1
|
Göz M, Pohl G, Steinecker SM, Walhorn V, Milting H, Anselmetti D. Arrhythmogenic cardiomyopathy-related cadherin variants affect desmosomal binding kinetics. J Mol Cell Cardiol 2024; 195:36-44. [PMID: 39079569 DOI: 10.1016/j.yjmcc.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/09/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024]
Abstract
Cadherins are calcium dependent adhesion proteins that establish and maintain the intercellular mechanical contact by bridging the gap between adjacent cells. Desmoglein-2 (Dsg2) and desmocollin-2 (Dsc2) are tissue specific cadherin isoforms of the cell-cell contact in cardiac desmosomes. Mutations in the DSG2-gene and in the DSC2-gene are related to arrhythmogenic right ventricular cardiomyopathy (ARVC) a rare but severe heart muscle disease. Here, several possible homophilic and heterophilic binding interactions of wild-type Dsg2, wild-type Dsc2, as well as one Dsg2- and two Dsc2-variants, each associated with ARVC, are investigated. Using single molecule force spectroscopy (SMFS) with atomic force microscopy (AFM) and applying Jarzynski's equality the kinetics and thermodynamics of Dsg2/Dsc2 interaction can be determined. The free energy landscape of Dsg2/Dsc2 dimerization exposes a high activation energy barrier, which is in line with the proposed strand-swapping binding motif. Although the binding motif is not affected by any of the mutations, the binding kinetics of the interactions differ significantly from the wild-type. While wild-type cadherins exhibit an average complex lifetime of approx. 0.3 s interactions involving a variant consistently show - lifetimes that are substantially larger. The lifetimes of the wild-type interactions give rise to the picture of a dynamic adhesion interface consisting of continuously dissociating and (re)associating molecular bonds, while the delayed binding kinetics of interactions involving an ARVC-associated variant might be part of the pathogenesis. Our data provide a comprehensive and consistent thermodynamic and kinetic description of cardiac cadherin binding, allowing detailed insight into the molecular mechanisms of cell adhesion.
Collapse
Affiliation(s)
- Manuel Göz
- Department of Physics, Experimental Biophysics and Applied Nanoscience, Bielefeld University, Universitätsstraße 25, Bielefeld, Germany
| | - Greta Pohl
- Erich & Hanna Klessmann Institute for Cardiovascular Research and Development, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstraße 11, Bad Oeynhausen, Germany
| | - Sylvia M Steinecker
- Department of Physics, Experimental Biophysics and Applied Nanoscience, Bielefeld University, Universitätsstraße 25, Bielefeld, Germany
| | - Volker Walhorn
- Department of Physics, Experimental Biophysics and Applied Nanoscience, Bielefeld University, Universitätsstraße 25, Bielefeld, Germany.
| | - Hendrik Milting
- Erich & Hanna Klessmann Institute for Cardiovascular Research and Development, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstraße 11, Bad Oeynhausen, Germany
| | - Dario Anselmetti
- Department of Physics, Experimental Biophysics and Applied Nanoscience, Bielefeld University, Universitätsstraße 25, Bielefeld, Germany
| |
Collapse
|
2
|
Minoshima M, Reja SI, Hashimoto R, Iijima K, Kikuchi K. Hybrid Small-Molecule/Protein Fluorescent Probes. Chem Rev 2024; 124:6198-6270. [PMID: 38717865 DOI: 10.1021/acs.chemrev.3c00549] [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: 05/23/2024]
Abstract
Hybrid small-molecule/protein fluorescent probes are powerful tools for visualizing protein localization and function in living cells. These hybrid probes are constructed by diverse site-specific chemical protein labeling approaches through chemical reactions to exogenous peptide/small protein tags, enzymatic post-translational modifications, bioorthogonal reactions for genetically incorporated unnatural amino acids, and ligand-directed chemical reactions. The hybrid small-molecule/protein fluorescent probes are employed for imaging protein trafficking, conformational changes, and bioanalytes surrounding proteins. In addition, fluorescent hybrid probes facilitate visualization of protein dynamics at the single-molecule level and the defined structure with super-resolution imaging. In this review, we discuss development and the bioimaging applications of fluorescent probes based on small-molecule/protein hybrids.
Collapse
Affiliation(s)
- Masafumi Minoshima
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita, Osaka 5650871, Japan
| | - Shahi Imam Reja
- Immunology Frontier Research Center, Osaka University, 2-1, Yamadaoka, Suita, Osaka 5650871, Japan
| | - Ryu Hashimoto
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita, Osaka 5650871, Japan
| | - Kohei Iijima
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita, Osaka 5650871, Japan
| | - Kazuya Kikuchi
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita, Osaka 5650871, Japan
| |
Collapse
|
3
|
Amati A, Moning SU, Javor S, Schär S, Deutschmann S, Reymond JL, von Ballmoos C. Overcoming Protein Orientation Mismatch Enables Efficient Nanoscale Light-Driven ATP Production. ACS Synth Biol 2024; 13:1355-1364. [PMID: 38569139 PMCID: PMC11036485 DOI: 10.1021/acssynbio.4c00058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 04/05/2024]
Abstract
Adenosine triphosphate (ATP)-producing modules energized by light-driven proton pumps are powerful tools for the bottom-up assembly of artificial cell-like systems. However, the maximum efficiency of such modules is prohibited by the random orientation of the proton pumps during the reconstitution process into lipid-surrounded nanocontainers. Here, we overcome this limitation using a versatile approach to uniformly orient the light-driven proton pump proteorhodopsin (pR) in liposomes. pR is post-translationally either covalently or noncovalently coupled to a membrane-impermeable protein domain guiding orientation during insertion into preformed liposomes. In the second scenario, we developed a novel bifunctional linker, trisNTA-SpyTag, that allows for the reversible connection of any SpyCatcher-containing protein and a HisTag-carrying protein. The desired protein orientations are verified by monitoring vectorial proton pumping and membrane potential generation. In conjunction with ATP synthase, highly efficient ATP production is energized by the inwardly pumping population. In comparison to other light-driven ATP-producing modules, the uniform orientation allows for maximal rates at economical protein concentrations. The presented technology is highly customizable and not limited to light-driven proton pumps but applicable to many membrane proteins and offers a general approach to overcome orientation mismatch during membrane reconstitution, requiring little to no genetic modification of the protein of interest.
Collapse
Affiliation(s)
| | | | - Sacha Javor
- Department of Chemistry, Biochemistry
and Pharmaceutical Sciences, University
of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Sandra Schär
- Department of Chemistry, Biochemistry
and Pharmaceutical Sciences, University
of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | | | - Jean-Louis Reymond
- Department of Chemistry, Biochemistry
and Pharmaceutical Sciences, University
of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Christoph von Ballmoos
- Department of Chemistry, Biochemistry
and Pharmaceutical Sciences, University
of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| |
Collapse
|
4
|
Göz M, Steinecker SM, Pohl GM, Walhorn V, Milting H, Anselmetti D. Cardiac desmosomal adhesion relies on ideal-, slip- and catch bonds. Sci Rep 2024; 14:2555. [PMID: 38297017 PMCID: PMC10830561 DOI: 10.1038/s41598-024-52725-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/23/2024] [Indexed: 02/02/2024] Open
Abstract
The cardiac muscle consists of individual cardiomyocytes that are mechanically linked by desmosomes. Desmosomal adhesion is mediated by densely packed and organized cadherins which, in presence of Ca2+, stretch out their extracellular domains (EC) and dimerize with opposing binding partners by exchanging an N-terminal tryptophan. The strand-swap binding motif of cardiac cadherins like desmocollin 2 (Dsc2) (and desmoglein2 alike) is highly specific but of low affinity with average bond lifetimes in the range of approximately 0.3 s. Notably, despite this comparatively weak interaction, desmosomes mediate a stable, tensile-resistant bond. In addition, force mediated dissociation of strand-swap dimers exhibit a reduced bond lifetime as external forces increase (slip bond). Using atomic force microscopy based single molecule force spectroscopy (AFM-SMFS), we demonstrate that Dsc2 has two further binding modes that, in addition to strand-swap dimers, most likely play a significant role in the integrity of the cardiac muscle. At short interaction times, the Dsc2 monomers associate only loosely, as can be seen from short-lived force-independent bonds. These ideal bonds are a precursor state and probably stabilize the formation of the self-inhibiting strand-swap dimer. The addition of tryptophan in the measurement buffer acts as a competitive inhibitor, preventing the N-terminal strand exchange. Here, Dsc2 dimerizes as X-dimer which clearly shows a tri-phasic slip-catch-slip type of dissociation. Within the force-mediated transition (catch) regime, Dsc2 dimers switch between a rather brittle low force and a strengthened high force adhesion state. As a result, we can assume that desmosomal adhesion is mediated not only by strand-swap dimers (slip) but also by their precursor states (ideal bond) and force-activated X-dimers (catch bond).
Collapse
Affiliation(s)
- Manuel Göz
- Department of Physics, Experimental Biophysics and Applied Nanoscience, Bielefeld University, Universitätstraße 25, 33615, Bielefeld, Germany
| | - Sylvia M Steinecker
- Department of Physics, Experimental Biophysics and Applied Nanoscience, Bielefeld University, Universitätstraße 25, 33615, Bielefeld, Germany
| | - Greta M Pohl
- Erich & Hanna Klessmann Institute for Cardiovascular Research and Development, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstraße 11, Bad Oeynhausen, Germany
| | - Volker Walhorn
- Department of Physics, Experimental Biophysics and Applied Nanoscience, Bielefeld University, Universitätstraße 25, 33615, Bielefeld, Germany.
| | - Hendrik Milting
- Erich & Hanna Klessmann Institute for Cardiovascular Research and Development, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstraße 11, Bad Oeynhausen, Germany
| | - Dario Anselmetti
- Department of Physics, Experimental Biophysics and Applied Nanoscience, Bielefeld University, Universitätstraße 25, 33615, Bielefeld, Germany
| |
Collapse
|
5
|
Paul S, Gupta M, Kumar Mahato A, Karak S, Basak A, Datta S, Banerjee R. Covalent Organic Frameworks for the Purification of Recombinant Enzymes and Heterogeneous Biocatalysis. J Am Chem Soc 2024; 146:858-867. [PMID: 38159294 DOI: 10.1021/jacs.3c11169] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Recombinant enzymes have gained prominence due to their diverse functionalities and specificity and are often a greener alternative in biocatalysis. This context makes purifying recombinant enzymes from host cells and other impurities crucial. The primary goal is to isolate the pure enzyme of interest and ensure its stability under ambient conditions. Covalent organic frameworks (COFs), renowned for their well-ordered structure and permeability, offer a promising approach for purifying histidine-tagged (His-tagged) enzymes. Furthermore, immobilizing enzymes within COFs represents a growing field in heterogeneous biocatalysis. In this study, we have developed a flow-based technology utilizing a nickel-infused covalent organic framework (Ni-TpBpy COF) to combine two distinct processes: the purification of His-tagged enzymes and the immobilization of enzymes simultaneously. Our work primarily focuses on the purification of three His-tagged enzymes β-glucosidase, cellobiohydrolase, and endoglucanase as well as two proteins with varying molecular weights, namely, green fluorescent protein (27 kDa) and BG Rho (88 kDa). We employed Ni-TpBpy as a column matrix to showcase the versatility of our system. Additionally, we successfully obtained a Ni-TpBpy COF immobilized with enzymes, which can serve as a heterogeneous catalyst for the hydrolysis of p-nitrophenyl-β-d-glucopyranoside and carboxymethylcellulose. These immobilized enzymes demonstrated catalytic activity comparable to that of their free counterparts, with the added advantages of recyclability and enhanced stability under ambient conditions for an extended period, ranging from 60 to 90 days. This contrasts with the free enzymes, which do not maintain their activity as effectively over time.
Collapse
Affiliation(s)
- Satyadip Paul
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
- Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
| | - Mani Gupta
- Department of Biological Sciences and Center for the Climate and Environmental Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
| | - Ashok Kumar Mahato
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
- Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
| | - Shayan Karak
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
- Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
| | - Ananda Basak
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
- Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
| | - Supratim Datta
- Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
- Department of Biological Sciences and Center for the Climate and Environmental Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
| | - Rahul Banerjee
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
- Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
| |
Collapse
|
6
|
Kirichuk O, Srimasorn S, Zhang X, Roberts ARE, Coche-Guerente L, Kwok JCF, Bureau L, Débarre D, Richter RP. Competitive Specific Anchorage of Molecules onto Surfaces: Quantitative Control of Grafting Densities and Contamination by Free Anchors. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:18410-18423. [PMID: 38049433 PMCID: PMC10734310 DOI: 10.1021/acs.langmuir.3c02567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/30/2023] [Accepted: 11/20/2023] [Indexed: 12/06/2023]
Abstract
The formation of surfaces decorated with biomacromolecules such as proteins, glycans, or nucleic acids with well-controlled orientations and densities is of critical importance for the design of in vitro models, e.g., synthetic cell membranes and interaction assays. To this effect, ligand molecules are often functionalized with an anchor that specifically binds to a surface with a high density of binding sites, providing control over the presentation of the molecules. Here, we present a method to robustly and quantitatively control the surface density of one or several types of anchor-bearing molecules by tuning the relative concentrations of target molecules and free anchors in the incubation solution. We provide a theoretical background that relates incubation concentrations to the final surface density of the molecules of interest and present effective guidelines toward optimizing incubation conditions for the quantitative control of surface densities. Focusing on the biotin anchor, a commonly used anchor for interaction studies, as a salient example, we experimentally demonstrate surface density control over a wide range of densities and target molecule sizes. Conversely, we show how the method can be adapted to quality control the purity of end-grafted biopolymers such as biotinylated glycosaminoglycans by quantifying the amount of residual free biotin reactant in the sample solution.
Collapse
Affiliation(s)
- Oksana Kirichuk
- School
of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K.
- School
of Physics and Astronomy, Faculty of Engineering and Physical Sciences,
Astbury Centre for Structural Molecular Biology, and Bragg Centre
for Materials Research, University of Leeds, Leeds LS2 9JT, U.K.
- Université
Grenoble-Alpes, CNRS, LIPhy, 38000 Grenoble, France
| | - Sumitra Srimasorn
- School
of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K.
- School
of Physics and Astronomy, Faculty of Engineering and Physical Sciences,
Astbury Centre for Structural Molecular Biology, and Bragg Centre
for Materials Research, University of Leeds, Leeds LS2 9JT, U.K.
| | - Xiaoli Zhang
- School
of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K.
- School
of Physics and Astronomy, Faculty of Engineering and Physical Sciences,
Astbury Centre for Structural Molecular Biology, and Bragg Centre
for Materials Research, University of Leeds, Leeds LS2 9JT, U.K.
| | - Abigail R. E. Roberts
- School
of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K.
- School
of Physics and Astronomy, Faculty of Engineering and Physical Sciences,
Astbury Centre for Structural Molecular Biology, and Bragg Centre
for Materials Research, University of Leeds, Leeds LS2 9JT, U.K.
| | - Liliane Coche-Guerente
- Département
de Chimie Moléculaire, Université
Grenoble-Alpes, CNRS, 38000 Grenoble, France
| | - Jessica C. F. Kwok
- School
of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K.
- Institute
of Experimental Medicine, Czech Academy of Sciences, Vídeňská 1083, 142 00 Prague, Czech Republic
| | - Lionel Bureau
- Université
Grenoble-Alpes, CNRS, LIPhy, 38000 Grenoble, France
| | | | - Ralf P. Richter
- School
of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K.
- School
of Physics and Astronomy, Faculty of Engineering and Physical Sciences,
Astbury Centre for Structural Molecular Biology, and Bragg Centre
for Materials Research, University of Leeds, Leeds LS2 9JT, U.K.
| |
Collapse
|
7
|
Jan S, Fratzke AP, Felgner J, Hernandez-Davies JE, Liang L, Nakajima R, Jasinskas A, Supnet M, Jain A, Felgner PL, Davies DH, Gregory AE. Multivalent vaccines demonstrate immunogenicity and protect against Coxiella burnetii aerosol challenge. Front Immunol 2023; 14:1192821. [PMID: 37533862 PMCID: PMC10390735 DOI: 10.3389/fimmu.2023.1192821] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/16/2023] [Indexed: 08/04/2023] Open
Abstract
Vaccines are among the most cost-effective public health measures for controlling infectious diseases. Coxiella burnetii is the etiological agent of Q fever, a disease with a wide clinical spectrum that ranges from mild symptoms, such as fever and fatigue, to more severe disease, such as pneumonia and endocarditis. The formalin-inactivated whole-cell vaccine Q-VAX® contains hundreds of antigens and confers lifelong protection in humans, but prior sensitization from infection or vaccination can result in deleterious reactogenic responses to vaccination. Consequently, there is great interest in developing non-reactogenic alternatives based on adjuvanted recombinant proteins. In this study, we aimed to develop a multivalent vaccine that conferred protection with reduced reactogenicity. We hypothesized that a multivalent vaccine consisting of multiple antigens would be more immunogenic and protective than a monovalent vaccine owing to the large number of potential protective antigens in the C. burnetii proteome. To address this, we identified immunogenic T and B cell antigens, and selected proteins were purified to evaluate with a combination adjuvant (IVAX-1), with or without C. burnetii lipopolysaccharide (LPS) in immunogenicity studies in vivo in mice and in a Hartley guinea pig intratracheal aerosol challenge model using C. burnetii strain NMI RSA 493. The data showed that multivalent vaccines are more immunogenic than monovalent vaccines and more closely emulate the protection achieved by Q-VAX. Although six antigens were the most immunogenic, we also discovered that multiplexing beyond four antigens introduces detectable reactogenicity, indicating that there is an upper limit to the number of antigens that can be safely included in a multivalent Q-fever vaccine. C. burnetii LPS also demonstrates efficacy as a vaccine antigen in conferring protection in an otherwise monovalent vaccine formulation, suggesting that its addition in multivalent vaccines, as demonstrated by a quadrivalent formulation, would improve protective responses.
Collapse
Affiliation(s)
- Sharon Jan
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Alycia P. Fratzke
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX, United States
- Department of Pathology, Charles River Laboratories, Reno, NV, United States
| | - Jiin Felgner
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Jenny E. Hernandez-Davies
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Li Liang
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Rie Nakajima
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Algimantas Jasinskas
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Medalyn Supnet
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Aarti Jain
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Philip L. Felgner
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - D. Huw Davies
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Anthony E. Gregory
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
8
|
Zheng Y, Wegner T, Di Iorio D, Pierau M, Glorius F, Wegner SV. NTA-Cholesterol Analogue for the Nongenetic Liquid-Ordered Phase-Specific Functionalization of Lipid Membranes with Proteins. ACS Chem Biol 2023; 18:1435-1443. [PMID: 37184283 DOI: 10.1021/acschembio.3c00180] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The nongenetic modification of cell membranes with proteins is a straightforward way of cellular engineering. In these processes, it is important to specifically address the proteins to liquid-ordered (Lo) or liquid-disordered (Ld) domains as this can largely affect their biological functions. Herein, we report a cholesterol analogue (CHIM) with a nitrilotriacetic acid (NTA) headgroup, named CHIM-NTA. CHIM-NTA integrates into lipid membranes similar to the widely used phospholipid-derived DGS-NTA and, when loaded with Ni2+, allows for specific membrane immobilization of any polyhistidine-tagged proteins of choice. Yet, unlike DGS-NTA, it localizes to the Lo phase in phase-separated giant unilamellar vesicles (GUVs) and allows addressing His-tagged proteins to Lo domains. Furthermore, CHIM-NTA readily integrates into the membranes of live cells and thus enables the nongenetic modification of the cell surface with proteins. Overall, CHIM-NTA provides a facile and flexible way to modify biological membranes, in particular Lo domains, with His-tagged proteins and can serve as a broadly applicable molecular tool for cell surface engineering.
Collapse
Affiliation(s)
- Yanjun Zheng
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Münster 48149, Germany
| | - Tristan Wegner
- University of Münster, Institute of Organic Chemistry, Münster 48149, Germany
| | - Daniele Di Iorio
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Münster 48149, Germany
| | - Marco Pierau
- University of Münster, Institute of Organic Chemistry, Münster 48149, Germany
| | - Frank Glorius
- University of Münster, Institute of Organic Chemistry, Münster 48149, Germany
| | - Seraphine V Wegner
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Münster 48149, Germany
| |
Collapse
|
9
|
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: 8.0] [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
|
10
|
Steiert F, Schultz P, Höfinger S, Müller TD, Schwille P, Weidemann T. Insights into receptor structure and dynamics at the surface of living cells. Nat Commun 2023; 14:1596. [PMID: 36949079 PMCID: PMC10033668 DOI: 10.1038/s41467-023-37284-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 03/10/2023] [Indexed: 03/24/2023] Open
Abstract
Evaluating protein structures in living cells remains a challenge. Here, we investigate Interleukin-4 receptor alpha (IL-4Rα) into which the non-canonical amino acid bicyclo[6.1.0]nonyne-lysine (BCNK) is incorporated by genetic code expansion. Bioorthogonal click labeling is performed with tetrazine-conjugated dyes. To quantify the reaction yield in situ, we develop brightness-calibrated ratiometric imaging, a protocol where fluorescent signals in confocal multi-color images are ascribed to local concentrations. Screening receptor mutants bearing BCNK in the extracellular domain uncovered site-specific variations of both click efficiency and Interleukin-4 binding affinity, indicating subtle well-defined structural perturbations. Molecular dynamics and continuum electrostatics calculations suggest solvent polarization to determine site-specific variations of BCNK reactivity. Strikingly, signatures of differential click efficiency, measured for IL-4Rα in ligand-bound and free form, mirror sub-angstrom deformations of the protein backbone at corresponding locations. Thus, click efficiency by itself represents a remarkably informative readout linked to protein structure and dynamics in the native plasma membrane.
Collapse
Affiliation(s)
- Frederik Steiert
- Department of Cellular and Molecular Biophysics, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152, Martinsried, Germany
- Department of Physics, Technical University Munich, 85748, Garching, Germany
| | - Peter Schultz
- Department of Cellular and Molecular Biophysics, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152, Martinsried, Germany
| | - Siegfried Höfinger
- VSC Research Center, TU Wien, Operngasse 11 / E057-09, 1040, Wien, Austria
- Department of Physics, Michigan Technological University, 1400 Townsend Drive, 49931, Houghton, MI, USA
| | - Thomas D Müller
- Biozentrum, Julius-von-Sachs-Institut für Biowissenschaften, Lehrstuhl für Molekulare Pflanzenphysiologie und Biophysik - Botanik I, Julius-von-Sachs-Platz 2, 97082, Würzburg, Germany
| | - Petra Schwille
- Department of Cellular and Molecular Biophysics, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152, Martinsried, Germany
| | - Thomas Weidemann
- Department of Cellular and Molecular Biophysics, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152, Martinsried, Germany.
| |
Collapse
|
11
|
Thimaradka V, Utsunomiya H, Tamura T, Hamachi I. Endogenous Cell-Surface Receptor Modification by Metal Chelation-Assisted Pyridinium Oxime Catalyst. Org Lett 2023; 25:2118-2122. [PMID: 36947590 DOI: 10.1021/acs.orglett.3c00541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Organocatalyst-mediated acyl transfer reactions hold promise for selective protein labeling in biological milieu. However, they often suffer from off-target reactions and high background signals because of the requirement of high concentrations of substrates. Here, we report a new catalytic protein acylation strategy promoted by the His-tag/NiNTA interaction. The recognition-assisted activation mechanism allows efficient protein labeling even with >10-fold lower substrate concentrations than conventional reactions, thereby enabling highly selective and efficient cell-surface receptor modification in live cells.
Collapse
Affiliation(s)
- Vikram Thimaradka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Hayata Utsunomiya
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Tomonori Tamura
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
- ERATO (Exploratory Research for Advanced Technology, JST), Sanbancho, Chiyodaku, Tokyo 102-0075, Japan
| |
Collapse
|
12
|
Badten A, Ramirez A, Hernandez-Davies JE, Albin TJ, Jain A, Nakajima R, Felgner J, Davies DH, Wang SW. Protein Nanoparticle-Mediated Delivery of Recombinant Influenza Hemagglutinin Enhances Immunogenicity and Breadth of the Antibody Response. ACS Infect Dis 2023; 9:239-252. [PMID: 36607269 PMCID: PMC9926493 DOI: 10.1021/acsinfecdis.2c00362] [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: 07/08/2022] [Indexed: 01/07/2023]
Abstract
The vast majority of seasonal influenza vaccines administered each year are derived from virus propagated in eggs using technology that has changed little since the 1930s. The immunogenicity, durability, and breadth of response would likely benefit from a recombinant nanoparticle-based approach. Although the E2 protein nanoparticle (NP) platform has been previously shown to promote effective cell-mediated responses to peptide epitopes, it has not yet been reported to deliver whole protein antigens. In this study, we synthesized a novel maleimido tris-nitrilotriacetic acid (NTA) linker to couple protein hemagglutinin (HA) from H1N1 influenza virus to the E2 NP, and we evaluated the HA-specific antibody responses using protein microarrays. We found that recombinant H1 protein alone is immunogenic in mice but requires two boosts for IgG to be detected and is strongly IgG1 (Th2) polarized. When conjugated to E2 NPs, IgG2c is produced leading to a more balanced Th1/Th2 response. Inclusion of the Toll-like receptor 4 agonist monophosphoryl lipid A (MPLA) significantly enhances the immunogenicity of H1-E2 NPs while retaining the Th1/Th2 balance. Interestingly, broader homo- and heterosubtypic cross-reactivity is also observed for conjugated H1-E2 with MPLA, compared to unconjugated H1 with or without MPLA. These results highlight the potential of an NP-based delivery of HA for tuning the immunogenicity, breadth, and Th1/Th2 balance generated by recombinant HA-based vaccination. Furthermore, the modularity of this protein-protein conjugation strategy may have utility for future vaccine development against other human pathogens.
Collapse
Affiliation(s)
- Alexander
J. Badten
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Aaron Ramirez
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Jenny E. Hernandez-Davies
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Tyler J. Albin
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Aarti Jain
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Rie Nakajima
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Jiin Felgner
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - D. Huw Davies
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Szu-Wen Wang
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| |
Collapse
|
13
|
Shen J, Dai Y, Xia F, Zhang X. Role of divalent metal ions in the function and application of hydrogels. Prog Polym Sci 2022. [DOI: 10.1016/j.progpolymsci.2022.101622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
14
|
Kolkman R, Michel-Souzy S, Wasserberg D, Segerink LI, Huskens J. Density Control over MBD2 Receptor-Coated Surfaces Provides Superselective Binding of Hypermethylated DNA. ACS APPLIED MATERIALS & INTERFACES 2022; 14:40579-40589. [PMID: 36052432 PMCID: PMC9478954 DOI: 10.1021/acsami.2c09641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/26/2022] [Indexed: 06/15/2023]
Abstract
Using the biomarker hypermethylated DNA (hmDNA) for cancer detection requires a pretreatment to isolate or concentrate hmDNA from nonmethylated DNA. Affinity chromatography using a methyl binding domain-2 (MBD2) protein can be used, but the relatively low enrichment selectivity of MBD2 limits its clinical applicability. Here, we developed a superselective, multivalent, MBD2-coated platform to improve the selectivity of hmDNA enrichment. The multivalent platform employs control over the MBD2 surface receptor density, which is shown to strongly affect the binding of DNA with varying degrees of methylation, improving both the selectivity and the affinity of DNAs with higher numbers of methylation sites. Histidine-10-tagged MBD2 was immobilized on gold surfaces with receptor density control by tuning the amount of nickel nitrilotriacetic acid (NiNTA)-functionalized thiols in a thiol-based self-assembled monolayer. The required MBD2 surface receptor densities for DNA surface binding decreases for DNA with higher degrees of methylation. Both higher degrees of superselectivity and surface coverages were observed upon DNA binding at increasing methylation levels. Adopting the findings of this study into hmDNA enrichment of clinical samples has the potential to become more selective and sensitive than current MBD2-based methods and, therefore, to improve cancer diagnostics.
Collapse
Affiliation(s)
- Ruben
W. Kolkman
- Molecular
Nanofabrication Group, Department for Molecules & Materials, MESA+
Institute, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
- BIOS
Lab on a Chip Group, MESA+ Institute and TechMed Centre, Max Planck
Institute for Complex Fluid Dynamics, Faculty of Electrical Engineering,
Mathematics and Computer Science, University
of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Sandra Michel-Souzy
- Biomolecular
Nanotechnology Group, Department for Molecules & Materials, MESA+
Institute, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Dorothee Wasserberg
- BIOS
Lab on a Chip Group, MESA+ Institute and TechMed Centre, Max Planck
Institute for Complex Fluid Dynamics, Faculty of Electrical Engineering,
Mathematics and Computer Science, University
of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Loes I. Segerink
- BIOS
Lab on a Chip Group, MESA+ Institute and TechMed Centre, Max Planck
Institute for Complex Fluid Dynamics, Faculty of Electrical Engineering,
Mathematics and Computer Science, University
of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Jurriaan Huskens
- Molecular
Nanofabrication Group, Department for Molecules & Materials, MESA+
Institute, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| |
Collapse
|
15
|
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: 4.0] [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
|
16
|
Lou J, Mooney DJ. Chemical strategies to engineer hydrogels for cell culture. Nat Rev Chem 2022; 6:726-744. [PMID: 37117490 DOI: 10.1038/s41570-022-00420-7] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2022] [Indexed: 12/12/2022]
Abstract
Two-dimensional and three-dimensional cell culture systems are widely used for biological studies, and are the basis of the organoid, tissue engineering and organ-on-chip research fields in applications such as disease modelling and drug screening. The natural extracellular matrix of tissues, a complex scaffold with varying chemical and mechanical properties, has a critical role in regulating important cellular functions such as spreading, migration, proliferation and differentiation, as well as tissue morphogenesis. Hydrogels are biomaterials that are used in cell culture systems to imitate critical features of a natural extracellular matrix. Chemical strategies to synthesize and tailor the properties of these hydrogels in a controlled manner, and manipulate their biological functions in situ, have been developed. In this Review, we provide the rational design criteria for predictably engineering hydrogels to mimic the properties of the natural extracellular matrix. We highlight the advances in using biocompatible strategies to engineer hydrogels for cell culture along with recent developments to dynamically control the cellular environment by exploiting stimuli-responsive chemistries. Finally, future opportunities to engineer hydrogels are discussed, in which the development of novel chemical methods will probably have an important role.
Collapse
|
17
|
Udupa S, Nagaraja V, Karambelkar S. Binding Affinity Quantifications of the Bacteriophage Mu DNA Modification Protein Mom Using Microscale Thermophoresis (MST). Bio Protoc 2022; 12:e4472. [PMID: 35978573 PMCID: PMC9350919 DOI: 10.21769/bioprotoc.4472] [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: 11/01/2021] [Revised: 05/15/2022] [Accepted: 06/11/2022] [Indexed: 12/29/2022] Open
Abstract
Epigenetic modifications play diverse roles in biological systems. Nucleic acid modifications control gene expression, protein synthesis, and sensitivity to nucleic acid-cleaving enzymes. However, the mechanisms underlying the biosynthesis of nucleic acid modifications can be challenging to identify. Studying protein-ligand interactions helps decipher biosynthetic and regulatory pathways underlying biological reactions. Here, we describe a fluorescence labeling-based quantitative method for unraveling the biomolecular interactions of bacteriophage Mu DNA modification protein Mom with its ligands, using microscale thermophoresis (MST). Compared to traditional methods for studying protein-biomolecular interactions, MST requires significantly lower sample amounts, volumes, and analysis time, thus allowing screening of a large number of candidates for interaction with a protein of interest. Another distinguishing feature of the method is that it obviates the need for protein purification, often a time- and resource-consuming step, and works well with whole or partially purified cell extracts. Importantly, the method is sensitive over a broad range of molecular affinities while offering great specificity and can be used to interrogate ligands ranging from metal ions to macromolecules. Although we established this method for a DNA modification protein, it can easily be adapted to study a variety of molecular interactions engaged by proteins.
Collapse
Affiliation(s)
- Shubha Udupa
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Valakunja Nagaraja
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
,
Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
,
*For correspondence:
;
| | - Shweta Karambelkar
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
,
Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
,
*For correspondence:
;
| |
Collapse
|
18
|
Xiong X, Blakely A, Kim JH, Menting JG, Schäfer IB, Schubert HL, Agrawal R, Gutmann T, Delaine C, Zhang YW, Artik GO, Merriman A, Eckert D, Lawrence MC, Coskun Ü, Fisher SJ, Forbes BE, Safavi-Hemami H, Hill CP, Chou DHC. Symmetric and asymmetric receptor conformation continuum induced by a new insulin. Nat Chem Biol 2022; 18:511-519. [PMID: 35289328 PMCID: PMC9248236 DOI: 10.1038/s41589-022-00981-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 01/24/2022] [Indexed: 02/02/2023]
Abstract
Cone snail venoms contain a wide variety of bioactive peptides, including insulin-like molecules with distinct structural features, binding modes and biochemical properties. Here, we report an active humanized cone snail venom insulin with an elongated A chain and a truncated B chain, and use cryo-electron microscopy (cryo-EM) and protein engineering to elucidate its interactions with the human insulin receptor (IR) ectodomain. We reveal how an extended A chain can compensate for deletion of B-chain residues, which are essential for activity of human insulin but also compromise therapeutic utility by delaying dissolution from the site of subcutaneous injection. This finding suggests approaches to developing improved therapeutic insulins. Curiously, the receptor displays a continuum of conformations from the symmetric state to a highly asymmetric low-abundance structure that displays coordination of a single humanized venom insulin using elements from both of the previously characterized site 1 and site 2 interactions.
Collapse
Affiliation(s)
- Xiaochun Xiong
- Department of Pediatrics, Division of Endocrinology and Diabetes, Stanford University, Stanford, CA, USA.,Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Alan Blakely
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Jin Hwan Kim
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - John G Menting
- WEHI, Parkville, Victoria, Australia.,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Ingmar B Schäfer
- Department of Structural Cell Biology, Max Planck Institute of Biochemistry, Munich, Germany
| | - Heidi L Schubert
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Rahul Agrawal
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Theresia Gutmann
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Carlie Delaine
- Discipline of Medical Biochemistry and Cell Biology, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Yi Wolf Zhang
- Department of Pediatrics, Division of Endocrinology and Diabetes, Stanford University, Stanford, CA, USA.,Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Gizem Olay Artik
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Membrane Biochemistry and Lipid Research, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Allanah Merriman
- Discipline of Medical Biochemistry and Cell Biology, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Debbie Eckert
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Michael C Lawrence
- WEHI, Parkville, Victoria, Australia.,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Ünal Coskun
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Membrane Biochemistry and Lipid Research, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Simon J Fisher
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA.,Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
| | - Briony E Forbes
- Discipline of Medical Biochemistry and Cell Biology, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Helena Safavi-Hemami
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA. .,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Christopher P Hill
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA.
| | - Danny Hung-Chieh Chou
- Department of Pediatrics, Division of Endocrinology and Diabetes, Stanford University, Stanford, CA, USA. .,Department of Biochemistry, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
19
|
|
20
|
Green CM, Mathur D, Susumu K, Oh E, Medintz IL, Díaz SA. Polyhistidine-Tag-Enabled Conjugation of Quantum Dots and Enzymes to DNA Nanostructures. Methods Mol Biol 2022; 2525:61-91. [PMID: 35836061 DOI: 10.1007/978-1-0716-2473-9_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
DNA nanostructures self-assemble into almost any arbitrary architecture, and when combined with their capability to precisely position and orient dyes, nanoparticles, and biological moieties, the technology reaches its potential. We present a simple yet multifaceted conjugation strategy based on metal coordination by a multi-histidine peptide tag (Histag). The versatility of the Histag as a means to conjugate to DNA nanostructures is shown by using Histags to capture semiconductor quantum dots (QDs) with numerical and positional precision onto a DNA origami breadboard. Additionally, Histag-expressing enzymes, such as the bioluminescent luciferase, can also be captured to the DNA origami breadboard with similar precision. DNA nanostructure conjugation of the QDs or luciferase is confirmed through imaging and/or energy transfer to organic dyes integrated into the DNA nanostructure.
Collapse
Affiliation(s)
- Christopher M Green
- Center for Bio/Molecular Science and Engineering, U.S. Naval Research Laboratory Code 6900, Washington, DC, USA
- National Research Council, Washington, DC, USA
| | - Divita Mathur
- Center for Bio/Molecular Science and Engineering, U.S. Naval Research Laboratory Code 6900, Washington, DC, USA
- College of Science, George Mason University, Fairfax, VA, USA
| | - Kimihiro Susumu
- Optical Sciences Division, Code 5600, U.S. Naval Research Laboratory, Washington, DC, USA
- Jacobs Corporation, Hanover, MD, USA
| | - Eunkeu Oh
- Optical Sciences Division, Code 5600, U.S. Naval Research Laboratory, Washington, DC, USA
| | - Igor L Medintz
- Center for Bio/Molecular Science and Engineering, U.S. Naval Research Laboratory Code 6900, Washington, DC, USA
| | - Sebastián A Díaz
- Center for Bio/Molecular Science and Engineering, U.S. Naval Research Laboratory Code 6900, Washington, DC, USA.
| |
Collapse
|
21
|
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: 34] [Impact Index Per Article: 11.3] [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
|
22
|
Liu L, Han C, Jiang M, Zhang T, Kang Q, Wang X, Wang P, Zhou F. Rapid and regenerable surface plasmon resonance determinations of biomarker concentration and biomolecular interaction based on tris-nitrilotriacetic acid chips. Anal Chim Acta 2021; 1170:338625. [PMID: 34090589 DOI: 10.1016/j.aca.2021.338625] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/01/2021] [Accepted: 05/04/2021] [Indexed: 12/22/2022]
Abstract
The tris-nitrilotriacetic acid (tris-NTA) chip has been used for surface plasmon resonance (SPR) kinetic studies involving histidine (His)-tagged proteins. However, its full potential, especially for analyte quantification in complex biological media, has not been realized due to a lack of systematic studies on the factors governing ligand immobilization, surface regeneration, and data analysis. We demonstrate that the tris-NTA chip not only retains His-tagged proteins more strongly than its mono-NTA counterpart, but also orients them more uniformly than protein molecules coupled to carboxymethylated dextran films. We accurately and rapidly quantified immunoglobulin (IgG) molecules in sera by using the initial association phase of their conjugation with His-tagged protein G densely immobilized onto the tris-NTA chip, and established criteria for selecting the optimal time for constructing the calibration curve. The method is highly reproducible (less than 2% RSD) and three orders of magnitude more sensitive than immunoturbidimetry. In addition, we found that the amount of His-protein immobilized is highly dependent on the protein isoelectric point (pI). Reliable kinetic data in a multi-channel SPR instrument can also be rapidly obtained by using a low density of immobilized His-tagged protein. The experimental parameters and procedures outlined in this study help expand the range of SPR applications involving His-tagged proteins.
Collapse
Affiliation(s)
- Luyao Liu
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, 250022, PR China
| | - Chaowei Han
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, 250022, PR China
| | - Meng Jiang
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, 250022, PR China
| | - Tiantian Zhang
- University Hospital, University of Jinan, Jinan, Shandong, 250022, PR China
| | - Qing Kang
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, 250022, PR China
| | - Xiaoying Wang
- State Key Laboratory of Biobased Materials and Green Papermaking, Qilu University of Technology, Jinan, Shandong, 250353, PR China
| | - Pengcheng Wang
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, 250022, PR China.
| | - Feimeng Zhou
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, 250022, PR China.
| |
Collapse
|
23
|
Mitscha-Baude G, Stadlbauer B, Howorka S, Heitzinger C. Protein Transport through Nanopores Illuminated by Long-Time-Scale Simulations. ACS NANO 2021; 15:9900-9912. [PMID: 34096722 PMCID: PMC8291773 DOI: 10.1021/acsnano.1c01078] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/28/2021] [Indexed: 06/12/2023]
Abstract
The transport of molecules through nanoscale confined space is relevant in biology, biosensing, and industrial filtration. Microscopically modeling transport through nanopores is required for a fundamental understanding and guiding engineering, but the short duration and low replica number of existing simulation approaches limit statistically relevant insight. Here we explore protein transport in nanopores with a high-throughput computational method that realistically simulates hundreds of up to seconds-long protein trajectories by combining Brownian dynamics and continuum simulation and integrating both driving forces of electroosmosis and electrophoresis. Ionic current traces are computed to enable experimental comparison. By examining three biological and synthetic nanopores, our study answers questions about the kinetics and mechanism of protein transport and additionally reveals insight that is inaccessible from experiments yet relevant for pore design. The discovery of extremely frequent unhindered passage can guide the improvement of biosensor pores to enhance desired biomolecular recognition by pore-tethered receptors. Similarly, experimentally invisible nontarget adsorption to pore walls highlights how to improve recently developed DNA nanopores. Our work can be expanded to pressure-driven flow to model industrial nanofiltration processes.
Collapse
Affiliation(s)
| | - Benjamin Stadlbauer
- Institute
of Analysis and Scientific Computing, TU
Wien, Vienna, 1040, Austria
| | - Stefan Howorka
- Department
of Chemistry, Institute of Structural Molecular Biology, University College London, London, WC1E 6BT, United Kingdom
- Institute
of Biophysics, Johannes Kepler University
Linz, Linz, 4020, Austria
| | - Clemens Heitzinger
- Institute
of Analysis and Scientific Computing, TU
Wien, Vienna, 1040, Austria
- School
of Mathematical and Statistical Sciences, Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|
24
|
Wolf P, Gavins G, Beck‐Sickinger AG, Seitz O. Strategies for Site-Specific Labeling of Receptor Proteins on the Surfaces of Living Cells by Using Genetically Encoded Peptide Tags. Chembiochem 2021; 22:1717-1732. [PMID: 33428317 PMCID: PMC8248378 DOI: 10.1002/cbic.202000797] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/08/2021] [Indexed: 12/14/2022]
Abstract
Fluorescence microscopy imaging enables receptor proteins to be investigated within their biological context. A key challenge is to site-specifically incorporate reporter moieties into proteins without interfering with biological functions or cellular networks. Small peptide tags offer the opportunity to combine inducible labeling with small tag sizes that avoid receptor perturbation. Herein, we review the current state of live-cell labeling of peptide-tagged cell-surface proteins. Considering their importance as targets in medicinal chemistry, we focus on membrane receptors such as G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs). We discuss peptide tags that i) are subject to enzyme-mediated modification reactions, ii) guide the complementation of reporter proteins, iii) form coiled-coil complexes, and iv) interact with metal complexes. Given our own contributions in the field, we place emphasis on peptide-templated labeling chemistry.
Collapse
Affiliation(s)
- Philipp Wolf
- Faculty of Life SciencesInstitute of BiochemistryLeipzig UniversityBrüderstrasse 3404103LeipzigGermany
| | - Georgina Gavins
- Faculty of Mathematics and Natural SciencesDepartment of ChemistryHumboldt-Universität zu BerlinBrook-Taylor-Str. 212489BerlinGermany
| | - Annette G. Beck‐Sickinger
- Faculty of Life SciencesInstitute of BiochemistryLeipzig UniversityBrüderstrasse 3404103LeipzigGermany
| | - Oliver Seitz
- Faculty of Mathematics and Natural SciencesDepartment of ChemistryHumboldt-Universität zu BerlinBrook-Taylor-Str. 212489BerlinGermany
| |
Collapse
|
25
|
Diederichs T, Tampé R. Single Cell-like Systems Reveal Active Unidirectional and Light-Controlled Transport by Nanomachineries. ACS NANO 2021; 15:6747-6755. [PMID: 33724767 PMCID: PMC8157534 DOI: 10.1021/acsnano.0c10139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Cellular life depends on transport and communication across membranes, which is emphasized by the fact that membrane proteins are prime drug targets. The cell-like environment of membrane proteins has gained increasing attention based on its important role in function and regulation. As a versatile scaffold for bottom-up synthetic biology and nanoscience, giant liposomes represent minimalistic models of living cells. Nevertheless, the incorporation of fragile multiprotein membrane complexes still remains a major challenge. Here, we report on an approach for the functional reconstitution of membrane assemblies exemplified by human and bacterial ATP-binding cassette (ABC) transporters. We reveal that these nanomachineries transport substrates unidirectionally against a steep concentration gradient. Active substrate transport can be spatiotemporally resolved in single cell-like compartments by light, enabling real-time tracking of substrate export and import in individual liposomes. This approach will help to construct delicate artificial cell-like systems.
Collapse
Affiliation(s)
- Tim Diederichs
- Institute of Biochemistry, Biocenter,
Goethe-University Frankfurt, Max-von Laue-Straße 9,
60438 Frankfurt a.M., Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter,
Goethe-University Frankfurt, Max-von Laue-Straße 9,
60438 Frankfurt a.M., Germany
| |
Collapse
|
26
|
Sánchez MF, Els-Heindl S, Beck-Sickinger AG, Wieneke R, Tampé R. Photoinduced receptor confinement drives ligand-independent GPCR signaling. Science 2021; 371:science.abb7657. [PMID: 33632896 DOI: 10.1126/science.abb7657] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 01/26/2021] [Indexed: 12/13/2022]
Abstract
Cell-cell communication relies on the assembly of receptor-ligand complexes at the plasma membrane. The spatiotemporal receptor organization has a pivotal role in evoking cellular responses. We studied the clustering of heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) and established a photoinstructive matrix with ultrasmall lock-and-key interaction pairs to control lateral membrane organization of hormone neuropeptide Y2 receptors in living cells by light. Within seconds, receptor clustering was modulated in size, location, and density. After in situ confinement, changes in cellular morphology, motility, and calcium signaling revealed ligand-independent receptor activation. This approach may enhance the exploration of mechanisms in cell signaling and mechanotransduction.
Collapse
Affiliation(s)
- M Florencia Sánchez
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Sylvia Els-Heindl
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, 04109 Leipzig, Germany
| | | | - Ralph Wieneke
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
27
|
Adler S, Motiei L, Mankovski N, Cohen H, Margulies D. Fluorescent Labelling of Cell Surface Proteins on a Solid Support. Isr J Chem 2021. [DOI: 10.1002/ijch.202100028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Sean Adler
- Department of Chemical and Structural Biology Weizmann Institute of Science Rehovot 7610001 Israel
| | - Leila Motiei
- Department of Chemical and Structural Biology Weizmann Institute of Science Rehovot 7610001 Israel
| | - Naama Mankovski
- Department of Chemical and Structural Biology Weizmann Institute of Science Rehovot 7610001 Israel
| | - Hagai Cohen
- Department of Chemical Research Support Weizmann Institute of Science Rehovot 7610001 Israel
| | - David Margulies
- Department of Chemical and Structural Biology Weizmann Institute of Science Rehovot 7610001 Israel
| |
Collapse
|
28
|
Hatai J, Prasad PK, Lahav-Mankovski N, Oppenheimer-Low N, Unger T, Sirkis YF, Dadosh T, Motiei L, Margulies D. Assessing changes in the expression levels of cell surface proteins with a turn-on fluorescent molecular probe. Chem Commun (Camb) 2021; 57:1875-1878. [PMID: 33427257 DOI: 10.1039/d0cc07095e] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tri-nitrilotriacetic acid (NTA)-based fluorescent probes were developed and used to image His-tagged-labelled outer membrane protein C (His-OmpC) in live Escherichia coli. One of these probes was designed to light up upon binding, which provided the means to assess changes in the His-OmpC expression levels by taking a simple fluorescence spectrum.
Collapse
Affiliation(s)
- Joydev Hatai
- Department of Organic Chemistry, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Füllbrunn N, Li Z, Jorde L, Richter CP, Kurre R, Langemeyer L, Yu C, Meyer C, Enderlein J, Ungermann C, Piehler J, You C. Nanoscopic anatomy of dynamic multi-protein complexes at membranes resolved by graphene-induced energy transfer. eLife 2021; 10:62501. [PMID: 33513092 PMCID: PMC7847308 DOI: 10.7554/elife.62501] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/28/2020] [Indexed: 11/30/2022] Open
Abstract
Insights into the conformational organization and dynamics of proteins complexes at membranes is essential for our mechanistic understanding of numerous key biological processes. Here, we introduce graphene-induced energy transfer (GIET) to probe axial orientation of arrested macromolecules at lipid monolayers. Based on a calibrated distance-dependent efficiency within a dynamic range of 25 nm, we analyzed the conformational organization of proteins and complexes involved in tethering and fusion at the lysosome-like yeast vacuole. We observed that the membrane-anchored Rab7-like GTPase Ypt7 shows conformational reorganization upon interactions with effector proteins. Ensemble and time-resolved single-molecule GIET experiments revealed that the HOPS tethering complex, when recruited via Ypt7 to membranes, is dynamically alternating between a ‘closed’ and an ‘open’ conformation, with the latter possibly interacting with incoming vesicles. Our work highlights GIET as a unique spectroscopic ruler to reveal the axial orientation and dynamics of macromolecular complexes at biological membranes with sub-nanometer resolution. Proteins are part of the building blocks of life and are essential for structure, function and regulation of every cell, tissue and organ of the body. Proteins adopt different conformations to work efficiently within the various environments of a cell. They can also switch between shapes. One way to monitor how proteins change their shapes involves energy transfer. This approach can measure how close two proteins, or two parts of the same protein, are, by using dye labels that respond to each other when they are close together. For example, in a method called FRET, one dye label absorbs light and transfers the energy to the other label, which emits it as a different color of light. However, FRET only works over short distances (less than 10nm apart or 1/100,000th of a millimeter), so it is not useful for larger proteins. Here, Füllbrunn, Li et al. developed a method called GIET that uses graphene to analyze the dynamic structures of proteins on membrane surfaces. Graphene is a type of carbon nanomaterial that can absorb energy from dye labels and could provide a way to study protein interactions over longer distances. Graphene was deposited on a glass surface where it was coated with single layer of membrane, which could then be used to capture specific proteins. The results showed that GIET worked over longer distances (up to 30 nm) than FRET and could be used to study proteins attached to the membrane around graphene. Füllbrunn, Li et al. used it to examine a specific complex of proteins called HOPS, which is linked to multiple diseases, including Ebola, measuring distances between the head or tail of HOPS and the membrane to understand protein shapes. This revealed that HOPS adopts an upright position on membranes and alternates between open and closed shapes. The study of Füllbrunn, Li et al. highlights the ability of GIET to address unanswered questions about the function of protein complexes on membrane surfaces and sheds new light on the structural dynamics of HOPS in living cells. As it allows protein interactions to be studied over much greater distances, GIET could be a powerful new tool for cell biology research. Moreover, graphene is also useful in electron microscopy and both approaches combined could achieve a detailed structural picture of proteins in action.
Collapse
Affiliation(s)
- Nadia Füllbrunn
- Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany.,Center of Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
| | - Zehao Li
- Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany.,Center of Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany.,College of Life Sciences, Beijing University of Chemical Technology, Beijing, China
| | - Lara Jorde
- Department of Physics, University of Osnabrück, Osnabrück, Germany
| | - Christian P Richter
- Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Rainer Kurre
- Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany.,Center of Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
| | - Lars Langemeyer
- Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Changyuan Yu
- College of Life Sciences, Beijing University of Chemical Technology, Beijing, China
| | - Carola Meyer
- Center of Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany.,Department of Physics, University of Osnabrück, Osnabrück, Germany
| | - Jörg Enderlein
- 3rd Institute of Physics - Biophysics, Georg August University, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), Georg August University, Göttingen, Germany
| | - Christian Ungermann
- Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany.,Center of Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
| | - Jacob Piehler
- Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany.,Center of Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
| | - Changjiang You
- Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany.,Center of Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
| |
Collapse
|
30
|
Thimaradka V, Hoon Oh J, Heroven C, Radu Aricescu A, Yuzaki M, Tamura T, Hamachi I. Site-specific covalent labeling of His-tag fused proteins with N-acyl-N-alkyl sulfonamide reagent. Bioorg Med Chem 2021; 30:115947. [PMID: 33360195 PMCID: PMC7610637 DOI: 10.1016/j.bmc.2020.115947] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 01/16/2023]
Abstract
The ability to incorporate a desired functionality into proteins of interest in a site-specific manner can provide powerful tools for investigating biological systems and creating therapeutic conjugates. However, there are not any universal methods that can be applied to all proteins, and it is thus important to explore the chemical strategy for protein modification. In this paper, we developed a new reactive peptide tag/probe pair system for site-specific covalent protein labeling. This method relies on the recognition-driven reaction of a peptide tag and a molecular probe, which comprises the lysine-containing short histidine tag (KH6 or H6K) and a binuclear nickel (II)- nitrilotriacetic acid (Ni2+-NTA) complex probe containing a lysine-reactive N-acyl-N-alkyl sulfonamide (NASA) group. The selective interaction of the His-tag and Ni2+-NTA propeles a rapid nucleophilic reaction between a lysine residue of the tag and the electrophilic NASA group of the probe by the proximity effect, resulting in the tag-site-specific functionalization of proteins. We characterized the reactive profile and site-specificity of this method using model peptides and proteins in vitro, and demonstrated the general utility for production of a nanobody-chemical probe conjugate without compromising its binding ability.
Collapse
Affiliation(s)
- Vikram Thimaradka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Jae Hoon Oh
- ERATO (Exploratory Research for Advanced Technology, JST), Sanbancho, Chiyodaku, Tokyo, 102-0075, Japan
| | - Christina Heroven
- Division of Structural Biology, University of Oxford, Oxford OX3 7BN, UK; Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - A Radu Aricescu
- Division of Structural Biology, University of Oxford, Oxford OX3 7BN, UK; Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Tomonori Tamura
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan.
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan; ERATO (Exploratory Research for Advanced Technology, JST), Sanbancho, Chiyodaku, Tokyo, 102-0075, Japan.
| |
Collapse
|
31
|
Prasad PK, Motiei L, Margulies D. Steps toward enhancing the fluorescence of small-molecule-based protein labels using supramolecular hosts. RESULTS IN CHEMISTRY 2021. [DOI: 10.1016/j.rechem.2021.100134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
32
|
Napiorkowska M, Pestalozzi L, Panke S, Held M, Schmitt S. High-Throughput Optimization of Recombinant Protein Production in Microfluidic Gel Beads. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2005523. [PMID: 33325637 DOI: 10.1002/smll.202005523] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 10/31/2020] [Indexed: 06/12/2023]
Abstract
Efficient production hosts are a key requirement for bringing biopharmaceutical and biotechnological innovations to the market. In this work, a truly universal high-throughput platform for optimization of microbial protein production is described. Using droplet microfluidics, large genetic libraries of strains are encapsulated into biocompatible gel beads that are engineered to selectively retain any protein of interest. Bead-retained products are then fluorescently labeled and strains with superior production titers are isolated using flow cytometry. The broad applicability of the platform is demonstrated by successfully culturing several industrially relevant bacterial and yeast strains and detecting peptides or proteins of interest that are secreted or released from the cell via autolysis. Lastly, the platform is applied to optimize cutinase secretion in Komagataella phaffii (Pichia pastoris) and a strain with 5.7-fold improvement is isolated. The platform permits the analysis of >106 genotypes per day and is readily applicable to any protein that can be equipped with a His6 -tag. It is envisioned that the platform will be useful for large screening campaigns that aim to identify improved hosts for large-scale production of biotechnologically relevant proteins, thereby accelerating the costly and time-consuming process of strain engineering.
Collapse
Affiliation(s)
- Marta Napiorkowska
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Rd, Cambridge, CB2 1GA, UK
| | - Luzius Pestalozzi
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Sven Panke
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Martin Held
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Steven Schmitt
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| |
Collapse
|
33
|
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: 2] [Impact Index Per Article: 0.5] [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
|
34
|
Hymel D, Liu F. Proximity‐driven, Regioselective Chemical Modification of Peptides and Proteins. ASIAN J ORG CHEM 2020. [DOI: 10.1002/ajoc.202000328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- David Hymel
- Discovery Chemistry Novo Nordisk Research Center Seattle, Inc. 500 Fairview Ave Seattle WA 98109 USA
| | - Fa Liu
- Focus-X Therapeutics, Inc 3541 223rd Ave SE Sammamish WA 98075 USA
| |
Collapse
|
35
|
Girodat D, Pati AK, Terry DS, Blanchard SC, Sanbonmatsu KY. Quantitative comparison between sub-millisecond time resolution single-molecule FRET measurements and 10-second molecular simulations of a biosensor protein. PLoS Comput Biol 2020; 16:e1008293. [PMID: 33151943 PMCID: PMC7643941 DOI: 10.1371/journal.pcbi.1008293] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
Molecular Dynamics (MD) simulations seek to provide atomic-level insights into conformationally dynamic biological systems at experimentally relevant time resolutions, such as those afforded by single-molecule fluorescence measurements. However, limitations in the time scales of MD simulations and the time resolution of single-molecule measurements have challenged efforts to obtain overlapping temporal regimes required for close quantitative comparisons. Achieving such overlap has the potential to provide novel theories, hypotheses, and interpretations that can inform idealized experimental designs that maximize the detection of the desired reaction coordinate. Here, we report MD simulations at time scales overlapping with in vitro single-molecule Förster (fluorescence) resonance energy transfer (smFRET) measurements of the amino acid binding protein LIV-BPSS at sub-millisecond resolution. Computationally efficient all-atom structure-based simulations, calibrated against explicit solvent simulations, were employed for sampling multiple cycles of LIV-BPSS clamshell-like conformational changes on the time scale of seconds, examining the relationship between these events and those observed by smFRET. The MD simulations agree with the smFRET measurements and provide valuable information on local dynamics of fluorophores at their sites of attachment on LIV-BPSS and the correlations between fluorophore motions and large-scale conformational changes between LIV-BPSS domains. We further utilize the MD simulations to inform the interpretation of smFRET data, including Förster radius (R0) and fluorophore orientation factor (κ2) determinations. The approach we describe can be readily extended to distinct biochemical systems, allowing for the interpretation of any FRET system conjugated to protein or ribonucleoprotein complexes, including those with more conformational processes, as well as those implementing multi-color smFRET. Förster (fluorescence) resonance energy transfer (FRET) has been used extensively by biophysicists as a molecular-scale ruler that yields fundamental structural and kinetic insights into transient processes including complex formation and conformational rearrangements required for biological function. FRET techniques require the identification of informative fluorophore labeling sites, spaced at defined distances to inform on a reaction coordinate of interest and consideration of noise sources that have the potential to obscure quantitative interpretations. Here, we describe an approach to leverage advancements in computationally efficient all-atom structure-based molecular dynamics simulations in which structural dynamics observed via FRET can be interpreted in full atomistic detail on commensurate time scales. We demonstrate the potential of this approach using a model FRET system, the amino acid binding protein LIV-BPSS conjugated to self-healing organic fluorophores. LIV-BPSS exhibits large scale, sub-millisecond clamshell-like conformational changes between open and closed conformations associated with ligand unbinding and binding, respectively. Our findings inform on the molecular basis of the dynamics observed by smFRET and on strategies to optimize fluorophore labeling sites, the manner of fluorophore attachment, and fluorophore composition.
Collapse
Affiliation(s)
- Dylan Girodat
- Theoretical Biology and Biophysics, Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Avik K Pati
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Daniel S Terry
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Scott C Blanchard
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Karissa Y Sanbonmatsu
- Theoretical Biology and Biophysics, Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America.,New Mexico Consortium, Los Alamos, New Mexico, United States of America
| |
Collapse
|
36
|
Shirzadi Z, Baharvand H, Nezhati MN, Sajedi RH. Synthesis of nonlinear polymer brushes on magnetic nanoparticles as an affinity adsorbent for His-tagged xylanase purification. Colloid Polym Sci 2020. [DOI: 10.1007/s00396-020-04749-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
37
|
Probing fibronectin adsorption on chemically defined surfaces by means of single molecule force microscopy. Sci Rep 2020; 10:15662. [PMID: 32973270 PMCID: PMC7518417 DOI: 10.1038/s41598-020-72617-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/02/2020] [Indexed: 12/18/2022] Open
Abstract
Atomic force microscope (AFM) based single molecule force spectroscopy (SMFS) and a quartz crystal microbalance (QCM) were respectively employed to probe interfacial characteristics of fibronectin fragment FNIII8–14 and full-length fibronectin (FN) on CH3–, OH–, COOH–, and NH2-terminated alkane-thiol self-assembled monolayers (SAMs). Force-distance curves acquired between hexahistidine-tagged FNIII8–14 immobilised on trisNTA-Ni2+ functionalized AFM cantilevers and the OH and COOH SAM surfaces were predominantly ‘loop-like’ (76% and 94% respectively), suggesting domain unfolding and preference for ‘end-on’ oriented binding, while those generated with NH2 and CH3 SAMs were largely ‘mixed type’ (81% and 86%, respectively) commensurate with unravelling and desorption, and ‘side-on’ binding. Time-dependent binding of FN to SAM-coated QCM crystals occurred in at least two phases: initial rapid coverage over the first 5 min; and variably diminishing adsorption thereafter (5–70 min). Loading profiles and the final hydrated surface concentrations reached (~ 950, ~ 1200, ~ 1400, ~ 1500 ng cm−2 for CH3, OH, COOH and NH2 SAMs) were consistent with: space-filling ‘side-on’ orientation and unfolding on CH3 SAM; greater numbers of FN molecules arranged ‘end-on’ on OH and especially COOH SAMs; and initial ‘side-on’ contact, followed by either (1) gradual tilting to a space-saving ‘end-on’ configuration, or (2) bi-/multi-layer adsorption on NH2 SAM.
Collapse
|
38
|
Scherr J, Tang Z, Küllmer M, Balser S, Scholz AS, Winter A, Parey K, Rittner A, Grininger M, Zickermann V, Rhinow D, Terfort A, Turchanin A. Smart Molecular Nanosheets for Advanced Preparation of Biological Samples in Electron Cryo-Microscopy. ACS NANO 2020; 14:9972-9978. [PMID: 32589396 DOI: 10.1021/acsnano.0c03052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Transmission electron cryo-microscopy (cryoEM) of vitrified biological specimens is a powerful tool for structural biology. Current preparation of vitrified biological samples starts off with sample isolation and purification, followed by the fixation in a freestanding layer of amorphous ice. Here, we demonstrate that ultrathin (∼10 nm) smart molecular nanosheets having specific biorecognition sites embedded in a biorepulsive layer covalently bound to a mechanically stable carbon nanomembrane allow for a much simpler isolation and structural analysis. We characterize in detail the engineering of these nanosheets and their biorecognition properties employing complementary methods such as X-ray photoelectron and infrared spectroscopy, atomic force microscopy as well as surface plasmon resonance measurements. The desired functionality of the developed nanosheets is demonstrated by in situ selection of a His-tagged protein from a mixture and its subsequent structural analysis by cryoEM.
Collapse
Affiliation(s)
- Julian Scherr
- Department of Chemistry, University of Frankfurt, Max-von-Laue-Str. 7, 60438 Frankfurt, Germany
| | - Zian Tang
- Institute of Physical Chemistry, Friedrich Schiller University Jena, Lessingstr. 10, 07743 Jena, Germany
| | - Maria Küllmer
- Institute of Physical Chemistry, Friedrich Schiller University Jena, Lessingstr. 10, 07743 Jena, Germany
| | - Sebastian Balser
- Department of Chemistry, University of Frankfurt, Max-von-Laue-Str. 7, 60438 Frankfurt, Germany
| | - Alexander Stefan Scholz
- Department of Chemistry, University of Frankfurt, Max-von-Laue-Str. 7, 60438 Frankfurt, Germany
| | - Andreas Winter
- Institute of Physical Chemistry, Friedrich Schiller University Jena, Lessingstr. 10, 07743 Jena, Germany
| | - Kristian Parey
- Department of Structural Biology, Max Planck Institute of Biophysics, Max-von-Laue-Str. 3, 60438 Frankfurt, Germany
| | - Alexander Rittner
- Institute of Organic Chemistry and Chemical Biology, Buchmann Institute for Molecular Life Sciences, University of Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt, Germany
| | - Martin Grininger
- Institute of Organic Chemistry and Chemical Biology, Buchmann Institute for Molecular Life Sciences, University of Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt, Germany
| | - Volker Zickermann
- Institute of Biochemistry II, Medical School, University of Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany
| | - Daniel Rhinow
- Department of Structural Biology, Max Planck Institute of Biophysics, Max-von-Laue-Str. 3, 60438 Frankfurt, Germany
| | - Andreas Terfort
- Department of Chemistry, University of Frankfurt, Max-von-Laue-Str. 7, 60438 Frankfurt, Germany
| | - Andrey Turchanin
- Institute of Physical Chemistry, Friedrich Schiller University Jena, Lessingstr. 10, 07743 Jena, Germany
| |
Collapse
|
39
|
Raducanu VS, Isaioglou I, Raducanu DV, Merzaban JS, Hamdan SM. Simplified detection of polyhistidine-tagged proteins in gels and membranes using a UV-excitable dye and a multiple chelator head pair. J Biol Chem 2020; 295:12214-12223. [PMID: 32647010 PMCID: PMC7443479 DOI: 10.1074/jbc.ra120.014132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/29/2020] [Indexed: 12/22/2022] Open
Abstract
The polyhistidine tag (His-tag) is one of the most popular protein tags used in the life sciences. Traditionally, the detection of His-tagged proteins relies on immunoblotting with anti-His antibodies. This approach is laborious for certain applications, such as protein purification, where time and simplicity are critical. The His-tag can also be directly detected by metal ion-loaded nickel-nitrilotriacetic acid-based chelator heads conjugated to fluorophores, which is a convenient alternative method to immunoblotting. Typically, such chelator heads are conjugated to either green or red fluorophores, the detection of which requires specialized excitation sources and detection systems. Here, we demonstrate that post-run staining is ideal for His-tag detection by metal ion-loaded and fluorescently labeled chelator heads in PAGE and blot membranes. Additionally, by comparing the performances of different chelator heads, we show how differences in microscopic affinity constants translate to macroscopic differences in the detection limits in environments with limited diffusion, such as PAGE. On the basis of these results, we devise a simple approach, called UVHis-PAGE, that uses metal ion-loaded and fluorescently labeled chelator heads to detect His-tagged proteins in PAGE and blot membranes. Our method uses a UV transilluminator as an excitation source, and the results can be visually inspected by the naked eye.
Collapse
Affiliation(s)
- Vlad-Stefan Raducanu
- Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Ioannis Isaioglou
- Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Daniela-Violeta Raducanu
- Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Jasmeen S Merzaban
- Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Samir M Hamdan
- Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia.
| |
Collapse
|
40
|
Janco M, Dedova I, Bryce NS, Hardeman EC, Gunning PW. Visualizing the in vitro assembly of tropomyosin/actin filaments using TIRF microscopy. Biophys Rev 2020; 12:879-885. [PMID: 32638329 PMCID: PMC7429660 DOI: 10.1007/s12551-020-00720-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 07/02/2020] [Indexed: 12/23/2022] Open
Abstract
Tropomyosins are elongated alpha-helical proteins that form co-polymers with most actin filaments within a cell and play important roles in the structural and functional diversification of the actin cytoskeleton. How the assembly of tropomyosins along an actin filament is regulated and the kinetics of tropomyosin association with an actin filament is yet to be fully determined. A recent series of publications have used total internal reflection fluorescence (TIRF) microscopy in combination with advanced surface and protein chemistry to visualise the molecular assembly of actin/tropomyosin filaments in vitro. Here, we review the use of the in vitro TIRF assay in the determination of kinetic data on tropomyosin filament assembly. This sophisticated approach has enabled generation of real-time single-molecule data to fill the gap between in vitro bulk assays and in vivo assays of tropomyosin function. The in vitro TIRF assays provide a new foundation for future studies involving multiple actin-binding proteins that will more accurately reflect the physiological protein-protein interactions in cells.
Collapse
Affiliation(s)
- Miro Janco
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Irina Dedova
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Nicole S Bryce
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Edna C Hardeman
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Peter W Gunning
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
41
|
Gutmann T, Schäfer IB, Poojari C, Brankatschk B, Vattulainen I, Strauss M, Coskun Ü. Cryo-EM structure of the complete and ligand-saturated insulin receptor ectodomain. J Cell Biol 2020; 219:jcb.201907210. [PMID: 31727777 PMCID: PMC7039211 DOI: 10.1083/jcb.201907210] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/19/2019] [Accepted: 10/20/2019] [Indexed: 12/20/2022] Open
Abstract
Glucose homeostasis and growth essentially depend on the hormone insulin engaging its receptor. Despite biochemical and structural advances, a fundamental contradiction has persisted in the current understanding of insulin ligand-receptor interactions. While biochemistry predicts two distinct insulin binding sites, 1 and 2, recent structural analyses have resolved only site 1. Using a combined approach of cryo-EM and atomistic molecular dynamics simulation, we present the structure of the entire dimeric insulin receptor ectodomain saturated with four insulin molecules. Complementing the previously described insulin-site 1 interaction, we present the first view of insulin bound to the discrete insulin receptor site 2. Insulin binding stabilizes the receptor ectodomain in a T-shaped conformation wherein the membrane-proximal domains converge and contact each other. These findings expand the current models of insulin binding to its receptor and of its regulation. In summary, we provide the structural basis for a comprehensive description of ligand-receptor interactions that ultimately will inform new approaches to structure-based drug design.
Collapse
Affiliation(s)
- Theresia Gutmann
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Hospital and Faculty of Medicine Carl Gustav Carus of Technische Universität Dresden, Dresden, Germany.,German Center for Diabetes Research, Neuherberg, Germany
| | - Ingmar B Schäfer
- Department of Structural Cell Biology, Max Planck Institute of Biochemistry, Munich, Germany
| | - Chetan Poojari
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Beate Brankatschk
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Hospital and Faculty of Medicine Carl Gustav Carus of Technische Universität Dresden, Dresden, Germany.,German Center for Diabetes Research, Neuherberg, Germany
| | - Ilpo Vattulainen
- Department of Physics, University of Helsinki, Helsinki, Finland.,Computational Physics Laboratory, Tampere University, Tampere, Finland
| | - Mike Strauss
- Department of Anatomy & Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Ünal Coskun
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Hospital and Faculty of Medicine Carl Gustav Carus of Technische Universität Dresden, Dresden, Germany.,German Center for Diabetes Research, Neuherberg, Germany
| |
Collapse
|
42
|
Cuellar-Camacho JL, Bhatia S, Reiter-Scherer V, Lauster D, Liese S, Rabe JP, Herrmann A, Haag R. Quantification of Multivalent Interactions between Sialic Acid and Influenza A Virus Spike Proteins by Single-Molecule Force Spectroscopy. J Am Chem Soc 2020; 142:12181-12192. [PMID: 32538085 DOI: 10.1021/jacs.0c02852] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Multivalency is a key principle in reinforcing reversible molecular interactions through the formation of multiple bonds. The influenza A virus deploys this strategy to bind strongly to cell surface receptors. We performed single-molecule force spectroscopy (SMFS) to investigate the rupture force required to break individual and multiple bonds formed between synthetic sialic acid (SA) receptors and the two principal spike proteins of the influenza A virus (H3N2): hemagglutinin (H3) and neuraminidase (N2). Kinetic parameters such as the rupture length (χβ) and dissociation rate (koff) are extracted using the model by Friddle, De Yoreo, and Noy. We found that a monovalent SA receptor binds to N2 with a significantly higher bond lifetime (270 ms) compared to that for H3 (36 ms). By extending the single-bond rupture analysis to a multibond system of n protein-receptor pairs, we provide an unprecedented quantification of the mechanistic features of multivalency between H3 and N2 with SA receptors and show that the stability of the multivalent connection increases with the number of bonds from tens to hundreds of milliseconds. Association rates (kon) are also provided, and an estimation of the dissociation constants (KD) between the SA receptors to both proteins indicate a 17-fold higher binding affinity for the SA-N2 bond with respect to that of SA-H3. An optimal designed multivalent SA receptor showed a higher binding stability to the H3 protein of the influenza A virus than to the monovalent SA receptor. Our study emphasizes the influence of the scaffold on the presentation of receptors during multivalent binding.
Collapse
Affiliation(s)
- Jose Luis Cuellar-Camacho
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany
| | - Sumati Bhatia
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany
| | - Valentin Reiter-Scherer
- Department of Physics & IRIS Adlershof, Humboldt-Universität zu Berlin, Newtonstraße 15, 12489 Berlin, Germany
| | - Daniel Lauster
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany.,Institute for Biology & IRI Life Sciences, Humboldt-Universität zu Berlin, Invalidenstraße 42, 10115 Berlin, Germany
| | - Susanne Liese
- Department of Mathematics, University of Oslo, Moltke Moes vei 35, 1053 Oslo, Norway
| | - Jürgen P Rabe
- Department of Physics & IRIS Adlershof, Humboldt-Universität zu Berlin, Newtonstraße 15, 12489 Berlin, Germany
| | - Andreas Herrmann
- Institute for Biology & IRI Life Sciences, Humboldt-Universität zu Berlin, Invalidenstraße 42, 10115 Berlin, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany
| |
Collapse
|
43
|
Koehler M, Lo Giudice C, Vogl P, Ebner A, Hinterdorfer P, Gruber HJ, Alsteens D. Control of Ligand-Binding Specificity Using Photocleavable Linkers in AFM Force Spectroscopy. NANO LETTERS 2020; 20:4038-4042. [PMID: 32320256 PMCID: PMC7252943 DOI: 10.1021/acs.nanolett.0c01426] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/21/2020] [Indexed: 05/26/2023]
Abstract
In recent decades, atomic force microscopy (AFM), in particular the force spectroscopy mode, has become a method of choice to study biomolecular interactions at the single-molecule level. However, grafting procedures as well as determining binding specificity remain challenging. We report here an innovative approach based on a photocleavable group that enables in situ release of the ligands bound to the AFM tip and thus allows direct assessment of the binding specificity. Applicable to a wide variety of molecules, the strategy presented here provides new opportunities to study specific interactions and deliver single molecules with high spatiotemporal resolution in a wide range of applications, including AFM-based cell biology.
Collapse
Affiliation(s)
- Melanie Koehler
- Louvain
Institute of Biomolecular Science and Technology, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | - Cristina Lo Giudice
- Louvain
Institute of Biomolecular Science and Technology, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | - Philipp Vogl
- Institute
of Biophysics, Johannes Kepler University, 4020 Linz, Austria
| | - Andreas Ebner
- Institute
of Biophysics, Johannes Kepler University, 4020 Linz, Austria
| | | | - Hermann J. Gruber
- Institute
of Biophysics, Johannes Kepler University, 4020 Linz, Austria
| | - David Alsteens
- Louvain
Institute of Biomolecular Science and Technology, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| |
Collapse
|
44
|
Qu JH, Horta S, Delport F, Sillen M, Geukens N, Sun DW, Vanhoorelbeke K, Declerck P, Lammertyn J, Spasic D. Expanding a Portfolio of (FO-) SPR Surface Chemistries with the Co(III)-NTA Oriented Immobilization of His 6-Tagged Bioreceptors for Applications in Complex Matrices. ACS Sens 2020; 5:960-969. [PMID: 32216277 DOI: 10.1021/acssensors.9b02227] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cobalt-nitrilotriacetic acid (Co(III)-NTA) chemistry is a recognized approach for oriented patterning of His6-tagged bioreceptors. We have applied the matching strategy for the first time on a surface plasmon resonance (SPR) platform, namely, the commercialized fiber optic (FO)-SPR. To accomplish this, His6-tagged bioreceptor (scFv-33H1F7) and its target PAI-1 were used as a model system, after scrutinizing the specificity of their interaction. When benchmarked to traditional carboxyl-based self-assembled monolayers (SAM), NTA allowed (1) more efficient FO-SPR surface coverage with bioreceptors compared with the former and (2) realization of thus far difficult-to-attain label-free bioassays on the FO-SPR platform in both buffer and 20-fold diluted human plasma. Moreover, Co(III)-NTA surface proved to be compatible with traditional gold nanoparticle-mediated signal amplification in the buffer as well as in 10-fold diluted human plasma, thus expanding the dynamic detection range to low ng/mL. Both types of bioassays revealed that scFv-33H1F7 immobilized on the FO-SPR surface using different concentrations (20, 10, or 5 μg/mL) had no impact on the bioassay sensitivity, accuracy, or reproducibility despite the lowest concentration effectively resulting in close to 20% fewer bioreceptors. Collectively, these results highlight the importance of Co(III)-NTA promoting the oriented patterning of bioreceptors on the FO-SPR sensor surface for securing robust and sensitive bioassays in complex matrices, both in label-free and labeled formats.
Collapse
Affiliation(s)
- Jia-Huan Qu
- Department of Biosystems, Biosensors Group, KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Sara Horta
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak, 8500 Kortrijk, Belgium
| | - Filip Delport
- FOx Biosystems, Bioville, Agoralaan Abis, 3590 Diepenbeek, Belgium
| | - Machteld Sillen
- Department of Pharmaceutical and Pharmacological Sciences, Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven, O&N II Herestraat 49, 3000 Leuven, Belgium
| | - Nick Geukens
- PharmAbs, KU Leuven, Herestraat 49,
Box 820, B 3000 Leuven, Belgium
| | - Da-Wen Sun
- School of Food Science and Engineering, South China University of Technology, 510641 Guangzhou, China
| | - Karen Vanhoorelbeke
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak, 8500 Kortrijk, Belgium
| | - Paul Declerck
- Department of Pharmaceutical and Pharmacological Sciences, Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven, O&N II Herestraat 49, 3000 Leuven, Belgium
| | - Jeroen Lammertyn
- Department of Biosystems, Biosensors Group, KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Dragana Spasic
- Department of Biosystems, Biosensors Group, KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| |
Collapse
|
45
|
Flesch J, Kappen M, Drees C, You C, Piehler J. Self-assembly of robust gold nanoparticle monolayer architectures for quantitative protein interaction analysis by LSPR spectroscopy. Anal Bioanal Chem 2020; 412:3413-3422. [PMID: 32198532 PMCID: PMC7214499 DOI: 10.1007/s00216-020-02551-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/13/2020] [Accepted: 02/26/2020] [Indexed: 11/30/2022]
Abstract
Localized surface plasmon resonance (LSPR) detection offers highly sensitive label-free detection of biomolecular interactions. Simple and robust surface architectures compatible with real-time detection in a flow-through system are required for broad application in quantitative interaction analysis. Here, we established self-assembly of a functionalized gold nanoparticle (AuNP) monolayer on a glass substrate for stable, yet reversible immobilization of Histidine-tagged proteins. To this end, one-step coating of glass substrates with poly-L-lysine graft poly(ethylene glycol) functionalized with ortho-pyridyl disulfide (PLL-PEG-OPSS) was employed as a reactive, yet biocompatible monolayer to self-assemble AuNP into a LSPR active monolayer. Site-specific, reversible immobilization of His-tagged proteins was accomplished by coating the AuNP monolayer with tris-nitrilotriacetic acid (trisNTA) PEG disulfide. LSPR spectroscopy detection of protein binding on these biocompatible functionalized AuNP monolayers confirms high stability under various harsh analytical conditions. These features were successfully employed to demonstrate unbiased kinetic analysis of cytokine-receptor interactions. Graphical abstract ![]()
Collapse
Affiliation(s)
- Julia Flesch
- Department of Biology/Chemistry, University of Osnabrück, Barbarastr. 11, 49076, Osnabrück, Germany
| | - Marie Kappen
- Department of Biology/Chemistry, University of Osnabrück, Barbarastr. 11, 49076, Osnabrück, Germany
| | - Christoph Drees
- Department of Biology/Chemistry, University of Osnabrück, Barbarastr. 11, 49076, Osnabrück, Germany
| | - Changjiang You
- Department of Biology/Chemistry, University of Osnabrück, Barbarastr. 11, 49076, Osnabrück, Germany.
- Center for Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Barbarastr. 11, 49076, Osnabrück, Germany.
| | - Jacob Piehler
- Department of Biology/Chemistry, University of Osnabrück, Barbarastr. 11, 49076, Osnabrück, Germany.
- Center for Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Barbarastr. 11, 49076, Osnabrück, Germany.
| |
Collapse
|
46
|
Brüchert S, Joest EF, Gatterdam K, Tampé R. Ultrafast in-gel detection by fluorescent super-chelator probes with HisQuick-PAGE. Commun Biol 2020; 3:138. [PMID: 32198384 PMCID: PMC7083852 DOI: 10.1038/s42003-020-0852-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/20/2020] [Indexed: 02/02/2023] Open
Abstract
Polyacrylamide gel electrophoresis (PAGE) and immunoblotting (Western blotting) are the most common methods in life science. In conjunction with these methods, the polyhistidine-tag has proven to be a superb fusion tag for protein purification as well as specific protein detection by immunoblotting, which led to a vast amount of commercially available antibodies. Nevertheless, antibody batch-to-batch variations and nonspecific binding complicate the laborious procedure. The interaction principle applied for His-tagged protein purification by metal-affinity chromatography using N-nitrilotriacetic acid (NTA) was employed to develop small high-affinity lock-and-key molecules coupled to a fluorophore. These multivalent NTA probes allow specific detection of His-tagged proteins by fluorescence. Here, we report on HisQuick-PAGE as a fast and versatile immunoblot alternative, using such high-affinity fluorescent super-chelator probes. The procedure allows direct, fast, and ultra-sensitive in-gel detection and analysis of soluble proteins as well as intact membrane protein complexes and macromolecular ribonucleoprotein particles. Brüchert et al. describe the application of high-affinity super-chelator probes that are linked to a fluorophore for the detection of His-tagged proteins using PAGE. Their system has the advantage over conventional antibody staining in the ease of application and ultra-sensitive detection.
Collapse
Affiliation(s)
- Stefan Brüchert
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt, Germany
| | - Eike F Joest
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt, Germany
| | - Karl Gatterdam
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt, Germany.,Institute of Structural Biology, Biomedical Center, University of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt, Germany.
| |
Collapse
|
47
|
X-ray structure of LeuT in an inward-facing occluded conformation reveals mechanism of substrate release. Nat Commun 2020; 11:1005. [PMID: 32081981 PMCID: PMC7035281 DOI: 10.1038/s41467-020-14735-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/24/2020] [Indexed: 12/21/2022] Open
Abstract
Neurotransmitter:sodium symporters (NSS) are conserved from bacteria to man and serve as targets for drugs, including antidepressants and psychostimulants. Here we report the X-ray structure of the prokaryotic NSS member, LeuT, in a Na+/substrate-bound, inward-facing occluded conformation. To obtain this structure, we were guided by findings from single-molecule fluorescence spectroscopy and molecular dynamics simulations indicating that L-Phe binding and mutation of the conserved N-terminal Trp8 to Ala both promote an inward-facing state. Compared to the outward-facing occluded conformation, our structure reveals a major tilting of the cytoplasmic end of transmembrane segment (TM) 5, which, together with release of the N-terminus but without coupled movement of TM1, opens a wide cavity towards the second Na+ binding site. The structure of this key intermediate in the LeuT transport cycle, in the context of other NSS structures, leads to the proposal of an intracellular release mechanism of substrate and ions in NSS proteins.
Collapse
|
48
|
Lindemann WR, Evans ED, Mijalis AJ, Saouaf OM, Pentelute BL, Ortony JH. Quantifying residue-specific conformational dynamics of a highly reactive 29-mer peptide. Sci Rep 2020; 10:2597. [PMID: 32054898 PMCID: PMC7018720 DOI: 10.1038/s41598-020-59047-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/22/2020] [Indexed: 11/19/2022] Open
Abstract
Understanding structural transitions within macromolecules remains an important challenge in biochemistry, with important implications for drug development and medicine. Insight into molecular behavior often requires residue-specific dynamics measurement at micromolar concentrations. We studied MP01-Gen4, a library peptide selected to rapidly undergo bioconjugation, by using electron paramagnetic resonance (EPR) to measure conformational dynamics. We mapped the dynamics of MP01-Gen4 with residue-specificity and identified the regions involved in a structural transformation related to the conjugation reaction. Upon reaction, the conformational dynamics of residues near the termini slow significantly more than central residues, indicating that the reaction induces a structural transition far from the reaction site. Arrhenius analysis demonstrates a nearly threefold decrease in the activation energy of conformational diffusion upon reaction (8.0 kBT to 3.4 kBT), which occurs across the entire peptide, independently of residue position. This novel approach to EPR spectral analysis provides insight into the positional extent of disorder and the nature of the energy landscape of a highly reactive, intrinsically disordered library peptide before and after conjugation.
Collapse
Affiliation(s)
- William R Lindemann
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts, 02139, United States
| | - Ethan D Evans
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts, 02139, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
| | - Alexander J Mijalis
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts, 02139, United States
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Olivia M Saouaf
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts, 02139, United States
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, California, 94305, United States
| | - Bradley L Pentelute
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts, 02139, United States
| | - Julia H Ortony
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts, 02139, United States.
| |
Collapse
|
49
|
Quantifying secondary transport at single-molecule resolution. Nature 2019; 575:528-534. [PMID: 31723269 DOI: 10.1038/s41586-019-1747-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 10/07/2019] [Indexed: 01/07/2023]
Abstract
Secondary active transporters, which are vital for a multitude of physiological processes, use the energy of electrochemical ion gradients to power substrate transport across cell membranes1,2. Efforts to investigate their mechanisms of action have been hampered by their slow transport rates and the inherent limitations of ensemble methods. Here we quantify the activity of individual MhsT transporters, which are representative of the neurotransmitter:sodium symporter family of secondary transporters3, by imaging the transport of individual substrate molecules across lipid bilayers at both single- and multi-turnover resolution. We show that MhsT is active only when physiologically oriented and that the rate-limiting step of the transport cycle varies with the nature of the transported substrate. These findings are consistent with an extracellular allosteric substrate-binding site that modulates the rate-limiting aspects of the transport mechanism4,5, including the rate at which the transporter returns to an outward-facing state after the transported substrate is released.
Collapse
|
50
|
Selective Derivatization of Hexahistidine-Tagged Recombinant Proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019. [PMID: 31347051 DOI: 10.1007/978-3-030-15950-4_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
Covalent modification of proteins is extensively used in research and industry for biosensing, medical diagnostics, targeted drug delivery, and many other practical applications. The conventional method for production of protein conjugates has changed little in the last 20 years mostly relying on reactions of side chains of cysteine and lysine residues. Due to the presence of large numbers of similar reactive amino acid residues in proteins, common synthetic methods generally produce complex mixtures of products, which are difficult to separate. An emerging alternative technology for covalent modification of proteins involves formation of a covalent bond with a hexahistidine affinity tag present in a majority of recombinant proteins without interfering with other amino acid residues. The approach is based on formation of a ternary complex of the hexahistidine sequence with a bivalent metal cation chelated by ligand bearing an electrophilic Baylis-Hillman ester group capable of subsequent formation of a covalent bond with one of the histidine residues of the tag. The reaction proceeds under mild reaction conditions in neutral aqueous solutions under high dilutions (10-5 to 10-4 M) providing a stable covalent bond between the label and an imidazole residue in a hexahistidine tag at either C- or N-terminus. Because hexahistidine affinity tag methodology is a de-facto standard for preparation of recombinant proteins our approach can be easily implemented for selective derivatization of these proteins with fluorescent groups, alkynyl groups for "click" reactions, or biotinylation.
Collapse
|