washington, Seattle, Washington 98105, United States.,Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States.,Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States.,Department of Chemistry, University of Washington, Seattle, Washington 98195, United States.,Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
15
|
Losada de la Lastra A, Hassan S, Tate EW. Deconvoluting the biology and druggability of protein lipidation using chemical proteomics. Curr Opin Chem Biol 2021; 60:97-112. [PMID: 33221680 DOI: 10.1016/j.cbpa.2020.10.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 01/13/2023]
Abstract
Lipids are indispensable cellular building blocks, and their post-translational attachment to proteins makes them important regulators of many biological processes. Dysfunction of protein lipidation is also implicated in many pathological states, yet its systematic analysis presents significant challenges. Thanks to innovations in chemical proteomics, lipidation can now be readily studied by metabolic tagging using functionalized lipid analogs, enabling global profiling of lipidated substrates using mass spectrometry. This has spearheaded the first deconvolution of their full scope in a range of contexts, from cells to pathogens and multicellular organisms. Protein N-myristoylation, S-acylation, and S-prenylation are the most well-studied lipid post-translational modifications because of their extensive contribution to the regulation of diverse cellular processes. In this review, we focus on recent advances in the study of these post-translational modifications, with an emphasis on how novel mass spectrometry methods have elucidated their roles in fundamental biological processes.
Collapse
Affiliation(s)
- Ana Losada de la Lastra
- Department of Chemistry, Molecular Sciences Research Hub, White City Campus, Wood Lane, London, W12 0BZ, UK
| | - Sarah Hassan
- Department of Chemistry, Molecular Sciences Research Hub, White City Campus, Wood Lane, London, W12 0BZ, UK
| | - Edward W Tate
- Department of Chemistry, Molecular Sciences Research Hub, White City Campus, Wood Lane, London, W12 0BZ, UK.
| |
Collapse
|
16
|
Boto A, González CC, Hernández D, Romero-Estudillo I, Saavedra CJ. Site-selective modification of peptide backbones. Org Chem Front 2021. [DOI: 10.1039/d1qo00892g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Exciting developments in the site-selective modification of peptide backbones are allowing an outstanding fine-tuning of peptide conformation, folding ability, and physico-chemical and biological properties.
Collapse
Affiliation(s)
- Alicia Boto
- Instituto de Productos Naturales y Agrobiología del CSIC, Avda. Astrofísico Francisco Sánchez 3, 38206-La Laguna, Tenerife, Spain
| | - Concepción C. González
- Instituto de Productos Naturales y Agrobiología del CSIC, Avda. Astrofísico Francisco Sánchez 3, 38206-La Laguna, Tenerife, Spain
| | - Dácil Hernández
- Instituto de Productos Naturales y Agrobiología del CSIC, Avda. Astrofísico Francisco Sánchez 3, 38206-La Laguna, Tenerife, Spain
| | - Iván Romero-Estudillo
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos. Av. Universidad 1001, Cuernavaca, Morelos 62209, Mexico
- Catedrático CONACYT-CIQ-UAEM, Mexico
| | - Carlos J. Saavedra
- Instituto de Productos Naturales y Agrobiología del CSIC, Avda. Astrofísico Francisco Sánchez 3, 38206-La Laguna, Tenerife, Spain
- Programa Agustín de Betancourt, Universidad de la Laguna, 38200 Tenerife, Spain
| |
Collapse
|
17
|
Gautam S, Lian J, R. Gonçales V, Vogel YB, Ciampi S, Tilley RD, Gooding JJ. Surface Patterning of Biomolecules Using Click Chemistry and Light‐Activated Electrochemistry to Locally Generate Cu(I). ChemElectroChem 2020. [DOI: 10.1002/celc.202001097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Shreedhar Gautam
- School of Chemistry Australian Centre of NanoMedicine and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology The University of New South Wales Sydney 2052 Australia
| | - Jiaxin Lian
- School of Chemistry Australian Centre of NanoMedicine and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology The University of New South Wales Sydney 2052 Australia
| | - Vinicius R. Gonçales
- School of Chemistry Australian Centre of NanoMedicine and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology The University of New South Wales Sydney 2052 Australia
| | - Yan B. Vogel
- School of Molecular and Life Sciences Curtin Institute of Functional Molecules and Interfaces Curtin University Bentley 6102 WA Australia
| | - Simone Ciampi
- School of Molecular and Life Sciences Curtin Institute of Functional Molecules and Interfaces Curtin University Bentley 6102 WA Australia
| | - Richard D. Tilley
- School of Chemistry Australian Centre of NanoMedicine and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology The University of New South Wales Sydney 2052 Australia
- Electron Microscope Unit Mark Wainwright Analytical Centre The University of New South Wales Sydney 2052 Australia
| | - J. Justin Gooding
- School of Chemistry Australian Centre of NanoMedicine and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology The University of New South Wales Sydney 2052 Australia
| |
Collapse
|
18
|
Bell AS, Yu Z, Hutton JA, Wright MH, Brannigan JA, Paape D, Roberts SM, Sutherell CL, Ritzefeld M, Wilkinson AJ, Smith DF, Leatherbarrow RJ, Tate EW. Novel Thienopyrimidine Inhibitors of Leishmania N-Myristoyltransferase with On-Target Activity in Intracellular Amastigotes. J Med Chem 2020; 63:7740-7765. [PMID: 32575985 PMCID: PMC7383931 DOI: 10.1021/acs.jmedchem.0c00570] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
![]()
The
leishmaniases, caused by Leishmania species
of protozoan parasites, are neglected tropical diseases with millions
of cases worldwide. Current therapeutic approaches are limited by
toxicity, resistance, and cost. N-Myristoyltransferase
(NMT), an enzyme ubiquitous and essential in all eukaryotes, has been
validated via genetic and pharmacological methods as a promising anti-leishmanial
target. Here we describe a comprehensive structure–activity
relationship (SAR) study of a thienopyrimidine series previously identified
in a high-throughput screen against Leishmania NMT,
across 68 compounds in enzyme- and cell-based assay formats. Using
a chemical tagging target engagement biomarker assay, we identify
the first inhibitor in this series with on-target NMT activity in
leishmania parasites. Furthermore, crystal structure analyses of 12
derivatives in complex with Leishmania major NMT revealed key factors important for future structure-guided optimization
delivering IMP-105 (43), a compound with modest activity
against Leishmania donovani intracellular
amastigotes and excellent selectivity (>660-fold) for Leishmania NMT over human NMTs.
Collapse
Affiliation(s)
- Andrew S Bell
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, U.K. W12 0BZ
| | - Zhiyong Yu
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, U.K. W12 0BZ
| | - Jennie A Hutton
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, U.K. W12 0BZ
| | - Megan H Wright
- School of Chemistry, University of Leeds, Leeds, U.K. LS2 9JT
| | - James A Brannigan
- Structural Biology Laboratory, York Biomedical Research Institute, Department of Chemistry, University of York, York, U.K. YO10 5DD
| | - Daniel Paape
- Centre for Immunology and Infection, York Biomedical Research Institute, Department of Biology, University of York, York, U.K. YO10 5NG
| | - Shirley M Roberts
- Structural Biology Laboratory, York Biomedical Research Institute, Department of Chemistry, University of York, York, U.K. YO10 5DD
| | - Charlotte L Sutherell
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, U.K. W12 0BZ
| | - Markus Ritzefeld
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, U.K. W12 0BZ
| | - Anthony J Wilkinson
- Structural Biology Laboratory, York Biomedical Research Institute, Department of Chemistry, University of York, York, U.K. YO10 5DD
| | - Deborah F Smith
- Centre for Immunology and Infection, York Biomedical Research Institute, Department of Biology, University of York, York, U.K. YO10 5NG
| | - Robin J Leatherbarrow
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, U.K. W12 0BZ
| | - Edward W Tate
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, U.K. W12 0BZ
| |
Collapse
|
19
|
Broncel M, Dominicus C, Vigetti L, Nofal SD, Bartlett EJ, Touquet B, Hunt A, Wallbank BA, Federico S, Matthews S, Young JC, Tate EW, Tardieux I, Treeck M. Profiling of myristoylation in Toxoplasma gondii reveals an N-myristoylated protein important for host cell penetration. eLife 2020; 9:e57861. [PMID: 32618271 PMCID: PMC7373427 DOI: 10.7554/elife.57861] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/27/2020] [Indexed: 12/26/2022] Open
Abstract
N-myristoylation is a ubiquitous class of protein lipidation across eukaryotes and N-myristoyl transferase (NMT) has been proposed as an attractive drug target in several pathogens. Myristoylation often primes for subsequent palmitoylation and stable membrane attachment, however, growing evidence suggests additional regulatory roles for myristoylation on proteins. Here we describe the myristoylated proteome of Toxoplasma gondii using chemoproteomic methods and show that a small-molecule NMT inhibitor developed against related Plasmodium spp. is also functional in Toxoplasma. We identify myristoylation on a transmembrane protein, the microneme protein 7 (MIC7), which enters the secretory pathway in an unconventional fashion with the myristoylated N-terminus facing the lumen of the micronemes. MIC7 and its myristoylation play a crucial role in the initial steps of invasion, likely during the interaction with and penetration of the host cell. Myristoylation of secreted eukaryotic proteins represents a substantial expansion of the functional repertoire of this co-translational modification.
Collapse
Affiliation(s)
- Malgorzata Broncel
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Caia Dominicus
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Luis Vigetti
- Institute for Advanced Biosciences, Team Membrane Dynamics of Parasite-Host Cell Interactions, CNRS UMR5309, INSERM U1209, Université Grenoble AlpesGrenobleFrance
| | - Stephanie D Nofal
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Edward J Bartlett
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City CampusLondonUnited Kingdom
| | - Bastien Touquet
- Institute for Advanced Biosciences, Team Membrane Dynamics of Parasite-Host Cell Interactions, CNRS UMR5309, INSERM U1209, Université Grenoble AlpesGrenobleFrance
| | - Alex Hunt
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Bethan A Wallbank
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Stefania Federico
- The Peptide Chemistry STP, The Francis Crick InstituteLondonUnited Kingdom
| | - Stephen Matthews
- Department of Life Sciences, Imperial College London, South KensingtonLondonUnited Kingdom
| | - Joanna C Young
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Edward W Tate
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City CampusLondonUnited Kingdom
| | - Isabelle Tardieux
- Institute for Advanced Biosciences, Team Membrane Dynamics of Parasite-Host Cell Interactions, CNRS UMR5309, INSERM U1209, Université Grenoble AlpesGrenobleFrance
| | - Moritz Treeck
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| |
Collapse
|
20
|
Lanyon-Hogg T, Ritzefeld M, Sefer L, Bickel JK, Rudolf AF, Panyain N, Bineva-Todd G, Ocasio CA, O'Reilly N, Siebold C, Magee AI, Tate EW. Acylation-coupled lipophilic induction of polarisation (Acyl-cLIP): a universal assay for lipid transferase and hydrolase enzymes. Chem Sci 2019; 10:8995-9000. [PMID: 31762980 PMCID: PMC6855259 DOI: 10.1039/c9sc01785b] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/16/2019] [Indexed: 12/12/2022] Open
Abstract
Posttranslational attachment of lipids to proteins is important for many cellular functions, and the enzymes responsible for these modifications are implicated in many diseases, from cancer to neurodegeneration. Lipid transferases and hydrolases are increasingly tractable therapeutic targets, but present unique challenges for high-throughput biochemical enzyme assays which hinder development of new inhibitors. We present Acylation-coupled Lipophilic Induction of Polarisation (Acyl-cLIP) as the first universally applicable biochemical lipidation assay, exploiting the hydrophobic nature of lipidated peptides to drive a polarised fluorescence readout. Acyl-cLIP allows sensitive, accurate, real-time measurement of S- or N-palmitoylation, N-myristoylation, S-farnesylation or S-geranylgeranylation. Furthermore, it is applicable to transfer and hydrolysis reactions, and we demonstrate its extension to a high-throughput screening format. We anticipate that Acyl-cLIP will greatly expedite future drug discovery efforts against these challenging targets.
Collapse
Affiliation(s)
- Thomas Lanyon-Hogg
- Department of Chemistry , Imperial College London , London , W12 0BZ , UK . ; ; Tel: +44 0207 5943752 ; Tel: +44 0207 5945821
| | - Markus Ritzefeld
- Department of Chemistry , Imperial College London , London , W12 0BZ , UK . ; ; Tel: +44 0207 5943752 ; Tel: +44 0207 5945821
| | - Lea Sefer
- Division of Structural Biology , Wellcome Centre for Human Genetics , University of Oxford , Oxford , OX3 7BN , UK
| | - Jasmine K Bickel
- Department of Chemistry , Imperial College London , London , W12 0BZ , UK . ; ; Tel: +44 0207 5943752 ; Tel: +44 0207 5945821
| | - Amalie F Rudolf
- Division of Structural Biology , Wellcome Centre for Human Genetics , University of Oxford , Oxford , OX3 7BN , UK
| | - Nattawadee Panyain
- Department of Chemistry , Imperial College London , London , W12 0BZ , UK . ; ; Tel: +44 0207 5943752 ; Tel: +44 0207 5945821
| | | | | | | | - Christian Siebold
- Division of Structural Biology , Wellcome Centre for Human Genetics , University of Oxford , Oxford , OX3 7BN , UK
| | - Anthony I Magee
- Molecular Medicine Section , National Heart & Lung Institute , Imperial College London , London , SW7 2AZ , UK
| | - Edward W Tate
- Department of Chemistry , Imperial College London , London , W12 0BZ , UK . ; ; Tel: +44 0207 5943752 ; Tel: +44 0207 5945821
| |
Collapse
|
21
|
Serwa RA, Sekine E, Brown J, Teo SHC, Tate EW, O’Hare P. Analysis of a fully infectious bio-orthogonally modified human virus reveals novel features of virus cell entry. PLoS Pathog 2019; 15:e1007956. [PMID: 31589653 PMCID: PMC6797222 DOI: 10.1371/journal.ppat.1007956] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/17/2019] [Accepted: 09/12/2019] [Indexed: 12/28/2022] Open
Abstract
We report the analysis of a complex enveloped human virus, herpes simplex virus (HSV), assembled after in vivo incorporation of bio-orthogonal methionine analogues homopropargylglycine (HPG) or azidohomoalanine (AHA). We optimised protocols for the production of virions incorporating AHA (termed HSVAHA), identifying conditions which resulted in normal yields of HSV and normal particle/pfu ratios. Moreover we show that essentially every single HSVAHA capsid-containing particle was detectable at the individual particle level by chemical ligation of azide-linked fluorochromes to AHA-containing structural proteins. This was a completely specific chemical ligation, with no capsids assembled under normal methionine-containing conditions detected in parallel. We demonstrate by quantitative mass spectrometric analysis that HSVAHA virions exhibit no qualitative or quantitative differences in the repertoires of structural proteins compared to virions assembled under normal conditions. Individual proteins and AHA incorporation sites were identified in capsid, tegument and envelope compartments, including major essential structural proteins. Finally we reveal novel aspects of entry pathways using HSVAHA and chemical fluorochrome ligation that were not apparent from conventional immunofluorescence. Since ligation targets total AHA-containing protein and peptides, our results demonstrate the presence of abundant AHA-labelled products in cytoplasmic macrodomains and tubules which no longer contain intact particles detectable by immunofluorescence. Although these do not co-localise with lysosomal markers, we propose they may represent sites of proteolytic virion processing. Analysis of HSVAHA also enabled the discrimination from primary entering from secondary assembling virions, demonstrating assembly and second round infection within 6 hrs of initial infection and dual infections of primary and secondary virus in spatially restricted cytoplasmic areas of the same cell. Together with other demonstrated applications e.g., in genome biology, lipid and protein trafficking, this work further exemplifies the utility and potential of bio-orthogonal chemistry for studies in many aspects of virus-host interactions.
Collapse
Affiliation(s)
- Remigiusz A. Serwa
- Department of Chemistry, Molecular Sciences Research Hub, White City Campus, London, United Kingdom
| | - Eiki Sekine
- Section of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Jonathan Brown
- Section of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Su Hui Catherine Teo
- Section of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Edward W. Tate
- Department of Chemistry, Molecular Sciences Research Hub, White City Campus, London, United Kingdom
| | - Peter O’Hare
- Section of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
22
|
Shadish JA, Strange AC, DeForest CA. Genetically Encoded Photocleavable Linkers for Patterned Protein Release from Biomaterials. J Am Chem Soc 2019; 141:15619-15625. [PMID: 31525979 DOI: 10.1021/jacs.9b07239] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Given the critical role that proteins play in almost all biological processes, there is great interest in controlling their presentation within and release from biomaterials. Despite such outstanding enthusiasm, previously developed strategies in this regard result in ill-defined and heterogeneous populations with substantially decreased activity, precluding their successful application to fragile species including growth factors. Here, we introduce a modular and scalable method for creating monodisperse, genetically encoded chimeras that enable bioactive proteins to be immobilized within and subsequently photoreleased from polymeric hydrogels. Building upon recent developments in chemoenzymatic reactions, bioorthogonal chemistry, and optogenetics, we tether fluorescent proteins, model enzymes, and growth factors site-specifically to gel biomaterials through a photocleavable protein (PhoCl) that undergoes irreversible backbone photoscission upon exposure to cytocompatible visible light (λ ≈ 400 nm) in a dose-dependent manner. Mask-based and laser-scanning lithographic strategies using commonly available light sources are employed to spatiotemporally pattern protein release from hydrogels while retaining their full activity. The photopatterned epidermal growth factor presentation is exploited to promote anisotropic cellular proliferation in 3D. We expect these methods to be broadly useful for applications in diagnostics, drug delivery, and regenerative medicine.
Collapse
Affiliation(s)
| | | | - Cole A DeForest
- Department of Bioengineering , University of Washington , Seattle , Washington 98105 , United States.,Institute for Stem Cell & Regenerative Medicine , University of Washington , Seattle , Washington 98109 , United States
| |
Collapse
|
23
|
Yuan Q, Cheng Y, Lou X, Xia F. Rational Fabrication and Biomedical Application of Biomolecule‐Conjugated AIEgens through Click Reaction. CHINESE J CHEM 2019. [DOI: 10.1002/cjoc.201900211] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Qiming Yuan
- Engineering Research Center of Nano‐Geomaterials of Ministry of Education, Faculty of Materials Science and ChemistryChina University of Geosciences Wuhan Hubei 430074 China
| | - Yong Cheng
- Engineering Research Center of Nano‐Geomaterials of Ministry of Education, Faculty of Materials Science and ChemistryChina University of Geosciences Wuhan Hubei 430074 China
- State Key Laboratory of Material Processing and Die & Mould Technology, School of Materials Science and EngineeringHuazhong University of Science and Technology Wuhan Hubei 430074 China
| | - Xiaoding Lou
- Engineering Research Center of Nano‐Geomaterials of Ministry of Education, Faculty of Materials Science and ChemistryChina University of Geosciences Wuhan Hubei 430074 China
- Zhejiang Institute, China University of Geosciences Hangzhou Zhejiang 311305 China
| | - Fan Xia
- Engineering Research Center of Nano‐Geomaterials of Ministry of Education, Faculty of Materials Science and ChemistryChina University of Geosciences Wuhan Hubei 430074 China
| |
Collapse
|
24
|
Ham TR, Cox DG, Leipzig ND. Concurrent Delivery of Soluble and Immobilized Proteins to Recruit and Differentiate Neural Stem Cells. Biomacromolecules 2019; 20:3445-3452. [PMID: 31460746 DOI: 10.1021/acs.biomac.9b00719] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Insufficient endogenous neural stem cell (NSC) migration to injury sites and incomplete replenishment of neurons complicates recovery following central nervous system (CNS) injury. Such insufficient migration can be addressed by delivering soluble chemotactic factors, such as stromal cell-derived factor 1-α (SDF-1α), to sites of injury. However, simply enhancing NSC migration is likely to result in insufficient regeneration, as the cells need to be given additional signals. Immobilized proteins, such as interferon-γ (IFN-γ) can encourage neurogenic differentiation of NSCs. Here, we combined both protein delivery paradigms: soluble SDF-1α delivery to enhance NSC migration alongside covalently tethered IFN-γ to differentiate the recruited NSCs into neurons. To slow the release of soluble SDF-1α, we copolymerized methacrylated heparin with methacrylamide chitosan (MAC), to which we tethered IFN-γ. We found that this hydrogel system could result in soft hydrogels with a ratio of up to 70:30 MAC/heparin by mass, which enabled the continuous release of SDF-1α over a period of 2 weeks. The hydrogels recruited NSCs in vitro over 2 weeks, proportional to their release rate: the 70:30 heparin gels recruited a consistent number of NSCs at each time point, while the formulations with less heparin recruited NSCs at only early time points. After remaining in contact with the hydrogels for 8 days, NSCs successfully differentiated into neurons. CNS regeneration is a complex challenge, and this system provides a foundation to address multiple aspects of that challenge.
Collapse
Affiliation(s)
- Trevor R Ham
- Department of Biomedical Engineering, Auburn Science and Engineering Center 275, West Tower , The University of Akron , Akron , Ohio 44325 , United States
| | - Dakotah G Cox
- Department of Chemical and Biomolecular Engineering, Whitby 211 , The University of Akron , Akron , Ohio 44325 , United States
| | - Nic D Leipzig
- Department of Biomedical Engineering, Auburn Science and Engineering Center 275, West Tower , The University of Akron , Akron , Ohio 44325 , United States.,Department of Chemical and Biomolecular Engineering, Whitby 211 , The University of Akron , Akron , Ohio 44325 , United States
| |
Collapse
|
25
|
Schlott AC, Mayclin S, Reers AR, Coburn-Flynn O, Bell AS, Green J, Knuepfer E, Charter D, Bonnert R, Campo B, Burrows J, Lyons-Abbott S, Staker BL, Chung CW, Myler PJ, Fidock DA, Tate EW, Holder AA. Structure-Guided Identification of Resistance Breaking Antimalarial N‑Myristoyltransferase Inhibitors. Cell Chem Biol 2019; 26:991-1000.e7. [PMID: 31080074 PMCID: PMC6658617 DOI: 10.1016/j.chembiol.2019.03.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/25/2019] [Accepted: 03/25/2019] [Indexed: 01/26/2023]
Abstract
The attachment of myristate to the N-terminal glycine of certain proteins is largely a co-translational modification catalyzed by N-myristoyltransferase (NMT), and involved in protein membrane-localization. Pathogen NMT is a validated therapeutic target in numerous infectious diseases including malaria. In Plasmodium falciparum, NMT substrates are important in essential processes including parasite gliding motility and host cell invasion. Here, we generated parasites resistant to a particular NMT inhibitor series and show that resistance in an in vitro parasite growth assay is mediated by a single amino acid substitution in the NMT substrate-binding pocket. The basis of resistance was validated and analyzed with a structure-guided approach using crystallography, in combination with enzyme activity, stability, and surface plasmon resonance assays, allowing identification of another inhibitor series unaffected by this substitution. We suggest that resistance studies incorporated early in the drug development process help selection of drug combinations to impede rapid evolution of parasite resistance.
Collapse
Affiliation(s)
- Anja C Schlott
- Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Molecular Sciences Research Hub, Imperial College, White City Campus Wood Lane, London W12 0BZ, UK.
| | - Stephen Mayclin
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA 98109, USA; UCB Pharma, 7869 NE Day Road West, Bainbridge Island, WA 98110, USA
| | - Alexandra R Reers
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA 98109, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, USA
| | - Olivia Coburn-Flynn
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Andrew S Bell
- Molecular Sciences Research Hub, Imperial College, White City Campus Wood Lane, London W12 0BZ, UK
| | - Judith Green
- Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ellen Knuepfer
- Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - David Charter
- Structural and Biophysical Sciences, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Roger Bonnert
- Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215 Geneva 15, Switzerland
| | - Brice Campo
- Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215 Geneva 15, Switzerland
| | - Jeremy Burrows
- Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215 Geneva 15, Switzerland
| | - Sally Lyons-Abbott
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA 98109, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, USA
| | - Bart L Staker
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA 98109, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, USA
| | - Chun-Wa Chung
- Structural and Biophysical Sciences, GlaxoSmithKline, Stevenage, Hertfordshire, UK; Crick-GSK Biomedical LinkLabs, GSK Medicines Research Centre, Stevenage, UK
| | - Peter J Myler
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA 98109, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, USA; Department of Biomedical Informatics & Medical Education, University of Washington, Seattle, USA; Department of Global Health, University of Washington, Seattle, USA
| | - David A Fidock
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY 10032, USA; Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Edward W Tate
- Molecular Sciences Research Hub, Imperial College, White City Campus Wood Lane, London W12 0BZ, UK.
| | | |
Collapse
|
26
|
Kallemeijn WW, Lueg GA, Faronato M, Hadavizadeh K, Goya Grocin A, Song OR, Howell M, Calado DP, Tate EW. Validation and Invalidation of Chemical Probes for the Human N-myristoyltransferases. Cell Chem Biol 2019; 26:892-900.e4. [PMID: 31006618 PMCID: PMC6593224 DOI: 10.1016/j.chembiol.2019.03.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/20/2019] [Accepted: 03/06/2019] [Indexed: 12/15/2022]
Abstract
On-target, cell-active chemical probes are of fundamental importance in chemical and cell biology, whereas poorly characterized probes often lead to invalid conclusions. Human N-myristoyltransferase (NMT) has attracted increasing interest as target in cancer and infectious diseases. Here we report an in-depth comparison of five compounds widely applied as human NMT inhibitors, using a combination of quantitative whole-proteome N-myristoylation profiling, biochemical enzyme assays, cytotoxicity, in-cell protein synthesis, and cell-cycle assays. We find that N-myristoylation is unaffected by 2-hydroxymyristic acid (100 μM), D-NMAPPD (30 μM), or Tris-DBA palladium (10 μM), with the latter compounds causing cytotoxicity through mechanisms unrelated to NMT. In contrast, drug-like inhibitors IMP-366 (DDD85646) and IMP-1088 delivered complete and specific inhibition of N-myristoylation in a range of cell lines at 1 μM and 100 nM, respectively. This study enables the selection of appropriate on-target probes for future studies and suggests the need for reassessment of previous studies that used off-target compounds.
Collapse
Affiliation(s)
- Wouter W Kallemeijn
- Department of Chemistry, Imperial College London, Molecular Research Science Hub, 80 Wood Lane, London W12 0BZ, UK
| | - Gregor A Lueg
- Department of Chemistry, Imperial College London, Molecular Research Science Hub, 80 Wood Lane, London W12 0BZ, UK; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Monica Faronato
- Department of Chemistry, Imperial College London, Molecular Research Science Hub, 80 Wood Lane, London W12 0BZ, UK; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Kate Hadavizadeh
- Department of Chemistry, Imperial College London, Molecular Research Science Hub, 80 Wood Lane, London W12 0BZ, UK
| | - Andrea Goya Grocin
- Department of Chemistry, Imperial College London, Molecular Research Science Hub, 80 Wood Lane, London W12 0BZ, UK
| | - Ok-Ryul Song
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Michael Howell
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Dinis P Calado
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London SE1 9RT, UK
| | - Edward W Tate
- Department of Chemistry, Imperial College London, Molecular Research Science Hub, 80 Wood Lane, London W12 0BZ, UK; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
27
|
Ejendal KFK, Fraseur JG, Kinzer-Ursem TL. Protein Labeling and Bioconjugation Using N-Myristoyltransferase. Methods Mol Biol 2019; 2033:149-165. [PMID: 31332753 DOI: 10.1007/978-1-4939-9654-4_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Methods that allow for labeling of proteins cotranslationally within protein expression systems have had wide-ranging applications in health, engineering, and medicine. Bioorthogonal chemistries that allow for conjugation of proteins or biomolecules of interest to substrates (fluorophores, gold nanoparticles, polymers, etc.) in living cells without prior enrichment or purification have likewise enabled advances in technology to study and engineer cellular and biomolecular systems. At the intersection of these, chemoenzymatic labeling of proteins at specific sites of interest and their subsequent selective bioconjugation to substrates without prior purification has dramatically streamlined workflows that allow proteins to reside in the native expression volumes as long as possible prior to conjugation, be readily isolated upon conjugation, and remain functionally active after conjugation. Here we present methods and protocols to express and label proteins of interest at the N-terminus with azide derivatives of myristic acid, a small, soluble, 14-carbon fatty acid, and conjugate the labeled protein to fluorophores and gold nanoparticle substrates. These methods can be extended to label proteins with other myristoyl derivatives and to conjugation to other solid or polymeric substrates of interest.
Collapse
Affiliation(s)
- Karin F K Ejendal
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Julia G Fraseur
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Tamara L Kinzer-Ursem
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
28
|
Zhang Y, Park KY, Suazo KF, Distefano MD. Recent progress in enzymatic protein labelling techniques and their applications. Chem Soc Rev 2018; 47:9106-9136. [PMID: 30259933 PMCID: PMC6289631 DOI: 10.1039/c8cs00537k] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Protein-based conjugates are valuable constructs for a variety of applications. Conjugation of proteins to fluorophores is commonly used to study their cellular localization and the protein-protein interactions. Modification of therapeutic proteins with either polymers or cytotoxic moieties greatly enhances their pharmacokinetics or potency. To label a protein of interest, conventional direct chemical reaction with the side-chains of native amino acids often yields heterogeneously modified products. This renders their characterization complicated, requires difficult separation steps and may impact protein function. Although modification can also be achieved via the insertion of unnatural amino acids bearing bioorthogonal functional groups, these methods can have lower protein expression yields, limiting large scale production. As a site-specific modification method, enzymatic protein labelling is highly efficient and robust under mild reaction conditions. Significant progress has been made over the last five years in modifying proteins using enzymatic methods for numerous applications, including the creation of clinically relevant conjugates with polymers, cytotoxins or imaging agents, fluorescent or affinity probes to study complex protein interaction networks, and protein-linked materials for biosensing. This review summarizes developments in enzymatic protein labelling over the last five years for a panel of ten enzymes, including sortase A, subtiligase, microbial transglutaminase, farnesyltransferase, N-myristoyltransferase, phosphopantetheinyl transferases, tubulin tyrosin ligase, lipoic acid ligase, biotin ligase and formylglycine generating enzyme.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | | | | | |
Collapse
|
29
|
Liu L, Shadish JA, Arakawa CK, Shi K, Davis J, DeForest CA. Cyclic Stiffness Modulation of Cell-Laden Protein-Polymer Hydrogels in Response to User-Specified Stimuli including Light. ADVANCED BIOSYSTEMS 2018; 2:1800240. [PMID: 34316509 PMCID: PMC8312699 DOI: 10.1002/adbi.201800240] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Indexed: 11/10/2022]
Abstract
Although mechanical signals presented by the extracellular matrix are known to regulate many essential cell functions, the specific effects of these interactions, particularly in response to dynamic and heterogeneous cues, remain largely unknown. Here, we introduce a modular semisynthetic approach to create protein-polymer hydrogel biomaterials that undergo reversible stiffening in response to user-specified inputs. Employing a novel dual-chemoenzymatic modification strategy, we create fusion protein-based gel crosslinkers that exhibit stimuli-dependent intramolecular association. Linkers based on calmodulin yield calcium-sensitive materials, while those containing the photosensitive LOV2 (light, oxygen, and voltage sensing domain 2) protein give phototunable constructs whose moduli can be cycled on demand with spatiotemporal control about living cells. We exploit these unique materials to demonstrate the significant role that cyclic mechanical loading plays on fibroblast-to-myofibroblast transdifferentiation in three-dimensional (3D) space. Our moduli-switchable materials should prove useful for studies in mechanobiology, providing new avenues to probe and direct matrix-driven changes in 4D cell physiology.
Collapse
Affiliation(s)
- Luman Liu
- Department of Chemical Engineering, University of Washington, 3781 Okanogan Lane NE, Seattle, WA, 98195, USA
| | - Jared A Shadish
- Department of Chemical Engineering, University of Washington, 3781 Okanogan Lane NE, Seattle, WA, 98195, USA
| | - Christopher K Arakawa
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA, 98105, USA
| | - Kevin Shi
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA, 98105, USA
| | - Jennifer Davis
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA, 98105, USA; Department of Pathology, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA; Institute of Stem Cell & Regenerative Medicine, University of Washington, 850 Republican St., Seattle, WA, 98109, USA
| | - Cole A DeForest
- Department of Chemical Engineering, University of Washington, 3781 Okanogan Lane NE, Seattle, WA, 98195, USA; Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA, 98105, USA; Institute of Stem Cell & Regenerative Medicine, University of Washington, 850 Republican St., Seattle, WA, 98109, USA; Molecular Engineering & Sciences Institute, University of Washington, 3946 W Stevens Way NE, Seattle, WA, 98195, USA
| |
Collapse
|
30
|
The Cellular Senescence-Inhibited Gene Is Essential for PPM1A Myristoylation To Modulate Transforming Growth Factor β Signaling. Mol Cell Biol 2018; 38:MCB.00414-18. [PMID: 30201805 DOI: 10.1128/mcb.00414-18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 09/05/2018] [Indexed: 12/16/2022] Open
Abstract
The cellular senescence-inhibited gene (CSIG) is implicated in important biological processes, including cellular senescence and apoptosis. Our work showed that CSIG is involved in the myristoylation of the serine/threonine protein phosphatase PPM1A. Previous research has shown that myristoylation is necessary for PPM1A to dephosphorylate Smad2 and Smad3. However, the control and the biological significance of the myristoylation remain poorly understood. In this study, we found that CSIG knockdown disturbs PPM1A myristoylation and reduces the dephosphorylation by PPM1A of its substrate Smad2. By regulating PPM1A myristoylation, CSIG is involved in modulating the signaling of transforming growth factor β (TGF-β). Further study of the mechanism indicated that CSIG facilitates the interaction between N-myristoyltransferase 1 (NMT1) and PPM1A. Taking the data together, we found that CSIG is a regulator of PPM1A myristoylation and TGF-β signaling. By promoting the myristoylation of PPM1A, CSIG enhanced the phosphatase activity of PPM1A and further inhibited TGF-β signaling. This work not only extends the biological significance of CSIG but also provides new ideas and a reference for the study of the regulatory mechanism of myristoylation.
Collapse
|
31
|
Next generation calmodulin affinity purification: Clickable calmodulin facilitates improved protein purification. PLoS One 2018; 13:e0197120. [PMID: 29864125 PMCID: PMC5986150 DOI: 10.1371/journal.pone.0197120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 04/26/2018] [Indexed: 11/29/2022] Open
Abstract
As the proteomics field continues to expand, scientists are looking to integrate cross-disciplinary tools for studying protein structure, function, and interactions. Protein purification remains a key tool for many characterization studies. Calmodulin (CaM) is a calcium-binding messenger protein with over a hundred downstream binding partners, and is involved in a host of physiological processes, from learning and memory to immune and cardiac function. To facilitate biophysical studies of calmodulin, researchers have designed a site-specific labeling process for use in bioconjugation applications while maintaining high levels of protein activity. Here, we present a platform for selective conjugation of calmodulin directly from clarified cell lysates under bioorthogonal reaction conditions. Using a chemoenzymatically modified calmodulin, we employ popular click chemistry reactions for the conjugation of calmodulin to Sepharose resin, thereby streamlining a previously multi-step purification and conjugation process. We show that this “next-generation” calmodulin-Sepharose resin is not only easy to produce, but is also able to purify more calmodulin-binding proteins per volume of resin than traditional calmodulin-Sepharose resins. We expect these methods to be translatable to other proteins of interest and to other conjugation applications such as surface-based assays for the characterization of protein-protein interaction dynamics.
Collapse
|
32
|
Lee TH, Hirst DJ, Kulkarni K, Del Borgo MP, Aguilar MI. Exploring Molecular-Biomembrane Interactions with Surface Plasmon Resonance and Dual Polarization Interferometry Technology: Expanding the Spotlight onto Biomembrane Structure. Chem Rev 2018; 118:5392-5487. [PMID: 29793341 DOI: 10.1021/acs.chemrev.7b00729] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The molecular analysis of biomolecular-membrane interactions is central to understanding most cellular systems but has emerged as a complex technical challenge given the complexities of membrane structure and composition across all living cells. We present a review of the application of surface plasmon resonance and dual polarization interferometry-based biosensors to the study of biomembrane-based systems using both planar mono- or bilayers or liposomes. We first describe the optical principals and instrumentation of surface plasmon resonance, including both linear and extraordinary transmission modes and dual polarization interferometry. We then describe the wide range of model membrane systems that have been developed for deposition on the chips surfaces that include planar, polymer cushioned, tethered bilayers, and liposomes. This is followed by a description of the different chemical immobilization or physisorption techniques. The application of this broad range of engineered membrane surfaces to biomolecular-membrane interactions is then overviewed and how the information obtained using these techniques enhance our molecular understanding of membrane-mediated peptide and protein function. We first discuss experiments where SPR alone has been used to characterize membrane binding and describe how these studies yielded novel insight into the molecular events associated with membrane interactions and how they provided a significant impetus to more recent studies that focus on coincident membrane structure changes during binding of peptides and proteins. We then discuss the emerging limitations of not monitoring the effects on membrane structure and how SPR data can be combined with DPI to provide significant new information on how a membrane responds to the binding of peptides and proteins.
Collapse
Affiliation(s)
- Tzong-Hsien Lee
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Daniel J Hirst
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Ketav Kulkarni
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Mark P Del Borgo
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| |
Collapse
|
33
|
Lubin AS, Rueda-Zubiaurre A, Matthews H, Baumann H, Fisher FR, Morales-Sanfrutos J, Hadavizadeh KS, Nardella F, Tate EW, Baum J, Scherf A, Fuchter MJ. Development of a Photo-Cross-Linkable Diaminoquinazoline Inhibitor for Target Identification in Plasmodium falciparum. ACS Infect Dis 2018; 4:523-530. [PMID: 29377668 DOI: 10.1021/acsinfecdis.7b00228] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Diaminoquinazolines represent a privileged scaffold for antimalarial discovery, including use as putative Plasmodium histone lysine methyltransferase inhibitors. Despite this, robust evidence for their molecular targets is lacking. Here we report the design and development of a small-molecule photo-cross-linkable probe to investigate the targets of our diaminoquinazoline series. We demonstrate the effectiveness of our designed probe for photoaffinity labeling of Plasmodium lysates and identify similarities between the target profiles of the probe and the representative diaminoquinazoline BIX-01294. Initial pull-down proteomics experiments identified 104 proteins from different classes, many of which are essential, highlighting the suitability of the developed probe as a valuable tool for target identification in Plasmodium falciparum.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Flore Nardella
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, 25-28 Rue du Dr Roux, Paris 75015, France
- CNRS ERL
9195, Paris 75015, France
- INSERM Unit U1201, Paris 75015, France
| | | | | | - Artur Scherf
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, 25-28 Rue du Dr Roux, Paris 75015, France
- CNRS ERL
9195, Paris 75015, France
- INSERM Unit U1201, Paris 75015, France
| | | |
Collapse
|
34
|
Albrow VE, Grimley RL, Clulow J, Rose CR, Sun J, Warmus JS, Tate EW, Jones LH, Storer RI. Design and development of histone deacetylase (HDAC) chemical probes for cell-based profiling. MOLECULAR BIOSYSTEMS 2017; 12:1781-9. [PMID: 27021930 DOI: 10.1039/c6mb00109b] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Histone deacetylases (HDACs) contribute to regulation of gene expression by mediating higher-order chromatin structures. They assemble into large multiprotein complexes that regulate activity and specificity. We report the development of small molecule probes with class IIa and pan-HDAC activity that contain photoreactive crosslinking groups and either a biotin reporter, or a terminal alkyne handle for subsequent bioorthogonal ligation. The probes retained inhibitory activity against recombinant HDAC proteins and caused an accumulation of acetylated histone and tubulin following cell treatment. The versatility of the probes has been demonstrated by their ability to photoaffinity modify HDAC targets in vitro. An affinity enrichment probe was used in conjunction with mass spectrometry proteomics to isolate HDACs and their interacting proteins in a native proteome. The performance of the probes in recombinant versus cell-based systems highlights issues for the development of chemoproteomic technologies targeting class IIa HDACs in particular.
Collapse
Affiliation(s)
- Victoria E Albrow
- Pfizer Ltd, The Portway Building, Granta Park, Great Abington, Cambridge, CB21 6GS, UK
| | - Rachel L Grimley
- Pfizer Ltd, The Portway Building, Granta Park, Great Abington, Cambridge, CB21 6GS, UK
| | - James Clulow
- Department of Chemistry and Institute of Chemical Biology, Imperial College London, London, SW7 2AZ, UK
| | - Colin R Rose
- Worldwide Medicinal Chemistry, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, USA
| | - Jianmin Sun
- Worldwide Medicinal Chemistry, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, USA
| | - Joseph S Warmus
- Worldwide Medicinal Chemistry, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, USA
| | - Edward W Tate
- Department of Chemistry and Institute of Chemical Biology, Imperial College London, London, SW7 2AZ, UK
| | - Lyn H Jones
- Worldwide Medicinal Chemistry, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| | - R Ian Storer
- Worldwide Medicinal Chemistry, Pfizer Ltd, The Portway Building, Granta Park, Great Abington, Cambridge, CB21 6GS, UK.
| |
Collapse
|
35
|
Sivashankar S, Sapsanis C, Agambayev S, Buttner U, Salama KN. Label-free detection of sex determining region Y (SRY) via capacitive biosensor. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2017; 2016:4349-4352. [PMID: 28269241 DOI: 10.1109/embc.2016.7591690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In this work, we present for the first time, the use of a simple fractal capacitive biosensor for the quantification and detection of sex-determining region Y (SRY) genes. This section of genetic code, which is found on the Y chromosome, finds importance for study as it causes fetuses to develop characteristics of male sex-like gonads when a mutation occurs. It is also an important genetic code in men, and disorders involving the SRY gene can cause infertility and sexual malfunction that lead to a variety of gene mutational disorders. We have therefore designed silicon-based, label-free fractal capacitive biosensors to quantify various proteins and genes. We take advantage of a good dielectric material, Parylene C for enhancing the performance of the sensors. We have integrated these sensors with a simple microchannel for easy handling of fluids on the detection area. The read-out value of an Agilent LCR meter used to measure capacitance of the sensor at a frequency of 1 MHz determined gene specificity and gene quantification. These data revealed that the capacitance measurement of the capacitive biosensor for the SRY gene depended on both the target and the concentration of DNA. The experimental outcomes in the present study can be used to detect DNA and its variations in crucial fields that have a great impact on our daily lives, such as clinical and veterinary diagnostics, industrial and environmental testing and forensic sciences.
Collapse
|
36
|
Wright MH, Sieber SA. Chemical proteomics approaches for identifying the cellular targets of natural products. Nat Prod Rep 2017; 33:681-708. [PMID: 27098809 PMCID: PMC5063044 DOI: 10.1039/c6np00001k] [Citation(s) in RCA: 280] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This review focuses on chemical probes to identify the protein binding partners of natural products in living systems.
Covering: 2010 up to 2016 Deconvoluting the mode of action of natural products and drugs remains one of the biggest challenges in chemistry and biology today. Chemical proteomics is a growing area of chemical biology that seeks to design small molecule probes to understand protein function. In the context of natural products, chemical proteomics can be used to identify the protein binding partners or targets of small molecules in live cells. Here, we highlight recent examples of chemical probes based on natural products and their application for target identification. The review focuses on probes that can be covalently linked to their target proteins (either via intrinsic chemical reactivity or via the introduction of photocrosslinkers), and can be applied “in situ” – in living systems rather than cell lysates. We also focus here on strategies that employ a click reaction, the copper-catalysed azide–alkyne cycloaddition reaction (CuAAC), to allow minimal functionalisation of natural product scaffolds with an alkyne or azide tag. We also discuss ‘competitive mode’ approaches that screen for natural products that compete with a well-characterised chemical probe for binding to a particular set of protein targets. Fuelled by advances in mass spectrometry instrumentation and bioinformatics, many modern strategies are now embracing quantitative proteomics to help define the true interacting partners of probes, and we highlight the opportunities this rapidly evolving technology provides in chemical proteomics. Finally, some of the limitations and challenges of chemical proteomics approaches are discussed.
Collapse
Affiliation(s)
- M H Wright
- Department of Chemistry, Technische Universität München, Lichtenbergstraße 4, 85748, Garching, Germany.
| | - S A Sieber
- Department of Chemistry, Technische Universität München, Lichtenbergstraße 4, 85748, Garching, Germany.
| |
Collapse
|
37
|
Abstract
The formation of well-defined protein bioconjugates is critical for many studies and technologies in chemical biology. Tried-and-true methods for accomplishing this typically involve the targeting of cysteine residues, but the rapid growth of contemporary bioconjugate applications has required an expanded repertoire of modification techniques. One very powerful set of strategies involves the modification of proteins at their N termini, as these positions are typically solvent exposed and provide chemically distinct sites for many protein targets. Several chemical techniques can be used to modify N-terminal amino acids directly or convert them into unique functional groups for further ligations. A growing number of N-terminus-specific enzymatic ligation strategies have provided additional possibilities. This Perspective provides an overview of N-terminal modification techniques and the chemical rationale governing each. Examples of specific N-terminal protein conjugates are provided, along with their uses in a number of diverse biological applications.
Collapse
|
38
|
Ham TR, Farrag M, Leipzig ND. Covalent growth factor tethering to direct neural stem cell differentiation and self-organization. Acta Biomater 2017; 53:140-151. [PMID: 28161574 DOI: 10.1016/j.actbio.2017.01.068] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/20/2017] [Accepted: 01/25/2017] [Indexed: 12/12/2022]
Abstract
Tethered growth factors offer exciting new possibilities for guiding stem cell behavior. However, many of the current methods present substantial drawbacks which can limit their application and confound results. In this work, we developed a new method for the site-specific covalent immobilization of azide-tagged growth factors and investigated its utility in a model system for guiding neural stem cell (NSC) behavior. An engineered interferon-γ (IFN-γ) fusion protein was tagged with an N-terminal azide group, and immobilized to two different dibenzocyclooctyne-functionalized biomimetic polysaccharides (chitosan and hyaluronan). We successfully immobilized azide-tagged IFN-γ under a wide variety of reaction conditions, both in solution and to bulk hydrogels. To understand the interplay between surface chemistry and protein immobilization, we cultured primary rat NSCs on both materials and showed pronounced biological effects. Expectedly, immobilized IFN-γ increased neuronal differentiation on both materials. Expression of other lineage markers varied depending on the material, suggesting that the interplay of surface chemistry and protein immobilization plays a large role in nuanced cell behavior. We also investigated the bioactivity of immobilized IFN-γ in a 3D environment in vivo and found that it sparked the robust formation of neural tube-like structures from encapsulated NSCs. These findings support a wide range of potential uses for this approach and provide further evidence that adult NSCs are capable of self-organization when exposed to the proper microenvironment. STATEMENT OF SIGNIFICANCE For stem cells to be used effectively in regenerative medicine applications, they must be provided with the appropriate cues and microenvironment so that they integrate with existing tissue. This study explores a new method for guiding stem cell behavior: covalent growth factor tethering. We found that adding an N-terminal azide-tag to interferon-γ enabled stable and robust Cu-free 'click' immobilization under a variety of physiologic conditions. We showed that the tagged growth factors retained their bioactivity when immobilized and were able to guide neural stem cell lineage commitment in vitro. We also showed self-organization and neurulation from neural stem cells in vivo. This approach will provide another tool for the orchestration of the complex signaling events required to guide stem cell integration.
Collapse
|
39
|
Demetriadou A, Morales-Sanfrutos J, Nearchou M, Baba O, Kyriacou K, Tate EW, Drousiotou A, Petrou PP. Mouse Stbd1 is N-myristoylated and affects ER-mitochondria association and mitochondrial morphology. J Cell Sci 2017; 130:903-915. [PMID: 28137759 PMCID: PMC5358331 DOI: 10.1242/jcs.195263] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 01/16/2017] [Indexed: 12/17/2022] Open
Abstract
Starch binding domain-containing protein 1 (Stbd1) is a carbohydrate-binding protein that has been proposed to be a selective autophagy receptor for glycogen. Here, we show that mouse Stbd1 is a transmembrane endoplasmic reticulum (ER)-resident protein with the capacity to induce the formation of organized ER structures in HeLa cells. In addition to bulk ER, Stbd1 was found to localize to mitochondria-associated membranes (MAMs), which represent regions of close apposition between the ER and mitochondria. We demonstrate that N-myristoylation and binding of Stbd1 to glycogen act as major determinants of its subcellular targeting. Moreover, overexpression of non-myristoylated Stbd1 enhanced the association between ER and mitochondria, and further induced prominent mitochondrial fragmentation and clustering. Conversely, shRNA-mediated Stbd1 silencing resulted in an increase in the spacing between ER and mitochondria, and an altered morphology of the mitochondrial network, suggesting elevated fusion and interconnectivity of mitochondria. Our data unravel the molecular mechanism underlying Stbd1 subcellular targeting, support and expand its proposed function as a selective autophagy receptor for glycogen and uncover a new role for the protein in the physical association between ER and mitochondria.
Collapse
Affiliation(s)
- Anthi Demetriadou
- Department of Biochemical Genetics, The Cyprus Institute of Neurology and Genetics, P. O. Box 23462, Nicosia 1683, Cyprus
- The Cyprus School of Molecular Medicine, P. O. Box 23462, Nicosia 1683, Cyprus
| | | | - Marianna Nearchou
- Department of Electron Microscopy / Molecular Pathology, The Cyprus Institute of Neurology and Genetics, P. O. Box 23462, Nicosia 1683, Cyprus
| | - Otto Baba
- Oral and Maxillofacial Anatomy, Faculty of Dentistry, Tokushima University, Tokushima 770-8504, Japan
| | - Kyriacos Kyriacou
- The Cyprus School of Molecular Medicine, P. O. Box 23462, Nicosia 1683, Cyprus
- Department of Electron Microscopy / Molecular Pathology, The Cyprus Institute of Neurology and Genetics, P. O. Box 23462, Nicosia 1683, Cyprus
| | - Edward W Tate
- Department of Chemistry, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - Anthi Drousiotou
- Department of Biochemical Genetics, The Cyprus Institute of Neurology and Genetics, P. O. Box 23462, Nicosia 1683, Cyprus
- The Cyprus School of Molecular Medicine, P. O. Box 23462, Nicosia 1683, Cyprus
| | - Petros P Petrou
- Department of Biochemical Genetics, The Cyprus Institute of Neurology and Genetics, P. O. Box 23462, Nicosia 1683, Cyprus
- The Cyprus School of Molecular Medicine, P. O. Box 23462, Nicosia 1683, Cyprus
| |
Collapse
|
40
|
|
41
|
Nonradioactive quantification of autophagic protein degradation with L-azidohomoalanine labeling. Nat Protoc 2017; 12:279-288. [PMID: 28079880 DOI: 10.1038/nprot.2016.160] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
At present, several assays that use radioisotope labeling to quantify the degradation of long-lived proteins have been developed to measure autophagic flux. Here, we describe a nonradioactive pulse-chase protocol using L-azidohomoalanine (AHA) labeling to quantify long-lived protein degradation during autophagy. AHA is used as a surrogate for L-methionine, and, when added to cultured cells grown in methionine-free medium, AHA is incorporated into proteins during de novo protein synthesis. After a chase period to remove short-lived proteins, autophagy is induced by starvation or other stimuli. Cells then undergo a 'click' reaction between the azide group of AHA and a fluorescently tagged alkyne probe. The AHA-containing proteins can then be detected by flow cytometry. This protocol is nonradioactive, sensitive and quantitative, and it is easy to perform. It is also applicable to various cell culture systems. The whole protocol is estimated to take 4-5 d to complete.
Collapse
|
42
|
Ozer I, Chilkoti A. Site-Specific and Stoichiometric Stealth Polymer Conjugates of Therapeutic Peptides and Proteins. Bioconjug Chem 2017; 28:713-723. [PMID: 27998056 DOI: 10.1021/acs.bioconjchem.6b00652] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
As potent and selective therapeutic agents, peptides and proteins are an important class of drugs, but they typically have suboptimal pharmacokinetic profiles. One approach to solve this problem is their conjugation with "stealth" polymers. Conventional methods for conjugation of this class of polymers to peptides and proteins are typically carried out by reactions that have poor yield and provide limited control over the site of conjugation and the stoichiometry of the conjugate. To address these limitations, new chemical and biological approaches have been developed that provide new molecular tools in the bioconjugation toolbox to create stealth polymer conjugates of peptides and proteins with exquisite control over their properties. This review article highlights these recent advances in the synthesis of therapeutic peptide- and protein-stealth polymer conjugates.
Collapse
Affiliation(s)
- Imran Ozer
- Department of Biomedical Engineering, Duke University , 101 Science Drive, Durham, North Carolina 27708, United States
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University , 101 Science Drive, Durham, North Carolina 27708, United States
| |
Collapse
|
43
|
Gao H, Sun W, Song Z, Yu Y, Wang L, Chen X, Zhang Q. A Method to Generate and Analyze Modified Myristoylated Proteins. Chembiochem 2017; 18:324-330. [PMID: 27925692 DOI: 10.1002/cbic.201600608] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Indexed: 11/07/2022]
Abstract
Covalent lipid modification of proteins is essential to their cellular localizations and functions. Engineered lipid motifs, coupled with bio-orthogonal chemistry, have been utilized to identify myristoylated or palmitoylated proteins in cells. However, whether modified proteins have similar properties as endogenous ones has not been well investigated mainly due to lack of methods to generate and analyze purified proteins. We have developed a method that utilizes metabolic interference and mass spectrometry to produce and analyze modified, myristoylated small GTPase ADP-ribosylation factor 1 (Arf1). The capacities of these recombinant proteins to bind liposomes and load and hydrolyze GTP were measured and compared with the unmodified myristoylated Arf1. The ketone-modified myristoylated Arf1 could be further labeled by fluorophore-coupled hydrazine and subsequently visualized through fluorescence imaging. This methodology provides an effective model system to characterize lipid-modified proteins with additional functions before applying them to cellular systems.
Collapse
Affiliation(s)
- Huanyao Gao
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, NC, 27599, USA
| | - Wei Sun
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, NC, 27599, USA
| | - Zhiquan Song
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, NC, 27599, USA
| | - Yanbao Yu
- Department of Biochemistry and Biophysics, School of Medicine, The University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC, 27599, USA
| | - Li Wang
- Department of Biochemistry and Biophysics, School of Medicine, The University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC, 27599, USA
| | - Xian Chen
- Department of Biochemistry and Biophysics, School of Medicine, The University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC, 27599, USA
| | - Qisheng Zhang
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, NC, 27599, USA
| |
Collapse
|
44
|
Rodgers U, Lanyon-Hogg T, Masumoto N, Ritzefeld M, Burke R, Blagg J, Magee AI, Tate EW. Characterization of Hedgehog Acyltransferase Inhibitors Identifies a Small Molecule Probe for Hedgehog Signaling by Cancer Cells. ACS Chem Biol 2016; 11:3256-3262. [PMID: 27779865 PMCID: PMC5349656 DOI: 10.1021/acschembio.6b00896] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 10/13/2016] [Indexed: 01/01/2023]
Abstract
The Sonic Hedgehog (Shh) signaling pathway plays a critical role during embryonic development and cancer progression. N-terminal palmitoylation of Shh by Hedgehog acyltransferase (Hhat) is essential for efficient signaling, raising interest in Hhat as a novel drug target. A recently identified series of dihydrothienopyridines has been proposed to function via this mode of action; however, the lead compound in this series (RUSKI-43) was subsequently shown to possess cytotoxic activity unrelated to canonical Shh signaling. To identify a selective chemical probe for cellular studies, we profiled three RUSKI compounds in orthogonal cell-based assays. We found that RUSKI-43 exhibits off-target cytotoxicity, masking its effect on Hhat-dependent signaling, hence results obtained with this compound in cells should be treated with caution. In contrast, RUSKI-201 showed no off-target cytotoxicity, and quantitative whole-proteome palmitoylation profiling with a bioorthogonal alkyne-palmitate reporter demonstrated specific inhibition of Hhat in cells. RUSKI-201 is the first selective Hhat chemical probe in cells and should be used in future studies of Hhat catalytic function.
Collapse
Affiliation(s)
- Ursula
R. Rodgers
- Molecular
Medicine Section, National Heart & Lung
Institute, Imperial College London, London SW7 2AZ, United
Kingdom
| | - Thomas Lanyon-Hogg
- Department
of Chemistry, Imperial College London, London SW7 2AZ, United Kingdom
| | - Naoko Masumoto
- Department
of Chemistry, Imperial College London, London SW7 2AZ, United Kingdom
| | - Markus Ritzefeld
- Department
of Chemistry, Imperial College London, London SW7 2AZ, United Kingdom
| | - Rosemary Burke
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London, SW7 3RP, United Kingdom
| | - Julian Blagg
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London, SW7 3RP, United Kingdom
| | - Anthony I. Magee
- Molecular
Medicine Section, National Heart & Lung
Institute, Imperial College London, London SW7 2AZ, United
Kingdom
| | - Edward W. Tate
- Department
of Chemistry, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
45
|
Ho SH, Tirrell DA. Chemoenzymatic Labeling of Proteins for Imaging in Bacterial Cells. J Am Chem Soc 2016; 138:15098-15101. [PMID: 27933886 DOI: 10.1021/jacs.6b07067] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Reliable methods to determine the subcellular localization of bacterial proteins are needed for the study of prokaryotic cell biology. We describe here a simple and general technique for imaging of bacterial proteins in situ by fluorescence microscopy. The method uses the eukaryotic enzyme N-myristoyltransferase to modify the N-terminus of the protein of interest with an azido fatty acid. Subsequent strain-promoted azide-alkyne cycloaddition allows conjugation of dyes and imaging of tagged proteins by confocal fluorescence microscopy. We demonstrate the method by labeling the chemotaxis proteins Tar and CheA and the cell division proteins FtsZ and FtsA in Escherichia coli. We observe distinct spatial patterns for each of these proteins in both fixed and live cells. The method should prove broadly useful for protein imaging in bacteria.
Collapse
Affiliation(s)
- Samuel H Ho
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Boulevard, Pasadena, California 91125, United States
| | - David A Tirrell
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Boulevard, Pasadena, California 91125, United States
| |
Collapse
|
46
|
Wright MH, Paape D, Price HP, Smith DF, Tate EW. Global Profiling and Inhibition of Protein Lipidation in Vector and Host Stages of the Sleeping Sickness Parasite Trypanosoma brucei. ACS Infect Dis 2016; 2:427-441. [PMID: 27331140 PMCID: PMC4906374 DOI: 10.1021/acsinfecdis.6b00034] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Indexed: 01/05/2023]
Abstract
The enzyme N-myristoyltransferase (NMT) catalyzes the essential fatty acylation of substrate proteins with myristic acid in eukaryotes and is a validated drug target in the parasite Trypanosoma brucei, the causative agent of African trypanosomiasis (sleeping sickness). N-Myristoylation typically mediates membrane localization of proteins and is essential to the function of many. However, only a handful of proteins are experimentally validated as N-myristoylated in T. brucei. Here, we perform metabolic labeling with an alkyne-tagged myristic acid analogue, enabling the capture of lipidated proteins in insect and host life stages of T. brucei. We further compare this with a longer chain palmitate analogue to explore the chain length-specific incorporation of fatty acids into proteins. Finally, we combine the alkynyl-myristate analogue with NMT inhibitors and quantitative chemical proteomics to globally define N-myristoylated proteins in the clinically relevant bloodstream form parasites. This analysis reveals five ARF family small GTPases, calpain-like proteins, phosphatases, and many uncharacterized proteins as substrates of NMT in the parasite, providing a global view of the scope of this important protein modification and further evidence for the crucial and pleiotropic role of NMT in the cell.
Collapse
Affiliation(s)
- Megan H. Wright
- Department of Chemistry, Imperial College London, London SW7 2AZ, United Kingdom
| | - Daniel Paape
- Centre for Immunology and Infection, Department
of Biology, University of York, York YO10 5DD, United Kingdom
| | - Helen P. Price
- Centre for Immunology and Infection, Department
of Biology, University of York, York YO10 5DD, United Kingdom
| | - Deborah F. Smith
- Centre for Immunology and Infection, Department
of Biology, University of York, York YO10 5DD, United Kingdom
| | - Edward W. Tate
- Department of Chemistry, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
47
|
Gerl MJ, Bittl V, Kirchner S, Sachsenheimer T, Brunner HL, Lüchtenborg C, Özbalci C, Wiedemann H, Wegehingel S, Nickel W, Haberkant P, Schultz C, Krüger M, Brügger B. Sphingosine-1-Phosphate Lyase Deficient Cells as a Tool to Study Protein Lipid Interactions. PLoS One 2016; 11:e0153009. [PMID: 27100999 PMCID: PMC4839656 DOI: 10.1371/journal.pone.0153009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 03/21/2016] [Indexed: 12/11/2022] Open
Abstract
Cell membranes contain hundreds to thousands of individual lipid species that are of structural importance but also specifically interact with proteins. Due to their highly controlled synthesis and role in signaling events sphingolipids are an intensely studied class of lipids. In order to investigate their metabolism and to study proteins interacting with sphingolipids, metabolic labeling based on photoactivatable sphingoid bases is the most straightforward approach. In order to monitor protein-lipid-crosslink products, sphingosine derivatives containing a reporter moiety, such as a radiolabel or a clickable group, are used. In normal cells, degradation of sphingoid bases via action of the checkpoint enzyme sphingosine-1-phosphate lyase occurs at position C2-C3 of the sphingoid base and channels the resulting hexadecenal into the glycerolipid biosynthesis pathway. In case the functionalized sphingosine looses the reporter moiety during its degradation, specificity towards sphingolipid labeling is maintained. In case degradation of a sphingosine derivative does not remove either the photoactivatable or reporter group from the resulting hexadecenal, specificity towards sphingolipid labeling can be achieved by blocking sphingosine-1-phosphate lyase activity and thus preventing sphingosine derivatives to be channeled into the sphingolipid-to-glycerolipid metabolic pathway. Here we report an approach using clustered, regularly interspaced, short palindromic repeats (CRISPR)-associated nuclease Cas9 to create a sphingosine-1-phosphate lyase (SGPL1) HeLa knockout cell line to disrupt the sphingolipid-to-glycerolipid metabolic pathway. We found that the lipid and protein compositions as well as sphingolipid metabolism of SGPL1 knock-out HeLa cells only show little adaptations, which validates these cells as model systems to study transient protein-sphingolipid interactions.
Collapse
Affiliation(s)
- Mathias J. Gerl
- Heidelberg University Biochemistry Center, Heidelberg, Germany
- * E-mail: (MJG); (BB)
| | - Verena Bittl
- Heidelberg University Biochemistry Center, Heidelberg, Germany
| | | | | | | | | | - Cagakan Özbalci
- Heidelberg University Biochemistry Center, Heidelberg, Germany
| | | | | | - Walter Nickel
- Heidelberg University Biochemistry Center, Heidelberg, Germany
| | - Per Haberkant
- European Molecular Biology Laboratory, Heidelberg, Germany
| | | | | | - Britta Brügger
- Heidelberg University Biochemistry Center, Heidelberg, Germany
- * E-mail: (MJG); (BB)
| |
Collapse
|
48
|
Broncel M, Serwa RA, Bunney TD, Katan M, Tate EW. Global Profiling of Huntingtin-associated protein E (HYPE)-Mediated AMPylation through a Chemical Proteomic Approach. Mol Cell Proteomics 2015; 15:715-25. [PMID: 26604261 DOI: 10.1074/mcp.o115.054429] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Indexed: 01/31/2023] Open
Abstract
AMPylation of mammalian small GTPases by bacterial virulence factors can be a key step in bacterial infection of host cells, and constitutes a potential drug target. This posttranslational modification also exists in eukaryotes, and AMP transferase activity was recently assigned to HYPE Filamentation induced by cyclic AMP domain containing protein (FICD) protein, which is conserved from Caenorhabditis elegans to humans. In contrast to bacterial AMP transferases, only a small number of HYPE substrates have been identified by immunoprecipitation and mass spectrometry approaches, and the full range of targets is yet to be determined in mammalian cells. We describe here the first example of global chemoproteomic screening and substrate validation for HYPE-mediated AMPylation in mammalian cell lysate. Through quantitative mass-spectrometry-based proteomics coupled with novel chemoproteomic tools providing MS/MS evidence of AMP modification, we identified a total of 25 AMPylated proteins, including the previously validated substrate endoplasmic reticulum (ER) chaperone BiP (HSPA5), and also novel substrates involved in pathways of gene expression, ATP biosynthesis, and maintenance of the cytoskeleton. This dataset represents the largest library of AMPylated human proteins reported to date and a foundation for substrate-specific investigations that can ultimately decipher the complex biological networks involved in eukaryotic AMPylation.
Collapse
Affiliation(s)
- Malgorzata Broncel
- From the ‡Department of Chemistry, Imperial College London, Exhibition Road, London SW7 2AZ, UK; ¶Current address: The Institute of Cancer Research, Division of Cancer Biology, 237 Fulham Road, London SW3 6JB, UK
| | - Remigiusz A Serwa
- From the ‡Department of Chemistry, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - Tom D Bunney
- §Division of Biosciences, Institute of Structural and Molecular Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Matilda Katan
- §Division of Biosciences, Institute of Structural and Molecular Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Edward W Tate
- From the ‡Department of Chemistry, Imperial College London, Exhibition Road, London SW7 2AZ, UK;
| |
Collapse
|
49
|
Kulkarni C, Lo M, Fraseur JG, Tirrell DA, Kinzer-Ursem TL. Bioorthogonal Chemoenzymatic Functionalization of Calmodulin for Bioconjugation Applications. Bioconjug Chem 2015; 26:2153-60. [PMID: 26431265 DOI: 10.1021/acs.bioconjchem.5b00449] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Calmodulin (CaM) is a widely studied Ca(2+)-binding protein that is highly conserved across species and involved in many biological processes, including vesicle release, cell proliferation, and apoptosis. To facilitate biophysical studies of CaM, researchers have tagged and mutated CaM at various sites, enabling its conjugation to fluorophores, microarrays, and other reactive partners. However, previous attempts to add a reactive label to CaM for downstream studies have generally employed nonselective labeling methods or resulted in diminished CaM function. Here we report the first engineered CaM protein that undergoes site-specific and bioorthogonal labeling while retaining wild-type activity levels. By employing a chemoenzymatic labeling approach, we achieved selective and quantitative labeling of the engineered CaM protein with an N-terminal 12-azidododecanoic acid tag; notably, addition of the tag did not interfere with the ability of CaM to bind Ca(2+) or a partner protein. The specificity of our chemoenzymatic labeling approach also allowed for selective conjugation of CaM to reactive partners in bacterial cell lysates, without intermediate purification of the engineered protein. Additionally, we prepared CaM-affinity resins that were highly effective in purifying a representative CaM-binding protein, demonstrating that the engineered CaM remains active even after surface capture. Beyond studies of CaM and CaM-binding proteins, the protein engineering and surface capture methods described here should be translatable to other proteins and other bioconjugation applications.
Collapse
Affiliation(s)
- Chethana Kulkarni
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States
| | - Megan Lo
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States
| | - Julia G Fraseur
- Weldon School of Biomedical Engineering, Purdue University , 206 South Martin Jischke Drive, West Lafayette, Indiana 47907, United States
| | - David A Tirrell
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States
| | - Tamara L Kinzer-Ursem
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States.,Weldon School of Biomedical Engineering, Purdue University , 206 South Martin Jischke Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
50
|
Guttery DS, Poulin B, Ramaprasad A, Wall RJ, Ferguson DJP, Brady D, Patzewitz EM, Whipple S, Straschil U, Wright MH, Mohamed AMAH, Radhakrishnan A, Arold ST, Tate EW, Holder AA, Wickstead B, Pain A, Tewari R. Genome-wide functional analysis of Plasmodium protein phosphatases reveals key regulators of parasite development and differentiation. Cell Host Microbe 2015; 16:128-40. [PMID: 25011111 PMCID: PMC4094981 DOI: 10.1016/j.chom.2014.05.020] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 04/17/2014] [Accepted: 05/27/2014] [Indexed: 12/24/2022]
Abstract
Reversible protein phosphorylation regulated by kinases and phosphatases controls many cellular processes. Although essential functions for the malaria parasite kinome have been reported, the roles of most protein phosphatases (PPs) during Plasmodium development are unknown. We report a functional analysis of the Plasmodium berghei protein phosphatome, which exhibits high conservation with the P. falciparum phosphatome and comprises 30 predicted PPs with differential and distinct expression patterns during various stages of the life cycle. Gene disruption analysis of P. berghei PPs reveals that half of the genes are likely essential for asexual blood stage development, whereas six are required for sexual development/sporogony in mosquitoes. Phenotypic screening coupled with transcriptome sequencing unveiled morphological changes and altered gene expression in deletion mutants of two N-myristoylated PPs. These findings provide systematic functional analyses of PPs in Plasmodium, identify how phosphatases regulate parasite development and differentiation, and can inform the identification of drug targets for malaria. Phylogenetic analysis identifies 30 Plasmodium berghei protein phosphatases (PPs) Functional analysis reveals role for six PPs in sexual development/sporogony Two N-myristoylated PPs play key roles in sex allocation and parasite transmission RNA-Seq highlights significantly altered gene clusters in the N-myristoylated PP mutants
Collapse
Affiliation(s)
- David S Guttery
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
| | - Benoit Poulin
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
| | - Abhinay Ramaprasad
- Computational Bioscience Research Center (CBRC), Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Richard J Wall
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
| | - David J P Ferguson
- Nuffield Department of Clinical Laboratory Science, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Declan Brady
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
| | - Eva-Maria Patzewitz
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
| | - Sarah Whipple
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
| | - Ursula Straschil
- Division of Cell and Molecular Biology, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - Megan H Wright
- Department of Chemistry, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - Alyaa M A H Mohamed
- Computational Bioscience Research Center (CBRC), Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Anand Radhakrishnan
- Computational Bioscience Research Center (CBRC), Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Stefan T Arold
- Computational Bioscience Research Center (CBRC), Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Edward W Tate
- Department of Chemistry, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - Anthony A Holder
- Division of Parasitology, MRC National Institute for Medical Research, Mill Hill, London NW7 1AA, UK.
| | - Bill Wickstead
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
| | - Arnab Pain
- Computational Bioscience Research Center (CBRC), Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Rita Tewari
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK.
| |
Collapse
|