1
|
Zhao X, Ou Y, Bai R, Yin J, Wang G, Wang J, Zhao X, Liang Y, Li Q. In situ cleavage-based sortase A-mediated site-specific immobilization of beta2-adrenoceptor on gold surface for surface plasmon resonance measurement. Biosens Bioelectron 2025; 281:117452. [PMID: 40228458 DOI: 10.1016/j.bios.2025.117452] [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] [Received: 01/02/2025] [Revised: 03/14/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
Surface Plasmon Resonance (SPR) is a pivotal technique for measuring biomolecular interactions, with the sensor surface typically made of gold or silver and requiring proteins to be immobilized in a controlled manner. Traditional methods, such as random crosslinking via covalent amide bonds (EDC/NHS strategy), resulting in diverse protein orientations. Alternatively, site-specific immobilization strategies offer better orientation control, they are still challenged by the purification needs for protein of interests and steric hindrance produced by bulk protein tags. To address these issues, we proposed a novel protein immobilization strategy relying on in situ cleavage and Sortase A (SrtA) to immobilize functional protein on SPR sensor chips. This strategy involves the β2-adrenoceptor (β2AR) as a model, incorporating an endogenous protease recognition site (EPRS) as a linker to fuse SrtA with β2AR, which contains an SrtA recognition sequence (LPXTG) at its C-terminal. When expressed in Escherichia coli (E. coli), the protease cleaves the EPRS, releasing SrtA and β2AR. When the lysate is mixed with an oligo-Gly or oligo-Gly-modified SPR chip, transpeptidation occurs, covalently immobilizing β2AR. The efficacy of the cleavage and transpeptidation reactions was validated through SDS-PAGE, Western blot, and chromatographic analysis. The SPR chip was characterized by scanning electron microscope (SEM), energy dispersive spectroscopy (EDS) mapping, X-ray photoelectron spectroscopy (XPS), and contact angle analysis, while β2AR activity was evaluated by SPR. When compared to the EDC/NHS-based random method and the haloalkane dehalogenase (HaloTag)-mediated site-specific strategy, β2AR immobilized through the SrtA-mediated method exhibited higher activity with ligands, demonstrating precision in binding affinity evaluations. This strategy meets the benchmarks for an optimal site-specific immobilization method and holds promise for applications involving the modification of other biological interfaces or biosensors.
Collapse
Affiliation(s)
- Xue Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Yuanyuan Ou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Ruoxue Bai
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Jiatai Yin
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Ge Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Jing Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Xinfeng Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Yinku Liang
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, 723001, China.
| | - Qian Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China.
| |
Collapse
|
2
|
Bjørlie M, Pontoppidan L, Yesiltas B, Sørensen ADM, Redwine J, Jacobsen C. Low pH affinity screening of peptides: An alternative to time-consuming storage experiments? Food Chem 2025; 480:143823. [PMID: 40117825 DOI: 10.1016/j.foodchem.2025.143823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 02/12/2025] [Accepted: 03/08/2025] [Indexed: 03/23/2025]
Abstract
Metal-catalyzed lipid oxidation substantially reduces the shelf life of mayonnaise, thereby increasing the demand for metal-chelating antioxidants. This study, for the first time, combined Surface Plasmon Resonance (SPR) and Immobilized Metal Affinity Chromatography (IMAC) at low pH with a 28-day storage experiment in mayonnaise to screen peptides for antioxidant activity. These novel techniques could potentially replace time-consuming storage experiments in the discovery of new antioxidants. However, the application of SPR at low pH was hindered by non-specific binding, despite attempts to address this issue through various strategies. Surprisingly, IMAC experiments showed a distinct difference in affinity for Ni2+ and Fe3+ among all peptides tested, suggesting that peptide-metal ion affinity varies significantly. Nonetheless, no peptides enhanced mayonnaise's oxidative stability; some even acted as prooxidants. These findings emphasize the complexity of predicting antioxidant efficacy in food matrices and highlight the urgent need for further research to refine low pH screening methodologies.
Collapse
Affiliation(s)
- Mads Bjørlie
- National Food Institute, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Laura Pontoppidan
- National Food Institute, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Betül Yesiltas
- National Food Institute, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | | | | | - Charlotte Jacobsen
- National Food Institute, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
3
|
Hossain M, Dodda SR, Das S, Aikat K, Mukhopadhyay SS. Catalytic tunnel engineering of thermostable endoglucanase of GH7 family (W356C) from Aspergillus fumigatus gains catalytic rate. Enzyme Microb Technol 2025; 187:110632. [PMID: 40139016 DOI: 10.1016/j.enzmictec.2025.110632] [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] [Received: 12/10/2024] [Revised: 02/20/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025]
Abstract
Tunnel engineering targets the access tunnels in enzymes, which is crucial for substrate binding and product release. Modifying the tunnels can lead to better biomass-degrading abilities of the lignocellulolytic enzymes. In this report, we have engineered the thermostable GH7 family endoglucanase from Aspergillus fumigatus (AfEgl7). The residues in the open tunnel having the highest bottleneck radius are mutated. Mutations are created (T229F, W356C) in the non-conserved region. The mutant W356C showed a 2-fold increase in product release rate (Vmax = 375.8 µM/min) and 2.5-fold higher catalytic activity (Kcat = 75.1 min-1) compared to wild-type (Vmax= 232 µM/min; Kcat = 30.9 min-1) using CM cellulose as substrate. The mutant T229F lost both catalytic activity and thermostability. Molecular dynamic simulations and docking studies of W356C revealed a change in structure near the product exit region, which may facilitate faster product release and account for the increased catalytic efficiency of the mutant. This study showed how redesigning the access pathways can be a promising strategy for protein engineering and de novo protein design by tailoring the open tunnel geometry to a ligand-specific manner.
Collapse
Affiliation(s)
- Musaddique Hossain
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal 713209, India
| | - Subba Reddy Dodda
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal 713209, India
| | - Shalini Das
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal 713209, India
| | - Kaustav Aikat
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal 713209, India
| | - Sudit S Mukhopadhyay
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal 713209, India.
| |
Collapse
|
4
|
Sato Y, Sumiyoshi R, Yamagishi M, Haraguchi T, Matsuda K, Sato S, Ito K, Yajima J. Reconstructing the Motility Driven by Membrane-Bound Myosin on the Inner Surface of Cell-Sized Droplets. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:10077-10084. [PMID: 40238146 DOI: 10.1021/acs.langmuir.4c04123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Molecular motors and the cytoskeleton perform essential cellular functions by interacting with the cell membrane. Reconstituting the behavior of motor proteins interacting with biological membranes and cytoskeletal filaments within a cell-sized space provides insights into their intracellular functions. A water-in-oil (W/O) emulsion droplet represents an invaluable experimental system because it offers an encapsulated space that mimics the intracellular environment. In this study, we aimed to reconstitute the actomyosin motility on the flat membrane surface of cell-sized W/O droplets using membrane-bound myosin I that anchors to and exerts force on both the cell membrane and actin cytoskeleton. Myosin IC or myosin ID, which binds specifically to the cell membrane phospholipid phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] together with the actin cytoskeleton, was encapsulated within W/O droplets. Myosin IC and myosin ID caused gliding of actin filaments on the inner bottom membrane of the hemispherical droplet containing PI(4,5)P2, and myosin-driven actin filament movement on the flat membrane surface was quantified. Fast motor myosin ID was more sensitive than slow motor myosin IC to the geometrical conditions and binding manner to the membrane. Therefore, the in-droplet actin filament gliding assay is a useful tool for elucidating the molecular mechanisms underlying cellular events occurring at the cell membrane, which are achieved through the concerted action of myosin and the actin cytoskeleton.
Collapse
Affiliation(s)
- Yusei Sato
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Rieko Sumiyoshi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Masahiko Yamagishi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
- Komaba Institute for Science, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Takeshi Haraguchi
- Department of Biology, Graduate School of Science, Chiba University, 1-33 Yayoicho, Inage-ku, Chiba 263-8522, Japan
| | - Kyohei Matsuda
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Suguru Sato
- Department of Biology, Graduate School of Science, Chiba University, 1-33 Yayoicho, Inage-ku, Chiba 263-8522, Japan
| | - Kohji Ito
- Department of Biology, Graduate School of Science, Chiba University, 1-33 Yayoicho, Inage-ku, Chiba 263-8522, Japan
| | - Junichiro Yajima
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
- Komaba Institute for Science, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
- Research Center for Complex Systems Biology, Universal Biology Institute, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| |
Collapse
|
5
|
Pawlik K, Ostrowska M, Gumienna-Kontecka E. Systematic Model Peptide Studies: A Crucial Step To Understand the Coordination Chemistry of Mn(II) and Fe(II) in Proteins. Inorg Chem 2025; 64:5472-5486. [PMID: 40067133 PMCID: PMC11938343 DOI: 10.1021/acs.inorgchem.4c05380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/12/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025]
Abstract
Pathogenic bacteria and all other species require Mn(II) and Fe(II) ions for proper growth. Microbes use a variety of assimilation pathways to obtain the necessary metal ions, and their metal homeostasis mechanisms are still not fully uncovered. The knowledge of the poorly discovered complexation chemistry of Mn(II) and Fe(II) ions could help us to understand the basis of those processes better. We have designed six model peptides (L1 - Ac-HHHHHH-NH2, L2 - Ac-HHHHHHHHH-NH2, L3 - Ac-HAHAHAHAH-NH2, L4 - Ac-HHAAAAAAAAAHHHH-NH2, L5 - Ac-HDHDHDHDH-NH2, and L6 - Ac-HEHEHEHEH-NH2) inspired by Mn(II) and Fe(II) binding motifs that are prevalent in nature, in order to clarify their coordination preferences. Spectrometric, spectroscopic, and potentiometric techniques were used to determine the thermodynamic and structural properties of the studied systems. All of the investigated ligands possess efficient Mn(II), Fe(II), and Zn(II) binding sites. Complex stability and metal affinity are significantly influenced by the length of the peptide sequences, as well as the location and quantity of coordinating amino acid residues like His, Asp, and Glu.
Collapse
Affiliation(s)
- Karolina Pawlik
- Faculty of Chemistry, University of Wrocław, Wrocław 50-383, Poland
| | | | | |
Collapse
|
6
|
Bilgin N, Hintzen JCJ, Mecinović J. Chemical tools for probing histidine modifications. Chem Commun (Camb) 2025; 61:3805-3820. [PMID: 39936705 DOI: 10.1039/d4cc06586g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Histidine is a unique amino acid with critical roles in protein structure and function, ranging from metal ion binding to enzyme catalysis. Histidine residues in proteins also undergo diverse posttranslational modifications, including methylation, phosphorylation and hydroxylation, by various enzymes, some of them being only recently identified and characterised. In this review, we describe the development of chemical tools for understanding the role of histidine residues in chemical and biological systems. We spotlight the application of histidine analogues in probing biomedically important posttranslational modifications of histidine residues in proteins, and we highlight novel bioconjugation methods that enable chemoselective modifications of histidine residues in peptides and proteins.
Collapse
Affiliation(s)
- Nurgül Bilgin
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark.
| | - Jordi C J Hintzen
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jasmin Mecinović
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark.
| |
Collapse
|
7
|
Shams A, Bousis S, Diamanti E, Elgaher WAM, Zeimetz L, Haupenthal J, Slotboom DJ, Hirsch AKH. Expression and characterization of pantothenate energy-coupling factor transporters as an anti-infective drug target. Protein Sci 2024; 33:e5195. [PMID: 39473025 PMCID: PMC11521937 DOI: 10.1002/pro.5195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/02/2024] [Accepted: 10/09/2024] [Indexed: 11/02/2024]
Abstract
This study investigates the potential of energy-coupling factor (ECF) transporters as promising anti-infective targets to combat antimicrobial resistance (AMR). ECF transporters, a subclass of ATP-binding cassette (ABC) transporters, facilitate the uptake of B-vitamins across bacterial membranes by utilizing ATP as an energy source. Vitamins are essential cofactors for bacterial metabolism and growth, and they can either be synthesized de novo or absorbed from the environment. These transporters are considered promising drug targets, underscoring the need for further research to harness their medicinal potential and develop selective inhibitors that block vitamin uptake in bacteria. Herein, we focused on the ECF transporter for pantothenate (vitamin B5) from Streptococcus pneumoniae and the ECF transporter for folate (vitamin B9) from Lactobacillus delbrueckii as a reference protein. We also included the energizing module for pantothenate along with both full transporter complexes. Initially, we transformed and purified the transporters, followed by an assessment of their thermal stability under various buffer composition, pH, and salt concentrations. Additionally, we monitored the melting temperature over six days to confirm their stability for further assays. We then measured the binding affinities of six ECF inhibitors using surface plasmon resonance (SPR) and evaluated their inhibitory effects through in vitro assays, including bacterial growth assay, whole-cell uptake, and transport-activity assays. After determining cytotoxicity in two human cell lines, we established an in vivo infection model using Galleria mellonella larvae to further validate our findings.
Collapse
Affiliation(s)
- Atanaz Shams
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Department of Drug Design and OptimizationSaarbrückenGermany
- Saarland UniversityDepartment of PharmacySaarbrückenGermany
| | - Spyridon Bousis
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Department of Drug Design and OptimizationSaarbrückenGermany
- Saarland UniversityDepartment of PharmacySaarbrückenGermany
- Stratingh Institute for Chemistry and Technology, Faculty of Science and EngineeringUniversity of GroningenGroningenThe Netherlands
| | - Eleonora Diamanti
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Department of Drug Design and OptimizationSaarbrückenGermany
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum—Università di BolognaBolognaItaly
| | - Walid A. M. Elgaher
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Department of Drug Design and OptimizationSaarbrückenGermany
| | - Lucie Zeimetz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Department of Drug Design and OptimizationSaarbrückenGermany
- Saarland UniversityDepartment of PharmacySaarbrückenGermany
| | - Jörg Haupenthal
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Department of Drug Design and OptimizationSaarbrückenGermany
| | - Dirk J. Slotboom
- Groningen Biomolecular Sciences and Biotechnology InstituteUniversity of GroningenGroningenThe Netherlands
| | - Anna K. H. Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Department of Drug Design and OptimizationSaarbrückenGermany
- Saarland UniversityDepartment of PharmacySaarbrückenGermany
| |
Collapse
|
8
|
Steinkühler J, Lipowsky R, Miettinen MS. Molecular Dynamics Simulations Show That Short Peptides Can Drive Synthetic Cell Division by Binding to the Inner Membrane Leaflet. J Phys Chem B 2024; 128:8782-8787. [PMID: 39223874 PMCID: PMC11403657 DOI: 10.1021/acs.jpcb.4c04358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
An important functionality of lifelike "synthetic cells" is to mimic cell division. Currently, specialized proteins that induce membrane fission in living cells are the primary candidates for dividing synthetic cells. However, interactions between lipid membranes and proteins that are not found in living cells may also be suitable. Here, we discuss the potential of short membrane-anchored peptides to induce cell division. Specifically, we used the coarse-grained MARTINI model to investigate the interaction between short membrane-anchored peptides and a lipid bilayer patch. The simulation revealed that the anchored peptide induces significant spontaneous curvature and suggests that the lipid-peptide complex can be considered as a conically shaped "bulky headgroup" lipid. By systematically increasing the electrostatic charge of the peptide, we find that membrane-anchored peptides may generate sufficiently large constriction forces even at dilute coverages. Finally, we show that when the peptide has an opposite charge to the membrane, the peptide may induce division by binding the inner membrane leaflet of a synthetic cell, that is, cell division from within.
Collapse
Affiliation(s)
- Jan Steinkühler
- Bio-Inspired Computation, Kiel University, Kaiserstraße 2, Kiel 24143, Germany
- Kiel Nano, Surface and Interface Science KiNSIS, Kiel University, Christian-Albrechts-Platz 4, Kiel 24118, Germany
| | - Reinhard Lipowsky
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, Potsdam 14476, Germany
| | - Markus S Miettinen
- Department of Chemistry, University of Bergen, Bergen 5007, Norway
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen 5008, Norway
| |
Collapse
|
9
|
Solomonov I, Locatelli I, Tortorella S, Unni M, Aharoni SL, Alchera E, Locatelli E, Maturi M, Venegoni C, Lucianò R, Salonia A, Corti A, Curnis F, Grasso V, Malamal G, Jose J, Comes Franchini M, Sagi I, Alfano M. Contrast enhanced photoacoustic detection of fibrillar collagen in the near infrared region-I. NANOSCALE ADVANCES 2024; 6:3655-3667. [PMID: 38989511 PMCID: PMC11232541 DOI: 10.1039/d4na00204k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/22/2024] [Indexed: 07/12/2024]
Abstract
Fibrillar collagen accumulation emerges as a promising biomarker in several diseases, such as desmoplastic tumors and unstable atherosclerotic plaque. Gold nanorods (GNRs) hold great potential as contrast agents in high-resolution, biomedically safe, and non-invasive photoacoustic imaging (PAI). This study presents the design and characterization of a specialized imaging tool which exploits GNR assisted targeted photoacoustic imaging that is tailored for the identification of fibrillar collagen. In addition to the photoacoustic characterization of collagen in the NIR 1 and 2 regions, we demonstrate the detailed steps of conjugating a decoy to GNRs. This study serves as a proof of concept, that demonstrates that conjugated collagenase-1 (MMP-1) generates a distinct and collagen-specific photoacoustic signal, facilitating real-time visualization in the wavelength range of 700-970 nm (NIR I). As most of the reported studies utilized the endogenous contrast of collagen in the NIR II wavelength that has major limitations to perform in vivo deep tissue imaging, the approach that we are proposing is unique and it highlights the promise of MMP-1 decoy-functionalized GNRs as novel contrast agents for photoacoustic imaging of collagen in the NIR 1 region. To our knowledge this is the first time functionalized GNRs are optimized for the detection of fibrillar collagen and utilized in the field of non-invasive photoacoustic imaging that can facilitate a better prognosis of desmoplastic tumors and broken atherosclerotic plaques.
Collapse
Affiliation(s)
- Inna Solomonov
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science Rehovot 76100 Israel
| | - Irene Locatelli
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele Milan Italy
| | - Silvia Tortorella
- Department of Industrial Chemistry "Toso Montanari", University of Bologna Via P. Gobetti 85 40129 Bologna Italy
| | - Manu Unni
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science Rehovot 76100 Israel
| | - Shay-Lee Aharoni
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science Rehovot 76100 Israel
| | - Elisa Alchera
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele Milan Italy
| | - Erica Locatelli
- Department of Industrial Chemistry "Toso Montanari", University of Bologna Via P. Gobetti 85 40129 Bologna Italy
| | - Mirko Maturi
- Department of Industrial Chemistry "Toso Montanari", University of Bologna Via P. Gobetti 85 40129 Bologna Italy
| | - Chiara Venegoni
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele Milan Italy
| | - Roberta Lucianò
- Department of Pathology, IRCCS San Raffaele Hospital and Scientific Institute Milan Italy
| | - Andrea Salonia
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele Milan Italy
- Vita-Salute San Raffaele University Milan Italy
| | - Angelo Corti
- Vita-Salute San Raffaele University Milan Italy
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute Milan Italy
| | - Flavio Curnis
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute Milan Italy
| | - Valeria Grasso
- FUJIFILM Visualsonics Inc. Amsterdam the Netherlands
- Faculty of Engineering, Institute for Materials Science, Christian-Albrecht University of Kiel Kiel Germany
| | | | - Jithin Jose
- FUJIFILM Visualsonics Inc. Amsterdam the Netherlands
| | - Mauro Comes Franchini
- Department of Industrial Chemistry "Toso Montanari", University of Bologna Via P. Gobetti 85 40129 Bologna Italy
| | - Irit Sagi
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science Rehovot 76100 Israel
| | - Massimo Alfano
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele Milan Italy
| |
Collapse
|
10
|
Weakly HMJ, Keller SL. Coupling liquid phases in 3D condensates and 2D membranes: Successes, challenges, and tools. Biophys J 2024; 123:1329-1341. [PMID: 38160256 PMCID: PMC11163299 DOI: 10.1016/j.bpj.2023.12.023] [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: 09/21/2023] [Revised: 12/05/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024] Open
Abstract
This review describes the major experimental challenges researchers meet when attempting to couple phase separation between membranes and condensates. Although it is well known that phase separation in a 2D membrane could affect molecules capable of forming a 3D condensate (and vice versa), few researchers have quantified the effects to date. The scarcity of these measurements is not due to a lack of intense interest or effort in the field. Rather, it reflects significant experimental challenges in manipulating coupled membranes and condensates to yield quantitative values. These challenges transcend many molecular details, which means they impact a wide range of systems. This review highlights recent exciting successes in the field, and it lays out a comprehensive list of tools that address potential pitfalls for researchers who are considering coupling membranes with condensates.
Collapse
Affiliation(s)
- Heidi M J Weakly
- Department of Chemistry, University of Washington - Seattle, Seattle, Washington
| | - Sarah L Keller
- Department of Chemistry, University of Washington - Seattle, Seattle, Washington.
| |
Collapse
|
11
|
Khare E, Gonzalez Obeso C, Martín-Moldes Z, Talib A, Kaplan DL, Holten-Andersen N, Blank KG, Buehler MJ. Heterogeneous and Cooperative Rupture of Histidine-Ni 2+ Metal-Coordination Bonds on Rationally Designed Protein Templates. ACS Biomater Sci Eng 2024; 10:2945-2955. [PMID: 38669114 DOI: 10.1021/acsbiomaterials.3c01819] [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: 04/28/2024]
Abstract
Metal-coordination bonds, a highly tunable class of dynamic noncovalent interactions, are pivotal to the function of a variety of protein-based natural materials and have emerged as binding motifs to produce strong, tough, and self-healing bioinspired materials. While natural proteins use clusters of metal-coordination bonds, synthetic materials frequently employ individual bonds, resulting in mechanically weak materials. To overcome this current limitation, we rationally designed a series of elastin-like polypeptide templates with the capability of forming an increasing number of intermolecular histidine-Ni2+ metal-coordination bonds. Using single-molecule force spectroscopy and steered molecular dynamics simulations, we show that templates with three histidine residues exhibit heterogeneous rupture pathways, including the simultaneous rupture of at least two bonds with more-than-additive rupture forces. The methodology and insights developed improve our understanding of the molecular interactions that stabilize metal-coordinated proteins and provide a general route for the design of new strong, metal-coordinated materials with a broad spectrum of dissipative time scales.
Collapse
Affiliation(s)
- Eesha Khare
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Laboratory for Atomistic and Molecular Mechanics (LAMM), Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Mechano(bio)chemistry, Max Planck Institute of Colloids and Interfaces, Am Muehlenberg 1, 14476 Potsdam, Germany
| | | | - Zaira Martín-Moldes
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Ayesha Talib
- Mechano(bio)chemistry, Max Planck Institute of Colloids and Interfaces, Am Muehlenberg 1, 14476 Potsdam, Germany
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Niels Holten-Andersen
- Department of Bioengineering and Materials Science and EngineeringLehigh University, 27 Memorial Dr W, Bethlehem, Pennsylvania 18015, United States
| | - Kerstin G Blank
- Mechano(bio)chemistry, Max Planck Institute of Colloids and Interfaces, Am Muehlenberg 1, 14476 Potsdam, Germany
- Department of Biomolecular & Selforganizing Matter, Institute of Experimental Physics, Johannes Kepler University, Altenberger Strasse 69, 4040 Linz, Austria
| | - Markus J Buehler
- Laboratory for Atomistic and Molecular Mechanics (LAMM), Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Center for Computational Science and Engineering, Schwarzman College of Computing, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
12
|
Echavarría JAC, El Hajj S, Irankunda R, Selmeczi K, Paris C, Udenigwe CC, Canabady-Rochelle L. Screening, separation and identification of metal-chelating peptides for nutritional, cosmetics and pharmaceutical applications. Food Funct 2024; 15:3300-3326. [PMID: 38488016 DOI: 10.1039/d3fo05765h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Metal-chelating peptides, which form metal-peptide coordination complexes with various metal ions, can be used as biofunctional ingredients notably to enhance human health and prevent diseases. This review aims to discuss recent insights into food-derived metal-chelating peptides, the strategies set up for their discovery, their study, and identification. After understanding the overall properties of metal-chelating peptides, their production from food-derived protein sources and their potential applications will be discussed, particularly in nutritional, cosmetics and pharmaceutical fields. In addition, the review provides an overview of the last decades of progress in discovering food-derived metal-chelating peptides, addressing several screening, separation and identification methodologies. Furthermore, it emphasizes the methods used to assess peptide-metal interaction, allowing for better understanding of chemical and thermodynamic parameters associated with the formation of peptide-metal coordination complexes, as well as the specific amino acid residues that play important roles in the metal ion coordination.
Collapse
Affiliation(s)
| | - Sarah El Hajj
- Université de Lorraine, CNRS, LRGP, F-54000 Nancy, France.
| | | | | | - Cédric Paris
- Université de Lorraine, LIBIO, F-54000 Nancy, France
| | - Chibuike C Udenigwe
- School of Nutrition Science, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada
| | | |
Collapse
|
13
|
Mohsenin H, Wagner HJ, Rosenblatt M, Kemmer S, Drepper F, Huesgen P, Timmer J, Weber W. Design of a Biohybrid Materials Circuit with Binary Decoder Functionality. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308092. [PMID: 38118057 DOI: 10.1002/adma.202308092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/05/2023] [Indexed: 12/22/2023]
Abstract
Synthetic biology applies concepts from electrical engineering and information processing to endow cells with computational functionality. Transferring the underlying molecular components into materials and wiring them according to topologies inspired by electronic circuit boards has yielded materials systems that perform selected computational operations. However, the limited functionality of available building blocks is restricting the implementation of advanced information-processing circuits into materials. Here, a set of protease-based biohybrid modules the bioactivity of which can either be induced or inhibited is engineered. Guided by a quantitative mathematical model and following a design-build-test-learn (DBTL) cycle, the modules are wired according to circuit topologies inspired by electronic signal decoders, a fundamental motif in information processing. A 2-input/4-output binary decoder for the detection of two small molecules in a material framework that can perform regulated outputs in form of distinct protease activities is designed. The here demonstrated smart material system is strongly modular and can be used for biomolecular information processing for example in advanced biosensing or drug delivery applications.
Collapse
Affiliation(s)
- Hasti Mohsenin
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
| | - Hanna J Wagner
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
| | - Marcus Rosenblatt
- Institute of Physics and Freiburg Center for Data Analysis and Modelling (FDM), University of Freiburg, Hermann-Herder-Straße 3, 79104, Freiburg, Germany
| | - Svenja Kemmer
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- Institute of Physics and Freiburg Center for Data Analysis and Modelling (FDM), University of Freiburg, Hermann-Herder-Straße 3, 79104, Freiburg, Germany
| | - Friedel Drepper
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
| | - Pitter Huesgen
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Jens Timmer
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Institute of Physics and Freiburg Center for Data Analysis and Modelling (FDM), University of Freiburg, Hermann-Herder-Straße 3, 79104, Freiburg, Germany
| | - Wilfried Weber
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
- Saarland University, Department of Materials Science and Engineering, Campus D2 2, 66123, Saarbrücken, Germany
| |
Collapse
|
14
|
Rola A, Gumienna-Kontecka E, Potocki S. Exploring binding preferences: Cu(II), Ni(II), and Zn(II) complexes of mycobacterial GroEL1 His-rich and Glu/His-rich domains. Dalton Trans 2024; 53:4054-4066. [PMID: 38305693 DOI: 10.1039/d3dt03579d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Mycobacterial histidine-rich GroEL1 protein significantly differs from the well-known methionine-glycine-rich GroEL chaperonin and most preferably participates in Cu(II) homeostasis. Some GroEL1 proteins, however, do not possess six but only three histidine residues and more acidic residues that can function as binding sites for metal ions. To evaluate the importance of this difference, we examined and compared the properties of GroEL1 His-rich or Glu/His-rich C-terminal domains as ligands for Cu(II), Ni(II), and Zn(II) ions. We studied the stoichiometry, stability, and binding sites of Cu(II)/Ni(II)/Zn(II) complexes of two model peptides: XEN = Ac-DKPEEEEDGHGHAH (M. xenopi) and ABS = Ac-DKPAEEADHGHGHHGHAH (M. abscessus) in the pH range 2-11. In the case of Cu(II), Ni(II), and Zn(II) complexes of XEN and ABS, ABS always formed more stable complexes. For XEN, there seemed to be no preference for Ni(II) or Zn(II) ions. In contrast, for ABS, Zn(II) formed a complex that was slightly more stable than the one formed by Ni(II). This may be due to the 6 His residues, which preferentially interact with Zn(II) rather than Ni(II). The study identified that an equilibrium of complexes-known as polymorphism-may occur in ABS complexes. Therefore, distinct sets of histidine residues may be involved in metal binding.
Collapse
Affiliation(s)
- Anna Rola
- Faculty of Chemistry, University of Wroclaw, 50-383 Wroclaw, Poland.
| | | | - Sławomir Potocki
- Faculty of Chemistry, University of Wroclaw, 50-383 Wroclaw, Poland.
| |
Collapse
|
15
|
Nyíri K, Gál E, Laczkovich M, Vértessy BG. Antirepressor specificity is shaped by highly efficient dimerization of the staphylococcal pathogenicity island regulating repressors: Stl repressor dimerization perturbed by dUTPases. Sci Rep 2024; 14:1953. [PMID: 38263343 PMCID: PMC10806181 DOI: 10.1038/s41598-024-51260-y] [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: 05/22/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
The excision and replication, thus the life cycle of pathogenicity islands in staphylococci are regulated by Stl master repressors that form strong dimers. It has been recently shown that SaPIbov1-Stl dimers are separated during the activation of the Staphylococcus aureus pathogenicity island (SaPI) transcription via helper phage proteins. To understand the mechanism of this regulation, a quantitative analysis of the dimerization characteristics is required. Due to the highly efficient dimerization process, such an analysis has to involve specific solutions that permit relevant experiments to be performed. In the present work, we focused on two staphylococcal Stls associated with high biomedical interest, namely Stl proteins of Staphylococcus aureus bov1 and Staphylococcus hominis ShoCI794_SEPI pathogenicity islands. Exploiting the interactions of these two Stl proteins with their antirepressor-mimicking interaction partners allowed precise determination of the Stl dimerization constant in the subnanomolar range.
Collapse
Affiliation(s)
- Kinga Nyíri
- Department of Applied Biotechnology and Food Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., Budapest, 1111, Hungary.
- Institute of Molecular Life Sciences, HUN-REN, Research Centre for Natural Sciences, Magyar Tudósok Krt 2., Budapest, 1117, Hungary.
| | - Enikő Gál
- Department of Applied Biotechnology and Food Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., Budapest, 1111, Hungary
- Institute of Molecular Life Sciences, HUN-REN, Research Centre for Natural Sciences, Magyar Tudósok Krt 2., Budapest, 1117, Hungary
| | - Máté Laczkovich
- Department of Applied Biotechnology and Food Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., Budapest, 1111, Hungary
- Institute of Molecular Life Sciences, HUN-REN, Research Centre for Natural Sciences, Magyar Tudósok Krt 2., Budapest, 1117, Hungary
| | - Beáta G Vértessy
- Department of Applied Biotechnology and Food Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., Budapest, 1111, Hungary
- Institute of Molecular Life Sciences, HUN-REN, Research Centre for Natural Sciences, Magyar Tudósok Krt 2., Budapest, 1117, Hungary
| |
Collapse
|
16
|
Tarabarova A, Lopukhov A, Fedorov AN, Yurkova MS. Novel His-tag Variants for Insertion Inside Polypeptide Chain. ACS OMEGA 2024; 9:858-865. [PMID: 38222536 PMCID: PMC10785306 DOI: 10.1021/acsomega.3c06682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 01/16/2024]
Abstract
His-tags are protein affinity tags ubiquitously used due to their convenience and effectiveness. However, in some individual cases, the attachment of His-tags to a protein's N- or C-termini resulted in impairment of the protein's structure or function, which led to attempts to include His-tags inside of polypeptide chains. In this work, we describe newly designed internal His-tags, where two triplets of histidine residues are separated by glycine residues to avoid steric hindrances and consequently minimize their impact on the protein structure. The applicability of these His-tags was tested with eGFP, a multifaceted reference protein, and GrAD207, a modified apical domain of GroEL chaperone, designed to stabilize in soluble form initially insoluble proteins. Both proteins are used as fusion partners for different purposes, and providing them with His-tags introduced into their polypeptide chains should conveniently broaden their functionality without involving the termini. We conclude that the insertable tags may be adjusted for the purification of proteins belonging to different structural classes.
Collapse
Affiliation(s)
- Anastasiia
G. Tarabarova
- A
N Bach Institute of Biochemistry of the Russian Academy of Sciences, Leninskii prosp 33/2, Moscow 119071, Russian Federation
| | - Anton Lopukhov
- Chemistry
Department, Lomonosov Moscow State University, GSP-1, Leninskie Gory 1/3, Moscow 119991, Russian Federation
| | - Alexey N. Fedorov
- FSI
Federal Research Centre Fundamentals of Biotechnology of the Russian
Academy of Sciences, Leninskii prosp 33/2, Moscow 119071, Russian Federation
| | - Maria S. Yurkova
- A
N Bach Institute of Biochemistry of the Russian Academy of Sciences, Leninskii prosp 33/2, Moscow 119071, Russian Federation
| |
Collapse
|
17
|
Lal M, Wachtel E, Pati S, Namboothiri INN, Patchornik G. His 1-tagged DM or DDM detergent micelles are reversibly conjugated by nickel ions. Sci Rep 2023; 13:17138. [PMID: 37816812 PMCID: PMC10564902 DOI: 10.1038/s41598-023-44236-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/05/2023] [Indexed: 10/12/2023] Open
Abstract
Specific conjugation of decyl β-D-maltoside (DM) or dodecyl β-D-maltoside (DDM) detergent micelles is accomplished between pH 7.0-8.5 in the presence of an amphiphilic analog of the amino acid histidine, bound to a 10-carbon hydrocarbon chain (His1-C10) and Ni2+ ions. Following addition of 10-15 wt% PEG-6000 as precipitant, phase separation in the form of oil-rich globules (30-600 µm) is observed by light microscopy. Other divalent cations: Zn2+, Fe2+, Cu2+ lead to dark precipitates rather than colorless globules; while Mg2+, Ca2+ do not promote any phase separation at all. Even in the absence of precipitant, dynamic light scattering (DLS) measurements demonstrate that DM micelles (hydrodynamic size ~ 6 nm) or DDM micelles (8 nm) self-associate into larger particles (9 nm and 411 nm for DM; 10 nm and 982 nm for DDM) in the presence of His1-C10 and nickel ions. Micellar conjugation is partially reversible in the presence of water soluble 50 mM EDTA, histidine or imidazole chelators. Cryo-transmission electron microscopy (cryo-TEM) imaging revealed the formation of non-uniformly dense detergent aggregates for both DM and DDM micelles in the presence of precipitant. The possible utility of such His1-tagged DM or DDM micelles for promoting crystallization of integral membrane proteins is discussed.
Collapse
Affiliation(s)
- Mitra Lal
- Department of Chemical Sciences, Ariel University, 70400, Ariel, Israel
| | - Ellen Wachtel
- Faculty of Chemistry, Weizmann Institute, 761001, Rehovot, Israel
| | - Soumyaranjan Pati
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India
| | - Irishi N N Namboothiri
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India
| | - Guy Patchornik
- Department of Chemical Sciences, Ariel University, 70400, Ariel, Israel.
| |
Collapse
|
18
|
Hamacek HSDR, Bresolin ITL, Fioravante IF, Bueno SMA. Synthesis and characterization of chitosan-polyacrylamide cryogels for the purification of human IgG by IMAC. Process Biochem 2023; 131:199-209. [DOI: 10.1016/j.procbio.2023.06.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
19
|
Zhu L, Chang Y, Li Y, Qiao M, Liu L. Biosensors Based on the Binding Events of Nitrilotriacetic Acid-Metal Complexes. BIOSENSORS 2023; 13:bios13050507. [PMID: 37232868 DOI: 10.3390/bios13050507] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023]
Abstract
Molecular immobilization and recognition are two key events for the development of biosensors. The general ways for the immobilization and recognition of biomolecules include covalent coupling reactions and non-covalent interactions of antigen-antibody, aptamer-target, glycan-lectin, avidin-biotin and boronic acid-diol. Tetradentate nitrilotriacetic acid (NTA) is one of the most common commercial ligands for chelating metal ions. The NTA-metal complexes show high and specific affinity toward hexahistidine tags. Such metal complexes have been widely utilized in protein separation and immobilization for diagnostic applications since most of commercialized proteins have been integrated with hexahistidine tags by synthetic or recombinant techniques. This review focused on the development of biosensors with NTA-metal complexes as the binding units, mainly including surface plasmon resonance, electrochemistry, fluorescence, colorimetry, surface-enhanced Raman scattering spectroscopy, chemiluminescence and so on.
Collapse
Affiliation(s)
- Lin Zhu
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang 455000, China
| | - Yong Chang
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang 455000, China
| | - Yingying Li
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang 455000, China
| | - Mingyi Qiao
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang 455000, China
| | - Lin Liu
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang 455000, China
| |
Collapse
|
20
|
Khare E, Grewal DS, Buehler MJ. Bond clusters control rupture force limit in shear loaded histidine-Ni 2+ metal-coordinated proteins. NANOSCALE 2023; 15:8578-8588. [PMID: 37092811 DOI: 10.1039/d3nr01287e] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Dynamic noncovalent interactions are pivotal to the structure and function of biological proteins and have been used in bioinspired materials for similar roles. Metal-coordination bonds, in particular, are especially tunable and enable control over static and dynamic properties when incorporated into synthetic materials. Despite growing efforts to engineer metal-coordination bonds to produce strong, tough, and self-healing materials, the systematic characterization of the exact contribution of these bonds towards mechanical strength and the effect of geometric arrangements is missing, limiting the full design potential of these bonds. In this work, we engineer the cooperative rupture of metal-coordination bonds to increase the rupture strength of metal-coordinated peptide dimers. Utilizing all-atom steered molecular dynamics simulations on idealized bidentate histidine-Ni2+ coordinated peptides, we show that histidine-Ni2+ bonds can rupture cooperatively in groups of two to three bonds. We find that there is a strength limit, where adding additional coordination bonds does not contribute to the additional increase in the protein rupture strength, likely due to the highly heterogeneous rupture behavior exhibited by the coordination bonds. Further, we show that this coordination bond limit is also found natural metal-coordinated biological proteins. Using these insights, we quantitatively suggest how other proteins can be rationally designed with dynamic noncovalent interactions to exhibit cooperative bond breaking behavior. Altogether, this work provides a quantitative analysis of the cooperativity and intrinsic strength limit for metal-coordination bonds with the aim of advancing clear guiding molecular principles for the mechanical design of metal-coordinated materials.
Collapse
Affiliation(s)
- Eesha Khare
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Laboratory for Atomistic and Molecular Mechanics, Massachusetts Institute of Technology, 33 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | - Darshdeep S Grewal
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Laboratory for Atomistic and Molecular Mechanics, Massachusetts Institute of Technology, 33 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | - Markus J Buehler
- Laboratory for Atomistic and Molecular Mechanics, Massachusetts Institute of Technology, 33 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
21
|
Vu TQ, Sant'Anna LE, Kamat NP. Tuning Targeted Liposome Avidity to Cells via Lipid Phase Separation. Biomacromolecules 2023; 24:1574-1584. [PMID: 36943688 PMCID: PMC10874583 DOI: 10.1021/acs.biomac.2c01338] [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] [Indexed: 03/23/2023]
Abstract
The addition of both cell-targeting moieties and polyethylene glycol (PEG) to nanoparticle (NP) drug delivery systems is a standard approach to improve the biodistribution, specificity, and uptake of therapeutic cargo. The spatial presentation of these molecules affects avidity of the NP to target cells in part through an interplay between the local ligand concentration and the steric hindrance imposed by PEG molecules. Here, we show that lipid phase separation in nanoparticles can modulate liposome avidity by changing the proximity of PEG and targeting protein molecules on a nanoparticle surface. Using lipid-anchored nickel-nitrilotriacetic acid (Ni-NTA) as a model ligand, we demonstrate that the attachment of lipid anchored Ni-NTA and PEG molecules to distinct lipid domains in nanoparticles can enhance liposome binding to cancer cells by increasing ligand clustering and reducing steric hindrance. We then use this technique to enhance the binding of RGD-modified liposomes, which can bind to integrins overexpressed on many cancer cells. These results demonstrate the potential of lipid phase separation to modulate the spatial presentation of targeting and shielding molecules on lipid nanocarriers, offering a powerful tool to enhance the efficacy of NP drug delivery systems.
Collapse
Affiliation(s)
- Timothy Q Vu
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center for Synthetic Biology, McCormick School of Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Lucas E Sant'Anna
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center for Synthetic Biology, McCormick School of Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Neha P Kamat
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center for Synthetic Biology, McCormick School of Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
22
|
Cheng H, Zheng L, Liu N, Huang C, Xu J, Lu Y, Cui X, Xu K, Hou Y, Tang J, Zhang Z, Li J, Ni X, Chen Y, Peng H, Wang HW. Dual-Affinity Graphene Sheets for High-Resolution Cryo-Electron Microscopy. J Am Chem Soc 2023; 145:8073-8081. [PMID: 37011903 PMCID: PMC10103130 DOI: 10.1021/jacs.3c00659] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
With the development of cryo-electron microscopy (cryo-EM), high-resolution structures of macromolecules can be reconstructed by the single particle method efficiently. However, challenges may still persist during the specimen preparation stage. Specifically, proteins tend to adsorb at the air-water interface and exhibit a preferred orientation in vitreous ice. To overcome these challenges, we have explored dual-affinity graphene (DAG) modified with two different affinity ligands as a supporting material for cryo-EM sample preparation. The ligands can bind to distinct sites on the corresponding tagged particles, which in turn generates various orientation distributions of particles and prevents the adsorption of protein particles onto the air-water interface. As expected, the DAG exhibited high binding specificity and affinity to target macromolecules, resulting in more balanced particle Euler angular distributions compared to single functionalized graphene on two different protein cases, including the SARS -CoV-2 spike glycoprotein. We anticipate that the DAG grids will enable facile and efficient three-dimensional (3D) reconstruction for cryo-EM structural determination, providing a robust and general technique for future studies.
Collapse
Affiliation(s)
- Hang Cheng
- Ministry of Education Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Shuimu BioSciences Ltd., Beijing 102206, China
| | - Liming Zheng
- Center for Nanochemistry, Beijing Science and Engineering Center for Nanocarbons, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Nan Liu
- Ministry of Education Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Frontier Research Center for Biological Structures, Tsinghua University, Beijing 100084, China
| | - Congyuan Huang
- Ministry of Education Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jie Xu
- Ministry of Education Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ye Lu
- Ministry of Education Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaoya Cui
- Ministry of Education Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Frontier Research Center for Biological Structures, Tsinghua University, Beijing 100084, China
| | - Kui Xu
- Ministry of Education Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yuan Hou
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Junchuan Tang
- Center for Nanochemistry, Beijing Science and Engineering Center for Nanocarbons, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Zhong Zhang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Jing Li
- Shuimu BioSciences Ltd., Beijing 102206, China
| | - Xiaodan Ni
- Shuimu BioSciences Ltd., Beijing 102206, China
| | - Yanan Chen
- School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Hailin Peng
- Center for Nanochemistry, Beijing Science and Engineering Center for Nanocarbons, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Beijing Graphene Institute (BGI), Beijing 100095, China
| | - Hong-Wei Wang
- Ministry of Education Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Frontier Research Center for Biological Structures, Tsinghua University, Beijing 100084, China
| |
Collapse
|
23
|
El Hajj S, Irankunda R, Camaño Echavarría JA, Arnoux P, Paris C, Stefan L, Gaucher C, Boschi-Muller S, Canabady-Rochelle L. Metal-chelating activity of soy and pea protein hydrolysates obtained after different enzymatic treatments from protein isolates. Food Chem 2023; 405:134788. [PMID: 36370575 DOI: 10.1016/j.foodchem.2022.134788] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 09/28/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022]
Abstract
Soy and pea proteins are two rich sources of essential amino acids. The hydrolysis of these proteins reveals functional and bioactive properties of the produced small peptide mixtures. In our study, we employed the hydrolysis of soy and pea protein isolates with the endopeptidases Alcalase® and Protamex®, used alone or followed by the exopeptidase Flavourzyme®. The sequential enzyme treatments were the most efficient regarding the degree of hydrolysis. Then, soy and pea protein hydrolysates (SPHs and PPHs, respectively) were ultrafiltrated in order to select peptides of molecular weight ≤ 1 kDa. Whatever the protein source or the hydrolysis treatment, the hydrolysates showed similar molecular weight distributions and amino acid compositions. In addition, all the ultrafiltrated hydrolysates possess metal-chelating activities, as determined by UV-spectrophotometry and Surface Plasmon Resonance (SPR). However, the SPR data revealed better chelating affinities in SPHs and PPHs when produced by sequential enzymatic treatment.
Collapse
Affiliation(s)
- Sarah El Hajj
- Université de Lorraine, CNRS, LRGP, F-54000 Nancy, France; Université de Lorraine, CITHEFOR, F-54505 Vandoeuvre Les Nancy, France.
| | | | | | | | - Cédric Paris
- Université de Lorraine, LIBio, F-54505 Vandoeuvre Les Nancy, France
| | - Loic Stefan
- Université de Lorraine, CNRS, LCPM, F-54000 Nancy, France
| | - Caroline Gaucher
- Université de Lorraine, CITHEFOR, F-54505 Vandoeuvre Les Nancy, France
| | | | | |
Collapse
|
24
|
Berntsson E, Vosough F, Svantesson T, Pansieri J, Iashchishyn IA, Ostojić L, Dong X, Paul S, Jarvet J, Roos PM, Barth A, Morozova-Roche LA, Gräslund A, Wärmländer SKTS. Residue-specific binding of Ni(II) ions influences the structure and aggregation of amyloid beta (Aβ) peptides. Sci Rep 2023; 13:3341. [PMID: 36849796 PMCID: PMC9971182 DOI: 10.1038/s41598-023-29901-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. AD brains display deposits of insoluble amyloid plaques consisting mainly of aggregated amyloid-β (Aβ) peptides, and Aβ oligomers are likely a toxic species in AD pathology. AD patients display altered metal homeostasis, and AD plaques show elevated concentrations of metals such as Cu, Fe, and Zn. Yet, the metal chemistry in AD pathology remains unclear. Ni(II) ions are known to interact with Aβ peptides, but the nature and effects of such interactions are unknown. Here, we use numerous biophysical methods-mainly spectroscopy and imaging techniques-to characterize Aβ/Ni(II) interactions in vitro, for different Aβ variants: Aβ(1-40), Aβ(1-40)(H6A, H13A, H14A), Aβ(4-40), and Aβ(1-42). We show for the first time that Ni(II) ions display specific binding to the N-terminal segment of full-length Aβ monomers. Equimolar amounts of Ni(II) ions retard Aβ aggregation and direct it towards non-structured aggregates. The His6, His13, and His14 residues are implicated as binding ligands, and the Ni(II)·Aβ binding affinity is in the low µM range. The redox-active Ni(II) ions induce formation of dityrosine cross-links via redox chemistry, thereby creating covalent Aβ dimers. In aqueous buffer Ni(II) ions promote formation of beta sheet structure in Aβ monomers, while in a membrane-mimicking environment (SDS micelles) coil-coil helix interactions appear to be induced. For SDS-stabilized Aβ oligomers, Ni(II) ions direct the oligomers towards larger sizes and more diverse (heterogeneous) populations. All of these structural rearrangements may be relevant for the Aβ aggregation processes that are involved in AD brain pathology.
Collapse
Affiliation(s)
- Elina Berntsson
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden.
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia.
| | - Faraz Vosough
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Teodor Svantesson
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Jonathan Pansieri
- Department of Medical Biochemistry and Biophysics, Umeå University, 901 87, Umeå, Sweden
| | - Igor A Iashchishyn
- Department of Medical Biochemistry and Biophysics, Umeå University, 901 87, Umeå, Sweden
| | - Lucija Ostojić
- Department of Medical Biochemistry and Biophysics, Umeå University, 901 87, Umeå, Sweden
| | - Xiaolin Dong
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Suman Paul
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Jüri Jarvet
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
- The National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Per M Roos
- Institute of Environmental Medicine, Karolinska Institutet, Nobels Väg 13, 171 77, Stockholm, Sweden
- Department of Clinical Physiology, Capio St. Göran Hospital, St. Göransplan 1, 112 19, Stockholm, Sweden
| | - Andreas Barth
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | | | - Astrid Gräslund
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | | |
Collapse
|
25
|
Chung YH, Volckaert BA, Steinmetz NF. Development of a Modular NTA:His Tag Viral Vaccine for Co-delivery of Antigen and Adjuvant. Bioconjug Chem 2023; 34:269-278. [PMID: 36608270 PMCID: PMC10545220 DOI: 10.1021/acs.bioconjchem.2c00601] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The SARS-CoV-2 pandemic has highlighted the need for vaccines that are effective, but quickly produced. Of note, vaccines with plug-and-play capabilities that co-deliver antigen and adjuvant to the same cell have shown remarkable success. Our approach of utilizing a nitrilotriacetic acid (NTA) histidine (His)-tag chemistry with viral adjuvants incorporates both of these characteristics: plug-and-play and co-delivery. We specifically utilize the cowpea mosaic virus (CPMV) and the virus-like particles from bacteriophage Qβ as adjuvants and bind the model antigen ovalbumin (OVA). Successful binding of the antigen to the adjuvant/carrier was verified by SDS-PAGE, western blot, and ELISA. Immunization in C57BL/6J mice demonstrates that with Qβ - but not CPMV - there is an improved antibody response against the target antigen using the Qβ-NiNTA:His-OVA versus a simple admixture of antigen and adjuvant. Antibody isotyping also shows that formulation of the vaccines can alter T helper biases; while the Qβ-NiNTA:His-OVA particle produces a balanced Th1/Th2 bias the admixture was strongly Th2. In a mouse model of B16F10-OVA, we further demonstrate improved survival and slower tumor growth in the vaccine groups compared to controls. The NiNTA:His chemistry demonstrates potential for rapid development of future generation vaccines enabling plug-and-play capabilities with effectiveness boosted by co-delivery to the same cell.
Collapse
Affiliation(s)
- Young Hun Chung
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, United States
| | - Britney A Volckaert
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Nicole F Steinmetz
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, United States
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, United States
- Department of Radiology, University of California, San Diego, La Jolla, California 92093, United States
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, California 92093, United States
- Center for Engineering in Cancer, Institute for Engineering in Medicine, University of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
26
|
Metal-Chelating Peptides Separation Using Immobilized Metal Ion Affinity Chromatography: Experimental Methodology and Simulation. SEPARATIONS 2022. [DOI: 10.3390/separations9110370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Metal-Chelating Peptides (MCPs), obtained from protein hydrolysates, present various applications in the field of nutrition, pharmacy, cosmetic etc. The separation of MCPs from hydrolysates mixture is challenging, yet, techniques based on peptide-metal ion interactions such as Immobilized Metal Ion Affinity Chromatography (IMAC) seem to be efficient. However, separation processes are time consuming and expensive, therefore separation prediction using chromatography modelling and simulation should be necessary. Meanwhile, the obtention of sorption isotherm for chromatography modelling is a crucial step. Thus, Surface Plasmon Resonance (SPR), a biosensor method efficient to screen MCPs in hydrolysates and with similarities to IMAC might be a good option to acquire sorption isotherm. This review highlights IMAC experimental methodology to separate MCPs and how, IMAC chromatography can be modelled using transport dispersive model and input data obtained from SPR for peptides separation simulation.
Collapse
|
27
|
Synthesis of 2,5-Dialkyl-1,3,4-oxadiazoles Bearing Carboxymethylamino Groups. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227687. [PMID: 36431787 PMCID: PMC9696334 DOI: 10.3390/molecules27227687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022]
Abstract
A series of new symmetrical 2,5-dialkyl-1,3,4-oxadiazoles containing substituted alkyl groups at the terminal positions with substituents, such as bromine, isopropyloxycarbonylmethylamino, and carboxymethylamino, were successfully synthesized. The developed multistep method employed commercially available acid chlorides differing in alkyl chain length and terminal substituent, hydrazine hydrate, and phosphorus oxychloride. The intermediate bromine-containing 2,5-dialkyl-1,3,4-oxadiazoles were easily substituted with diisopropyl iminodiacetate, followed by hydrolysis in aqueous methanol solution giving the corresponding 1,3,4-oxadiazoles bearing carboxymethylamino substituents. The structure of all products was confirmed by conventional spectroscopic methods including 1H NMR, 13C NMR, and HRMS.
Collapse
|
28
|
Cong ATQ, Witter TL, Schellenberg MJ. High-efficiency recombinant protein purification using mCherry and YFP nanobody affinity matrices. Protein Sci 2022; 31:e4383. [PMID: 36040252 PMCID: PMC9413470 DOI: 10.1002/pro.4383] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/28/2022] [Accepted: 04/05/2022] [Indexed: 11/11/2022]
Abstract
Mammalian cell lines are important expression systems for large proteins and protein complexes, particularly when the acquisition of post-translational modifications in the protein's native environment is desired. However, low or variable transfection efficiencies are challenges that must be overcome to use such an expression system. Expression of recombinant proteins as a fluorescent protein fusion enables real-time monitoring of protein expression, and also provides an affinity handle for one-step protein purification using a suitable affinity reagent. Here, we describe a panel of anti-GFP and anti-mCherry nanobody affinity matrices and their efficacy for purification of GFP/YFP or mCherry fusion proteins. We define the molecular basis by which they bind their target proteins using X-ray crystallography. From these analyses, we define an optimal pair of nanobodies for purification of recombinant protein tagged with GFP/YFP or mCherry, and demonstrate these nanobody-sepharose supports are stable to many rounds of cleaning and extended incubation in denaturing conditions. Finally, we demonstrate the utility of the mCherry-tag system by using it to purify recombinant human topoisomerase 2α expressed in HEK293F cells. The mCherry-tag and GFP/YFP-tag expression systems can be utilized for recombinant protein expression individually or in tandem for mammalian protein expression systems where real-time monitoring of protein expression levels and a high-efficiency purification step is needed.
Collapse
Affiliation(s)
- Anh T. Q. Cong
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesotaUSA
| | - Taylor L. Witter
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesotaUSA
| | | |
Collapse
|
29
|
Pramanik S, Steinkühler J, Dimova R, Spatz J, Lipowsky R. Binding of His-tagged fluorophores to lipid bilayers of giant vesicles. SOFT MATTER 2022; 18:6372-6383. [PMID: 35975692 DOI: 10.1039/d2sm00915c] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
His-tagged molecules can be attached to lipid bilayers via certain anchor lipids, a method that has been widely used for the biofunctionalization of membranes and vesicles. To observe the membrane-bound molecules, it is useful to consider His-tagged molecules that are fluorescent as well. Here, we study two such molecules, green fluorescence protein (GFP) and green-fluorescent fluorescein isothiocyanate (FITC), both of which are tagged with a chain of six histidines (6H) that bind to the anchor lipids within the bilayers. The His-tag 6H is much smaller than the GFP molecule but somewhat larger than the FITC dye. The lipid bilayers form giant unilamellar vesicles (GUVs), the behavior of which can be directly observed in the optical microscope. We apply and compare three well-established preparation methods for GUVs: electroformation on platinum wire, polyvinyl alcohol (PVA) hydrogel swelling, and electroformation on indium tin oxide (ITO) glass. Microfluidics is used to expose the GUVs to a constant fluorophore concentration in the exterior solution. The brightness of membrane-bound 6H-GFP exceeds the brightness of membrane-bound 6H-FITC, in contrast to the quantum yields of the two fluorophores in solution. In fact, 6H-FITC is observed to be strongly quenched by the anchor lipids which bind the fluorophores via Ni2+ ions. For both 6H-GFP and 6H-FITC, the membrane fluorescence is measured as a function of the fluorophores' molar concentration. The theoretical analysis of these data leads to the equilibrium dissociation constants Kd = 37.5 nM for 6H-GFP and Kd = 18.5 nM for 6H-FITC. We also observe a strong pH-dependence of the membrane fluorescence.
Collapse
Affiliation(s)
- Shreya Pramanik
- Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany.
| | - Jan Steinkühler
- Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany.
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Rumiana Dimova
- Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany.
| | - Joachim Spatz
- Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Reinhard Lipowsky
- Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany.
| |
Collapse
|
30
|
Wa Tły J, Hecel A, Wieczorek R, Rowińska-Żyrek M, Kozłowski H. Poly-Gly Region Regulates the Accessibility of Metal Binding Sites in Snake Venom Peptides. Inorg Chem 2022; 61:14247-14251. [PMID: 36039984 PMCID: PMC9472272 DOI: 10.1021/acs.inorgchem.2c02584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
![]()
It is supposed that the presence of poly-His regions
in close proximity
to poly-Gly domains in snake venoms is related to their biological
activity; poly-His/poly-Gly (pHpG) peptides inhibit the activity of
metalloproteinases during venom storage via the chelation metal ions,
necessary for their proper functioning. This work shows that only
the histidyl residues from the N-terminal VDHDHDH motif (but not from
the poly-His tag) were the primary Zn(II) binding sites and that the
poly-Gly domain situated in the proximity of a central proline residue
may play a regulatory role in venom gland protection. The proline
induces a kink of the peptide, resulting in steric hindrance, which
may modulate the accessibility of potential metal binding sites in
the poly-His domain and may, in turn, be one of the regulators of
Zn(II) accessibility in the venom gland and therefore a modulator
of metalloproteinase activity during venom storage. The proline induces a kink of the peptide, resulting in
a steric hindrance, which may modulate the accessibility of potential
metal binding sites in the poly-His domain and may, in turn, be one
of the regulators of Zn(II) accessibility in the venom gland and therefore
a modulator of metalloproteinase activity during venom storage.
Collapse
Affiliation(s)
- Joanna Wa Tły
- Faculty of Chemistry, University of Wrocław, F. Joliot-Curie 14, Wrocław 50-383, Poland
| | - Aleksandra Hecel
- Faculty of Chemistry, University of Wrocław, F. Joliot-Curie 14, Wrocław 50-383, Poland
| | - Robert Wieczorek
- Faculty of Chemistry, University of Wrocław, F. Joliot-Curie 14, Wrocław 50-383, Poland
| | | | - Henryk Kozłowski
- Faculty of Chemistry, University of Wrocław, F. Joliot-Curie 14, Wrocław 50-383, Poland.,Institute of Health Sciences, University of Opole, 68 Katowicka Street, Opole 45-060, Poland
| |
Collapse
|
31
|
Mondal AK, Xu D, Wu S, Zou Q, Lin W, Huang F, Ni Y. Lignin-containing hydrogels with anti-freezing, excellent water retention and super-flexibility for sensor and supercapacitor applications. Int J Biol Macromol 2022; 214:77-90. [PMID: 35691432 DOI: 10.1016/j.ijbiomac.2022.06.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 11/05/2022]
Abstract
We developed a highly conductive and flexible, anti-freezing sulfonated lignin (SL)-containing polyacrylic acid (PAA) (SL-g-PAA-Ni) hydrogel, with a high concentration of NiCl2. Ni2+ contributes multi-functions to the preparation of the hydrogel and its final properties, such as fast polymerization reaction as a result of the presence of redox pairs of Ni3+/Ni2+ and hydroquinone/quinone, and anti-freezing properties of the hydrogel due to the salt effects of NiCl2 so that at -20 °C the hydrogel shows similar properties to those at the room temperature. Thanks to the effective coordinations of Ni2+ with catecholic groups and carboxylic groups, as well as the rich hydrogen bonding capacity, the resultant hydrogel possesses excellent mechanical properties. High ionic conductivity (6.85 S·m-1) of the hydrogel is obtained due to the supply of high concentration of Ni2+. Moreover, the ionic solvation effect of NiCl2 in the hydrogel imparts excellent water retention ability, with water retention of ~93 % after 21-day storage. The SL-g-PAA-Ni hydrogel can accurately detect various human motions at -20 °C. The supercapacitor assembled from SL-g-PAA-Al hydrogel at -20 °C manifests a high specific capacitance of 252 F·g-1, with maximum energy density of 26.97 Wh·kg-1, power density of 2667 W·kg-1, and capacitance retention of 96.7 % after 3000 consecutive charge-discharge cycles.
Collapse
Affiliation(s)
- Ajoy Kanti Mondal
- College of Material Engineering, Fujian Agriculture and Forestry University, Fuzhou 350108, China; Institute of Fuel Research and Development, Bangladesh Council of Scientific and Industrial Research, Dhaka 1205, Bangladesh
| | - Dezhong Xu
- College of Material Engineering, Fujian Agriculture and Forestry University, Fuzhou 350108, China
| | - Shuai Wu
- College of Material Engineering, Fujian Agriculture and Forestry University, Fuzhou 350108, China
| | - Qiuxia Zou
- College of Material Engineering, Fujian Agriculture and Forestry University, Fuzhou 350108, China
| | - Weijie Lin
- College of Material Engineering, Fujian Agriculture and Forestry University, Fuzhou 350108, China
| | - Fang Huang
- College of Material Engineering, Fujian Agriculture and Forestry University, Fuzhou 350108, China.
| | - Yonghao Ni
- College of Material Engineering, Fujian Agriculture and Forestry University, Fuzhou 350108, China; Department of Chemical Engineering, University of New Brunswick, Fredericton E3B 5A3, Canada.
| |
Collapse
|
32
|
Tanaka YK, Shimazaki S, Fukumoto Y, Ogra Y. Detection of Histidine-Tagged Protein in Escherichia coli by Single-Cell Inductively Coupled Plasma-Mass Spectrometry. Anal Chem 2022; 94:7952-7959. [PMID: 35617709 DOI: 10.1021/acs.analchem.2c00774] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We have developed a rapid and precise quantification method for a histidine (His)-tagged recombinant protein produced in Escherichia coli (E. coli) by single-cell inductively coupled plasma-mass spectrometry (SC-ICP-MS). Plasmid vector containing enhanced green fluorescent protein (EGFP) or red fluorescent protein (mCherry) gene fused with His-tag was transformed into E. coli. The transformed E. coli was exposed to nickel (Ni) chloride or cobalt (Co) chloride for labeling His-tag with the Ni or Co ion. Then, E. coli was analyzed by SC-ICP-MS to determine the amount of EGFP or mCherry protein on the basis of the signal of Ni or Co bound to His-tag. By comparing Ni and Co contents in E. coli expressing His-tagged mCherry with those in nontagged mCherry, the specific binding of Co to His-tag was more clearly detected than that of Ni. The Co contents were increased until 6 h after the protein induction, and this observation was coincident with the increases in fluorescence intensity of EGFP or mCherry measured by a flow cytometer. However, the Co contents were decreased for EGFP and kept at a constant level for mCherry from 6 to 24 h despite the continuous increase in the fluorescence intensity through incubation. The fluorescent proteins were mainly recovered in the insoluble fraction 24 h after the induction. This can be explained by the fact that the overexpressed fluorescent proteins with His-tag are transferred into inclusion bodies, which hampers the binding of the fluorescent proteins to the Co ion. SC-ICP-MS can be a useful technique to precisely quantify soluble recombinant proteins in E. coli without the extraction and purification process.
Collapse
Affiliation(s)
- Yu-Ki Tanaka
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo, Chiba 260-8675, Japan
| | - Shunsuke Shimazaki
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo, Chiba 260-8675, Japan
| | - Yasunori Fukumoto
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo, Chiba 260-8675, Japan
| | - Yasumitsu Ogra
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo, Chiba 260-8675, Japan
| |
Collapse
|
33
|
Vermeer B, Schmid S. Can DyeCycling break the photobleaching limit in single-molecule FRET? NANO RESEARCH 2022; 15:9818-9830. [PMID: 35582137 PMCID: PMC9101981 DOI: 10.1007/s12274-022-4420-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 05/03/2023]
Abstract
Biomolecular systems, such as proteins, crucially rely on dynamic processes at the nanoscale. Detecting biomolecular nanodynamics is therefore key to obtaining a mechanistic understanding of the energies and molecular driving forces that control biomolecular systems. Single-molecule fluorescence resonance energy transfer (smFRET) is a powerful technique to observe in real-time how a single biomolecule proceeds through its functional cycle involving a sequence of distinct structural states. Currently, this technique is fundamentally limited by irreversible photobleaching, causing the untimely end of the experiment and thus, a narrow temporal bandwidth of ≤ 3 orders of magnitude. Here, we introduce "DyeCycling", a measurement scheme with which we aim to break the photobleaching limit in smFRET. We introduce the concept of spontaneous dye replacement by simulations, and as an experimental proof-of-concept, we demonstrate the intermittent observation of a single biomolecule for one hour with a time resolution of milliseconds. Theoretically, DyeCycling can provide > 100-fold more information per single molecule than conventional smFRET. We discuss the experimental implementation of DyeCycling, its current and fundamental limitations, and specific biological use cases. Given its general simplicity and versatility, DyeCycling has the potential to revolutionize the field of time-resolved smFRET, where it may serve to unravel a wealth of biomolecular dynamics by bridging from milliseconds to the hour range. Electronic Supplementary Material Supplementary material is available for this article at 10.1007/s12274-022-4420-5 and is accessible for authorized users.
Collapse
Affiliation(s)
- Benjamin Vermeer
- NanoDynamicsLab, Laboratory of Biophysics, Wageningen University, Stippeneng 4, 6708WE Wageningen, The Netherlands
| | - Sonja Schmid
- NanoDynamicsLab, Laboratory of Biophysics, Wageningen University, Stippeneng 4, 6708WE Wageningen, The Netherlands
| |
Collapse
|
34
|
Basso A, Brown MS, Cruz-Izquierdo A, Martinez CA, Serban S. Optimization of Metal Affinity Ketoreductase Immobilization for Application in Batch and Flow Processes. Org Process Res Dev 2022. [DOI: 10.1021/acs.oprd.1c00483] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Alessandra Basso
- Unit D, Purolite Ltd., Llantrisant Business Park, Llantrisant CF72 8LF, U.K
| | - Maria S. Brown
- Pfizer Inc., 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | | | - Carlos A. Martinez
- Pfizer Inc., 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Simona Serban
- Unit D, Purolite Ltd., Llantrisant Business Park, Llantrisant CF72 8LF, U.K
| |
Collapse
|
35
|
Forstater JH, Grosser ST. Data-rich process development of immobilized biocatalysts in flow. REACT CHEM ENG 2022. [DOI: 10.1039/d1re00298h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The authors describe an automated, data-rich screening and process development method for rapid discovery, development, and optimization of immobilized enzymes, critical to many biocatalytic processes.
Collapse
Affiliation(s)
- Jacob H. Forstater
- Process Research and Development, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Shane T. Grosser
- Process Research and Development, Merck & Co., Inc., Rahway, NJ 07065, USA
| |
Collapse
|
36
|
Gao J, Yuan X, Zheng X, Zhao X, Wang T, Liang Q, Xiao C, Wang J, Li Q, Zhao X. Two-point immobilization of a conformation-specific beta 2-adrenoceptor for recognizing the receptor agonists or antagonists inspired by binding-induced DNA assembly. Biomater Sci 2021; 9:7934-7943. [PMID: 34704989 DOI: 10.1039/d1bm01222c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Immobilized protein has advanced in many areas like drug discovery. While this field evolved rapidly over the last three decades, the immobilization platform for the G-protein-coupled receptor (GPCR) remains unpromising due to its instability under the relatively harsh conditions of current methodologies. Taking beta2-adrenoceptor (β2-AR) as an example, we presented here a general strategy for immobilization of GPCRs by combining the His6-tag trap system, conformation-specific aptamer, and target binding induced DNA hybridization. Morphology characterization by diverse assays confirmed a monolayer of β2-AR on the microsphere surface. The radio-ligand binding assay and immuno-transmission electron microscopy showed desirable ligand- and antibody-binding activities. A case study of chromatography using the immobilized receptor as a stationary phase exhibited a demonstrable conformation specificity that enables the selective recognition of the receptor agonists or antagonists. Owing to the competitive strand displacement during the immobilization, the method proved to be capable of sensitively and directly determining the receptor density on the surface which enormously challenges most of the reported assays. This method is possible to turn into a general strategy for the immobilization of GPCRs with a defined orientation, conformation, function, and density, thus paving the way for precisely realizing the receptor-ligand binding interaction and screening the receptor agonist or antagonist with high efficiency.
Collapse
Affiliation(s)
- Juan Gao
- College of Life Sciences, Northwest University, Xi'an 710069, China.
| | - Xinyi Yuan
- College of Life Sciences, Northwest University, Xi'an 710069, China.
| | - Xinxin Zheng
- College of Life Sciences, Northwest University, Xi'an 710069, China.
| | - Xue Zhao
- College of Life Sciences, Northwest University, Xi'an 710069, China.
| | - Taotao Wang
- College of Life Sciences, Northwest University, Xi'an 710069, China.
| | - Qi Liang
- College of Life Sciences, Northwest University, Xi'an 710069, China.
| | - Chaoni Xiao
- College of Life Sciences, Northwest University, Xi'an 710069, China.
| | - Jing Wang
- College of Life Sciences, Northwest University, Xi'an 710069, China.
| | - Qian Li
- College of Life Sciences, Northwest University, Xi'an 710069, China.
| | - Xinfeng Zhao
- College of Life Sciences, Northwest University, Xi'an 710069, China.
| |
Collapse
|
37
|
López-Laguna H, Sánchez JM, Carratalá JV, Rojas-Peña M, Sánchez-García L, Parladé E, Sánchez-Chardi A, Voltà-Durán E, Serna N, Cano-Garrido O, Flores S, Ferrer-Miralles N, Nolan V, de Marco A, Roher N, Unzueta U, Vazquez E, Villaverde A. Biofabrication of functional protein nanoparticles through simple His-tag engineering. ACS SUSTAINABLE CHEMISTRY & ENGINEERING 2021; 9:12341-12354. [PMID: 34603855 PMCID: PMC8483566 DOI: 10.1021/acssuschemeng.1c04256] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/16/2021] [Indexed: 05/03/2023]
Abstract
We have developed a simple, robust, and fully transversal approach for the a-la-carte fabrication of functional multimeric nanoparticles with potential biomedical applications, validated here by a set of diverse and unrelated polypeptides. The proposed concept is based on the controlled coordination between Zn2+ ions and His residues in His-tagged proteins. This approach results in a spontaneous and reproducible protein assembly as nanoscale oligomers that keep the original functionalities of the protein building blocks. The assembly of these materials is not linked to particular polypeptide features, and it is based on an environmentally friendly and sustainable approach. The resulting nanoparticles, with dimensions ranging between 10 and 15 nm, are regular in size, are architecturally stable, are fully functional, and serve as intermediates in a more complex assembly process, resulting in the formation of microscale protein materials. Since most of the recombinant proteins produced by biochemical and biotechnological industries and intended for biomedical research are His-tagged, the green biofabrication procedure proposed here can be straightforwardly applied to a huge spectrum of protein species for their conversion into their respective nanostructured formats.
Collapse
Affiliation(s)
- Hèctor López-Laguna
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- Departament
de Genètica i de Microbiologia, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- CIBER
de Bioingeniería, Biomateriales y
Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, Madrid 28029, Spain
| | - Julieta M. Sánchez
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- Departament
de Genètica i de Microbiologia, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- Universidad
Nacional de Córdoba, Facultad de
Ciencias Exactas, Físicas y Naturales, ICTA and Departamento
de Química, Cátedra de Química
Biológica, Av. Vélez Sársfield
1611, Córdoba 5016, Argentina
- CONICET-Universidad
Nacional de Córdoba, Instituto de Investigaciones Biológicas y Tecnológicas
(IIByT), Av. Velez Sarsfield
1611, Córdoba, 5016, Argentina
| | - José Vicente Carratalá
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- Departament
de Genètica i de Microbiologia, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- CIBER
de Bioingeniería, Biomateriales y
Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, Madrid 28029, Spain
| | - Mauricio Rojas-Peña
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Laura Sánchez-García
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- Departament
de Genètica i de Microbiologia, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- CIBER
de Bioingeniería, Biomateriales y
Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, Madrid 28029, Spain
| | - Eloi Parladé
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- Departament
de Genètica i de Microbiologia, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- CIBER
de Bioingeniería, Biomateriales y
Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, Madrid 28029, Spain
| | - Alejandro Sánchez-Chardi
- Servei de
Microscòpia, Universitat Autònoma
de Barcelona, Bellaterra, Barcelona 08193, Spain
- Departament
de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat
de Biologia, Universitat de Barcelona, Av. Diagonal 643, Barcelona 08028, Spain
| | - Eric Voltà-Durán
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- Departament
de Genètica i de Microbiologia, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- CIBER
de Bioingeniería, Biomateriales y
Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, Madrid 28029, Spain
| | - Naroa Serna
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- Departament
de Genètica i de Microbiologia, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- CIBER
de Bioingeniería, Biomateriales y
Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, Madrid 28029, Spain
| | - Olivia Cano-Garrido
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- Departament
de Genètica i de Microbiologia, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- CIBER
de Bioingeniería, Biomateriales y
Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, Madrid 28029, Spain
| | - Sandra Flores
- Universidad
Nacional de Córdoba, Facultad de
Ciencias Exactas, Físicas y Naturales, ICTA and Departamento
de Química, Cátedra de Química
Biológica, Av. Vélez Sársfield
1611, Córdoba 5016, Argentina
- CONICET-Universidad
Nacional de Córdoba, Instituto de Investigaciones Biológicas y Tecnológicas
(IIByT), Av. Velez Sarsfield
1611, Córdoba, 5016, Argentina
| | - Neus Ferrer-Miralles
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- Departament
de Genètica i de Microbiologia, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- CIBER
de Bioingeniería, Biomateriales y
Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, Madrid 28029, Spain
| | - Verónica Nolan
- Universidad
Nacional de Córdoba, Facultad de
Ciencias Exactas, Físicas y Naturales, ICTA and Departamento
de Química, Cátedra de Química
Biológica, Av. Vélez Sársfield
1611, Córdoba 5016, Argentina
- CONICET-Universidad
Nacional de Córdoba, Instituto de Investigaciones Biológicas y Tecnológicas
(IIByT), Av. Velez Sarsfield
1611, Córdoba, 5016, Argentina
| | - Ario de Marco
- Laboratory
for Environmental and Life Sciences, University
of Nova Gorica, Nova Gorica 5000, Slovenia
| | - Nerea Roher
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- CIBER
de Bioingeniería, Biomateriales y
Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, Madrid 28029, Spain
- Departament
de Biologia Cel·lular, Fisiologia Animal i Immunologia, Universitat Autònoma de Barcelona, Barcelona 08193, Spain
| | - Ugutz Unzueta
- Departament
de Genètica i de Microbiologia, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- CIBER
de Bioingeniería, Biomateriales y
Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, Madrid 28029, Spain
- Biomedical
Research Institute Sant Pau (IIB Sant Pau), Sant Antoni Ma Claret 167, Barcelona 08025, Spain
| | - Esther Vazquez
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- Departament
de Genètica i de Microbiologia, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- CIBER
de Bioingeniería, Biomateriales y
Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, Madrid 28029, Spain
| | - Antonio Villaverde
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- Departament
de Genètica i de Microbiologia, Universitat
Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- CIBER
de Bioingeniería, Biomateriales y
Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, Madrid 28029, Spain
| |
Collapse
|
38
|
Wilks LR, Joshi G, Grisham MR, Gill HS. Tyrosine-Based Cross-Linking of Peptide Antigens to Generate Nanoclusters with Enhanced Immunogenicity: Demonstration Using the Conserved M2e Peptide of Influenza A. ACS Infect Dis 2021; 7:2723-2735. [PMID: 34432416 DOI: 10.1021/acsinfecdis.1c00219] [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: 01/19/2023]
Abstract
A method of creating nanoclusters (NCs) from soluble peptide molecules is described utilizing an approach based on a tyrosine-tyrosine cross-linking reaction. A reactive tag comprising histidine and tyrosine residues was introduced at the termini of the peptide molecules. The cross-linking reaction led to the creation of dityrosine bonds within the tag, which allowed for the generation of peptide NCs. We show that it is essential for the reactive tag to be present at both the "N" and "C" termini of the peptide for cluster formation to occur. Additionally, the cross-linking reaction was systematically characterized to show the importance of reaction conditions on final cluster diameter, allowing us to generate NCs of various sizes. To demonstrate the immunogenic potential of the peptide clusters, we chose to study the conserved influenza peptide, M2e, as the antigen. M2e NCs were formulated using the cross-linking reaction. We show the ability of the clusters to generate protective immunity in a dose, size, and frequency dependent manner against a lethal influenza A challenge in BALB/c mice. Taken together, the data presented suggest this new cluster formation technique can generate highly immunogenic peptide NCs in a simple and controllable manner.
Collapse
Affiliation(s)
- Logan R. Wilks
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Gaurav Joshi
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Megan R. Grisham
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| |
Collapse
|
39
|
López-Laguna H, Voltà-Durán E, Parladé E, Villaverde A, Vázquez E, Unzueta U. Insights on the emerging biotechnology of histidine-rich peptides. Biotechnol Adv 2021; 54:107817. [PMID: 34418503 DOI: 10.1016/j.biotechadv.2021.107817] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/16/2021] [Accepted: 08/16/2021] [Indexed: 02/07/2023]
Abstract
In the late 70's, the discovery of the restriction enzymes made possible the biological production of functional proteins by recombinant DNA technologies, a fact that largely empowered both biotechnological and pharmaceutical industries. Short peptides or small protein domains, with specific molecular affinities, were developed as purification tags in downstream processes to separate the target protein from the culture media or cell debris, upon breaking the producing cells. Among these tags, and by exploiting the interactivity of the imidazole ring of histidine residues, the hexahistidine peptide (H6) became a gold standard. Although initially used almost exclusively in protein production, H6 and related His-rich peptides are progressively proving a broad applicability in novel utilities including enzymatic processes, advanced drug delivery systems and diagnosis, through a so far unsuspected adaptation of their binding capabilities. In this context, the coordination of histidine residues and metals confers intriguing functionalities to His-rich sequences useable in the forward-thinking design of protein-based nano- and micro-materials and devices, through strategies that are comprehensively presented here.
Collapse
Affiliation(s)
- Hèctor López-Laguna
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Eric Voltà-Durán
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Eloi Parladé
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| | - Ugutz Unzueta
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain; Biomedical Research Institute Sant Pau (IIB Sant Pau), Sant Antoni Mª Claret 167, 08025 Barcelona, Spain.
| |
Collapse
|
40
|
El Hajj S, Sepúlveda Rincón CT, Girardet JM, Cakir-Kiefer C, Stefan L, Zapata Montoya JE, Boschi-Muller S, Gaucher C, Canabady-Rochelle L. Electrically Switchable Nanolever Technology for the Screening of Metal-Chelating Peptides in Hydrolysates. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:8819-8827. [PMID: 34324321 DOI: 10.1021/acs.jafc.1c02199] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Metal-chelating peptides (MCP) are considered as indirect antioxidants due to their capacity to inhibit radical chain reaction and oxidation. Here, we propose a new proof of concept for the screening of MCPs present in protein hydrolysates for valorizing their antioxidant properties by using the emerging time-resolved molecular dynamics technology, switchSENSE. This method unveils possible interactions between MCPs and immobilized nickel ions using fluorescence and electro-switchable DNA chips. The switchSENSE method was first set up on synthetic peptides known for their metal-chelating properties. Then, it was applied to soy and tilapia viscera protein hydrolysates. Their Cu2+-chelation capacity was, in addition, determined by UV-visible spectrophotometry as a reference method. The switchSENSE method has displayed a high sensitivity to evidence the presence of MCPs in both hydrolysates. Hence, we demonstrate for the first time that this newly introduced technology is a convenient methodology to screen protein hydrolysates in order to determine the presence of MCPs before launching time-consuming separations.
Collapse
Affiliation(s)
- Sarah El Hajj
- Université de Lorraine, CNRS, LRGP, Nancy F-54000, France
- Université de Lorraine, CITHEFOR, Vandoeuvre Les Nancy F-54505, France
| | - Cindy Tatiana Sepúlveda Rincón
- Université de Lorraine, CNRS, LRGP, Nancy F-54000, France
- Nutrition & Food Technology Group, Universidad de Antioquia, Medellín 050010, Colombia
| | | | | | - Loic Stefan
- Université de Lorraine, CNRS, LCPM, Nancy F-54000, France
| | | | | | - Caroline Gaucher
- Université de Lorraine, CITHEFOR, Vandoeuvre Les Nancy F-54505, France
| | | |
Collapse
|
41
|
Drozd M, Karoń S, Malinowska E. Recent Advancements in Receptor Layer Engineering for Applications in SPR-Based Immunodiagnostics. SENSORS (BASEL, SWITZERLAND) 2021; 21:3781. [PMID: 34072572 PMCID: PMC8198293 DOI: 10.3390/s21113781] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022]
Abstract
The rapid progress in the development of surface plasmon resonance-based immunosensing platforms offers wide application possibilities in medical diagnostics as a label-free alternative to enzyme immunoassays. The early diagnosis of diseases or metabolic changes through the detection of biomarkers in body fluids requires methods characterized by a very good sensitivity and selectivity. In the case of the SPR technique, as well as other surface-sensitive detection strategies, the quality of the transducer-immunoreceptor interphase is crucial for maintaining the analytical reliability of an assay. In this work, an overview of general approaches to the design of functional SPR-immunoassays is presented. It covers both immunosensors, the design of which utilizes well-known and often commercially available substrates, as well as the latest solutions developed in-house. Various approaches employing chemical and passive binding, affinity-based antibody immobilization, and the introduction of nanomaterial-based surfaces are discussed. The essence of their influence on the improvement of the main analytical parameters of a given immunosensor is explained. Particular attention is paid to solutions compatible with the latest trends in the development of label-free immunosensors, such as platforms dedicated to real-time monitoring in a quasi-continuous mode, the use of in situ-generated receptor layers (elimination of the regeneration step), and biosensors using recombinant and labelled protein receptors.
Collapse
Affiliation(s)
- Marcin Drozd
- Faculty of Chemistry, The Chair of Medical Biotechnology, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland;
- Center for Advanced Materials and Technologies, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| | - Sylwia Karoń
- Faculty of Chemistry, The Chair of Medical Biotechnology, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland;
- Center for Advanced Materials and Technologies, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| | - Elżbieta Malinowska
- Faculty of Chemistry, The Chair of Medical Biotechnology, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland;
- Center for Advanced Materials and Technologies, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| |
Collapse
|
42
|
Abad JM, Puertas S, Pérez D, Sánchez-Espinel C. Design and Development of Antibody Functionalized Gold Nanoparticles for Biomedical Applications. JOURNAL OF NANOSCIENCE AND NANOTECHNOLOGY 2021; 21:2834-2840. [PMID: 33653448 DOI: 10.1166/jnn.2021.19057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Antibody-functionalized gold nanoparticle constitutes a powerful interface biosystem for biomedical applications where the properties of gold nanoparticles and the specificity of antibody-antigen interactions are combined. This study provides insight into the key factors for the development of antibody functionalized gold nanoparticles focusing on the immobilization of the antibody. Here, we address an oriented antibody immobilization procedure on gold nanoparticles. It comprises chelatemodified gold nanoparticles that are designed for oriented immobilization of IgG antibodies (end on spatial orientation) through the metal-chelation to histidine-rich metal binding site in the heavy chain (Fc) of the antibody.
Collapse
Affiliation(s)
- José M Abad
- Nanoimmunotech, S.L. Edificio CITEXVI Fonte das Abelleiras s/n, Campus Universitario de Vigo, 36310 Vigo, Pontevedra, Spain
| | - Sara Puertas
- Nanoimmunotech, S.L. Edificio CITEXVI Fonte das Abelleiras s/n, Campus Universitario de Vigo, 36310 Vigo, Pontevedra, Spain
| | - Daniel Pérez
- Nanoimmunotech, S.L. Edificio CITEXVI Fonte das Abelleiras s/n, Campus Universitario de Vigo, 36310 Vigo, Pontevedra, Spain
| | - Christian Sánchez-Espinel
- Nanoimmunotech, S.L. Edificio CITEXVI Fonte das Abelleiras s/n, Campus Universitario de Vigo, 36310 Vigo, Pontevedra, Spain
| |
Collapse
|
43
|
Ardini M, Bellelli A, Williams DL, Di Leandro L, Giansanti F, Cimini A, Ippoliti R, Angelucci F. Taking Advantage of the Morpheein Behavior of Peroxiredoxin in Bionanotechnology. Bioconjug Chem 2021; 32:43-62. [PMID: 33411522 PMCID: PMC8023583 DOI: 10.1021/acs.bioconjchem.0c00621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
![]()
Morpheeins
are proteins that reversibly assemble into different
oligomers, whose architectures are governed by conformational changes
of the subunits. This property could be utilized in bionanotechnology
where the building of nanometric and new high-ordered structures is
required. By capitalizing on the adaptability of morpheeins to create
patterned structures and exploiting their inborn affinity toward inorganic
and living matter, “bottom-up” creation of nanostructures
could be achieved using a single protein building block, which may
be useful as such or as scaffolds for more complex materials. Peroxiredoxins
represent the paradigm of a morpheein that can be applied to bionanotechnology.
This review describes the structural and functional transitions that
peroxiredoxins undergo to form high-order oligomers, e.g., rings,
tubes, particles, and catenanes, and reports on the chemical and genetic
engineering approaches to employ them in the generation of responsive
nanostructures and nanodevices. The usefulness of the morpheeins’
behavior is emphasized, supporting their use in future applications.
Collapse
Affiliation(s)
- Matteo Ardini
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, Piazzale Salvatore Tommasi 1, 67100 L'Aquila, Italy
| | - Andrea Bellelli
- Department of Biochemical Sciences "A. Rossi Fanelli", University of Roma "Sapienza", Piazzale Aldo Moro 5, 00185 Roma, Italy
| | - David L Williams
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois 60612, United States
| | - Luana Di Leandro
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, Piazzale Salvatore Tommasi 1, 67100 L'Aquila, Italy
| | - Francesco Giansanti
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, Piazzale Salvatore Tommasi 1, 67100 L'Aquila, Italy
| | - Annamaria Cimini
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, Piazzale Salvatore Tommasi 1, 67100 L'Aquila, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, Piazzale Salvatore Tommasi 1, 67100 L'Aquila, Italy
| | - Francesco Angelucci
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, Piazzale Salvatore Tommasi 1, 67100 L'Aquila, Italy
| |
Collapse
|
44
|
Krishnarjuna B, Sunanda P, Villegas-Moreno J, Csoti A, A V Morales R, Wai DCC, Panyi G, Prentis P, Norton RS. A disulfide-stabilised helical hairpin fold in acrorhagin I: An emerging structural motif in peptide toxins. J Struct Biol 2020; 213:107692. [PMID: 33387653 DOI: 10.1016/j.jsb.2020.107692] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/14/2020] [Accepted: 12/24/2020] [Indexed: 12/22/2022]
Abstract
Acrorhagin I (U-AITX-Aeq5a) is a disulfide-rich peptide identified in the aggressive organs (acrorhagi) of the sea anemone Actinia equina. Previous studies (Toxicon 2005, 46:768-74) found that the peptide is toxic in crabs, although the structural and functional properties of acrorhagin I have not been reported. In this work, an Escherichia coli (BL21 strain) expression system was established for the preparation of 13C,15N-labelled acrorhagin I, and the solution structure was determined using NMR spectroscopy. Structurally, acrorhagin I is similar to B-IV toxin from the marine worm Cerebratulus lacteus (PDB id 1VIB), with a well-defined helical hairpin structure stabilised by four intramolecular disulfide bonds. The recombinant peptide was tested in patch-clamp electrophysiology assays against voltage-gated potassium and sodium channels, and in bacterial and fungal growth inhibitory assays and haemolytic assays. Acrorhagin I was not active against any of the ion channels tested and showed no activity in functional assays, indicating that this peptide may possess a different biological function. Metal ion interaction studies using NMR spectroscopy showed that acrorhagin I bound zinc and nickel, suggesting that its function might be modulated by metal ions or that it may be involved in regulating metal ion levels and their transport. The similarity between the structure of acrorhagin I and that of B-IV toxin from a marine worm suggests that this fold may prove to be a recurring motif in disulfide-rich peptides from marine organisms.
Collapse
Affiliation(s)
- Bankala Krishnarjuna
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Punnepalli Sunanda
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Jessica Villegas-Moreno
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia; Centro de Investigaciones Químicas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | - Agota Csoti
- Department of Biophysics and Cell Biology, University of Debrecen, 4032 Debrecen, Hungary; MTA-DE-NAP B Ion Channel Structure-Function Research Group, RCMM, University of Debrecen, 4032 Debrecen, Hungary
| | - Rodrigo A V Morales
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Dorothy C C Wai
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, University of Debrecen, 4032 Debrecen, Hungary; MTA-DE-NAP B Ion Channel Structure-Function Research Group, RCMM, University of Debrecen, 4032 Debrecen, Hungary
| | - Peter Prentis
- School of Earth, Environmental and Biological Sciences, Science and Engineering Faculty, Queensland University of Technology, Brisbane, Australia; Institute for Future Environments, Queensland University of Technology, Brisbane, Australia
| | - Raymond S Norton
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia; ARC Centre for Fragment-Based Design, Monash University, Parkville, Victoria 3052, Australia.
| |
Collapse
|
45
|
Tsitkov S, Song Y, Rodriguez JB, Zhang Y, Hess H. Kinesin-Recruiting Microtubules Exhibit Collective Gliding Motion while Forming Motor Trails. ACS NANO 2020; 14:16547-16557. [PMID: 33054177 DOI: 10.1021/acsnano.0c03263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Microtubules gliding on surfaces coated with kinesin motors are minimalist experimental systems for studying collective behavior. Collective behavior in these systems arises from interactions between filaments, for example, from steric interactions, depletion forces, or cross-links. To maximize the utilization of system components and the production of work, it is desirable to achieve mutualistic interactions leading to the congregations of both types of agents, that is, cytoskeletal filaments and molecular motors. To this end, we used a microtubule-kinesin system, where motors reversibly bind to the surface via an interaction between a hexahistidine (His6) tag on the motor and a Ni(II)-nitrilotriacetic acid (Ni-NTA) moiety on the surface. The surface density of binding sites for kinesin motors was increased relative to our earlier work, driving the motors from the solution to the surface. Characterization of the motor-surface interactions in the absence of microtubules yielded kinetic parameters consistent with previous data and revealed the capacity of the surface to support two-dimensional motor diffusion. The motor density gradually fell over 2 h, presumably due to the stripping of Ni(II) from the NTA moieties on the surface. Microtubules gliding on these reversibly bound motors were unable to cross each other and at high enough densities began to align and form long, dense bundles. The kinesin motors accumulated in trails surrounding the microtubule bundles and participated in microtubule transport.
Collapse
Affiliation(s)
- Stanislav Tsitkov
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Yuchen Song
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
- Department of Biomedical Engineering, Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Juan B Rodriguez
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Yifei Zhang
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Henry Hess
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| |
Collapse
|
46
|
Gunnarsson A, Stubbs CJ, Rawlins PB, Taylor-Newman E, Lee WC, Geschwindner S, Hytönen V, Holdgate G, Jha R, Dahl G. Regenerable Biosensors for Small-Molecule Kinetic Characterization Using SPR. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2020; 26:730-739. [PMID: 33289457 DOI: 10.1177/2472555220975358] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A key activity in small-molecule drug discovery is the characterization of compound-target interactions. Surface plasmon resonance (SPR) is a flexible technique for this purpose, with a wide affinity range (micromoles to picomoles), low protein requirements, and the ability to characterize the kinetics of compound binding. However, a key requirement of SPR is the immobilization of the target protein to the surface of the sensor chip. The most commonly used immobilization techniques (covalent immobilization, streptavidin-biotin) are irreversible in nature, which can afford excellent baseline stability but impose limitations throughput for slowly dissociating compounds or unstable targets. Reversible immobilization (e.g., His-tag-Ni-NTA) is possible but typically precludes accurate quantification of slow dissociation kinetics due to baseline drift.Here we present our investigation of three immobilization strategies (dual-His-tagged target protein, His-tagged streptavidin, and switchavidin) that combine the robustness of irreversible immobilization with the flexibility of reversible immobilization. Each has its own advantages and limitations, and while a universal immobilization procedure remains to be found, these strategies add to the immobilization toolbox that enables previously out-of-scope applications. Such applications are highlighted in two examples that greatly increased throughput for the kinetic characterization of potent kinase inhibitors and kinetic profiling of covalent inhibitors.
Collapse
Affiliation(s)
- Anders Gunnarsson
- Structure, Biophysics and Fragment-based Lead Generation, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Christopher J Stubbs
- Structure, Biophysics and Fragment-based Lead Generation, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Philip B Rawlins
- Structure, Biophysics and Fragment-based Lead Generation, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Eleanor Taylor-Newman
- Structure, Biophysics and Fragment-based Lead Generation, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK.,Deptartment of Chemistry, University of Leicester, Leicester, UK
| | - Wei-Chao Lee
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Stefan Geschwindner
- Structure, Biophysics and Fragment-based Lead Generation, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Vesa Hytönen
- Faculty of Medicine and Health Technology, University of Tampere, Tampere, Pirkanmaa, Finland
| | - Geoffrey Holdgate
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Alderley Park, UK
| | - Rupam Jha
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Göran Dahl
- Structure, Biophysics and Fragment-based Lead Generation, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
47
|
Buddingh' BC, Llopis-Lorente A, Abdelmohsen LKEA, van Hest JCM. Dynamic spatial and structural organization in artificial cells regulates signal processing by protein scaffolding. Chem Sci 2020; 11:12829-12834. [PMID: 34094478 PMCID: PMC8163283 DOI: 10.1039/d0sc03933k] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/04/2020] [Indexed: 01/25/2023] Open
Abstract
Structural and spatial organization are fundamental properties of biological systems that allow cells to regulate a wide range of biochemical processes. This organization is often transient and governed by external cues that initiate dynamic self-assembly processes. The construction of synthetic cell-like materials with similar properties requires the hierarchical and reversible organization of selected functional components on molecular scaffolds to dynamically regulate signaling pathways. The realization of such transient molecular programs in synthetic cells, however, remains underexplored due to the associated complexity of such hierarchical platforms. In this contribution, we effectuate dynamic spatial organization of effector protein subunits in a synthetic biomimetic compartment, a giant unilamellar vesicle (GUV), by associating in a reversible manner two fragments of a split luciferase to the membrane. This induces their structural dimerization, which consequently leads to the activation of enzymatic signaling. Importantly, such organization and activation are dynamic processes, and can be autonomously regulated - thus opening up avenues toward continuous spatiotemporal control over supramolecular organization and signaling in an artificial cell.
Collapse
Affiliation(s)
- Bastiaan C Buddingh'
- Department of Chemical Engineering and Chemistry, Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology PO Box 513 5600 MB Eindhoven The Netherlands
| | - Antoni Llopis-Lorente
- Department of Chemical Engineering and Chemistry, Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology PO Box 513 5600 MB Eindhoven The Netherlands
| | - Loai K E A Abdelmohsen
- Department of Chemical Engineering and Chemistry, Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology PO Box 513 5600 MB Eindhoven The Netherlands
| | - Jan C M van Hest
- Department of Chemical Engineering and Chemistry, Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology PO Box 513 5600 MB Eindhoven The Netherlands
| |
Collapse
|
48
|
Roos N, Breiner B, Preuss L, Lilie H, Hipp K, Herrmann H, Horn T, Biener R, Iftner T, Simon C. Optimized production strategy of the major capsid protein HPV 16L1 non-assembly variant in E. coli. Protein Expr Purif 2020; 175:105690. [DOI: 10.1016/j.pep.2020.105690] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 12/31/2022]
|
49
|
Leeman M, Albers WM, Bombera R, Kuncova-Kallio J, Tuppurainen J, Nilsson L. Asymmetric flow field-flow fractionation coupled to surface plasmon resonance detection for analysis of therapeutic proteins in blood serum. Anal Bioanal Chem 2020; 413:117-127. [PMID: 33098467 PMCID: PMC7801359 DOI: 10.1007/s00216-020-03011-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022]
Abstract
Coupling of surface plasmon resonance (SPR) detection to asymmetric flow field-flow fractionation (AF4) offers the possibility to study active fractions of bio-separations on real samples, such as serum and saliva, including the assessment of activity of possibly aggregated species. The coupling of SPR with AF4 requires the possibility to select fractions from a fractogram and redirect them to the SPR. The combination of SPR with chromatography-like methods also requires a mechanism for regeneration of the receptor immobilised onto the SPR sensor surface. In recent work, the combination of size exclusion chromatography (SEC) with SPR was pioneered as a successful methodology for identification, characterisation and quantification of active biocomponents in biological samples. In this study, the approach using AF4 is evaluated for the antibody trastuzumab in buffer and serum. The particular object of this study was to test the feasibility of using AF4 in combination with SPR to detect and quantify proteins and aggregates in complex samples such as blood serum. Also, in the investigation, three different immobilisation methods for the receptor HER-2 were compared, which involved (1) direct binding via EDC/NHS, the standard approach; (2) immobilisation via NTA-Ni-Histag complexation; and (3) biotin/avidin-linked chemistry using a regenerable form of avidin. The highest specific activity was obtained for the biotin-avidin method, while the lowest specific activity was observed for the NTA-Ni-Histag linkage. The data show that AF4 can separate trastuzumab monomers and aggregates in blood serum and that SPR has the ability to selectively monitor the elution. This is an encouraging result for automated analysis of complex biological samples using AF4-SPR.
Collapse
Affiliation(s)
- Mats Leeman
- SOLVE Research and Consultancy AB, Medicon village, 22381, Lund, Sweden
| | | | | | | | | | - Lars Nilsson
- Department of Food Technology, Engineering and Nutrition, Faculty of Engineering LTH, Lund University, 22100, Lund, Sweden.
| |
Collapse
|
50
|
Development of a Novel SPR Assay to Study CXCR4-Ligand Interactions. BIOSENSORS-BASEL 2020; 10:bios10100150. [PMID: 33096938 PMCID: PMC7589327 DOI: 10.3390/bios10100150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/15/2020] [Accepted: 10/21/2020] [Indexed: 01/16/2023]
Abstract
G protein-coupled receptors (GPCRs) are involved in a plethora of different diseases. Consequently, these proteins are considered as an important class of drug targets. Measuring detailed kinetic information on these types of proteins has been challenging. Surface plasmon resonance (SPR) can provide this information, however, the use of SPR on GPCRs remains a complex issue. Here, we report an SPR assay to investigate the interactions between the full-length chemokine receptor CXCR4 and nanobody-Fc (Nb-Fc) ligands. Nb-Fcs consist of two monovalent VHH domains fused with an Fc domain of a human IgG molecule. The CXCR4 protein used in this assay was produced with a C-terminal 10x-histidine tag and was immobilized on a nitrilotriacetic acid chip. In order to verify the sensitivity and effectiveness of this assay, the results were compared to data obtained from cellular assays as well as from another SPR assay using CXCR4 virus-like particles (VLPs). CXCR4 remained intact and stable for at least 12 h, and the kinetic results correlated well with both the cellular assays and the VLP SPR assay results. Apart from determining the binding kinetics of Nb-Fc with CXCR4, our results contributed to understanding CXCR4 interaction dynamics. In conclusion, this assay provides a viable experimental platform that has high potential to be expanded for studying other molecules as well as other histidine-tagged GPCRs.
Collapse
|