51
|
Sobrinos-Sanguino M, Vélez M, Richter RP, Rivas G. Reversible Membrane Tethering by ZipA Determines FtsZ Polymerization in Two and Three Dimensions. Biochemistry 2019; 58:4003-4015. [PMID: 31390865 DOI: 10.1021/acs.biochem.9b00378] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In most bacteria, the early step of septum formation implies the association of soluble FtsZ polymers with the cytoplasmic membrane. ZipA, together with FtsA, provides membrane tethering to FtsZ in Escherichia coli, forming a dynamic proto-ring that serves as an assembly scaffold for the remaining elements of the divisome. Despite their importance for bacterial cell division, multivalent interactions between proto-ring elements at membrane surfaces remain poorly characterized in quantitative terms. We measured the binding of FtsZ to ZipA incorporated in supported lipid bilayers at controlled densities by using a combination of biophysical surface-sensitive techniques (quartz crystal microbalance and spectroscopic ellipsometry) and analyzed how ZipA density and FtsZ concentration control the state of assembly of FtsZ. We found that ZipA attachment enables FtsZ-GMPCPP (where GMPCPP is a GTP analogue with a reduced level of hydrolysis) to assemble in several distinct ways: (i) two-dimensional polymerization at the membrane and (ii) three-dimensional polymerization from the membrane into the solution phase where this may be associated with the formation of higher-order complexes. In these processes, ZipA is required to enrich FtsZ at the surface but the FtsZ bulk concentration defines which morphology is being formed. Moreover, we report a strong effect of the nucleotide (GDP vs GMPCPP/GTP) on the kinetics of ZipA association/dissociation of FtsZ. These results provide insights into the mode of interaction of proto-ring elements in minimal membrane systems and contribute to the completion of our understanding of the initial events of bacterial division.
Collapse
Affiliation(s)
- Marta Sobrinos-Sanguino
- Centro de Investigaciones Biológicas , Consejo Superior de Investigaciones Científicas (CSIC) , 28040 Madrid , Spain.,School of Biomedical Sciences, Faculty of Biological Sciences, School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, and Astbury Centre for Structural Molecular Biology , University of Leeds , Leeds LS2 9JT , United Kingdom
| | - Marisela Vélez
- Instituto de Catálisis y Petroleoquímica , CSIC , 28049 Madrid , Spain
| | - Ralf P Richter
- School of Biomedical Sciences, Faculty of Biological Sciences, School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, and Astbury Centre for Structural Molecular Biology , University of Leeds , Leeds LS2 9JT , United Kingdom.,Biosurfaces Lab , CIC biomaGUNE , 20014 San Sebastian , Spain
| | - Germán Rivas
- Centro de Investigaciones Biológicas , Consejo Superior de Investigaciones Científicas (CSIC) , 28040 Madrid , Spain
| |
Collapse
|
52
|
Kryštůfek R, Šácha P. An iBody-based lateral flow assay for semi-quantitative determination of His-tagged protein concentration. J Immunol Methods 2019; 473:112640. [PMID: 31400409 DOI: 10.1016/j.jim.2019.112640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/24/2019] [Accepted: 08/06/2019] [Indexed: 10/26/2022]
Abstract
The polyhistidine tag (His-tag) is one of the most commonly used epitope tags in protein engineering. While His-tagged proteins can be detected reliably using immunological methods such as ELISA and Western blot, these methods are costly and time-intensive, necessitating more facile solutions for preliminary qualitative determination and concentration estimation. To this end, we present a rapid test strip assay based on iBody antibody mimetics that target the His-tag. We compare this strategy to commercial antibody-based assays and discuss the advantages and caveats of lateral flow assay design. Our test strip detected a panel of His-tagged proteins with different tag attachment strategies with a visual detection limit of 1 μM and densitometric detection limit of 0.5 μM. Due to its chemical nature, the presented assay exhibits wide reagent compatibility in comparison to antibody-based assays.
Collapse
Affiliation(s)
- Robin Kryštůfek
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Flemingovo n. 2, 16610 Prague 6, Czech Republic
| | - Pavel Šácha
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Flemingovo n. 2, 16610 Prague 6, Czech Republic.
| |
Collapse
|
53
|
LeVine MV, Terry DS, Khelashvili G, Siegel ZS, Quick M, Javitch JA, Blanchard SC, Weinstein H. The allosteric mechanism of substrate-specific transport in SLC6 is mediated by a volumetric sensor. Proc Natl Acad Sci U S A 2019; 116:15947-15956. [PMID: 31324743 PMCID: PMC6689989 DOI: 10.1073/pnas.1903020116] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neurotransmitter:sodium symporters (NSSs) in the SLC6 family terminate neurotransmission by coupling the thermodynamically favorable transport of ions to the thermodynamically unfavorable transport of neurotransmitter back into presynaptic neurons. Results from many structural, functional, and computational studies on LeuT, a bacterial NSS homolog, have provided critical insight into the mechanism of sodium-coupled transport, but the mechanism underlying substrate-specific transport rates is still not understood. We present a combination of molecular dynamics simulations, single-molecule fluorescence resonance energy transfer (smFRET) imaging, and measurements of Na+ binding and substrate transport that reveals an allosteric substrate specificity mechanism. In this mechanism, residues F259 and I359 in the substrate binding pocket couple the binding of substrate to Na+ release from the Na2 site by allosterically modulating the stability of a partially open, inward-facing state. We propose a model for transport selectivity in which residues F259 and I359 act as a volumetric sensor that inhibits the transport of bulky amino acids.
Collapse
Affiliation(s)
- Michael V LeVine
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065;
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021
| | - Daniel S Terry
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065
| | - George Khelashvili
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021
| | - Zarek S Siegel
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021
| | - Matthias Quick
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032
| | - Jonathan A Javitch
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032
- Department of Pharmacology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Scott C Blanchard
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065
| | - Harel Weinstein
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065;
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021
| |
Collapse
|
54
|
Chen W, Guo J, Cai Y, Fu Q, Chen B, Chou JJ. Unidirectional Presentation of Membrane Proteins in Nanoparticle-Supported Liposomes. Angew Chem Int Ed Engl 2019; 58:9866-9870. [PMID: 30990942 PMCID: PMC6660371 DOI: 10.1002/anie.201903093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Indexed: 12/30/2022]
Abstract
Presentation of membrane proteins to host immune systems has been a challenging problem owing to complexity arising from the poor in vivo stability of the membrane-mimetic media often used for solubilizing the membrane proteins. The use of functionalized, biocompatible nanoparticles as substrates is shown to guide the formation of proteoliposomes, which can present many copies of membrane proteins in a unidirectional manner. The approach was demonstrated to present the membrane-proximal region of the HIV-1 envelope glycoprotein. These nanoparticle-supported liposomes are broadly applicable as membrane antigen vehicles for inducing host immune responses.
Collapse
Affiliation(s)
- Wen Chen
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, United States
| | - Junling Guo
- Department of Biomass Science and Engineering, Sichuan University, 252 Shuncheng Street, Chengdu, Sichuan 610065, China
| | - Yongfei Cai
- Division of Molecular Medicine, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, 3 Blackfan Street, Boston, MA 02115, United States
| | - Qingshan Fu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, United States
| | - Bing Chen
- Division of Molecular Medicine, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, 3 Blackfan Street, Boston, MA 02115, United States
| | - James J. Chou
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, United States
| |
Collapse
|
55
|
Chen W, Cai Y, Fu Q, Chen B, Guo J, Chou JJ. Unidirectional Presentation of Membrane Proteins in Nanoparticle‐Supported Liposomes. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201903093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Wen Chen
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical School 250 Longwood Avenue Boston MA 02115 USA
| | - Yongfei Cai
- Division of Molecular MedicineBoston Children's HospitalDepartment of PediatricsHarvard Medical School 3 Blackfan Street Boston MA 02115 USA
| | - Qingshan Fu
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical School 250 Longwood Avenue Boston MA 02115 USA
| | - Bing Chen
- Division of Molecular MedicineBoston Children's HospitalDepartment of PediatricsHarvard Medical School 3 Blackfan Street Boston MA 02115 USA
| | - Junling Guo
- Department of Biomass Science and EngineeringSichuan University 24 South Section Yihuan Road Chengdu Sichuan 610065 China
| | - James J. Chou
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical School 250 Longwood Avenue Boston MA 02115 USA
| |
Collapse
|
56
|
Fu Q, Piai A, Chen W, Xia K, Chou JJ. Structure determination protocol for transmembrane domain oligomers. Nat Protoc 2019; 14:2483-2520. [PMID: 31270510 DOI: 10.1038/s41596-019-0188-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 04/30/2019] [Indexed: 01/08/2023]
Abstract
The transmembrane (TM) anchors of cell surface proteins have been one of the 'blind spots' in structural biology because they are generally very hydrophobic, sometimes dynamic, and thus difficult targets for structural characterization. A plethora of examples show these membrane anchors are not merely anchors but can multimerize specifically to activate signaling receptors on the cell surface or to stabilize envelope proteins in viruses. Through a series of studies of the TM domains (TMDs) of immune receptors and viral membrane proteins, we have established a robust protocol for determining atomic-resolution structures of TM oligomers by NMR in bicelles that closely mimic a lipid bilayer. Our protocol overcomes hurdles typically encountered by structural biology techniques such as X-ray crystallography and cryo-electron microscopy (cryo-EM) when studying small TMDs. Here, we provide the details of the protocol, covering five major technical aspects: (i) a general method for producing isotopically labeled TM or membrane-proximal (MP) protein fragments that involves expression of the protein (which is fused to TrpLE) into inclusion bodies and releasing the target protein by cyanogen bromide (CNBr) cleavage; (ii) determination of the oligomeric state of TMDs in bicelles; (iii) detection of intermolecular contacts using nuclear Overhauser effect (NOE) experiments; (iv) structure determination; and (v) paramagnetic probe titration (PPT) to characterize the membrane partition of the TM oligomers. This protocol is broadly applicable for filling structural gaps of many type I/II membrane proteins. The procedures may take 3-6 months to complete, depending on the complexity and stability of the protein sample.
Collapse
Affiliation(s)
- Qingshan Fu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Alessandro Piai
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Wen Chen
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Ke Xia
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - James J Chou
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
57
|
Zenmyo N, Tokumaru H, Uchinomiya S, Fuchida H, Tabata S, Hamachi I, Shigemoto R, Ojida A. Optimized Reaction Pair of the CysHis Tag and Ni(II)-NTA Probe for Highly Selective Chemical Labeling of Membrane Proteins. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2019. [DOI: 10.1246/bcsj.20190034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Naoki Zenmyo
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroki Tokumaru
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Shohei Uchinomiya
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirokazu Fuchida
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Shigekazu Tabata
- Institute of Science and Technology Austria, A-3400 Klosterneuburg, Austria
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Ryuichi Shigemoto
- Institute of Science and Technology Austria, A-3400 Klosterneuburg, Austria
| | - Akio Ojida
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
58
|
Hanspach G, Trucks S, Hengesbach M. Strategic labelling approaches for RNA single-molecule spectroscopy. RNA Biol 2019; 16:1119-1132. [PMID: 30874475 DOI: 10.1080/15476286.2019.1593093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Most single-molecule techniques observing RNA in vitro or in vivo require fluorescent labels that have to be connected to the RNA of interest. In recent years, a plethora of methods has been developed to achieve site-specific labelling, in many cases under near-native conditions. Here, we review chemical as well as enzymatic labelling methods that are compatible with single-molecule fluorescence spectroscopy or microscopy and show how these can be combined to offer a large variety of options to site-specifically place one or more labels in an RNA of interest. By either chemically forming a covalent bond or non-covalent hybridization, these techniques are prerequisites to perform state-of-the-art single-molecule experiments.
Collapse
Affiliation(s)
- Gerd Hanspach
- a Goethe-University Frankfurt, Institute for Organic Chemistry and Chemical Biology , Frankfurt , Germany
| | - Sven Trucks
- a Goethe-University Frankfurt, Institute for Organic Chemistry and Chemical Biology , Frankfurt , Germany
| | - Martin Hengesbach
- a Goethe-University Frankfurt, Institute for Organic Chemistry and Chemical Biology , Frankfurt , Germany
| |
Collapse
|
59
|
Baldering TN, Dietz MS, Gatterdam K, Karathanasis C, Wieneke R, Tampé R, Heilemann M. Synthetic and genetic dimers as quantification ruler for single-molecule counting with PALM. Mol Biol Cell 2019; 30:1369-1376. [PMID: 30969885 PMCID: PMC6724688 DOI: 10.1091/mbc.e18-10-0661] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
How membrane proteins oligomerize determines their function. Superresolution microscopy can report on protein clustering and extract quantitative molecular information. Here, we evaluate the blinking kinetics of four photoactivatable fluorescent proteins for quantitative single-molecule microscopy. We identified mEos3.2 and mMaple3 to be suitable for molecular quantification through blinking histogram analysis. We designed synthetic and genetic dimers of mEos3.2 as well as fusion proteins of monomeric and dimeric membrane proteins as reference structures, and we demonstrate their versatile use for quantitative superresolution imaging in vitro and in situ. We further found that the blinking behavior of mEos3.2 and mMaple3 is modified by a reducing agent, offering the possibility to adjust blinking parameters according to experimental needs.
Collapse
Affiliation(s)
- Tim N Baldering
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany
| | - Marina S Dietz
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany
| | - Karl Gatterdam
- Institute of Biochemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany
| | - Christos Karathanasis
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany
| | - Ralph Wieneke
- Institute of Biochemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany
| | - Robert Tampé
- Institute of Biochemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany
| | - Mike Heilemann
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany
| |
Collapse
|
60
|
Wieneke R, Tampé R. Multivalent Chelators for In Vivo Protein Labeling. Angew Chem Int Ed Engl 2019; 58:8278-8290. [PMID: 30919542 DOI: 10.1002/anie.201811293] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Indexed: 01/09/2023]
Abstract
With the advent of single-molecule methods, chemoselective and site-specific labeling of proteins evolved to become a central aspect in chemical biology as well as cell biology. Protein labeling demands high specificity, rapid as well as efficient conjugation, while maintaining low concentration and biocompatibility under physiological conditions. Generic methods that do not interfere with the function, dynamics, subcellular localization of proteins, and crosstalk with other factors are crucial to probe and image proteins in vitro and in living cells. Alternatives to enzyme-based tags or autofluorescent proteins are short peptide-based recognition tags. These tags provide high specificity, enhanced binding rates, bioorthogonality, and versatility. Here, we report on recent applications of multivalent chelator heads, recognizing oligohistidine-tagged proteins. The striking features of this system has facilitated the analysis of protein complexes by single-molecule approaches.
Collapse
Affiliation(s)
- Ralph Wieneke
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue Str. 9, 60438, Frankfurt/M., Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue Str. 9, 60438, Frankfurt/M., Germany
| |
Collapse
|
61
|
Wieneke R, Tampé R. Multivalent Chelators for In Vivo Protein Labeling. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201811293] [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]
Affiliation(s)
- Ralph Wieneke
- Institute of BiochemistryBiocenterGoethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt/M. Germany
| | - Robert Tampé
- Institute of BiochemistryBiocenterGoethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt/M. Germany
| |
Collapse
|
62
|
Schwaminger SP, Fraga-García P, Blank-Shim SA, Straub T, Haslbeck M, Muraca F, Dawson KA, Berensmeier S. Magnetic One-Step Purification of His-Tagged Protein by Bare Iron Oxide Nanoparticles. ACS OMEGA 2019; 4:3790-3799. [PMID: 31459591 PMCID: PMC6648446 DOI: 10.1021/acsomega.8b03348] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/04/2019] [Indexed: 05/21/2023]
Abstract
Magnetic separation is a promising alternative to conventional methods in downstream processing. This can facilitate easier handling, fewer processing steps, and more sustainable processes. Target materials can be extracted directly from crude cell lysates in a single step by magnetic nanoadsorbents with high-gradient magnetic fishing (HGMF). Additionally, the use of hazardous consumables for reducing downstream processing steps can be avoided. Here, we present proof of principle of one-step magnetic fishing from crude Escherichia coli cell lysate of a green fluorescent protein (GFP) with an attached hexahistidine (His6)-tag, which is used as the model target molecule. The focus of this investigation is the upscale to a liter scale magnetic fishing process in which a purity of 91% GFP can be achieved in a single purification step from cleared cell lysate. The binding through the His6-tag can be demonstrated, since no significant binding of nontagged GFP toward bare iron oxide nanoparticles (BIONs) can be observed. Nonfunctionalized BIONs with primary particle diameters of around 12 nm, as used in the process, can be produced with a simple and low-cost coprecipitation synthesis. Thus, HGMF with BIONs might pave the way for a new and greener era of downstream processing.
Collapse
Affiliation(s)
- Sebastian P. Schwaminger
- Bioseparation
Engineering Group, Department of Mechanical Engineering and Department of
Chemistry, Technical University of Munich, Garching 85748, Germany
- Centre
for BioNano Interactions, School of Chemistry and Chemical Biology
and Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin D14 YH57, Ireland
| | - Paula Fraga-García
- Bioseparation
Engineering Group, Department of Mechanical Engineering and Department of
Chemistry, Technical University of Munich, Garching 85748, Germany
| | - Silvia A. Blank-Shim
- Bioseparation
Engineering Group, Department of Mechanical Engineering and Department of
Chemistry, Technical University of Munich, Garching 85748, Germany
| | - Tamara Straub
- Bioseparation
Engineering Group, Department of Mechanical Engineering and Department of
Chemistry, Technical University of Munich, Garching 85748, Germany
| | - Martin Haslbeck
- Bioseparation
Engineering Group, Department of Mechanical Engineering and Department of
Chemistry, Technical University of Munich, Garching 85748, Germany
| | - Francesco Muraca
- Centre
for BioNano Interactions, School of Chemistry and Chemical Biology
and Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin D14 YH57, Ireland
| | - Kenneth A. Dawson
- Centre
for BioNano Interactions, School of Chemistry and Chemical Biology
and Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin D14 YH57, Ireland
| | - Sonja Berensmeier
- Bioseparation
Engineering Group, Department of Mechanical Engineering and Department of
Chemistry, Technical University of Munich, Garching 85748, Germany
| |
Collapse
|
63
|
Pan L, Fu TM, Zhao W, Zhao L, Chen W, Qiu C, Liu W, Liu Z, Piai A, Fu Q, Chen S, Wu H, Chou JJ. Higher-Order Clustering of the Transmembrane Anchor of DR5 Drives Signaling. Cell 2019; 176:1477-1489.e14. [PMID: 30827683 DOI: 10.1016/j.cell.2019.02.001] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 11/14/2018] [Accepted: 01/29/2019] [Indexed: 12/18/2022]
Abstract
Receptor clustering on the cell membrane is critical in the signaling of many immunoreceptors, and this mechanism has previously been attributed to the extracellular and/or the intracellular interactions. Here, we report an unexpected finding that for death receptor 5 (DR5), a receptor in the tumor necrosis factor receptor superfamily, the transmembrane helix (TMH) alone in the receptor directly assembles a higher-order structure to drive signaling and that this structure is inhibited by the unliganded ectodomain. Nuclear magnetic resonance structure of the TMH in bicelles shows distinct trimerization and dimerization faces, allowing formation of dimer-trimer interaction networks. Single-TMH mutations that disrupt either trimerization or dimerization abolish ligand-induced receptor activation. Surprisingly, proteolytic removal of the DR5 ectodomain can fully activate downstream signaling in the absence of ligand. Our data suggest a receptor activation mechanism in which binding of ligand or antibodies to overcome the pre-ligand autoinhibition allows TMH clustering and thus signaling.
Collapse
Affiliation(s)
- Liqiang Pan
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Tian-Min Fu
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Wenbin Zhao
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Linlin Zhao
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Wen Chen
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Chixiao Qiu
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Wenhui Liu
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Zhijun Liu
- National Facility for Protein Science in Shanghai, ZhangJiang Lab, Chinese Academy of Sciences, 201210 Shanghai, China
| | - Alessandro Piai
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Qingshan Fu
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Shuqing Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 310058 Hangzhou, China.
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA.
| | - James J Chou
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
64
|
Di Russo J, Young JL, Balakrishnan A, Benk AS, Spatz JP. NTA-Co3+-His6 versus NTA-Ni2+-His6 mediated E-Cadherin surface immobilization enhances cellular traction. Biomaterials 2019; 192:171-178. [DOI: 10.1016/j.biomaterials.2018.10.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 10/23/2018] [Accepted: 10/28/2018] [Indexed: 01/09/2023]
|
65
|
Mortensen MR, Nielsen NL, Palmfeldt J, Gothelf KV. Imidazole carbamate probes for affinity guided azide-transfer to metal-binding proteins. Org Biomol Chem 2019; 17:1379-1383. [DOI: 10.1039/c8ob03017k] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Probes for affinity guided conjugation have shown great promise for the preparation of high-quality protein conjugates.
Collapse
Affiliation(s)
- Michael Rosholm Mortensen
- Center for Multifunctional Biomolecular Drug Design at the Interdisciplinary Nanoscience Center
- Aarhus University
- Aarhus C
- Denmark
- Department of Chemistry
| | - Nanna Louise Nielsen
- Center for Multifunctional Biomolecular Drug Design at the Interdisciplinary Nanoscience Center
- Aarhus University
- Aarhus C
- Denmark
- Department of Chemistry
| | - Johan Palmfeldt
- Research Unit for Molecular Medicine
- Aarhus University
- 8200 Aarhus N
- Denmark
| | - Kurt Vesterager Gothelf
- Center for Multifunctional Biomolecular Drug Design at the Interdisciplinary Nanoscience Center
- Aarhus University
- Aarhus C
- Denmark
- Department of Chemistry
| |
Collapse
|
66
|
Wang X, Liu Q, Tan X, Liu L, Zhou F. Covalent affixation of histidine-tagged proteins tethered onto Ni-nitrilotriacetic acid sensors for enhanced surface plasmon resonance detection of small molecule drugs and kinetic studies of antibody/antigen interactions. Analyst 2019; 144:587-593. [DOI: 10.1039/c8an01794h] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Covalent affixation of histidine-tagged proteins tethered onto Ni-nitrilotriacetic acid sensors for enhanced surface plasmon resonance detection.
Collapse
Affiliation(s)
- Xiaoying Wang
- College of Chemistry and Chemical Engineering
- Central South University
- Changsha
- P. R. China
| | - Qinghua Liu
- College of Chemistry and Chemical Engineering
- Central South University
- Changsha
- P. R. China
| | - Xiaofeng Tan
- Institute of Surface Analysis and Chemical Biology
- University of Jinan
- Jinan
- P. R. China
| | - Luyao Liu
- Institute of Surface Analysis and Chemical Biology
- University of Jinan
- Jinan
- P. R. China
| | - Feimeng Zhou
- Institute of Surface Analysis and Chemical Biology
- University of Jinan
- Jinan
- P. R. China
| |
Collapse
|
67
|
Tang Q, Zhao D, Yang H, Wang L, Zhang X. A pH-responsive self-healing hydrogel based on multivalent coordination of Ni2+ with polyhistidine-terminated PEG and IDA-modified oligochitosan. J Mater Chem B 2019; 7:30-42. [DOI: 10.1039/c8tb02360c] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A multivalent Ni2+ coordination hydrogel based on polyhistidine-terminated PEG and IDA-modified oligochitosan with enhanced neutral stability and mild-acid responsiveness is reported herein.
Collapse
Affiliation(s)
- Quan Tang
- CAS Key Laboratory of Soft Matter Chemistry
- School of Chemistry and Materials Science
- University of Science and Technology of China
- Hefei
- P. R. China
| | - Dinglei Zhao
- CAS Key Laboratory of Soft Matter Chemistry
- School of Chemistry and Materials Science
- University of Science and Technology of China
- Hefei
- P. R. China
| | - Haiyang Yang
- CAS Key Laboratory of Soft Matter Chemistry
- School of Chemistry and Materials Science
- University of Science and Technology of China
- Hefei
- P. R. China
| | - Lijun Wang
- CAS Key Laboratory of Soft Matter Chemistry
- School of Chemistry and Materials Science
- University of Science and Technology of China
- Hefei
- P. R. China
| | - Xingyuan Zhang
- CAS Key Laboratory of Soft Matter Chemistry
- School of Chemistry and Materials Science
- University of Science and Technology of China
- Hefei
- P. R. China
| |
Collapse
|
68
|
Hoogerheide DP, Noskov SY, Kuszak AJ, Buchanan SK, Rostovtseva TK, Nanda H. Structure of voltage-dependent anion channel-tethered bilayer lipid membranes determined using neutron reflectivity. Acta Crystallogr D Struct Biol 2018; 74:1219-1232. [PMID: 30605136 PMCID: PMC6317592 DOI: 10.1107/s2059798318011749] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 08/20/2018] [Indexed: 01/05/2023] Open
Abstract
Neutron reflectivity (NR) has emerged as a powerful technique to study the structure and behavior of membrane proteins at planar lipid interfaces. Integral membrane proteins (IMPs) remain a significant challenge for NR owing to the difficulty of forming complete bilayers with sufficient protein density for scattering techniques. One strategy to achieve high protein density on a solid substrate is the capture of detergent-stabilized, affinity-tagged IMPs on a nitrilotriacetic acid (NTA)-functionalized self-assembled monolayer (SAM), followed by reconstitution into the lipids of interest. Such protein-tethered bilayer lipid membranes (ptBLMs) have the notable advantage of a uniform IMP orientation on the substrate. Here, NR is used to provide a structural characterization of the ptBLM process from formation of the SAM to capture of the detergent-stabilized IMP and lipid reconstitution. The mitochondrial outer-membrane voltage-dependent anion channel (VDAC), which controls the exchange of bioenergetic metabolites between mitochondria and the cytosol, was used as a model β-barrel IMP. Molecular dynamics simulations were used for comparison with the experimental results and to inform the parameters of the physical models describing the NR data. The detailed structure of the SAM is shown to depend on the density of the NTA chelating groups. The relative content of detergent and protein in surface-immobilized, detergent-stabilized VDAC is measured, while the reconstituted lipid bilayer is shown to be complete to within a few percent, using the known atomic structure of VDAC. Finally, excess lipid above the reconstituted bilayer, which is of consequence for more indirect structural and functional studies, is shown to be present.
Collapse
Affiliation(s)
- David P. Hoogerheide
- Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, MD 20878, USA
| | - Sergei Yu. Noskov
- Centre for Molecular Simulations and Department of Biological Sciences, University of Calgary, Calgary T2N 1N4, Canada
| | - Adam J. Kuszak
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Susan K. Buchanan
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tatiana K. Rostovtseva
- Section on Molecular Transport, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hirsh Nanda
- Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, MD 20878, USA
- Physics Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
69
|
Kubota R, Nomura W, Iwasaka T, Ojima K, Kiyonaka S, Hamachi I. Chemogenetic Approach Using Ni(II) Complex-Agonist Conjugates Allows Selective Activation of Class A G-Protein-Coupled Receptors. ACS CENTRAL SCIENCE 2018; 4:1211-1221. [PMID: 30276255 PMCID: PMC6161059 DOI: 10.1021/acscentsci.8b00390] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Indexed: 05/04/2023]
Abstract
Investigating individual G-protein-coupled receptors (GPCRs) involved in various signaling cascades can unlock a myriad of invaluable physiological findings. One of the promising strategies for addressing the activity of each subtype of receptor is to design chemical turn-on switches on the target receptors. However, valid methods to selectively control class A GPCRs, the largest receptor family encoded in the human genome, remain limited. Here, we describe a novel approach to chemogenetically manipulate activity of engineered class A GPCRs carrying a His4 tag, using metal complex-agonist conjugates (MACs). This manipulation is termed coordination tethering. With the assistance of coordination bonds, MACs showed 10-100-fold lower EC50 values in the engineered receptors, compared with wild-type receptors. Such coordination tethering enabled selective activation of β2-adrenoceptors and muscarinic acetylcholine receptors, without loss of natural receptor responses, in living mammalian cells, including primary cultured astrocytes. Our generalized, modular chemogenetic approach should facilitate more precise control and deeper understanding of individual GPCR signaling pathways in living systems.
Collapse
Affiliation(s)
- Ryou Kubota
- Department
of Synthetic Chemistry and Biological Chemistry, Graduate School of
Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Wataru Nomura
- Department
of Synthetic Chemistry and Biological Chemistry, Graduate School of
Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Takuma Iwasaka
- Department
of Synthetic Chemistry and Biological Chemistry, Graduate School of
Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Kento Ojima
- Department
of Synthetic Chemistry and Biological Chemistry, Graduate School of
Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Shigeki Kiyonaka
- 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
- Core
Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), 5 Sanbancho, Chiyoda-ku, Tokyo 102-0075, Japan
- E-mail:
| |
Collapse
|
70
|
Gatterdam K, Joest EF, Gatterdam V, Tampé R. The Scaffold Design of Trivalent Chelator Heads Dictates Affinity and Stability for Labeling His-tagged Proteins in vitro and in Cells. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201802746] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Karl Gatterdam
- Institute of Biochemistry; Biocenter; Goethe University Frankfurt; Max-von-Laue-Str. 9 60438 Frankfurt/M Germany
| | - Eike F. Joest
- Institute of Biochemistry; Biocenter; Goethe University Frankfurt; Max-von-Laue-Str. 9 60438 Frankfurt/M Germany
| | - Volker Gatterdam
- Institute of Biochemistry; Biocenter; Goethe University Frankfurt; Max-von-Laue-Str. 9 60438 Frankfurt/M Germany
| | - Robert Tampé
- Institute of Biochemistry; Biocenter; Goethe University Frankfurt; Max-von-Laue-Str. 9 60438 Frankfurt/M Germany
| |
Collapse
|
71
|
Gatterdam K, Joest EF, Gatterdam V, Tampé R. The Scaffold Design of Trivalent Chelator Heads Dictates Affinity and Stability for Labeling His-tagged Proteins in vitro and in Cells. Angew Chem Int Ed Engl 2018; 57:12395-12399. [PMID: 29845721 DOI: 10.1002/anie.201802746] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/01/2018] [Indexed: 11/08/2022]
Abstract
Small chemical/biological interaction pairs are at the forefront in tracing protein function and interaction at high signal-to-background ratios in cellular pathways. However, the optimal design of scaffold, linker, and chelator head still deserve systematic investigation to achieve the highest affinity and kinetic stability for in vitro and especially cellular applications. We report on a library of N-nitrilotriacetic acid (NTA)-based multivalent chelator heads (MCHs) built on linear, cyclic, and dendritic scaffolds and compare these with regard to their binding affinity and stability for the labeling of cellular His-tagged proteins. Furthermore, we describe a new approach for tracing cellular target proteins at picomolar probe concentrations in cells. Finally, we outline fundamental differences between the MCH scaffolds and define a cyclic trisNTA chelator that displays the highest affinity and kinetic stability of all reported reversible, low-molecular-weight interaction pairs.
Collapse
Affiliation(s)
- Karl Gatterdam
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt/M, Germany
| | - Eike F Joest
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt/M, Germany
| | - Volker Gatterdam
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt/M, Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt/M, Germany
| |
Collapse
|
72
|
Endogenous Membrane Receptor Labeling by Reactive Cytokines and Growth Factors to Chase Their Dynamics in Live Cells. Chem 2018. [DOI: 10.1016/j.chempr.2018.03.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
73
|
Ananth A, Genua M, Aissaoui N, Díaz L, Eisele NB, Frey S, Dekker C, Richter RP, Görlich D. Reversible Immobilization of Proteins in Sensors and Solid-State Nanopores. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1703357. [PMID: 29611258 DOI: 10.1002/smll.201703357] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 02/13/2018] [Indexed: 06/08/2023]
Abstract
The controlled functionalization of surfaces with proteins is crucial for many analytical methods in life science research and biomedical applications. Here, a coating for silica-based surfaces is established which enables stable and selective immobilization of proteins with controlled orientation and tunable surface density. The coating is reusable, retains functionality upon long-term storage in air, and is applicable to surfaces of complex geometry. The protein anchoring method is validated on planar surfaces, and then a method is developed to measure the anchoring process in real time using silicon nitride solid-state nanopores. For surface attachment, polyhistidine tags that are site specifically introduced into recombinant proteins are exploited, and the yeast nucleoporin Nsp1 is used as model protein. Contrary to the commonly used covalent thiol chemistry, the anchoring of proteins via polyhistidine tag is reversible, permitting to take proteins off and replace them by other ones. Such switching in real time in experiments on individual nanopores is monitored using ion conductivity. Finally, it is demonstrated that silica and gold surfaces can be orthogonally functionalized to accommodate polyhistidine-tagged proteins on silica but prevent protein binding to gold, which extends the applicability of this surface functionalization method to even more complex sensor devices.
Collapse
Affiliation(s)
- Adithya Ananth
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands
| | - María Genua
- CIC biomaGUNE, Biosurfaces Lab, Paseo Miramon 182, 20014, San Sebastian, Spain
| | - Nesrine Aissaoui
- CIC biomaGUNE, Biosurfaces Lab, Paseo Miramon 182, 20014, San Sebastian, Spain
| | - Leire Díaz
- CIC biomaGUNE, Biosurfaces Lab, Paseo Miramon 182, 20014, San Sebastian, Spain
| | - Nico B Eisele
- CIC biomaGUNE, Biosurfaces Lab, Paseo Miramon 182, 20014, San Sebastian, Spain
| | - Steffen Frey
- Department for Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Cees Dekker
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands
| | - Ralf P Richter
- CIC biomaGUNE, Biosurfaces Lab, Paseo Miramon 182, 20014, San Sebastian, Spain
- Faculty of Biological Sciences, School of Biomedical Sciences, Faculty of Mathematics and Physical Sciences, School of Physics and Astronomy, and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Dirk Görlich
- Department for Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| |
Collapse
|
74
|
Gatterdam K, Joest EF, Dietz MS, Heilemann M, Tampé R. Super-Chelators for Advanced Protein Labeling in Living Cells. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201800827] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Karl Gatterdam
- Institute of Biochemistry; Biocenter; Goethe-University Frankfurt; Max-von-Laue-Str. 9 60438 Frankfurt/M Germany
| | - Eike F. Joest
- Institute of Biochemistry; Biocenter; Goethe-University Frankfurt; Max-von-Laue-Str. 9 60438 Frankfurt/M Germany
| | - Marina S. Dietz
- Institute of Physical and Theoretical Chemistry; Goethe-University Frankfurt; Max-von-Laue-Str. 9 60438 Frankfurt/M Germany
| | - Mike Heilemann
- Institute of Physical and Theoretical Chemistry; Goethe-University Frankfurt; Max-von-Laue-Str. 9 60438 Frankfurt/M Germany
| | - Robert Tampé
- Institute of Biochemistry; Biocenter; Goethe-University Frankfurt; Max-von-Laue-Str. 9 60438 Frankfurt/M Germany
| |
Collapse
|
75
|
Gatterdam K, Joest EF, Dietz MS, Heilemann M, Tampé R. Super-Chelators for Advanced Protein Labeling in Living Cells. Angew Chem Int Ed Engl 2018; 57:5620-5625. [PMID: 29464841 DOI: 10.1002/anie.201800827] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 02/17/2018] [Indexed: 12/17/2022]
Abstract
Live-cell labeling, super-resolution microscopy, single-molecule applications, protein localization, or chemically induced assembly are emerging approaches, which require specific and very small interaction pairs. The minimal disturbance of protein function is essential to derive unbiased insights into cellular processes. Herein, we define a new class of hexavalent N-nitrilotriacetic acid (hexaNTA) chelators, displaying the highest affinity and stability of all NTA-based small interaction pairs described so far. Coupled to bright organic fluorophores with fine-tuned photophysical properties, the super-chelator probes were delivered into human cells by chemically gated nanopores. These super-chelators permit kinetic profiling, multiplexed labeling of His6 - and His12 -tagged proteins as well as single-molecule-based super-resolution imaging.
Collapse
Affiliation(s)
- Karl Gatterdam
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt/M, Germany
| | - Eike F Joest
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt/M, Germany
| | - Marina S Dietz
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt/M, Germany
| | - Mike Heilemann
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt/M, Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt/M, Germany
| |
Collapse
|
76
|
Chen W, Dev J, Mezhyrova J, Pan L, Piai A, Chou JJ. The Unusual Transmembrane Partition of the Hexameric Channel of the Hepatitis C Virus. Structure 2018; 26:627-634.e4. [PMID: 29551287 PMCID: PMC5884736 DOI: 10.1016/j.str.2018.02.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 01/24/2018] [Accepted: 02/09/2018] [Indexed: 01/14/2023]
Abstract
The p7 protein of the hepatitis C virus (HCV) can oligomerize in membrane to form cation channels. Previous studies showed that the channel assembly in detergent micelles adopts a unique flower-shaped oligomer, but the unusual architecture also presented problems for understanding how this viroporin resides in the membrane. Moreover, the oligomeric state of p7 remains controversial, as both hexamer and heptamer have been proposed. Here we address the above issues using p7 reconstituted in bicelles that mimic a lipid bilayer. We found, using a recently developed oligomer-labeling method, that p7 forms hexamers in the bicelles. Solvent paramagnetic relaxation enhancement analyses showed that the bilayer thickness around the HCV ion channel is substantially smaller than expected, and thus a significant portion of the previously assigned membrane-embedded region is solvent exposed. Our study provides an effective approach for characterizing the transmembrane partition of small ion channels in near lipid bilayer environment.
Collapse
Affiliation(s)
- Wen Chen
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Jyoti Dev
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Julija Mezhyrova
- Institute of Biophysical Chemistry, Centre for Biomolecular Magnetic Resonance, J.W. Goethe-University, Frankfurt am Main, Germany
| | - Liqiang Pan
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Alessandro Piai
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - James J Chou
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
77
|
Posch S, Obser T, König G, Schneppenheim R, Tampé R, Hinterdorfer P. Interaction of von Willebrand factor domains with collagen investigated by single molecule force spectroscopy. J Chem Phys 2018; 148:123310. [DOI: 10.1063/1.5007313] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Sandra Posch
- Institute of Biophysics, Johannes Kepler University, Linz, Austria
| | - Tobias Obser
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gesa König
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Reinhard Schneppenheim
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | | |
Collapse
|
78
|
Bartoschik T, Galinec S, Kleusch C, Walkiewicz K, Breitsprecher D, Weigert S, Muller YA, You C, Piehler J, Vercruysse T, Daelemans D, Tschammer N. Near-native, site-specific and purification-free protein labeling for quantitative protein interaction analysis by MicroScale Thermophoresis. Sci Rep 2018; 8:4977. [PMID: 29563556 PMCID: PMC5862892 DOI: 10.1038/s41598-018-23154-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/05/2018] [Indexed: 12/21/2022] Open
Abstract
MicroScale Thermophoresis (MST) is a frequently used method for the quantitative characterization of intermolecular interactions with several advantages over other technologies. One of these is its capability to determine equilibrium constants in solution including complex biological matrices such as cell lysates. MST requires one binding partner to be fluorescent, which is typically achieved by labeling target proteins with a suitable fluorophore. Here, we present a near-native, site-specific in situ labeling strategy for MST experiments that enables reliable measurements in cell lysates and that has distinct advantages over routine covalent labeling techniques. To this end, we exploited the high-affinity interaction of tris-NTA with oligohistidine-tags, which are popular for purification, immobilization or detection of recombinant proteins. We used various DYE-tris-NTA conjugates to successfully label His-tagged proteins that were either purified or a component of cell lysate. The RED-tris-NTA was identified as the optimal dye conjugate with a high affinity towards oligohistidine-tags, a high fluorescence signal and an optimal signal-to-noise ratio in MST binding experiments. Owing to its emission in the red region of the spectrum, it also enables reliable measurements in complex biological matrices such as cell lysates allowing a more physiologically realistic assessment and eliminating the need for protein purification.
Collapse
Affiliation(s)
- Tanja Bartoschik
- NanoTemper Technologies GmbH, Floessergasse 4, 81069, München, Germany
| | - Stefanie Galinec
- NanoTemper Technologies GmbH, Floessergasse 4, 81069, München, Germany
| | - Christian Kleusch
- NanoTemper Technologies GmbH, Floessergasse 4, 81069, München, Germany
| | | | | | - Sebastian Weigert
- Division of Biotechnology, Department of Biology, Friedrich-Alexander University Erlangen, Nuremberg Henkestr 91, 91052, Erlangen, Germany
| | - Yves A Muller
- Division of Biotechnology, Department of Biology, Friedrich-Alexander University Erlangen, Nuremberg Henkestr 91, 91052, Erlangen, Germany
| | - Changjiang You
- Division of Biophysics, Department of Biology, University Osnabrück, Barbarastr 11, 49076, Osnabrück, Germany
| | - Jacob Piehler
- Division of Biophysics, Department of Biology, University Osnabrück, Barbarastr 11, 49076, Osnabrück, Germany
| | - Thomas Vercruysse
- KU Leuven Department of Immunology and Microbiology, Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Herestraat 49, 3000, Leuven, Belgium
| | - Dirk Daelemans
- KU Leuven Department of Immunology and Microbiology, Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Herestraat 49, 3000, Leuven, Belgium
| | - Nuska Tschammer
- NanoTemper Technologies GmbH, Floessergasse 4, 81069, München, Germany.
| |
Collapse
|
79
|
Evans ED, Pentelute BL. Discovery of a 29-Amino-Acid Reactive Abiotic Peptide for Selective Cysteine Arylation. ACS Chem Biol 2018; 13:527-532. [PMID: 29283243 DOI: 10.1021/acschembio.7b00520] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The regio- and chemoselective modification of proteins or peptides with chemical reagents is often challenging. One approach to overcome this problem involves identifying abiotic polypeptide sequences that react with specific small molecules. Toward this goal, we profiled ∼5 × 1013 randomized 30-mer peptides using mRNA display and high-throughput sequencing in search of polypeptides that can undergo cysteine arylation with a water-soluble perfluoroarene. Within this vast chemical space, we discovered a cysteine-containing sequence with a second-order rate constant of 0.29 M-1 s-1 for arylation. An N- and C-terminal truncation reduced the reaction rate, as did the addition of denaturants. When the reactive peptide was covalently fused to the enzyme Sortase A, we observed regiospecific arylation at a single cysteine site, leaving the enzyme's active site cysteine unchanged. Taken together, these results demonstrate that long polypeptides of defined sequence, when matched with the appropriate reactive group, can be used for selective arylation of cysteine in water.
Collapse
Affiliation(s)
- Ethan D. Evans
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Bradley L. Pentelute
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
80
|
Knoops B, Becker S, Poncin MA, Glibert J, Derclaye S, Clippe A, Alsteens D. Specific Interactions Measured by AFM on Living Cells between Peroxiredoxin-5 and TLR4: Relevance for Mechanisms of Innate Immunity. Cell Chem Biol 2018; 25:550-559.e3. [PMID: 29551349 DOI: 10.1016/j.chembiol.2018.02.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/08/2018] [Accepted: 02/07/2018] [Indexed: 12/14/2022]
Abstract
Inflammation is a pathophysiological response of innate immunity to infection or tissue damage. This response is among others triggered by factors released by damaged or dying cells, termed damage-associated molecular pattern (DAMP) molecules that act as danger signals. DAMPs interact with pattern recognition receptors (PRRs) to contribute to the induction of inflammation. However, how released peroxiredoxins (PRDXs) are able to activate PRRs, such as Toll-like receptors (TLRs), remains elusive. Here, we used force-distance curve-based atomic force microscopy to investigate the molecular mechanisms by which extracellular human PRDX5 can activate a proinflammatory response. Single-molecule experiments demonstrated that PRDX5 binds to purified TLR4 receptors, on macrophage-differentiated THP-1 cells, and on human TLR4-transfected CHO cells. These findings suggest that extracellular PRDX5 can specifically trigger a proinflammatory response. Moreover, our work also revealed that PRDX5 binding induces a cellular mechanoresponse. Collectively, this study provides insights into the role of extracellular PRDX5 in innate immunity.
Collapse
Affiliation(s)
- Bernard Knoops
- Université catholique de Louvain, Institut des Sciences de la Vie (ISV), 1348 Louvain-la-Neuve, Belgium.
| | - Sarah Becker
- Université catholique de Louvain, Institut des Sciences de la Vie (ISV), 1348 Louvain-la-Neuve, Belgium
| | - Mégane Anne Poncin
- Université catholique de Louvain, Institut des Sciences de la Vie (ISV), 1348 Louvain-la-Neuve, Belgium
| | - Julien Glibert
- Université catholique de Louvain, Institut des Sciences de la Vie (ISV), 1348 Louvain-la-Neuve, Belgium
| | - Sylvie Derclaye
- Université catholique de Louvain, Institut des Sciences de la Vie (ISV), 1348 Louvain-la-Neuve, Belgium
| | - André Clippe
- Université catholique de Louvain, Institut des Sciences de la Vie (ISV), 1348 Louvain-la-Neuve, Belgium
| | - David Alsteens
- Université catholique de Louvain, Institut des Sciences de la Vie (ISV), 1348 Louvain-la-Neuve, Belgium.
| |
Collapse
|
81
|
Delsuc N, Uchinomiya S, Ojida A, Hamachi I. A host-guest system based on collagen-like triple-helix hybridization. Chem Commun (Camb) 2018; 53:6856-6859. [PMID: 28604910 DOI: 10.1039/c7cc03055j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A strategy inspired by tweezer receptors has been employed to develop a new host-guest system. The hybridization into a collagen-like triple helix is the driving force for the recognition that occurs with high affinity and selectivity. Several systems have been screened to find the best host-guest pair and this strategy may be implemented for tag fused protein recognition.
Collapse
Affiliation(s)
- N Delsuc
- Laboratoire des Biomolécules, Département de Chimie, Ecole Normale Supérieure, PSL Research University, Sorbonne Universités, UPMC Univ Paris 06, CNRS, 24, rue Lhomond, 75005 Paris, France.
| | | | | | | |
Collapse
|
82
|
Lam ATC, Tsitkov S, Zhang Y, Hess H. Reversibly Bound Kinesin-1 Motor Proteins Propelling Microtubules Demonstrate Dynamic Recruitment of Active Building Blocks. NANO LETTERS 2018; 18:1530-1534. [PMID: 29318877 DOI: 10.1021/acs.nanolett.7b05361] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Biological materials and systems often dynamically self-assemble and disassemble, forming temporary structures as needed and allowing for dynamic responses to stimuli and changing environmental conditions. However, this dynamic interplay of localized component recruitment and release has been difficult to achieve in artificial molecular-scale systems, which are usually designed to have long-lasting, stable bonds. Here, we report the experimental realization of a molecular-scale system that dynamically assembles and disassembles its building blocks while retaining functionality. In our system, filaments (microtubules) recruit biomolecular motors (kinesins) to a surface engineered to allow for the reversible binding of the kinesin-1 motors. These recruited motors work to propel the cytoskeletal filaments along the surface. After the microtubules leave the motors behind, the trail of motors disassembles, releasing the motors back into solution. Engineering such dynamic systems may allow us to create materials that mimic the way in which biological systems achieve self-healing and adaptation.
Collapse
Affiliation(s)
- Amy Tsui-Chi Lam
- Department of Biomedical Engineering, Columbia University , New York City, New York 10027, United States
| | - Stanislav Tsitkov
- Department of Biomedical Engineering, Columbia University , New York City, New York 10027, United States
| | - Yifei Zhang
- Department of Biomedical Engineering, Columbia University , New York City, New York 10027, United States
| | - Henry Hess
- Department of Biomedical Engineering, Columbia University , New York City, New York 10027, United States
| |
Collapse
|
83
|
Terry DS, Kolster RA, Quick M, LeVine MV, Khelashvili G, Zhou Z, Weinstein H, Javitch JA, Blanchard SC. A partially-open inward-facing intermediate conformation of LeuT is associated with Na + release and substrate transport. Nat Commun 2018; 9:230. [PMID: 29335402 PMCID: PMC5768729 DOI: 10.1038/s41467-017-02202-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 11/12/2017] [Indexed: 12/20/2022] Open
Abstract
Neurotransmitter:sodium symporters (NSS), targets of antidepressants and psychostimulants, clear neurotransmitters from the synaptic cleft through sodium (Na+)-coupled transport. Substrate and Na+ are thought to be transported from the extracellular to intracellular space through an alternating access mechanism by coordinated conformational rearrangements in the symporter that alternately expose the binding sites to each side of the membrane. However, the mechanism by which the binding of ligands coordinates conformational changes occurring on opposite sides of the membrane is not well understood. Here, we report the use of single-molecule fluorescence resonance energy transfer (smFRET) techniques to image transitions between distinct conformational states on both the extracellular and intracellular sides of the prokaryotic NSS LeuT, including partially open intermediates associated with transport activity. The nature and functional context of these hitherto unidentified intermediate states shed new light on the allosteric mechanism that couples substrate and Na+ symport by the NSS family through conformational dynamics.
Collapse
Affiliation(s)
- Daniel S Terry
- Department of Physiology and Biophysics, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Rachel A Kolster
- Department of Psychiatry, Division of Molecular Therapeutics, Columbia University College of Physicians and Surgeons and New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA
| | - Matthias Quick
- Department of Psychiatry, Division of Molecular Therapeutics, Columbia University College of Physicians and Surgeons and New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA
| | - Michael V LeVine
- Department of Physiology and Biophysics, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, 1305 York Avenue, New York, NY, 10021, USA
| | - George Khelashvili
- Department of Physiology and Biophysics, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, 1305 York Avenue, New York, NY, 10021, USA
| | - Zhou Zhou
- Department of Physiology and Biophysics, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Harel Weinstein
- Department of Physiology and Biophysics, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, 1305 York Avenue, New York, NY, 10021, USA
| | - Jonathan A Javitch
- Department of Psychiatry, Division of Molecular Therapeutics, Columbia University College of Physicians and Surgeons and New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA.
- Department of Pharmacology, Columbia University College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY, 10032, USA.
| | - Scott C Blanchard
- Department of Physiology and Biophysics, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
84
|
Bartelt SM, Chervyachkova E, Steinkühler J, Ricken J, Wieneke R, Tampé R, Dimova R, Wegner SV. Dynamic blue light-switchable protein patterns on giant unilamellar vesicles. Chem Commun (Camb) 2018; 54:948-951. [DOI: 10.1039/c7cc08758f] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The photoswitchable iLID/Nano interaction allows for specific, non-invasive, reversible and dynamic protein photopatterning on GUVs with high spatiotemporal control.
Collapse
Affiliation(s)
- S. M. Bartelt
- Max Planck Institute for Polymer Research
- Mainz
- Germany
| | | | - J. Steinkühler
- Department of Theory and Biosystems
- Max Planck Institute of Colloids and Interfaces
- Potsdam
- Germany
| | - J. Ricken
- Max Planck Institute for Polymer Research
- Mainz
- Germany
| | - R. Wieneke
- Institut für Biochemie, Biozentrum
- Cluster of Excellence Frankfurt
- Goethe-Universität Frankfurt
- Frankfurt
- Germany
| | - R. Tampé
- Institut für Biochemie, Biozentrum
- Cluster of Excellence Frankfurt
- Goethe-Universität Frankfurt
- Frankfurt
- Germany
| | - R. Dimova
- Department of Theory and Biosystems
- Max Planck Institute of Colloids and Interfaces
- Potsdam
- Germany
| | - S. V. Wegner
- Max Planck Institute for Polymer Research
- Mainz
- Germany
| |
Collapse
|
85
|
Abstract
Virus-like particle (VLP) technologies are based on virus-inspired artificial structures and the intrinsic ability of viral proteins to self-assemble at controlled conditions. Therefore, the basic knowledge about the mechanisms of viral particle formation is highly important for designing of industrial applications. As an alternative to genetic and chemical processes, different physical methods are frequently used for VLP construction, including well characterized protein complexes for introduction of foreign molecules in VLP structures.This chapter shortly discusses the mechanisms how the viruses form their perfectly ordered structures as well as the principles and most interesting application examples, how to exploit the structural and assembly/disassembly properties of viral structures for creation of new nanomaterials.
Collapse
Affiliation(s)
- Andris Zeltins
- Latvian Biomedical Research and Study Centre, Riga, Latvia.
| |
Collapse
|
86
|
Schartner J, Güldenhaupt J, Katharina Gaßmeyer S, Rosga K, Kourist R, Gerwert K, Kötting C. Highly stable protein immobilizationviamaleimido-thiol chemistry to monitor enzymatic activity. Analyst 2018; 143:2276-2284. [DOI: 10.1039/c8an00301g] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Combining a novel protein immobilisation method with multivariate curve resolution enables the direct observation of biocatalysis by ATR-FTIR spectroscopy.
Collapse
Affiliation(s)
- Jonas Schartner
- Department of Biophysics
- Ruhr-Universität Bochum
- 44801 Bochum
- Germany
| | - Jörn Güldenhaupt
- Department of Biophysics
- Ruhr-Universität Bochum
- 44801 Bochum
- Germany
| | | | - Katharina Rosga
- Department of Biophysics
- Ruhr-Universität Bochum
- 44801 Bochum
- Germany
| | - Robert Kourist
- Junior Research Group for Microbial Biotechnology
- Ruhr-Universität Bochum
- 44801 Bochum
- Germany
| | - Klaus Gerwert
- Department of Biophysics
- Ruhr-Universität Bochum
- 44801 Bochum
- Germany
| | - Carsten Kötting
- Department of Biophysics
- Ruhr-Universität Bochum
- 44801 Bochum
- Germany
| |
Collapse
|
87
|
The molecular basis of phosphite and hypophosphite recognition by ABC-transporters. Nat Commun 2017; 8:1746. [PMID: 29170493 PMCID: PMC5700983 DOI: 10.1038/s41467-017-01226-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 08/30/2017] [Indexed: 11/10/2022] Open
Abstract
Inorganic phosphate is the major bioavailable form of the essential nutrient phosphorus. However, the concentration of phosphate in most natural habitats is low enough to limit microbial growth. Under phosphate-depleted conditions some bacteria utilise phosphite and hypophosphite as alternative sources of phosphorus, but the molecular basis of reduced phosphorus acquisition from the environment is not fully understood. Here, we present crystal structures and ligand binding affinities of periplasmic binding proteins from bacterial phosphite and hypophosphite ATP-binding cassette transporters. We reveal that phosphite and hypophosphite specificity results from a combination of steric selection and the presence of a P-H…π interaction between the ligand and a conserved aromatic residue in the ligand-binding pocket. The characterisation of high affinity and specific transporters has implications for the marine phosphorus redox cycle, and might aid the use of phosphite as an alternative phosphorus source in biotechnological, industrial and agricultural applications. Some bacteria can use inorganic phosphite and hypophosphite as sources of inorganic phosphorus. Here, the authors report crystal structures of the periplasmic proteins that bind these reduced phosphorus species and show that a P-H…π interaction between the ligand and binding site determines their specificity.
Collapse
|
88
|
Dyla M, Terry DS, Kjaergaard M, Sørensen TLM, Lauwring Andersen J, Andersen JP, Rohde Knudsen C, Altman RB, Nissen P, Blanchard SC. Dynamics of P-type ATPase transport revealed by single-molecule FRET. Nature 2017; 551:346-351. [PMID: 29144454 DOI: 10.1038/nature24296] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 09/18/2017] [Indexed: 01/05/2023]
Abstract
Phosphorylation-type (P-type) ATPases are ubiquitous primary transporters that pump cations across cell membranes through the formation and breakdown of a phosphoenzyme intermediate. Structural investigations suggest that the transport mechanism is defined by conformational changes in the cytoplasmic domains of the protein that are allosterically coupled to transmembrane helices so as to expose ion binding sites to alternate sides of the membrane. Here, we have used single-molecule fluorescence resonance energy transfer to directly observe conformational changes associated with the functional transitions in the Listeria monocytogenes Ca2+-ATPase (LMCA1), an orthologue of eukaryotic Ca2+-ATPases. We identify key intermediates with no known crystal structures and show that Ca2+ efflux by LMCA1 is rate-limited by phosphoenzyme formation. The transport process involves reversible steps and an irreversible step that follows release of ADP and extracellular release of Ca2+.
Collapse
Affiliation(s)
- Mateusz Dyla
- Centre for Membrane Pumps in Cells and Disease - PUMPKIN, Danish National Research Foundation & Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, DK-8000 Aarhus C, Denmark.,Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C, Denmark.,Interdisciplinary Nanoscience Center (iNANO), Aarhus University, DK-8000 Aarhus C, Denmark
| | - Daniel S Terry
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, New York 10021, USA
| | - Magnus Kjaergaard
- Centre for Membrane Pumps in Cells and Disease - PUMPKIN, Danish National Research Foundation & Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, DK-8000 Aarhus C, Denmark.,Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C, Denmark.,Interdisciplinary Nanoscience Center (iNANO), Aarhus University, DK-8000 Aarhus C, Denmark.,Aarhus Institute of Advanced Studies (AIAS), Aarhus University, DK-8000 Aarhus C, Denmark
| | - Thomas L-M Sørensen
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK
| | - Jacob Lauwring Andersen
- Centre for Membrane Pumps in Cells and Disease - PUMPKIN, Danish National Research Foundation & Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, DK-8000 Aarhus C, Denmark.,Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Jens P Andersen
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | | | - Roger B Altman
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, New York 10021, USA
| | - Poul Nissen
- Centre for Membrane Pumps in Cells and Disease - PUMPKIN, Danish National Research Foundation & Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, DK-8000 Aarhus C, Denmark.,Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C, Denmark.,Interdisciplinary Nanoscience Center (iNANO), Aarhus University, DK-8000 Aarhus C, Denmark
| | - Scott C Blanchard
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, New York 10021, USA
| |
Collapse
|
89
|
Li C, Wu G, Ma R, Liu Y, Liu Y, Lv J, An Y, Shi L. Nitrilotriacetic Acid (NTA) and Phenylboronic Acid (PBA) Functionalized Nanogels for Efficient Encapsulation and Controlled Release of Insulin. ACS Biomater Sci Eng 2017; 4:2007-2017. [DOI: 10.1021/acsbiomaterials.7b00546] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Chang Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Gang Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Rujiang Ma
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yong Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Ying Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Juan Lv
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yingli An
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| |
Collapse
|
90
|
Wasserberg D, Cabanas-Danés J, Prangsma J, O’Mahony S, Cazade PA, Tromp E, Blum C, Thompson D, Huskens J, Subramaniam V, Jonkheijm P. Controlling Protein Surface Orientation by Strategic Placement of Oligo-Histidine Tags. ACS NANO 2017; 11:9068-9083. [PMID: 28850777 PMCID: PMC5618149 DOI: 10.1021/acsnano.7b03717] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/29/2017] [Indexed: 05/24/2023]
Abstract
We report oriented immobilization of proteins using the standard hexahistidine (His6)-Ni2+:NTA (nitrilotriacetic acid) methodology, which we systematically tuned to give control of surface coverage. Fluorescence microscopy and surface plasmon resonance measurements of self-assembled monolayers (SAMs) of red fluorescent proteins (TagRFP) showed that binding strength increased by 1 order of magnitude for each additional His6-tag on the TagRFP proteins. All TagRFP variants with His6-tags located on only one side of the barrel-shaped protein yielded a 1.5 times higher surface coverage compared to variants with His6-tags on opposite sides of the so-called β-barrel. Time-resolved fluorescence anisotropy measurements supported by polarized infrared spectroscopy verified that the orientation (and thus coverage and functionality) of proteins on surfaces can be controlled by strategic placement of a His6-tag on the protein. Molecular dynamics simulations show how the differently tagged proteins reside at the surface in "end-on" and "side-on" orientations with each His6-tag contributing to binding. Also, not every dihistidine subunit in a given His6-tag forms a full coordination bond with the Ni2+:NTA SAMs, which varied with the position of the His6-tag on the protein. At equal valency but different tag positions on the protein, differences in binding were caused by probing for Ni2+:NTA moieties and by additional electrostatic interactions between different fractions of the β-barrel structure and charged NTA moieties. Potential of mean force calculations indicate there is no specific single-protein interaction mode that provides a clear preferential surface orientation, suggesting that the experimentally measured preference for the end-on orientation is a supra-protein, not a single-protein, effect.
Collapse
Affiliation(s)
- Dorothee Wasserberg
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Jordi Cabanas-Danés
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Jord Prangsma
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Shane O’Mahony
- Department
of Physics, Bernal Institute, University
of Limerick, Limerick V94 T9PX, Ireland
| | - Pierre-Andre Cazade
- Department
of Physics, Bernal Institute, University
of Limerick, Limerick V94 T9PX, Ireland
| | - Eldrich Tromp
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Christian Blum
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Damien Thompson
- Department
of Physics, Bernal Institute, University
of Limerick, Limerick V94 T9PX, Ireland
| | - Jurriaan Huskens
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Vinod Subramaniam
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
- Free
University of Amsterdam, De Boelelaan 1105, 1081 HV Amsterdam, The Netherlands
| | - Pascal Jonkheijm
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| |
Collapse
|
91
|
Bührmann M, Wiedemann BM, Müller MP, Hardick J, Ecke M, Rauh D. Structure-based design, synthesis and crystallization of 2-arylquinazolines as lipid pocket ligands of p38α MAPK. PLoS One 2017; 12:e0184627. [PMID: 28892510 PMCID: PMC5593189 DOI: 10.1371/journal.pone.0184627] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/28/2017] [Indexed: 12/27/2022] Open
Abstract
In protein kinase research, identifying and addressing small molecule binding sites other than the highly conserved ATP-pocket are of intense interest because this line of investigation extends our understanding of kinase function beyond the catalytic phosphotransfer. Such alternative binding sites may be involved in altering the activation state through subtle conformational changes, control cellular enzyme localization, or in mediating and disrupting protein-protein interactions. Small organic molecules that target these less conserved regions might serve as tools for chemical biology research and to probe alternative strategies in targeting protein kinases in disease settings. Here, we present the structure-based design and synthesis of a focused library of 2-arylquinazoline derivatives to target the lipophilic C-terminal binding pocket in p38α MAPK, for which a clear biological function has yet to be identified. The interactions of the ligands with p38α MAPK was analyzed by SPR measurements and validated by protein X-ray crystallography.
Collapse
Affiliation(s)
- Mike Bührmann
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Bianca M. Wiedemann
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Matthias P. Müller
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Julia Hardick
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Maria Ecke
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Daniel Rauh
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
- * E-mail:
| |
Collapse
|
92
|
Laskowski PR, Pfreundschuh M, Stauffer M, Ucurum Z, Fotiadis D, Müller DJ. High-Resolution Imaging and Multiparametric Characterization of Native Membranes by Combining Confocal Microscopy and an Atomic Force Microscopy-Based Toolbox. ACS NANO 2017; 11:8292-8301. [PMID: 28745869 DOI: 10.1021/acsnano.7b03456] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
To understand how membrane proteins function requires characterizing their structure, assembly, and inter- and intramolecular interactions in physiologically relevant conditions. Conventionally, such multiparametric insight is revealed by applying different biophysical methods. Here we introduce the combination of confocal microscopy, force-distance curve-based (FD-based) atomic force microscopy (AFM), and single-molecule force spectroscopy (SMFS) for the identification of native membranes and the subsequent multiparametric analysis of their membrane proteins. As a well-studied model system, we use native purple membrane from Halobacterium salinarum, whose membrane protein bacteriorhodopsin was His-tagged to bind nitrilotriacetate (NTA) ligands. First, by confocal microscopy we localize the extracellular and cytoplasmic surfaces of purple membrane. Then, we apply AFM to image single bacteriorhodopsins approaching sub-nanometer resolution. Afterwards, the binding of NTA ligands to bacteriorhodopsins is localized and quantified by FD-based AFM. Finally, we apply AFM-based SMFS to characterize the (un)folding of the membrane protein and to structurally map inter- and intramolecular interactions. The multimethodological approach is generally applicable to characterize biological membranes and membrane proteins at physiologically relevant conditions.
Collapse
Affiliation(s)
- Pawel R Laskowski
- Department of Biosystems Science and Engineering, ETH Zurich , 4058 Basel, Switzerland
| | - Moritz Pfreundschuh
- Department of Biosystems Science and Engineering, ETH Zurich , 4058 Basel, Switzerland
| | - Mirko Stauffer
- Institute of Biochemistry and Molecular Medicine, University of Bern , 3012 Bern, Switzerland
| | - Zöhre Ucurum
- Institute of Biochemistry and Molecular Medicine, University of Bern , 3012 Bern, Switzerland
| | - Dimitrios Fotiadis
- Institute of Biochemistry and Molecular Medicine, University of Bern , 3012 Bern, Switzerland
| | - Daniel J Müller
- Department of Biosystems Science and Engineering, ETH Zurich , 4058 Basel, Switzerland
| |
Collapse
|
93
|
Development of an ON/OFF switchable fluorescent probe targeting His tag fused proteins in living cells. Bioorg Med Chem Lett 2017. [DOI: 10.1016/j.bmcl.2017.05.087] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
94
|
Okitsu K, Hattori T, Misawa T, Shoda T, Kurihara M, Naito M, Demizu Y. Development of a Small Hybrid Molecule That Mediates Degradation of His-Tag Fused Proteins. J Med Chem 2017; 61:576-582. [DOI: 10.1021/acs.jmedchem.7b00413] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Koyo Okitsu
- Division
of Organic Chemistry, National Institute of Health Sciences, 1-18-1,
Kamiyoga, Setagaya, Tokyo 158-8501, Japan
- Graduate
School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Takayuki Hattori
- Division
of Molecular Target and Gene Therapy Products, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya, Tokyo 158-8501, Japan
| | - Takashi Misawa
- Division
of Organic Chemistry, National Institute of Health Sciences, 1-18-1,
Kamiyoga, Setagaya, Tokyo 158-8501, Japan
| | - Takuji Shoda
- Division
of Organic Chemistry, National Institute of Health Sciences, 1-18-1,
Kamiyoga, Setagaya, Tokyo 158-8501, Japan
| | - Masaaki Kurihara
- Division
of Organic Chemistry, National Institute of Health Sciences, 1-18-1,
Kamiyoga, Setagaya, Tokyo 158-8501, Japan
| | - Mikihiko Naito
- Division
of Molecular Target and Gene Therapy Products, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya, Tokyo 158-8501, Japan
| | - Yosuke Demizu
- Division
of Organic Chemistry, National Institute of Health Sciences, 1-18-1,
Kamiyoga, Setagaya, Tokyo 158-8501, Japan
| |
Collapse
|
95
|
Gilles P, Wenck K, Stratmann I, Kirsch M, Smolin DA, Schaller T, de Groot H, Kraft A, Schrader T. High-Affinity Copolymers Inhibit Digestive Enzymes by Surface Recognition. Biomacromolecules 2017; 18:1772-1784. [DOI: 10.1021/acs.biomac.7b00162] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | | | | | | | | | | | - Arno Kraft
- Heriot-Watt University, Edinburgh, Scotland, United Kingdom EH14 4AS
| | | |
Collapse
|
96
|
Kankate L, Aguf A, Großmann H, Schnietz M, Tampé R, Turchanin A, Gölzhäuser A. Vapor Phase Exchange of Self-Assembled Monolayers for Engineering of Biofunctional Surfaces. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:3847-3854. [PMID: 28340533 DOI: 10.1021/acs.langmuir.6b04207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
We show that 4'-nitro-1,1'-biphenyl-4-thiol self-assembled monolayers (NBPT SAMs) on gold can be exchanged with 11-(mercaptoundecyl)triethylene glycol (C11EG3OH) SAMs via vapor deposition (VD). The pristine and the exchanged SAMs obtained by VD as well as solution method (SM) were characterized by X-ray photoelectron spectroscopy (XPS) and polarization modulation infrared reflection-absorption spectroscopy (PM-IRRAS). Using surface plasmon resonance (SPR), it is shown that C11EG3OH SAMs on gold obtained by vapor deposition exchange (VDEx) have the same protein resistivity as SAMs obtained by the direct self-assembly process. As expected, the cross-linked NBPT SAM are found to be resistive to both exchange processes, VDEx and solution method exchange (SMEx). In this way, VDEx opens up an elegant and new approach of patterning SAM surfaces in situ at vacuum conditions without using any solvents. By combining electron irradiation-induced chemical lithography of NBPT SAMs with VDEx, biofunctional patterned substrates were engineered and used for immobilization of protein arrays.
Collapse
Affiliation(s)
- L Kankate
- Department of Physics, Physics of Supramolecular Systems and Surfaces, University of Bielefeld , 33615 Bielefeld, Germany
- Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195 Berlin, Germany
| | - A Aguf
- Department of Physics, Physics of Supramolecular Systems and Surfaces, University of Bielefeld , 33615 Bielefeld, Germany
| | - H Großmann
- Institute of Biochemistry, Biocenter, Goethe-University , 60438 Frankfurt, Germany
| | - M Schnietz
- Department of Physics, Physics of Supramolecular Systems and Surfaces, University of Bielefeld , 33615 Bielefeld, Germany
| | - R Tampé
- Institute of Biochemistry, Biocenter, Goethe-University , 60438 Frankfurt, Germany
| | - A Turchanin
- Institute of Physical Chemistry, Friedrich Schiller University Jena , Lessingstr. 10, 07743 Jena, Germany
| | - A Gölzhäuser
- Department of Physics, Physics of Supramolecular Systems and Surfaces, University of Bielefeld , 33615 Bielefeld, Germany
| |
Collapse
|
97
|
Hager R, Halilovic A, Burns JR, Schäffler F, Howorka S. Arrays of Individual DNA Molecules on Nanopatterned Substrates. Sci Rep 2017; 7:42075. [PMID: 28198806 PMCID: PMC5304548 DOI: 10.1038/srep42075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/03/2017] [Indexed: 02/02/2023] Open
Abstract
Arrays of individual molecules can combine the advantages of microarrays and single-molecule studies. They miniaturize assays to reduce sample and reagent consumption and increase throughput, and additionally uncover static and dynamic heterogeneity usually masked in molecular ensembles. However, realizing single-DNA arrays must tackle the challenge of capturing structurally highly dynamic strands onto defined substrate positions. Here, we create single-molecule arrays by electrostatically adhering single-stranded DNA of gene-like length onto positively charged carbon nanoislands. The nanosites are so small that only one molecule can bind per island. Undesired adsorption of DNA to the surrounding non-target areas is prevented via a surface-passivating film. Of further relevance, the DNA arrays are of tunable dimensions, and fabricated on optically transparent substrates that enable singe-molecule detection with fluorescence microscopy. The arrays are hence compatible with a wide range of bioanalytical, biophysical, and cell biological studies where individual DNA strands are either examined in isolation, or interact with other molecules or cells.
Collapse
Affiliation(s)
- Roland Hager
- Center for Advanced Bioanalysis GmbH, 4020 Linz, Austria
| | - Alma Halilovic
- Institute for Semiconductor and Solid State Physics, Johannes Kepler University, 4040 Linz, Austria
| | - Jonathan R. Burns
- Department of Chemistry, Institute of Structural and Molecular Biology, University College London, London, England, United Kingdom
| | - Friedrich Schäffler
- Institute for Semiconductor and Solid State Physics, Johannes Kepler University, 4040 Linz, Austria
| | - Stefan Howorka
- Center for Advanced Bioanalysis GmbH, 4020 Linz, Austria
- Department of Chemistry, Institute of Structural and Molecular Biology, University College London, London, England, United Kingdom
| |
Collapse
|
98
|
Cai K, Tan Y, Tan C, Wu J, Wu P, Liang J, Liu S, Zhang B, Jiang Y. An iminodiacetate-modified conjugated polyelectrolyte for fluorescent labeling of histidine-tagged proteins. Chem Commun (Camb) 2017; 53:4191-4194. [DOI: 10.1039/c7cc00850c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An iminodiacetate-modified conjugated polyelectrolyte was used in the fluorescent labeling of hexahistidine-tagged proteins.
Collapse
Affiliation(s)
- Kai Cai
- Department of Chemistry
- Tsinghua University
- Beijing 100084
- P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology
| | - Ying Tan
- Department of Chemistry
- Tsinghua University
- Beijing 100084
- P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology
| | - Chunyan Tan
- Department of Chemistry
- Tsinghua University
- Beijing 100084
- P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology
| | - Jiatao Wu
- Department of Chemistry
- Tsinghua University
- Beijing 100084
- P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology
| | - Pan Wu
- Department of Chemistry
- Tsinghua University
- Beijing 100084
- P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology
| | - Jiamei Liang
- Department of Chemistry
- Tsinghua University
- Beijing 100084
- P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology
| | - Shuwen Liu
- Department of Chemistry
- Tsinghua University
- Beijing 100084
- P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology
| | - Bibo Zhang
- Department of Chemistry
- Tsinghua University
- Beijing 100084
- P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology
| | - Yuyang Jiang
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology
- the Graduate School at Shenzhen
- Tsinghua University
- Shenzhen 518055
- P. R. China
| |
Collapse
|
99
|
Salimi K, Usta DD, Koçer İ, Çelik E, Tuncel A. Highly selective magnetic affinity purification of histidine-tagged proteins by Ni2+ carrying monodisperse composite microspheres. RSC Adv 2017. [DOI: 10.1039/c6ra27736e] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A magnetic sorbent based on monodisperse-porous silica microspheres was developed for His-tagged protein purification by immobilized metal affinity chromatography.
Collapse
Affiliation(s)
- Kouroush Salimi
- Chemical Engineering Department
- Hacettepe University
- Ankara
- Turkey
| | - Duygu Deniz Usta
- Department of Medical Biology and Genetics
- Gazi University
- Ankara
- Turkey
- Department of Medical Biology
| | - İlkay Koçer
- Chemical Engineering Department
- Hacettepe University
- Ankara
- Turkey
| | - Eda Çelik
- Chemical Engineering Department
- Hacettepe University
- Ankara
- Turkey
- Institute of Science
| | - Ali Tuncel
- Chemical Engineering Department
- Hacettepe University
- Ankara
- Turkey
- Division of Nanotechnology and Nanomedicine
| |
Collapse
|
100
|
Chao A, Jiang N, Yang Y, Li H, Sun H. A Ni-NTA-based red fluorescence probe for protein labelling in live cells. J Mater Chem B 2017; 5:1166-1173. [DOI: 10.1039/c6tb02848a] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The great success of a His6-Ni-nitrilotriaceate (Ni-NTA) system in protein purification has inspired scientists to develop novel Ni-NTA based fluoresent probes for imaging of proteins in live cells.
Collapse
Affiliation(s)
- Ailun Chao
- Department of Chemistry
- The University of Hong Kong
- Hong Kong
- P. R. China
| | - Nan Jiang
- Department of Chemistry
- The University of Hong Kong
- Hong Kong
- P. R. China
| | - Ya Yang
- Department of Chemistry
- The University of Hong Kong
- Hong Kong
- P. R. China
| | - Hongyan Li
- Department of Chemistry
- The University of Hong Kong
- Hong Kong
- P. R. China
| | - Hongzhe Sun
- Department of Chemistry
- The University of Hong Kong
- Hong Kong
- P. R. China
| |
Collapse
|