1
|
Peacock AFA. Coiled coils as ligands for inclusion in the inorganic chemist's toolbox - For advances in MRI contrast agent design. J Inorg Biochem 2025; 268:112903. [PMID: 40169349 DOI: 10.1016/j.jinorgbio.2025.112903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 04/03/2025]
Abstract
Ligands are essential tools in synthetic inorganic chemistry, enabling the fine-tuning of metal ion properties to optimize performance. Spanning from small molecules to macromolecular proteins, ligands vary widely in structure and function. De novo designed coiled coils serve as a unique bridge between these extremes, offering precise control over metal coordination. Here, we explore the application of coiled coil ligands in MRI contrast agent design, leveraging their versatility to systematically modulate the coordination chemistry and hydration state of gadolinium - the metal used in most clinical MRI contrast agents. This novel class of gadolinium-based agents demonstrates superior performance compared to existing clinical agents, highlighting the potential of coiled coil ligands. Furthermore, when coordinated to copper, these ligands form complexes that challenge the conventional notion that copper is unsuitable for MRI contrast agents. These findings establish coiled coil ligands as a powerful platform for advancing contrast agent design.
Collapse
Affiliation(s)
- Anna F A Peacock
- School of Chemistry, University of Birmingham, Edgbaston B15 2TT, UK.
| |
Collapse
|
2
|
Prakash D, Mitra S, Sony S, Murphy M, Andi B, Ashley L, Prasad P, Chakraborty S. Controlling outer-sphere solvent reorganization energy to turn on or off the function of artificial metalloenzymes. Nat Commun 2025; 16:3048. [PMID: 40155633 PMCID: PMC11953277 DOI: 10.1038/s41467-025-57904-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 03/05/2025] [Indexed: 04/01/2025] Open
Abstract
Metalloenzymes play essential roles in biology. However, unraveling how outer-sphere interactions can be predictably controlled to influence their functions remains a significant challenge. Inspired by Cu enzymes, we demonstrate how variations in the primary, secondary, and outer coordination-sphere interactions of de novo designed artificial copper proteins (ArCuPs) within trimeric (3SCC) and tetrameric (4SCC) self-assemblies-featuring a trigonal Cu(His)3 and a square pyramidal Cu(His)4(OH2) coordination-influence their catalytic and electron transfer properties. While 3SCC electrocatalyzes C-H oxidation, 4SCC does not. CuI-3SCC reacts more rapidly with H2O2 than O2, whereas 4SCC is less active. Electron transfer, reorganization energies, and extended H2O-mediated hydrogen bonding patterns provide insights into the observed reactivity differences. The inactivity of 4SCC is attributed to a significant solvent reorganization energy barrier mediated by a specific His---Glu hydrogen bond. When this hydrogen bond is disrupted, the solvent reorganization energy is reduced, and C-H peroxidation activity is restored.
Collapse
Affiliation(s)
- Divyansh Prakash
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, USA
- Northwestern University, Evanston, IL, USA
| | - Suchitra Mitra
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Simran Sony
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, USA
| | - Morgan Murphy
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, USA
| | - Babak Andi
- Center for BioMolecular Structure, National Synchrotron Light Source II, Brookhaven National Laboratory, Upton, NY, USA
| | - Landon Ashley
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, USA
| | - Pallavi Prasad
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, USA
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Saumen Chakraborty
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, USA.
| |
Collapse
|
3
|
Di Costanzo LF, Sgueglia G, Orlando C, Polentarutti M, Leone L, La Gatta S, De Fenza M, De Gioia L, Lombardi A, Arrigoni F, Chino M. Structural insights into temperature-dependent dynamics of METPsc1, a miniaturized electron-transfer protein. J Inorg Biochem 2025; 264:112810. [PMID: 39689412 DOI: 10.1016/j.jinorgbio.2024.112810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/30/2024] [Accepted: 12/07/2024] [Indexed: 12/19/2024]
Abstract
The design of protein-metal complexes is rapidly advancing, with applications spanning catalysis, sensing, and bioremediation. We report a comprehensive investigation of METPsc1, a Miniaturized Electron Transfer Protein, in complex with cadmium. This study elucidates the impact of metal coordination on protein folding and structural dynamics across temperatures from 100 K to 300 K. Our findings reveal that METPsc1, composed of two similar halves stabilized by intramolecular hydrogen bonds, exhibits a unique "clothespin-like" recoil mechanism. This allows it to adapt to metal ions of varying radii, mirroring the flexibility observed in natural rubredoxins. High-resolution crystallography and molecular dynamics simulations unveil concerted backbone motions and subtle temperature-dependent shifts in side-chain conformations, particularly for residues involved in crystal packing. Notably, CdS bond lengths increase with temperature, correlating with anisotropic motions of the sulfur atoms involved in second-shell hydrogen bonding. This suggests a dynamic role of protein matrix upon redox cycling. These insights into METPsc1 highlight its potential for catalysis and contribute to the designing of artificial metalloproteins with functional plasticity.
Collapse
Affiliation(s)
- Luigi F Di Costanzo
- Department of Agricultural Sciences, University of Naples Federico II, Via Università 100, 80055 Portici, NA, Italy.
| | - Gianmattia Sgueglia
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, 80126 Napoli, Italy
| | - Carla Orlando
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | | | - Linda Leone
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, 80126 Napoli, Italy
| | - Salvatore La Gatta
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, 80126 Napoli, Italy
| | - Maria De Fenza
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, 80126 Napoli, Italy
| | - Luca De Gioia
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Angela Lombardi
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, 80126 Napoli, Italy
| | - Federica Arrigoni
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Marco Chino
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, 80126 Napoli, Italy.
| |
Collapse
|
4
|
Muthahari YA, Magnus L, Laurino P. From duplication to fusion: Expanding Dayhoff's model of protein evolution. Protein Sci 2025; 34:e70054. [PMID: 39969106 PMCID: PMC11837038 DOI: 10.1002/pro.70054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/20/2025]
Abstract
Dayhoff's hypothesis suggests that complex proteins emerged from simpler peptides or domains, which duplicated and fused to create more complex proteins and novel functions. These processes expanded and diversified the protein repertoire within organisms. Extensive studies and reviews over the past two decades have highlighted the impact of gene duplication on protein evolution. However, the role of fusion in this evolutionary narrative remains less understood. This perspective seeks to address this gap by emphasizing the role of fusion in evolution. Fusion is critical in determining the evolutionary fate of duplicated protomers, either preserving their ancestral function or evolving entirely new functions. It complements mutations, insertions, and deletions as evolutionary steps to enhance protein evolvability by expanding the capacity of the protein to explore new structural and functional space.
Collapse
Affiliation(s)
| | - Lilian Magnus
- Protein Engineering and Evolution UnitOkinawa Institute of Science and TechnologyOkinawaJapan
| | - Paola Laurino
- Protein Engineering and Evolution UnitOkinawa Institute of Science and TechnologyOkinawaJapan
- Institute for Protein ResearchOsaka UniversitySuitaJapan
| |
Collapse
|
5
|
Petrenas R, Hawkins OA, Jones JF, Scott DA, Fletcher JM, Obst U, Lombardi L, Pirro F, Leggett GJ, Oliver TA, Woolfson DN. Confinement and Catalysis within De Novo Designed Peptide Barrels. J Am Chem Soc 2025; 147:3796-3803. [PMID: 39813445 PMCID: PMC11783595 DOI: 10.1021/jacs.4c16633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/18/2025]
Abstract
De novo protein design has advanced such that many peptide assemblies and protein structures can be generated predictably and quickly. The drive now is to bring functions to these structures, for example, small-molecule binding and catalysis. The formidable challenge of binding and orienting multiple small molecules to direct chemistry is particularly important for paving the way to new functionalities. To address this, here we describe the design, characterization, and application of small-molecule:peptide ternary complexes in aqueous solution. This uses α-helical barrel (αHB) peptide assemblies, which comprise 5 or more α helices arranged around central channels. These channels are solvent accessible, and their internal dimensions and chemistries can be altered predictably. Thus, αHBs are analogous to "molecular flasks" made in supramolecular, polymer, and materials chemistry. Using Förster resonance energy transfer as a readout, we demonstrate that specific αHBs can accept two different organic dyes, 1,6-diphenyl-1,3,5-hexatriene and Nile red, in close proximity. In addition, two anthracene molecules can be accommodated within an αHB to promote anthracene photodimerization. However, not all ternary complexes are productive, either in energy transfer or photodimerization, illustrating the control that can be exerted by judicious choice and design of the αHB.
Collapse
Affiliation(s)
- Rokas Petrenas
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
| | - Olivia A. Hawkins
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
| | - Jacob F. Jones
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
| | - D. Arne Scott
- Rosa
Biotech, Science Creates St Philips, Albert Road, Bristol BS2 0XJ, U.K.
| | - Jordan M. Fletcher
- Rosa
Biotech, Science Creates St Philips, Albert Road, Bristol BS2 0XJ, U.K.
| | - Ulrike Obst
- Rosa
Biotech, Science Creates St Philips, Albert Road, Bristol BS2 0XJ, U.K.
| | - Lucia Lombardi
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
- Department
of Chemical Engineering, Imperial College
London, London SW7 2AZ, U.K.
| | - Fabio Pirro
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
| | - Graham J. Leggett
- School
of Mathematical and Physical Sciences, University
of Sheffield, Brook Hill, Sheffield S3 7HF, U.K.
| | - Thomas A.A. Oliver
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
| | - Derek N. Woolfson
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
- Max
Planck-Bristol Centre for Minimal Biology, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
- Bristol BioDesign
Institute, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
- School
of Biochemistry, University of Bristol, Medical Sciences Building, Bristol BS8 1TD, U.K.
| |
Collapse
|
6
|
Häge F, Schwan M, Conde González MR, Huber J, Germer S, Macrì M, Kopp J, Sinning I, Thomas F. Strand-Swapped SH3 Domain Dimer with Superoxide Dismutase Activity. ACS CENTRAL SCIENCE 2025; 11:157-166. [PMID: 39866698 PMCID: PMC11758493 DOI: 10.1021/acscentsci.4c01347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025]
Abstract
The design of metalloproteins allows us to better understand metal complexation in proteins and the resulting function. In this study, we incorporated a Cu2+-binding site into a natural protein domain, the 58 amino acid c-Crk-SH3, to create a miniaturized superoxide dismutase model, termed SO1. The resulting low complexity metalloprotein was characterized for structure and function by circular dichroism and UV spectroscopy as well as EPR spectroscopy and X-ray crystallography. The miniprotein was found to be a strand-swapped dimer with an unusual coupled binuclear Type 2-like copper center in each protomer. SO1-Cu was found to be SOD-active with an activity only 1 order of magnitude lower than that of natural SOD enzymes and 1 to 2 orders of magnitude higher than that of other low-complexity SOD protein models of similar size. This serendipitous design provides us with a new structural template for future designs of binuclear metalloproteins with different metal ions and functions.
Collapse
Affiliation(s)
- Florian
R. Häge
- Institute
of Organic Chemistry, Heidelberg University, Im Neuenheimer Feld 270, 69120 Heidelberg, Germany
| | - Merlin Schwan
- Biochemistry
Center, Heidelberg University, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany
| | - Marcos Rafael Conde González
- Institute
of Organic Chemistry, Heidelberg University, Im Neuenheimer Feld 270, 69120 Heidelberg, Germany
- Max
Planck School Matter to Life, Jahnstraße 29, 69120 Heidelberg, Germany
| | - Jonas Huber
- Institute
of Organic Chemistry, Heidelberg University, Im Neuenheimer Feld 270, 69120 Heidelberg, Germany
| | - Stefan Germer
- Institute
of Organic Chemistry, Heidelberg University, Im Neuenheimer Feld 270, 69120 Heidelberg, Germany
| | - Matilde Macrì
- Institute
of Organic Chemistry, Heidelberg University, Im Neuenheimer Feld 270, 69120 Heidelberg, Germany
| | - Jürgen Kopp
- Biochemistry
Center, Heidelberg University, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany
| | - Irmgard Sinning
- Biochemistry
Center, Heidelberg University, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany
| | - Franziska Thomas
- Institute
of Organic Chemistry, Heidelberg University, Im Neuenheimer Feld 270, 69120 Heidelberg, Germany
| |
Collapse
|
7
|
Williams JC, Faillace MS, Gonzalez EJ, Dominguez RE, Knappenberger K, Heredia DA, Moore TA, Moore AL, Allen JP. Mn-porphyrins in a four-helix bundle participate in photo-induced electron transfer with a bacterial reaction center. PHOTOSYNTHESIS RESEARCH 2024; 162:1-14. [PMID: 37910331 DOI: 10.1007/s11120-023-01051-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/18/2023] [Indexed: 11/03/2023]
Abstract
Hybrid complexes incorporating synthetic Mn-porphyrins into an artificial four-helix bundle domain of bacterial reaction centers created a system to investigate new electron transfer pathways. The reactions were initiated by illumination of the bacterial reaction centers, whose primary photochemistry involves electron transfer from the bacteriochlorophyll dimer through a series of electron acceptors to the quinone electron acceptors. Porphyrins with diphenyl, dimesityl, or fluorinated substituents were synthesized containing either Mn or Zn. Electrochemical measurements revealed potentials for Mn(III)/Mn(II) transitions that are ~ 0.4 V higher for the fluorinated Mn-porphyrins than the diphenyl and dimesityl Mn-porphyrins. The synthetic porphyrins were introduced into the proteins by binding to a four-helix bundle domain that was genetically fused to the reaction center. Light excitation of the bacteriochlorophyll dimer of the reaction center resulted in new derivative signals, in the 400 to 450 nm region of light-minus-dark spectra, that are consistent with oxidation of the fluorinated Mn(II) porphyrins and reduction of the diphenyl and dimesityl Mn(III) porphyrins. These features recovered in the dark and were not observed in the Zn(II) porphyrins. The amplitudes of the signals were dependent upon the oxidation/reduction midpoint potentials of the bacteriochlorophyll dimer. These results are interpreted as photo-induced charge-separation processes resulting in redox changes of the Mn-porphyrins, demonstrating the utility of the hybrid artificial reaction center system to establish design guidelines for novel electron transfer reactions.
Collapse
Affiliation(s)
- J C Williams
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - M S Faillace
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - E J Gonzalez
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - R E Dominguez
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - K Knappenberger
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - D A Heredia
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - T A Moore
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - A L Moore
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - J P Allen
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
8
|
Leone L, De Fenza M, Esposito A, Maglio O, Nastri F, Lombardi A. Peptides and metal ions: A successful marriage for developing artificial metalloproteins. J Pept Sci 2024; 30:e3606. [PMID: 38719781 DOI: 10.1002/psc.3606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 10/12/2024]
Abstract
The mutual relationship between peptides and metal ions enables metalloproteins to have crucial roles in biological systems, including structural, sensing, electron transport, and catalytic functions. The effort to reproduce or/and enhance these roles, or even to create unprecedented functions, is the focus of protein design, the first step toward the comprehension of the complex machinery of nature. Nowadays, protein design allows the building of sophisticated scaffolds, with novel functions and exceptional stability. Recent progress in metalloprotein design has led to the building of peptides/proteins capable of orchestrating the desired functions of different metal cofactors. The structural diversity of peptides allows proper selection of first- and second-shell ligands, as well as long-range electrostatic and hydrophobic interactions, which represent precious tools for tuning metal properties. The scope of this review is to discuss the construction of metal sites in de novo designed and miniaturized scaffolds. Selected examples of mono-, di-, and multi-nuclear binding sites, from the last 20 years will be described in an effort to highlight key artificial models of catalytic or electron-transfer metalloproteins. The authors' goal is to make readers feel like guests at the marriage between peptides and metal ions while offering sources of inspiration for future architects of innovative, artificial metalloproteins.
Collapse
Affiliation(s)
- Linda Leone
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Maria De Fenza
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Alessandra Esposito
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Ornella Maglio
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | - Flavia Nastri
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Angela Lombardi
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
9
|
Kano R, Oohora K, Hayashi T. Photo-induced imine reduction by a photoredox biocatalyst consisting of a pentapeptide and a Ru bipyridine terpyridine complex. J Inorg Biochem 2024; 259:112657. [PMID: 38981409 DOI: 10.1016/j.jinorgbio.2024.112657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/17/2024] [Accepted: 06/27/2024] [Indexed: 07/11/2024]
Abstract
Imine reduction is a useful reaction in the preparation of amine derivatives. Various catalysts have been reported to promote this reaction and photoredox catalysts are promising candidates for sustainable amine synthesis. Improvement of this reaction using biomolecule-based reaction scaffolds is expected to increase the utility of the reaction. In this context, we have recently investigated photoredox Ru complexes with pentapeptide scaffolds via coordination bonds as catalysts for photoreduction of dihydroisoquinoline derivatives. First, Ru bipyridine terpyridine complexes coordinated with five different pentapeptides (XVHVV: X = V, F, W, Y, C) were prepared and characterized by mass spectrometry. Catalytic activities of the Ru complexes with XVHVV were evaluated for photoreduction of dihydroisoquinoline derivatives in the presence of ascorbate and thiol compounds as sacrificial reagents and hydrogen sources. Interestingly, the turnover number of the Ru complex with VVHVV is 531, which is two-fold higher than that of a simple Ru complex with an imidazole ligand. The detailed emission lifetime measurements indicate that the enhanced catalytic activity provided by the peptide scaffold is caused by an efficient reaction with the thiol derivative to accelerate reductive quenching of Ru complex. The quenching behavior suggests formation of an active species such as a Ru(I) complex. These findings reveal that the simple pentapeptide serves as an effective scaffold to enhance the photocatalytic activity of a photoactive Ru complex.
Collapse
Affiliation(s)
- Ryusei Kano
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka 565-0871, Japan
| | - Koji Oohora
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka 565-0871, Japan; Innovative Catalysis Science Division, Institute for Open and Transdisciplinary Research Initiatives (ICS-OTRI), Osaka University, Suita, Osaka 565-0871, Japan.
| | - Takashi Hayashi
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
10
|
Hardy BJ, Dubiel P, Bungay EL, Rudin M, Williams C, Arthur CJ, Guberman‐Pfeffer MJ, Sofia Oliveira A, Curnow P, Anderson JLR. Delineating redox cooperativity in water-soluble and membrane multiheme cytochromes through protein design. Protein Sci 2024; 33:e5113. [PMID: 38980168 PMCID: PMC11232281 DOI: 10.1002/pro.5113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024]
Abstract
Nature has evolved diverse electron transport proteins and multiprotein assemblies essential to the generation and transduction of biological energy. However, substantially modifying or adapting these proteins for user-defined applications or to gain fundamental mechanistic insight can be hindered by their inherent complexity. De novo protein design offers an attractive route to stripping away this confounding complexity, enabling us to probe the fundamental workings of these bioenergetic proteins and systems, while providing robust, modular platforms for constructing completely artificial electron-conducting circuitry. Here, we use a set of de novo designed mono-heme and di-heme soluble and membrane proteins to delineate the contributions of electrostatic micro-environments and dielectric properties of the surrounding protein medium on the inter-heme redox cooperativity that we have previously reported. Experimentally, we find that the two heme sites in both the water-soluble and membrane constructs have broadly equivalent redox potentials in isolation, in agreement with Poisson-Boltzmann Continuum Electrostatics calculations. BioDC, a Python program for the estimation of electron transfer energetics and kinetics within multiheme cytochromes, also predicts equivalent heme sites, and reports that burial within the low dielectric environment of the membrane strengthens heme-heme electrostatic coupling. We conclude that redox cooperativity in our diheme cytochromes is largely driven by heme electrostatic coupling and confirm that this effect is greatly strengthened by burial in the membrane. These results demonstrate that while our de novo proteins present minimalist, new-to-nature constructs, they enable the dissection and microscopic examination of processes fundamental to the function of vital, yet complex, bioenergetic assemblies.
Collapse
Affiliation(s)
| | | | | | - May Rudin
- School of BiochemistryUniversity of BristolBristolUK
| | | | | | | | | | - Paul Curnow
- School of BiochemistryUniversity of BristolBristolUK
| | | |
Collapse
|
11
|
Renzi E, Esposito A, Leone L, Chávez M, Pineda T, Lombardi A, Nastri F. Biohybrid materials comprising an artificial peroxidase and differently shaped gold nanoparticles. NANOSCALE ADVANCES 2024; 6:3533-3542. [PMID: 38989515 PMCID: PMC11232542 DOI: 10.1039/d4na00344f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/01/2024] [Indexed: 07/12/2024]
Abstract
The immobilization of biocatalysts on inorganic supports allows the development of bio-nanohybrid materials with defined functional properties. Gold nanomaterials (AuNMs) are the main players in this field, due to their fascinating shape-dependent properties that account for their versatility. Even though incredible progress has been made in the preparation of AuNMs, few studies have been carried out to analyze the impact of particle morphology on the behavior of immobilized biocatalysts. Herein, the artificial peroxidase Fe(iii)-Mimochrome VI*a (FeMC6*a) was conjugated to two different anisotropic gold nanomaterials, nanorods (AuNRs) and triangular nanoprisms (AuNTs), to investigate how the properties of the nanosupport can affect the functional behavior of FeMC6*a. The conjugation of FeMC6*a to AuNMs was performed by a click-chemistry approach, using FeMC6*a modified with pegylated aza-dibenzocyclooctyne (FeMC6*a-PEG4@DBCO), which was allowed to react with azide-functionalized AuNRs and AuNTs, synthesized from citrate-capped AuNMs. To this end, a literature protocol for depleting CTAB from AuNRs was herein reported for the first time to prepare citrate-AuNTs. The overall results suggest that the nanomaterial shape influences the nanoconjugate functional properties. Besides giving new insights into the effect of the surfaces on the artificial peroxidase properties, these results open up the way for creating novel nanostructures with potential applications in the field of sensing devices.
Collapse
Affiliation(s)
- Emilia Renzi
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte S. Angelo via Cintia Naples 80126 Italy
| | - Alessandra Esposito
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte S. Angelo via Cintia Naples 80126 Italy
| | - Linda Leone
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte S. Angelo via Cintia Naples 80126 Italy
| | - Miriam Chávez
- Department of Physical Chemistry and Applied Thermodynamics, Institute of Chemistry for Energy and Environment, University of Cordoba, Campus Rabanales Ed. Marie Curie Córdoba E-14014 Spain
| | - Teresa Pineda
- Department of Physical Chemistry and Applied Thermodynamics, Institute of Chemistry for Energy and Environment, University of Cordoba, Campus Rabanales Ed. Marie Curie Córdoba E-14014 Spain
| | - Angela Lombardi
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte S. Angelo via Cintia Naples 80126 Italy
| | - Flavia Nastri
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte S. Angelo via Cintia Naples 80126 Italy
| |
Collapse
|
12
|
Schnettler JD, Wang MS, Gantz M, Bunzel HA, Karas C, Hollfelder F, Hecht MH. Selection of a promiscuous minimalist cAMP phosphodiesterase from a library of de novo designed proteins. Nat Chem 2024; 16:1200-1208. [PMID: 38702405 PMCID: PMC11230910 DOI: 10.1038/s41557-024-01490-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 02/27/2024] [Indexed: 05/06/2024]
Abstract
The ability of unevolved amino acid sequences to become biological catalysts was key to the emergence of life on Earth. However, billions of years of evolution separate complex modern enzymes from their simpler early ancestors. To probe how unevolved sequences can develop new functions, we use ultrahigh-throughput droplet microfluidics to screen for phosphoesterase activity amidst a library of more than one million sequences based on a de novo designed 4-helix bundle. Characterization of hits revealed that acquisition of function involved a large jump in sequence space enriching for truncations that removed >40% of the protein chain. Biophysical characterization of a catalytically active truncated protein revealed that it dimerizes into an α-helical structure, with the gain of function accompanied by increased structural dynamics. The identified phosphodiesterase is a manganese-dependent metalloenzyme that hydrolyses a range of phosphodiesters. It is most active towards cyclic AMP, with a rate acceleration of ~109 and a catalytic proficiency of >1014 M-1, comparable to larger enzymes shaped by billions of years of evolution.
Collapse
Affiliation(s)
| | - Michael S Wang
- Department of Chemistry, Princeton University, Princeton, USA
| | - Maximilian Gantz
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - H Adrian Bunzel
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Christina Karas
- Department of Molecular Biology, Princeton University, Princeton, USA
| | | | - Michael H Hecht
- Department of Chemistry, Princeton University, Princeton, USA.
| |
Collapse
|
13
|
Borghesani V, Zastrow ML, Tolbert AE, Deb A, Penner-Hahn JE, Pecoraro VL. Co(II) Substitution Enhances the Esterase Activity of a de Novo Designed Zn(II) Carbonic Anhydrase. Chemistry 2024; 30:e202304367. [PMID: 38377169 PMCID: PMC11045307 DOI: 10.1002/chem.202304367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 02/22/2024]
Abstract
Carbonic Anhydrases (CAs) have been a target for de novo protein designers due to the simplicity of the active site and rapid rate of the reaction. The first reported mimic contained a Zn(II) bound to three histidine imidazole nitrogens and an exogenous water molecule, hence closely mimicking the native enzymes' first coordination sphere. Co(II) has served as an alternative metal to interrogate CAs due to its d7 electronic configuration for more detailed solution characterization. We present here the Co(II) substituted [Co(II)(H2O/OH-)]N(TRIL2WL23H)3 n+ that behaves similarly to native Co(II) substituted human-CAs. Like the Zn(II) analogue, the cobalt-derivative at slightly basic pH is incapable of hydrolyzing p-nitrophenylacetate (pNPA); however, as the pH is increased a significant activity develops, which at pH values above 10 eventually yields a catalytic efficiency that exceeds that of the [Zn(II)(OH-)]N(TRIL2WL23H)3 + peptide complex. X-ray absorption analysis is consistent with an octahedral species at pH 7.5 that converts to a 5-coordinate species by pH 11. UV-vis spectroscopy can monitor this transition, giving a pKa for the conversion of 10.3. We assign this conversion to the formation of a 5-coordinate Co(II)(Nimid)3(OH)(H2O) species. The pH dependent kinetic analysis indicates the maximal rate (kcat), and thus the catalytic efficiency (kcat/Km), follow the same pH profile as the spectroscopic conversion to the pentacoordinate species. This correlation suggests that the chemically irreversible ester hydrolysis corresponds to the rate determining process.
Collapse
Affiliation(s)
- Valentina Borghesani
- Department of Chemistry and Biophysics, University of Michigan, Ann Arbor, MI-48109-1055, United States
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle, Scienze 11A, 43124, Parma, Italy
| | - Melissa L Zastrow
- Department of Chemistry and Biophysics, University of Michigan, Ann Arbor, MI-48109-1055, United States
- Department of Chemistry, University of Houston, 3585 Cullen Blvd, Houston, TX-77204, United States
| | - Audrey E Tolbert
- Department of Chemistry and Biophysics, University of Michigan, Ann Arbor, MI-48109-1055, United States
| | - Aniruddha Deb
- Department of Chemistry and Biophysics, University of Michigan, Ann Arbor, MI-48109-1055, United States
| | - James E Penner-Hahn
- Department of Chemistry and Biophysics, University of Michigan, Ann Arbor, MI-48109-1055, United States
| | - Vincent L Pecoraro
- Department of Chemistry and Biophysics, University of Michigan, Ann Arbor, MI-48109-1055, United States
| |
Collapse
|
14
|
Sgueglia G, Vrettas MD, Chino M, De Simone A, Lombardi A. MetalHawk: Enhanced Classification of Metal Coordination Geometries by Artificial Neural Networks. J Chem Inf Model 2024; 64:2356-2367. [PMID: 37956388 PMCID: PMC11005052 DOI: 10.1021/acs.jcim.3c00873] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/29/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023]
Abstract
The chemical properties of metal complexes are strongly dependent on the number and geometrical arrangement of ligands coordinated to the metal center. Existing methods for determining either coordination number or geometry rely on a trade-off between accuracy and computational costs, which hinders their application to the study of large structure data sets. Here, we propose MetalHawk (https://github.com/vrettasm/MetalHawk), a machine learning-based approach to perform simultaneous classification of metal site coordination number and geometry through artificial neural networks (ANNs), which were trained using the Cambridge Structural Database (CSD) and Metal Protein Data Bank (MetalPDB). We demonstrate that the CSD-trained model can be used to classify sites belonging to the most common coordination numbers and geometry classes with balanced accuracy equal to 96.51% for CSD-deposited metal sites. The CSD-trained model was also found to be capable of classifying bioinorganic metal sites from the MetalPDB database, with balanced accuracy equal to 84.29% on the whole PDB data set and to 91.66% on manually reviewed sites in the PDB validation set. Moreover, we report evidence that the output vectors of the CSD-trained model can be considered as a proxy indicator of metal-site distortions, showing that these can be interpreted as a low-dimensional representation of subtle geometrical features present in metal site structures.
Collapse
Affiliation(s)
- Gianmattia Sgueglia
- Department
of Chemical Sciences, University of Naples
Federico II, Via Cintia 21, 80126 Napoli, Italy
| | - Michail D. Vrettas
- Department
of Pharmacy, University of Naples Federico
II, Via Domenico Montesano
49, 80131 Napoli, Italy
| | - Marco Chino
- Department
of Chemical Sciences, University of Naples
Federico II, Via Cintia 21, 80126 Napoli, Italy
| | - Alfonso De Simone
- Department
of Pharmacy, University of Naples Federico
II, Via Domenico Montesano
49, 80131 Napoli, Italy
| | - Angela Lombardi
- Department
of Chemical Sciences, University of Naples
Federico II, Via Cintia 21, 80126 Napoli, Italy
| |
Collapse
|
15
|
Bell EL, Hutton AE, Burke AJ, O'Connell A, Barry A, O'Reilly E, Green AP. Strategies for designing biocatalysts with new functions. Chem Soc Rev 2024; 53:2851-2862. [PMID: 38353665 PMCID: PMC10946311 DOI: 10.1039/d3cs00972f] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Indexed: 03/19/2024]
Abstract
The engineering of natural enzymes has led to the availability of a broad range of biocatalysts that can be used for the sustainable manufacturing of a variety of chemicals and pharmaceuticals. However, for many important chemical transformations there are no known enzymes that can serve as starting templates for biocatalyst development. These limitations have fuelled efforts to build entirely new catalytic sites into proteins in order to generate enzymes with functions beyond those found in Nature. This bottom-up approach to enzyme development can also reveal new fundamental insights into the molecular origins of efficient protein catalysis. In this tutorial review, we will survey the different strategies that have been explored for designing new protein catalysts. These methods will be illustrated through key selected examples, which demonstrate how highly proficient and selective biocatalysts can be developed through experimental protein engineering and/or computational design. Given the rapid pace of development in the field, we are optimistic that designer enzymes will begin to play an increasingly prominent role as industrial biocatalysts in the coming years.
Collapse
Affiliation(s)
- Elizabeth L Bell
- Renewable Resources and Enabling Sciences Center, National Renewable Energy Laboratory, Golden, CO 80401, USA
- Manchester Institute of Biotechnology, School of Chemistry, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK.
| | - Amy E Hutton
- Manchester Institute of Biotechnology, School of Chemistry, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK.
| | - Ashleigh J Burke
- Manchester Institute of Biotechnology, School of Chemistry, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK.
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA 92093, USA
| | - Adam O'Connell
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Amber Barry
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Elaine O'Reilly
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Anthony P Green
- Manchester Institute of Biotechnology, School of Chemistry, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK.
| |
Collapse
|
16
|
López-Domene R, Manteca A, Rodriguez-Abetxuko A, Beloqui A, Cortajarena AL. In vitro Production of Hemin-Based Artificial Metalloenzymes. Chemistry 2024; 30:e202303254. [PMID: 38145337 DOI: 10.1002/chem.202303254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 12/26/2023]
Abstract
Developing enzyme alternatives is pivotal to improving and enabling new processes in biotechnology and industry. Artificial metalloenzymes (ArMs) are combinations of protein scaffolds with metal elements, such as metal nanoclusters or metal-containing molecules with specific catalytic properties, which can be customized. Here, we engineered an ArM based on the consensus tetratricopeptide repeat (CTPR) scaffold by introducing a unique histidine residue to coordinate the hemin cofactor. Our results show that this engineered system exhibits robust peroxidase-like catalytic activity driven by the hemin. The expression of the scaffold and subsequent coordination of hemin was achieved by recombinant expression in bulk and through in vitro transcription and translation systems in water-in-oil drops. The ability to synthesize this system in emulsio paves the way to improve its properties by means of droplet microfluidic screenings, facilitating the exploration of the protein combinatorial space to discover improved or novel catalytic activities.
Collapse
Affiliation(s)
- Rocío López-Domene
- Centre for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, E-20014, Spain
- POLYMAT and Department of Applied Chemistry, Faculty of Chemistry, University of the Basque Country UPV/EHU, Donostia-San Sebastián, E-20018, Spain
| | - Aitor Manteca
- Centre for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, E-20014, Spain
| | - Andoni Rodriguez-Abetxuko
- POLYMAT and Department of Applied Chemistry, Faculty of Chemistry, University of the Basque Country UPV/EHU, Donostia-San Sebastián, E-20018, Spain
| | - Ana Beloqui
- POLYMAT and Department of Applied Chemistry, Faculty of Chemistry, University of the Basque Country UPV/EHU, Donostia-San Sebastián, E-20018, Spain
- IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, E-48009, Bilbao, Spain
| | - Aitziber L Cortajarena
- Centre for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, E-20014, Spain
- IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, E-48009, Bilbao, Spain
| |
Collapse
|
17
|
Yu K, Zhang K, Jakob RP, Maier T, Ward TR. An artificial nickel chlorinase based on the biotin-streptavidin technology. Chem Commun (Camb) 2024; 60:1944-1947. [PMID: 38277163 PMCID: PMC10863421 DOI: 10.1039/d3cc05847f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/17/2024] [Indexed: 01/27/2024]
Abstract
Herein, we report on an artificial nickel chlorinase (ANCase) resulting from anchoring a biotinylated nickel-based cofactor within streptavidin (Sav). The resulting ANCase was efficient for the chlorination of diverse C(sp3)-H bonds. Guided by the X-ray analysis of the ANCase, the activity of the artificial chlorinase could be significantly improved. This approach opens interesting perspectives for late-stage functionalization of organic intermediates as it complements biocatalytic chlorination strategies.
Collapse
Affiliation(s)
- Kun Yu
- Department of Chemistry, University of Basel, Mattenstrasse 22, Basel, CH-4058, Switzerland.
| | - Kailin Zhang
- Department of Chemistry, University of Basel, Mattenstrasse 22, Basel, CH-4058, Switzerland.
| | - Roman P Jakob
- Biozentrum, University of Basel, Spitalstrasse 41, Basel, CH-4056, Switzerland
| | - Timm Maier
- Biozentrum, University of Basel, Spitalstrasse 41, Basel, CH-4056, Switzerland
| | - Thomas R Ward
- Department of Chemistry, University of Basel, Mattenstrasse 22, Basel, CH-4058, Switzerland.
| |
Collapse
|
18
|
Kurihara K, Umezawa K, Donnelly AE, Sperling B, Liao G, Hecht MH, Arai R. Crystal structure and activity of a de novo enzyme, ferric enterobactin esterase Syn-F4. Proc Natl Acad Sci U S A 2023; 120:e2218281120. [PMID: 37695900 PMCID: PMC10515146 DOI: 10.1073/pnas.2218281120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 08/07/2023] [Indexed: 09/13/2023] Open
Abstract
Producing novel enzymes that are catalytically active in vitro and biologically functional in vivo is a key goal of synthetic biology. Previously, we reported Syn-F4, the first de novo protein that meets both criteria. Syn-F4 hydrolyzed the siderophore ferric enterobactin, and expression of Syn-F4 allowed an inviable strain of Escherichia coli (Δfes) to grow in iron-limited medium. Here, we describe the crystal structure of Syn-F4. Syn-F4 forms a dimeric 4-helix bundle. Each monomer comprises two long α-helices, and the loops of the Syn-F4 dimer are on the same end of the bundle (syn topology). Interestingly, there is a penetrated hole in the central region of the Syn-F4 structure. Extensive mutagenesis experiments in a previous study showed that five residues (Glu26, His74, Arg77, Lys78, and Arg85) were essential for enzymatic activity in vivo. All these residues are located around the hole in the central region of the Syn-F4 structure, suggesting a putative active site with a catalytic dyad (Glu26-His74). The complete inactivity of purified proteins with mutations at the five residues supports the putative active site and reaction mechanism. Molecular dynamics and docking simulations of the ferric enterobactin siderophore binding to the Syn-F4 structure demonstrate the dynamic property of the putative active site. The structure and active site of Syn-F4 are completely different from native enterobactin esterase enzymes, thereby demonstrating that proteins designed de novo can provide life-sustaining catalytic activities using structures and mechanisms dramatically different from those that arose in nature.
Collapse
Affiliation(s)
- Kodai Kurihara
- Department of Applied Biology, Faculty of Textile Science and Technology, Shinshu University, Ueda, Nagano386-8567, Japan
| | - Koji Umezawa
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Minami-minowa, Kami-ina, Nagano399-4598, Japan
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Nagano390-8621, Japan
| | - Ann E. Donnelly
- Department of Chemistry, Princeton University, Princeton, NJ08544
| | - Brendan Sperling
- Department of Chemistry, Princeton University, Princeton, NJ08544
| | - Guanyu Liao
- Department of Chemistry, Princeton University, Princeton, NJ08544
| | - Michael H. Hecht
- Department of Chemistry, Princeton University, Princeton, NJ08544
| | - Ryoichi Arai
- Department of Applied Biology, Faculty of Textile Science and Technology, Shinshu University, Ueda, Nagano386-8567, Japan
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Nagano390-8621, Japan
| |
Collapse
|
19
|
Sučec I, Mammeri NE, Dregni AJ, Hong M. Rapid Determination of the Topology of Oligomeric α-Helical Membrane Proteins by Water- and Lipid-Edited Methyl NMR. J Phys Chem B 2023; 127:7518-7530. [PMID: 37606918 PMCID: PMC10893779 DOI: 10.1021/acs.jpcb.3c05295] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
Single-span oligomeric α-helical transmembrane proteins are common in virus ion channels, which are targets of antiviral drugs. Knowledge about the high-resolution structures of these oligomeric α-helical bundles is so far scarce. Structure determination of these membrane proteins by solid-state NMR traditionally requires resolving and assigning protein chemical shifts and measuring many interhelical distances, which are time-consuming. To accelerate experimental structure determination, here we introduce a simple solid-state NMR approach that uses magnetization transfer from water and lipid protons to the protein. By detecting the water- and lipid-transferred intensities of the high-sensitivity methyl 13C signals of Leu, Val, and Ile residues, which are highly enriched in these membrane proteins, we can derive models of the topology of these homo-oligomeric helical bundles. The topology is specified by the positions of amino acid residues in heptad repeats and the orientations of residues relative to the channel pore, lipids, and the helical interface. We demonstrate this water- and lipid-edited methyl NMR approach on the envelope (E) protein of SARS-CoV-2, the causative agent of the COVID-19 pandemic. We show that water-edited and lipid-edited 2D 13C-13C correlation spectra can be measured with sufficient sensitivity. Even without resolving multiple residues of the same type in the NMR spectra, we can obtain the helical bundle topology. We apply these experiments to the structurally unknown E proteins of the MERS coronavirus and the human coronavirus NL63. The resulting structural topologies show interesting differences in the positions of the aromatic residues in these three E proteins, suggesting that these viroporins may have different mechanisms of activation and ion conduction.
Collapse
Affiliation(s)
- Iva Sučec
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA 02139
| | - Nadia El Mammeri
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA 02139
| | - Aurelio J. Dregni
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA 02139
| | - Mei Hong
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA 02139
| |
Collapse
|
20
|
Yu K, Zou Z, Igareta NV, Tachibana R, Bechter J, Köhler V, Chen D, Ward TR. Artificial Metalloenzyme-Catalyzed Enantioselective Amidation via Nitrene Insertion in Unactivated C( sp3)-H Bonds. J Am Chem Soc 2023; 145:16621-16629. [PMID: 37471698 PMCID: PMC10401721 DOI: 10.1021/jacs.3c03969] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Indexed: 07/22/2023]
Abstract
Enantioselective C-H amidation offers attractive means to assemble C-N bonds to synthesize high-added value, nitrogen-containing molecules. In recent decades, complementary enzymatic and homogeneous-catalytic strategies for C-H amidation have been reported. Herein, we report on an artificial metalloenzyme (ArM) resulting from anchoring a biotinylated Ir-complex within streptavidin (Sav). The resulting ArM catalyzes the enantioselective amidation of unactivated C(sp3)-H bonds. Chemogenetic optimization of the Ir cofactor and Sav led to significant improvement in both the activity and enantioselectivity. Up to >700 TON and 92% ee for the amidation of unactivated C(sp3)-H bonds was achieved. The single crystal X-ray analysis of the artificial nitrene insertase (ANIase) combined with quantum mechanics-molecular mechanics (QM-MM) calculations sheds light on critical second coordination sphere contacts leading to improved catalytic performance.
Collapse
Affiliation(s)
- Kun Yu
- Department
of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, Basel CH-4058, Switzerland
| | - Zhi Zou
- Department
of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, Basel CH-4058, Switzerland
| | - Nico V. Igareta
- Department
of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, Basel CH-4058, Switzerland
| | - Ryo Tachibana
- Department
of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, Basel CH-4058, Switzerland
| | - Julia Bechter
- Department
of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, Basel CH-4058, Switzerland
| | - Valentin Köhler
- Department
of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, Basel CH-4058, Switzerland
| | - Dongping Chen
- Department
of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, Basel CH-4058, Switzerland
| | - Thomas R. Ward
- Department
of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, Basel CH-4058, Switzerland
| |
Collapse
|
21
|
D'Alonzo D, De Fenza M, Pavone V, Lombardi A, Nastri F. Selective Oxidation of Halophenols Catalyzed by an Artificial Miniaturized Peroxidase. Int J Mol Sci 2023; 24:ijms24098058. [PMID: 37175773 PMCID: PMC10178546 DOI: 10.3390/ijms24098058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
The development of artificial enzymes for application in sustainable technologies, such as the transformation of environmental pollutants or biomass, is one of the most challenging goals in metalloenzyme design. In this work, we describe the oxidation of mono-, di-, tri- and penta-halogenated phenols catalyzed by the artificial metalloenzyme Fe-MC6*a. It promoted the dehalogenation of 4-fluorophenol into the corresponding 1,4-benzoquinone, while under the same experimental conditions, 4-chloro, 4-bromo and 4-iodophenol were selectively converted into higher molecular weight compounds. Analysis of the 4-chlorophenol oxidation products clarified that oligomers based on C-O bonds were exclusively formed in this case. All results show that Fe-MC6*a holds intriguing enzymatic properties, as it catalyzes halophenol oxidation with substrate-dependent chemoselectivity.
Collapse
Affiliation(s)
- Daniele D'Alonzo
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia, 80126 Napoli, Italy
| | - Maria De Fenza
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia, 80126 Napoli, Italy
| | - Vincenzo Pavone
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia, 80126 Napoli, Italy
| | - Angela Lombardi
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia, 80126 Napoli, Italy
| | - Flavia Nastri
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia, 80126 Napoli, Italy
| |
Collapse
|
22
|
Hanreich S, Bonandi E, Drienovská I. Design of Artificial Enzymes: Insights into Protein Scaffolds. Chembiochem 2023; 24:e202200566. [PMID: 36418221 DOI: 10.1002/cbic.202200566] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
The design of artificial enzymes has emerged as a promising tool for the generation of potent biocatalysts able to promote new-to-nature reactions with improved catalytic performances, providing a powerful platform for wide-ranging applications and a better understanding of protein functions and structures. The selection of an appropriate protein scaffold plays a key role in the design process. This review aims to give a general overview of the most common protein scaffolds that can be exploited for the generation of artificial enzymes. Several examples are discussed and categorized according to the strategy used for the design of the artificial biocatalyst, namely the functionalization of natural enzymes, the creation of a new catalytic site in a protein scaffold bearing a wide hydrophobic pocket and de novo protein design. The review is concluded by a comparison of these different methods and by our perspective on the topic.
Collapse
Affiliation(s)
- Stefanie Hanreich
- Department of Chemistry and Pharmaceutical Sciences Vrije Universiteit, Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam (The, Netherlands
| | - Elisa Bonandi
- Department of Chemistry and Pharmaceutical Sciences Vrije Universiteit, Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam (The, Netherlands
| | - Ivana Drienovská
- Department of Chemistry and Pharmaceutical Sciences Vrije Universiteit, Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam (The, Netherlands
| |
Collapse
|
23
|
Abdul Aziz SFN, Rahim ASMA, Normi YM, Alang Ahmad SA, Salleh AB. Rational design of mini protein mimicking uricase: Encapsulation in ZIF-8 for uric acid detection. Proteins 2023. [PMID: 36908223 DOI: 10.1002/prot.26485] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 02/20/2023] [Accepted: 02/28/2023] [Indexed: 03/14/2023]
Abstract
Five mini proteins mimicking uricase comprising 20, 40, 60, 80, and 100 amino acids were designed based on the conserved active site residues within the same dimer, using the crystal structure of tetrameric uricase from Arthrobacter globiformis (PDB ID: 2yzb) as a template. Based on molecular docking analysis, the smallest mini protein, mp20, shared similar residues to that of native uricase that formed hydrogen bonds with uric acid and was chosen for further studies. Although purified recombinant mp20 did not exhibit uricase activity, it showed specific binding towards uric acid and evinced excellent thermotolerance and structural stability at temperatures ranging from 10°C to 100°C, emulating its natural origin. To explore the potential of mp20 as a bioreceptor in uric acid sensing, mp20 was encapsulated within zeolitic imidazolate framework-8 (mp20@ZIF-8) followed by the modification on rGO-screen printed electrode (rGO/SPCE) to maintain the structural stability. An irreversible anodic peak and increased semicircular arcs of the Nyquist plot with an increase of the analyte concentrations were observed by utilizing cyclic voltammetry and electrochemical impedance spectroscopy (EIS), suggesting the detection of uric acid occurred, which is based on substrate-mp20 interaction.
Collapse
Affiliation(s)
| | - Arilla Sri Masayu Abd Rahim
- Enzyme and Microbial Technology Research Centre (EMTech), Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Yahaya M Normi
- Enzyme and Microbial Technology Research Centre (EMTech), Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Institute of Nanoscience and Nanotechnology (ION2), Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Shahrul Ainliah Alang Ahmad
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Institute of Nanoscience and Nanotechnology (ION2), Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia
| | - Abu Bakar Salleh
- Enzyme and Microbial Technology Research Centre (EMTech), Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| |
Collapse
|
24
|
Prasad P, Hunt LA, Pall AE, Ranasinghe M, Williams AE, Stemmler TL, Demeler B, Hammer NI, Chakraborty S. Photocatalytic Hydrogen Evolution by a De Novo Designed Metalloprotein that Undergoes Ni-Mediated Oligomerization Shift. Chemistry 2023; 29:e202202902. [PMID: 36440875 PMCID: PMC10308963 DOI: 10.1002/chem.202202902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 11/29/2022]
Abstract
De novo metalloprotein design involves the construction of proteins guided by specific repeat patterns of polar and apolar residues, which, upon self-assembly, provide a suitable environment to bind metals and produce artificial metalloenzymes. While a wide range of functionalities have been realized in de novo designed metalloproteins, the functional repertoire of such constructs towards alternative energy-relevant catalysis is currently limited. Here we show the application of de novo approach to design a functional H2 evolving protein. The design involved the assembly of an amphiphilic peptide featuring cysteines at tandem a/d sites of each helix. Intriguingly, upon NiII addition, the oligomers shift from a major trimeric assembly to a mix of dimers and trimers. The metalloprotein produced H2 photocatalytically with a bell-shape pH dependence, having a maximum activity at pH 5.5. Transient absorption spectroscopy is used to determine the timescales of electron transfer as a function of pH. Selective outer sphere mutations are made to probe how the local environment tunes activity. A preferential enhancement of activity is observed via steric modulation above the NiII site, towards the N-termini, compared to below the NiII site towards the C-termini.
Collapse
Affiliation(s)
- Pallavi Prasad
- Department of Chemistry and Biochemistry, The University of Mississippi, University, MS, 38677 (USA)
| | - Leigh Anna Hunt
- Department of Chemistry and Biochemistry, The University of Mississippi, University, MS, 38677 (USA)
| | - Ashley E. Pall
- Department of Pharmaceutical Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201-2417 (USA)
| | - Maduni Ranasinghe
- Department of Chemistry and Biochemistry, University of Lethbridge, 4401,University Dr W, Lethbridge, AB T1K 6T5 (CA)
| | - Ashley E. Williams
- Department of Chemistry and Biochemistry, The University of Mississippi, University, MS, 38677 (USA)
| | - Timothy L. Stemmler
- Department of Pharmaceutical Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201-2417 (USA)
| | - Borries Demeler
- Department of Chemistry and Biochemistry, University of Lethbridge, 4401,University Dr W, Lethbridge, AB T1K 6T5 (CA)
| | - Nathan I. Hammer
- Department of Chemistry and Biochemistry, The University of Mississippi, University, MS, 38677 (USA)
| | - Saumen Chakraborty
- Department of Chemistry and Biochemistry, The University of Mississippi, University, MS, 38677 (USA)
| |
Collapse
|
25
|
Wang Z, Wang M, Zhao Z, Zheng P. Quantification of carboxylate-bridged di-zinc site stability in protein due ferri by single-molecule force spectroscopy. Protein Sci 2023; 32:e4583. [PMID: 36718829 PMCID: PMC9926469 DOI: 10.1002/pro.4583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/16/2023] [Accepted: 01/27/2023] [Indexed: 02/01/2023]
Abstract
Carboxylate-bridged diiron proteins belong to a protein family involved in different physiological processes. These proteins share the conservative EXXH motif, which provides the carboxylate bridge and is critical for metal binding. Here, we choose de novo-designed single-chain due ferri protein (DFsc), a four-helical protein with two EXXH motifs as a model protein, to study the stability of the carboxylate-bridged di-metal binding site. The mechanical and kinetic properties of the di-Zn site in DFsc were obtained by atomic force microscopy-based single-molecule force spectroscopy. Zn-DFsc showed a considerable rupture force of ~200 pN, while the apo-protein is mechanically labile. In addition, multiple rupture pathways were observed with different probabilities, indicating the importance of the EXXH-based carboxylate-bridged metal site. These results demonstrate carboxylate-bridged di-metal site is mechanically stable and improve our understanding of this important type of metalloprotein.
Collapse
Affiliation(s)
- Zhiyi Wang
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical EngineeringNanjing UniversityNanjingPeople's Republic of China
| | - Mengdie Wang
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical EngineeringNanjing UniversityNanjingPeople's Republic of China
| | - Zhongxin Zhao
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical EngineeringNanjing UniversityNanjingPeople's Republic of China
| | - Peng Zheng
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical EngineeringNanjing UniversityNanjingPeople's Republic of China
| |
Collapse
|
26
|
Curti M, Maffeis V, Teixeira Alves Duarte LG, Shareef S, Hallado LX, Curutchet C, Romero E. Engineering excitonically coupled dimers in an artificial protein for light harvesting via computational modeling. Protein Sci 2023; 32:e4579. [PMID: 36715022 PMCID: PMC9951196 DOI: 10.1002/pro.4579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023]
Abstract
In photosynthesis, pigment-protein complexes achieve outstanding photoinduced charge separation efficiencies through a set of strategies in which excited states delocalization over multiple pigments ("excitons") and charge-transfer states play key roles. These concepts, and their implementation in bioinspired artificial systems, are attracting increasing attention due to the vast potential that could be tapped by realizing efficient photochemical reactions. In particular, de novo designed proteins provide a diverse structural toolbox that can be used to manipulate the geometric and electronic properties of bound chromophore molecules. However, achieving excitonic and charge-transfer states requires closely spaced chromophores, a non-trivial aspect since a strong binding with the protein matrix needs to be maintained. Here, we show how a general-purpose artificial protein can be optimized via molecular dynamics simulations to improve its binding capacity of a chlorophyll derivative, achieving complexes in which chromophores form two closely spaced and strongly interacting dimers. Based on spectroscopy results and computational modeling, we demonstrate each dimer is excitonically coupled, and propose they display signatures of charge-transfer state mixing. This work could open new avenues for the rational design of chromophore-protein complexes with advanced functionalities.
Collapse
Affiliation(s)
- Mariano Curti
- Institute of Chemical Research of Catalonia (ICIQ), Barcelona Institute of Science and Technology (BIST)TarragonaSpain
| | - Valentin Maffeis
- Institute of Chemical Research of Catalonia (ICIQ), Barcelona Institute of Science and Technology (BIST)TarragonaSpain
- Laboratoire de Chimie, UMR 5182, ENS Lyon, CNRSUniversité Lyon 1LyonFrance
| | | | - Saeed Shareef
- Institute of Chemical Research of Catalonia (ICIQ), Barcelona Institute of Science and Technology (BIST)TarragonaSpain
- Departament de Química Física i InorgànicaUniversitat Rovira i VirgiliTarragonaSpain
| | - Luisa Xiomara Hallado
- Institute of Chemical Research of Catalonia (ICIQ), Barcelona Institute of Science and Technology (BIST)TarragonaSpain
- Departament de Química Física i InorgànicaUniversitat Rovira i VirgiliTarragonaSpain
| | - Carles Curutchet
- Departament de Farmàcia i Tecnologia Farmacèutica i Fisicoquímica, Facultat de Farmàcia i Ciències de l'AlimentacióUniversitat de Barcelona (UB)BarcelonaSpain
- Institut de Química Teòrica i Computacional (IQTCUB), Universitat de Barcelona (UB)BarcelonaSpain
| | - Elisabet Romero
- Institute of Chemical Research of Catalonia (ICIQ), Barcelona Institute of Science and Technology (BIST)TarragonaSpain
| |
Collapse
|
27
|
Guffy SL, Pulavarti SVSRK, Harrison J, Fleming D, Szyperski T, Kuhlman B. Inside-Out Design of Zinc-Binding Proteins with Non-Native Backbones. Biochemistry 2023; 62:770-781. [PMID: 36634348 PMCID: PMC9939277 DOI: 10.1021/acs.biochem.2c00595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The de novo design of functional proteins requires specification of tertiary structure and incorporation of molecular binding sites. Here, we develop an inside-out design strategy in the molecular modeling program Rosetta that begins with amino acid side chains from one or two α-helices making well-defined contacts with a ligand. A full-sized protein is then built around the ligand by adding additional helices that promote the formation of a protein core and allow additional contacts with the ligand. The protocol was tested by designing 12 zinc-binding proteins, each with 4-5 helices. Four of the designs were folded and bound to zinc with equilibrium dissociation constants varying between 95 nM and 1.1 μM. The design with the tightest affinity for zinc, N12, adopts a unique conformation in the folded state as assessed with nuclear magnetic resonance (NMR) and the design model closely matches (backbone root-mean-square deviation (RMSD) < 1 Å) an AlphaFold model of the sequence. Retrospective analysis with AlphaFold suggests that the sequences of many of the failed designs did not encode the desired tertiary packing.
Collapse
Affiliation(s)
- Sharon L. Guffy
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | | | - Joseph Harrison
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Drew Fleming
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Thomas Szyperski
- Department of Chemistry, State University of New York, Buffalo, NY, 14260, USA
| | - Brian Kuhlman
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
28
|
Yeh AHW, Norn C, Kipnis Y, Tischer D, Pellock SJ, Evans D, Ma P, Lee GR, Zhang JZ, Anishchenko I, Coventry B, Cao L, Dauparas J, Halabiya S, DeWitt M, Carter L, Houk KN, Baker D. De novo design of luciferases using deep learning. Nature 2023; 614:774-780. [PMID: 36813896 PMCID: PMC9946828 DOI: 10.1038/s41586-023-05696-3] [Citation(s) in RCA: 162] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 01/03/2023] [Indexed: 02/24/2023]
Abstract
De novo enzyme design has sought to introduce active sites and substrate-binding pockets that are predicted to catalyse a reaction of interest into geometrically compatible native scaffolds1,2, but has been limited by a lack of suitable protein structures and the complexity of native protein sequence-structure relationships. Here we describe a deep-learning-based 'family-wide hallucination' approach that generates large numbers of idealized protein structures containing diverse pocket shapes and designed sequences that encode them. We use these scaffolds to design artificial luciferases that selectively catalyse the oxidative chemiluminescence of the synthetic luciferin substrates diphenylterazine3 and 2-deoxycoelenterazine. The designed active sites position an arginine guanidinium group adjacent to an anion that develops during the reaction in a binding pocket with high shape complementarity. For both luciferin substrates, we obtain designed luciferases with high selectivity; the most active of these is a small (13.9 kDa) and thermostable (with a melting temperature higher than 95 °C) enzyme that has a catalytic efficiency on diphenylterazine (kcat/Km = 106 M-1 s-1) comparable to that of native luciferases, but a much higher substrate specificity. The creation of highly active and specific biocatalysts from scratch with broad applications in biomedicine is a key milestone for computational enzyme design, and our approach should enable generation of a wide range of luciferases and other enzymes.
Collapse
Affiliation(s)
- Andy Hsien-Wei Yeh
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA.
| | - Christoffer Norn
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Yakov Kipnis
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Doug Tischer
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Samuel J Pellock
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Declan Evans
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Pengchen Ma
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
- School of Chemistry, Xi'an Key Laboratory of Sustainable Energy Materials Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, Xi'an Jiaotong University, Xi'an, China
| | - Gyu Rie Lee
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jason Z Zhang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Ivan Anishchenko
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Brian Coventry
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Longxing Cao
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Justas Dauparas
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Samer Halabiya
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Michelle DeWitt
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Lauren Carter
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - K N Houk
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
29
|
Lu H, Cheng Z, Hu Y, Tang LV. What Can De Novo Protein Design Bring to the Treatment of Hematological Disorders? BIOLOGY 2023; 12:166. [PMID: 36829445 PMCID: PMC9952452 DOI: 10.3390/biology12020166] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
Protein therapeutics have been widely used to treat hematological disorders. With the advent of de novo protein design, protein therapeutics are not limited to ameliorating natural proteins but also produce novel protein sequences, folds, and functions with shapes and functions customized to bind to the therapeutic targets. De novo protein techniques have been widely used biomedically to design novel diagnostic and therapeutic drugs, novel vaccines, and novel biological materials. In addition, de novo protein design has provided new options for treating hematological disorders. Scientists have designed protein switches called Colocalization-dependent Latching Orthogonal Cage-Key pRoteins (Co-LOCKR) that perform computations on the surface of cells. De novo designed molecules exhibit a better capacity than the currently available tyrosine kinase inhibitors in chronic myeloid leukemia therapy. De novo designed protein neoleukin-2/15 enhances chimeric antigen receptor T-cell activity. This new technique has great biomedical potential, especially in exploring new treatment methods for hematological disorders. This review discusses the development of de novo protein design and its biological applications, with emphasis on the treatment of hematological disorders.
Collapse
Affiliation(s)
| | | | | | - Liang V. Tang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
30
|
Pirro F, La Gatta S, Arrigoni F, Famulari A, Maglio O, Del Vecchio P, Chiesa M, De Gioia L, Bertini L, Chino M, Nastri F, Lombardi A. A De Novo-Designed Type 3 Copper Protein Tunes Catechol Substrate Recognition and Reactivity. Angew Chem Int Ed Engl 2023; 62:e202211552. [PMID: 36334012 DOI: 10.1002/anie.202211552] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Indexed: 11/07/2022]
Abstract
De novo metalloprotein design is a remarkable approach to shape protein scaffolds toward specific functions. Here, we report the design and characterization of Due Rame 1 (DR1), a de novo designed protein housing a di-copper site and mimicking the Type 3 (T3) copper-containing polyphenol oxidases (PPOs). To achieve this goal, we hierarchically designed the first and the second di-metal coordination spheres to engineer the di-copper site into a simple four-helix bundle scaffold. Spectroscopic, thermodynamic, and functional characterization revealed that DR1 recapitulates the T3 copper site, supporting different copper redox states, and being active in the O2 -dependent oxidation of catechols to o-quinones. Careful design of the residues lining the substrate access site endows DR1 with substrate recognition, as revealed by Hammet analysis and computational studies on substituted catechols. This study represents a premier example in the construction of a functional T3 copper site into a designed four-helix bundle protein.
Collapse
Affiliation(s)
- Fabio Pirro
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126, Naples, Italy
| | - Salvatore La Gatta
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126, Naples, Italy
| | - Federica Arrigoni
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Antonino Famulari
- Department of Chemistry, University of Torino, Via Giuria 9, 10125, Torino, Italy.,Department of Condensed Matter Physics, University of of Zaragoza, Calle Pedro Cerbuna 12, 50009, Zaragoza, Spain
| | - Ornella Maglio
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126, Naples, Italy.,Institute of Biostructures and Bioimaging (IBB), National Research Council (CNR), Via Pietro Castellino 111, 80131, Napoli, Italy
| | - Pompea Del Vecchio
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126, Naples, Italy
| | - Mario Chiesa
- Department of Chemistry, University of Torino, Via Giuria 9, 10125, Torino, Italy
| | - Luca De Gioia
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Luca Bertini
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Marco Chino
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126, Naples, Italy
| | - Flavia Nastri
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126, Naples, Italy
| | - Angela Lombardi
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126, Naples, Italy
| |
Collapse
|
31
|
Vallapurackal J, Stucki A, Liang AD, Klehr J, Dittrich PS, Ward TR. Ultrahigh-Throughput Screening of an Artificial Metalloenzyme using Double Emulsions. Angew Chem Int Ed Engl 2022; 61:e202207328. [PMID: 36130864 PMCID: PMC9828110 DOI: 10.1002/anie.202207328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Indexed: 01/12/2023]
Abstract
The potential for ultrahigh-throughput compartmentalization renders droplet microfluidics an attractive tool for the directed evolution of enzymes. Importantly, it ensures maintenance of the phenotype-genotype linkage, enabling reliable identification of improved mutants. Herein, we report an approach for ultrahigh-throughput screening of an artificial metalloenzyme in double emulsion droplets (DEs) using commercially available fluorescence-activated cell sorters (FACS). This protocol was validated by screening a 400 double-mutant streptavidin library for ruthenium-catalyzed deallylation of an alloc-protected aminocoumarin. The most active variants, identified by next-generation sequencing, were in good agreement with hits obtained using a 96-well plate procedure. These findings pave the way for the systematic implementation of FACS for the directed evolution of (artificial) enzymes and will significantly expand the accessibility of ultrahigh-throughput DE screening protocols.
Collapse
Affiliation(s)
- Jaicy Vallapurackal
- Department of ChemistryUniversity of BaselMattenstrasse 24a4058BaselSwitzerland,National Competence Center in Research (NCCR) Molecular Systems EngineeringBaselSwitzerland
| | - Ariane Stucki
- Department of Biosystems Science and EngineeringETH ZurichMattenstrasse 264058BaselSwitzerland,National Competence Center in Research (NCCR) Molecular Systems EngineeringBaselSwitzerland
| | - Alexandria Deliz Liang
- Department of ChemistryUniversity of BaselMattenstrasse 24a4058BaselSwitzerland,National Competence Center in Research (NCCR) Molecular Systems EngineeringBaselSwitzerland
| | - Juliane Klehr
- Department of ChemistryUniversity of BaselMattenstrasse 24a4058BaselSwitzerland,National Competence Center in Research (NCCR) Molecular Systems EngineeringBaselSwitzerland
| | - Petra S. Dittrich
- Department of Biosystems Science and EngineeringETH ZurichMattenstrasse 264058BaselSwitzerland,National Competence Center in Research (NCCR) Molecular Systems EngineeringBaselSwitzerland
| | - Thomas R. Ward
- Department of ChemistryUniversity of BaselMattenstrasse 24a4058BaselSwitzerland,National Competence Center in Research (NCCR) Molecular Systems EngineeringBaselSwitzerland
| |
Collapse
|
32
|
Yang X, Wu W, Chen X, Wu F, Fan S, Yu P, Mao L. A versatile artificial metalloenzyme scaffold enabling direct bioelectrocatalysis in solution. SCIENCE ADVANCES 2022; 8:eabo3315. [PMID: 36322668 PMCID: PMC9629707 DOI: 10.1126/sciadv.abo3315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 09/13/2022] [Indexed: 06/16/2023]
Abstract
Artificial metalloenzymes (ArMs) are commonly designed with protein scaffolds containing buried coordination pockets to achieve substrate specificity and product selectivity for homogeneous reactions. However, their reactivities toward heterogeneous transformations are limited because interfacial electron transfers are hampered by the backbone shells. Here, we introduce bacterial small laccase (SLAC) as a new protein scaffold for constructing ArMs to directly catalyze electrochemical transformations. We use molecular dynamics simulation, x-ray crystallography, spectroscopy, and computation to illustrate the scaffold-directed assembly of an oxo-bridged dicobalt motif on protein surface. The resulting ArM in aqueous phase catalyzes electrochemical water oxidation without mediators or electrode modifications. Mechanistic investigation reveals the role of SLAC scaffold in defining the four-electron transfer pathway from water to oxygen. Furthermore, we demonstrate that SLAC-based ArMs implemented with Ni2+, Mn2+, Ru3+, Pd2+, or Ir3+ also enable direct bioelectrocatalysis of water electrolysis. Our study provides a versatile and generalizable route to complement heterogeneous repertoire of ArMs for expanded applications.
Collapse
Affiliation(s)
- Xiaoti Yang
- Beijing National Laboratory for Molecular Science, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenjie Wu
- Beijing National Laboratory for Molecular Science, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiling Chen
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Fei Wu
- Beijing National Laboratory for Molecular Science, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shilong Fan
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ping Yu
- Beijing National Laboratory for Molecular Science, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lanqun Mao
- Beijing National Laboratory for Molecular Science, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
33
|
Naudin EA, Albanese KI, Smith AJ, Mylemans B, Baker EG, Weiner OD, Andrews DM, Tigue N, Savery NJ, Woolfson DN. From peptides to proteins: coiled-coil tetramers to single-chain 4-helix bundles. Chem Sci 2022; 13:11330-11340. [PMID: 36320580 PMCID: PMC9533478 DOI: 10.1039/d2sc04479j] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 08/24/2022] [Indexed: 11/21/2022] Open
Abstract
The design of completely synthetic proteins from first principles-de novo protein design-is challenging. This is because, despite recent advances in computational protein-structure prediction and design, we do not understand fully the sequence-to-structure relationships for protein folding, assembly, and stabilization. Antiparallel 4-helix bundles are amongst the most studied scaffolds for de novo protein design. We set out to re-examine this target, and to determine clear sequence-to-structure relationships, or design rules, for the structure. Our aim was to determine a common and robust sequence background for designing multiple de novo 4-helix bundles. In turn, this could be used in chemical and synthetic biology to direct protein-protein interactions and as scaffolds for functional protein design. Our approach starts by analyzing known antiparallel 4-helix coiled-coil structures to deduce design rules. In terms of the heptad repeat, abcdefg -i.e., the sequence signature of many helical bundles-the key features that we identify are: a = Leu, d = Ile, e = Ala, g = Gln, and the use of complementary charged residues at b and c. Next, we implement these rules in the rational design of synthetic peptides to form antiparallel homo- and heterotetramers. Finally, we use the sequence of the homotetramer to derive in one step a single-chain 4-helix-bundle protein for recombinant production in E. coli. All of the assembled designs are confirmed in aqueous solution using biophysical methods, and ultimately by determining high-resolution X-ray crystal structures. Our route from peptides to proteins provides an understanding of the role of each residue in each design.
Collapse
Affiliation(s)
- Elise A Naudin
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
| | - Katherine I Albanese
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
- Max Planck-Bristol Centre for Minimal Biology, University of Bristol Cantock's Close Bristol BS8 1TS UK
| | - Abigail J Smith
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk Bristol BS8 1TD UK
| | - Bram Mylemans
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
- Max Planck-Bristol Centre for Minimal Biology, University of Bristol Cantock's Close Bristol BS8 1TS UK
| | - Emily G Baker
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk Bristol BS8 1TD UK
| | - Orion D Weiner
- Cardiovascular Research Institute, Department of Biochemistry and Biophysics, University of California 555 Mission Bay Blvd. South San Francisco CA 94158 USA
| | - David M Andrews
- Oncology R&D, AstraZeneca Cambridge Science Park, Darwin Building Cambridge CB4 0WG UK
| | - Natalie Tigue
- BioPharmaceuticals R&D, AstraZeneca Granta Park Cambridge CB21 6GH UK
| | - Nigel J Savery
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk Bristol BS8 1TD UK
- BrisEngBio, School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
| | - Derek N Woolfson
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
- Max Planck-Bristol Centre for Minimal Biology, University of Bristol Cantock's Close Bristol BS8 1TS UK
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk Bristol BS8 1TD UK
- BrisEngBio, School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
| |
Collapse
|
34
|
Qing R, Hao S, Smorodina E, Jin D, Zalevsky A, Zhang S. Protein Design: From the Aspect of Water Solubility and Stability. Chem Rev 2022; 122:14085-14179. [PMID: 35921495 PMCID: PMC9523718 DOI: 10.1021/acs.chemrev.1c00757] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Indexed: 12/13/2022]
Abstract
Water solubility and structural stability are key merits for proteins defined by the primary sequence and 3D-conformation. Their manipulation represents important aspects of the protein design field that relies on the accurate placement of amino acids and molecular interactions, guided by underlying physiochemical principles. Emulated designer proteins with well-defined properties both fuel the knowledge-base for more precise computational design models and are used in various biomedical and nanotechnological applications. The continuous developments in protein science, increasing computing power, new algorithms, and characterization techniques provide sophisticated toolkits for solubility design beyond guess work. In this review, we summarize recent advances in the protein design field with respect to water solubility and structural stability. After introducing fundamental design rules, we discuss the transmembrane protein solubilization and de novo transmembrane protein design. Traditional strategies to enhance protein solubility and structural stability are introduced. The designs of stable protein complexes and high-order assemblies are covered. Computational methodologies behind these endeavors, including structure prediction programs, machine learning algorithms, and specialty software dedicated to the evaluation of protein solubility and aggregation, are discussed. The findings and opportunities for Cryo-EM are presented. This review provides an overview of significant progress and prospects in accurate protein design for solubility and stability.
Collapse
Affiliation(s)
- Rui Qing
- State
Key Laboratory of Microbial Metabolism, School of Life Sciences and
Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Media
Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- The
David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Shilei Hao
- Media
Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Key
Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Eva Smorodina
- Department
of Immunology, University of Oslo and Oslo
University Hospital, Oslo 0424, Norway
| | - David Jin
- Avalon GloboCare
Corp., Freehold, New Jersey 07728, United States
| | - Arthur Zalevsky
- Laboratory
of Bioinformatics Approaches in Combinatorial Chemistry and Biology, Shemyakin−Ovchinnikov Institute of Bioorganic
Chemistry RAS, Moscow 117997, Russia
| | - Shuguang Zhang
- Media
Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
35
|
Ennist NM, Stayrook SE, Dutton PL, Moser CC. Rational design of photosynthetic reaction center protein maquettes. Front Mol Biosci 2022; 9:997295. [PMID: 36213121 PMCID: PMC9532970 DOI: 10.3389/fmolb.2022.997295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/18/2022] [Indexed: 11/20/2022] Open
Abstract
New technologies for efficient solar-to-fuel energy conversion will help facilitate a global shift from dependence on fossil fuels to renewable energy. Nature uses photosynthetic reaction centers to convert photon energy into a cascade of electron-transfer reactions that eventually produce chemical fuel. The design of new reaction centers de novo deepens our understanding of photosynthetic charge separation and may one day allow production of biofuels with higher thermodynamic efficiency than natural photosystems. Recently, we described the multi-step electron-transfer activity of a designed reaction center maquette protein (the RC maquette), which can assemble metal ions, tyrosine, a Zn tetrapyrrole, and heme into an electron-transport chain. Here, we detail our modular strategy for rational protein design and show that the intended RC maquette design agrees with crystal structures in various states of assembly. A flexible, dynamic apo-state collapses by design into a more ordered holo-state upon cofactor binding. Crystal structures illustrate the structural transitions upon binding of different cofactors. Spectroscopic assays demonstrate that the RC maquette binds various electron donors, pigments, and electron acceptors with high affinity. We close with a critique of the present RC maquette design and use electron-tunneling theory to envision a path toward a designed RC with a substantially higher thermodynamic efficiency than natural photosystems.
Collapse
Affiliation(s)
- Nathan M. Ennist
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Protein Design, University of Washington, Seattle, WA, United States
- Department of Biochemistry, University of Washington, Seattle, WA, United States
- *Correspondence: Nathan M. Ennist,
| | - Steven E. Stayrook
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, United States
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, United States
- Yale Cancer Biology Institute, Yale University West Campus, West Haven, CT, United States
| | - P. Leslie Dutton
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, United States
| | - Christopher C. Moser
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
36
|
Ennist NM, Zhao Z, Stayrook SE, Discher BM, Dutton PL, Moser CC. De novo protein design of photochemical reaction centers. Nat Commun 2022; 13:4937. [PMID: 35999239 PMCID: PMC9399245 DOI: 10.1038/s41467-022-32710-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/12/2022] [Indexed: 11/09/2022] Open
Abstract
Natural photosynthetic protein complexes capture sunlight to power the energetic catalysis that supports life on Earth. Yet these natural protein structures carry an evolutionary legacy of complexity and fragility that encumbers protein reengineering efforts and obfuscates the underlying design rules for light-driven charge separation. De novo development of a simplified photosynthetic reaction center protein can clarify practical engineering principles needed to build new enzymes for efficient solar-to-fuel energy conversion. Here, we report the rational design, X-ray crystal structure, and electron transfer activity of a multi-cofactor protein that incorporates essential elements of photosynthetic reaction centers. This highly stable, modular artificial protein framework can be reconstituted in vitro with interchangeable redox centers for nanometer-scale photochemical charge separation. Transient absorption spectroscopy demonstrates Photosystem II-like tyrosine and metal cluster oxidation, and we measure charge separation lifetimes exceeding 100 ms, ideal for light-activated catalysis. This de novo-designed reaction center builds upon engineering guidelines established for charge separation in earlier synthetic photochemical triads and modified natural proteins, and it shows how synthetic biology may lead to a new generation of genetically encoded, light-powered catalysts for solar fuel production.
Collapse
Affiliation(s)
- Nathan M Ennist
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, 19104-6058, USA. .,Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA. .,Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA.
| | - Zhenyu Zhao
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, 19104-6058, USA
| | - Steven E Stayrook
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, 19104-6058, USA.,Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Yale Cancer Biology Institute, Yale University West Campus, West Haven, CT, 06516, USA
| | - Bohdana M Discher
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, 19104-6058, USA
| | - P Leslie Dutton
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, 19104-6058, USA
| | - Christopher C Moser
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, 19104-6058, USA
| |
Collapse
|
37
|
Wang J, Lisanza S, Juergens D, Tischer D, Watson JL, Castro KM, Ragotte R, Saragovi A, Milles LF, Baek M, Anishchenko I, Yang W, Hicks DR, Expòsit M, Schlichthaerle T, Chun JH, Dauparas J, Bennett N, Wicky BIM, Muenks A, DiMaio F, Correia B, Ovchinnikov S, Baker D. Scaffolding protein functional sites using deep learning. Science 2022; 377:387-394. [PMID: 35862514 PMCID: PMC9621694 DOI: 10.1126/science.abn2100] [Citation(s) in RCA: 205] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The binding and catalytic functions of proteins are generally mediated by a small number of functional residues held in place by the overall protein structure. Here, we describe deep learning approaches for scaffolding such functional sites without needing to prespecify the fold or secondary structure of the scaffold. The first approach, "constrained hallucination," optimizes sequences such that their predicted structures contain the desired functional site. The second approach, "inpainting," starts from the functional site and fills in additional sequence and structure to create a viable protein scaffold in a single forward pass through a specifically trained RoseTTAFold network. We use these two methods to design candidate immunogens, receptor traps, metalloproteins, enzymes, and protein-binding proteins and validate the designs using a combination of in silico and experimental tests.
Collapse
Affiliation(s)
- Jue Wang
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
| | - Sidney Lisanza
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
- Graduate program in Biological Physics, Structure and
Design, University of Washington, Seattle, WA 98105, USA
| | - David Juergens
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
- Molecular Engineering Graduate Program, University of
Washington, Seattle, WA 98105, USA
| | - Doug Tischer
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
| | - Joseph L. Watson
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
| | - Karla M. Castro
- Institute of Bioengineering, École Polytechnique
Fédérale de Lausanne, Lausanne CH-1015, Switzerland
| | - Robert Ragotte
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
| | - Amijai Saragovi
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
| | - Lukas F. Milles
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
| | - Minkyung Baek
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
| | - Ivan Anishchenko
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
| | - Wei Yang
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
| | - Derrick R. Hicks
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
| | - Marc Expòsit
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
- Molecular Engineering Graduate Program, University of
Washington, Seattle, WA 98105, USA
| | - Thomas Schlichthaerle
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
| | - Jung-Ho Chun
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
- Graduate program in Biological Physics, Structure and
Design, University of Washington, Seattle, WA 98105, USA
| | - Justas Dauparas
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
| | - Nathaniel Bennett
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
- Molecular Engineering Graduate Program, University of
Washington, Seattle, WA 98105, USA
| | - Basile I. M. Wicky
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
| | - Andrew Muenks
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
| | - Frank DiMaio
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
| | - Bruno Correia
- Institute of Bioengineering, École Polytechnique
Fédérale de Lausanne, Lausanne CH-1015, Switzerland
| | - Sergey Ovchinnikov
- FAS Division of Science, Harvard University, Cambridge, MA
02138, USA
- John Harvard Distinguished Science Fellowship Program,
Harvard University, Cambridge, MA 02138, USA
| | - David Baker
- Department of Biochemistry, University of Washington,
Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington,
Seattle, WA 98105, USA
- Howard Hughes Medical Institute, University of Washington,
Seattle, WA 98105, USA
| |
Collapse
|
38
|
An artificial metalloprotein with metal-adaptive coordination sites and Ni-dependent quercetinase activity. J Inorg Biochem 2022; 235:111914. [PMID: 35841720 DOI: 10.1016/j.jinorgbio.2022.111914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 06/20/2022] [Accepted: 07/03/2022] [Indexed: 11/23/2022]
Abstract
Engineering non-native metal active sites into proteins using canonical amino acids offers many advantages but is hampered by significant challenges. The TIM barrel protein, imidazole glycerol phosphate synthase from the hyperthermophilic organism Thermotoga maritima (tHisF), is well-suited for the construction of artificial metalloenzymes by this approach. To this end, we have generated a tHisF variant (tHisFEHH) with a Glu/His/His motif for metal ion coordination. Crystal structures of ZnII:tHisFEHH and NiII:tHisFEHH reveal that both metal ions bind to the engineered histidines. However, the two metals bind at distinct sites with different geometries, demonstrating the adaptability of tHisF. Only ZnII additionally ligates the Glu residue and adopts a tetrahedral geometry. The pseudo-octahedral NiII site comprises the two His and a native Ser residue. NiII:tHisFEHH catalyzes the oxidative cleavage of the flavanols quercetin and myricetin, providing an unprecedented example of an artificial metalloprotein with quercetinase activity.
Collapse
|
39
|
Synthesis of metalloporphyrin complexes based on chlorophyllin. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
40
|
Abstract
The ability to design efficient enzymes from scratch would have a profound effect on chemistry, biotechnology and medicine. Rapid progress in protein engineering over the past decade makes us optimistic that this ambition is within reach. The development of artificial enzymes containing metal cofactors and noncanonical organocatalytic groups shows how protein structure can be optimized to harness the reactivity of nonproteinogenic elements. In parallel, computational methods have been used to design protein catalysts for diverse reactions on the basis of fundamental principles of transition state stabilization. Although the activities of designed catalysts have been quite low, extensive laboratory evolution has been used to generate efficient enzymes. Structural analysis of these systems has revealed the high degree of precision that will be needed to design catalysts with greater activity. To this end, emerging protein design methods, including deep learning, hold particular promise for improving model accuracy. Here we take stock of key developments in the field and highlight new opportunities for innovation that should allow us to transition beyond the current state of the art and enable the robust design of biocatalysts to address societal needs.
Collapse
|
41
|
Tanaka T, Kuroiwa K. Supramolecular Hybrids from Cyanometallate Complexes and Diblock Copolypeptide Amphiphiles in Water. Molecules 2022; 27:3262. [PMID: 35630738 PMCID: PMC9143414 DOI: 10.3390/molecules27103262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/20/2022] Open
Abstract
The self-assembly of discrete cyanometallates has attracted significant interest due to the potential of these materials to undergo soft metallophilic interactions as well as their optical properties. Diblock copolypeptide amphiphiles have also been investigated concerning their capacity for self-assembly into morphologies such as nanostructures. The present work combined these two concepts by examining supramolecular hybrids comprising cyanometallates with diblock copolypeptide amphiphiles in aqueous solutions. Discrete cyanometallates such as [Au(CN)2]-, [Ag(CN)2]-, and [Pt(CN)4]2- dispersed at the molecular level in water cannot interact with each other at low concentrations. However, the results of this work demonstrate that the addition of diblock copolypeptide amphiphiles such as poly-(L-lysine)-block-(L-cysteine) (Lysm-b-Cysn) to solutions of these complexes induces the supramolecular assembly of the discrete cyanometallates, resulting in photoluminescence originating from multinuclear complexes with metal-metal interactions. Electron microscopy images confirmed the formation of nanostructures of several hundred nanometers in size that grew to form advanced nanoarchitectures, including those resembling the original nanostructures. This concept of combining diblock copolypeptide amphiphiles with discrete cyanometallates allows the design of flexible and functional supramolecular hybrid systems in water.
Collapse
Affiliation(s)
| | - Keita Kuroiwa
- Department of Nanoscience, Faculty of Engineering, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan;
| |
Collapse
|
42
|
Choi TS, Tezcan FA. Overcoming universal restrictions on metal selectivity by protein design. Nature 2022; 603:522-527. [PMID: 35236987 PMCID: PMC9157509 DOI: 10.1038/s41586-022-04469-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 01/21/2022] [Indexed: 11/09/2022]
Abstract
Selective metal coordination is central to the functions of metalloproteins:1,2 each metalloprotein must pair with its cognate metallocofactor to fulfil its biological role3. However, achieving metal selectivity solely through a three-dimensional protein structure is a great challenge, because there is a limited set of metal-coordinating amino acid functionalities and proteins are inherently flexible, which impedes steric selection of metals3,4. Metal-binding affinities of natural proteins are primarily dictated by the electronic properties of metal ions and follow the Irving-Williams series5 (Mn2+ < Fe2+ < Co2+ < Ni2+ < Cu2+ > Zn2+) with few exceptions6,7. Accordingly, metalloproteins overwhelmingly bind Cu2+ and Zn2+ in isolation, regardless of the nature of their active sites and their cognate metal ions1,3,8. This led organisms to evolve complex homeostatic machinery and non-equilibrium strategies to achieve correct metal speciation1,3,8-10. Here we report an artificial dimeric protein, (AB)2, that thermodynamically overcomes the Irving-Williams restrictions in vitro and in cells, favouring the binding of lower-Irving-Williams transition metals over Cu2+, the most dominant ion in the Irving-Williams series. Counter to the convention in molecular design of achieving specificity through structural preorganization, (AB)2 was deliberately designed to be flexible. This flexibility enabled (AB)2 to adopt mutually exclusive, metal-dependent conformational states, which led to the discovery of structurally coupled coordination sites that disfavour Cu2+ ions by enforcing an unfavourable coordination geometry. Aside from highlighting flexibility as a valuable element in protein design, our results illustrate design principles for constructing selective metal sequestration agents.
Collapse
Affiliation(s)
- Tae Su Choi
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA
| | - F Akif Tezcan
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
43
|
Bromberg Y, Aptekmann AA, Mahlich Y, Cook L, Senn S, Miller M, Nanda V, Ferreiro DU, Falkowski PG. Quantifying structural relationships of metal-binding sites suggests origins of biological electron transfer. SCIENCE ADVANCES 2022; 8:eabj3984. [PMID: 35030025 PMCID: PMC8759750 DOI: 10.1126/sciadv.abj3984] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 11/22/2021] [Indexed: 06/07/2023]
Abstract
Biological redox reactions drive planetary biogeochemical cycles. Using a novel, structure-guided sequence analysis of proteins, we explored the patterns of evolution of enzymes responsible for these reactions. Our analysis reveals that the folds that bind transition metal–containing ligands have similar structural geometry and amino acid sequences across the full diversity of proteins. Similarity across folds reflects the availability of key transition metals over geological time and strongly suggests that transition metal–ligand binding had a small number of common peptide origins. We observe that structures central to our similarity network come primarily from oxidoreductases, suggesting that ancestral peptides may have also facilitated electron transfer reactions. Last, our results reveal that the earliest biologically functional peptides were likely available before the assembly of fully functional protein domains over 3.8 billion years ago.Thus, life is a special, very complex form of motion of matter, but this form did not always exist, and it is not separated from inorganic nature by an impassable abyss; rather, it arose from inorganic nature as a new property in the process of evolution of the world. We must study the history of this evolution if we want to solve the problem of the origin of life. [A. I. Oparin (1)]
Collapse
Affiliation(s)
- Yana Bromberg
- Department of Biochemistry and Microbiology, Rutgers University, 76 Lipman Dr, New Brunswick, NJ 08873, USA
| | - Ariel A. Aptekmann
- Department of Biochemistry and Microbiology, Rutgers University, 76 Lipman Dr, New Brunswick, NJ 08873, USA
| | - Yannick Mahlich
- Department of Biochemistry and Microbiology, Rutgers University, 76 Lipman Dr, New Brunswick, NJ 08873, USA
| | - Linda Cook
- Program in Applied and Computational Math, Princeton University, Princeton, NJ 08540, USA
| | - Stefan Senn
- Department of Biochemistry and Microbiology, Rutgers University, 76 Lipman Dr, New Brunswick, NJ 08873, USA
| | - Maximillian Miller
- Department of Biochemistry and Microbiology, Rutgers University, 76 Lipman Dr, New Brunswick, NJ 08873, USA
| | - Vikas Nanda
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, and Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ 08854, USA
| | - Diego U. Ferreiro
- Protein Physiology Lab, Departamento de Química Biológica, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Paul G. Falkowski
- Environmental Biophysics and Molecular Ecology Program, Department of Marine and Coastal Sciences, Rutgers University, New Brunswick, NJ 08901, USA
| |
Collapse
|
44
|
Zambrano G, Sekretareva A, D'Alonzo D, Leone L, Pavone V, Lombardi A, Nastri F. Oxidative dehalogenation of trichlorophenol catalyzed by a promiscuous artificial heme-enzyme. RSC Adv 2022; 12:12947-12956. [PMID: 35527726 PMCID: PMC9067433 DOI: 10.1039/d2ra00811d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/13/2022] [Indexed: 01/21/2023] Open
Abstract
The miniaturized metalloenzyme Fe(iii)-mimochrome VI*a (Fe(iii)-MC6*a) acts as an excellent biocatalyst in the H2O2-mediated oxidative dehalogenation of the well-known pesticide and biocide 2,4,6-trichlorophenol (TCP). The artificial enzyme can oxidize TCP with a catalytic efficiency (kcat/KTCPm = 150 000 mM−1 s−1) up to 1500-fold higher than the most active natural metalloenzyme horseradish peroxidase (HRP). UV-visible and EPR spectroscopies were used to provide indications of the catalytic mechanism. One equivalent of H2O2 fully converts Fe(iii)-MC6*a into the oxoferryl-porphyrin radical cation intermediate [(Fe(iv)
Created by potrace 1.16, written by Peter Selinger 2001-2019
]]>
O)por˙+], similarly to peroxidase compound I (Cpd I). Addition of TCP to Cpd I rapidly leads to the formation of the corresponding quinone, while Cpd I decays back to the ferric resting state in the absence of substrate. EPR data suggest a catalytic mechanism involving two consecutive one-electron reactions. All results highlight the value of the miniaturization strategy for the development of chemically stable, highly efficient artificial metalloenzymes as powerful catalysts for the oxidative degradation of toxic pollutants. The artificial metalloenzyme FeMC6*a is able to perform the H2O2-mediated dechlorination of 2,4,6-trichlorophenol with unrivalled catalytic efficiency, highlighting its potential application for the removal of toxic pollutants.![]()
Collapse
Affiliation(s)
- Gerardo Zambrano
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia, 80126 Napoli, Italy
| | - Alina Sekretareva
- Department of Chemistry – Ångström, Uppsala University, Lägerhyddsvägen 1, 75120 Uppsala, Sweden
| | - Daniele D'Alonzo
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia, 80126 Napoli, Italy
| | - Linda Leone
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia, 80126 Napoli, Italy
| | - Vincenzo Pavone
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia, 80126 Napoli, Italy
| | - Angela Lombardi
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia, 80126 Napoli, Italy
| | - Flavia Nastri
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia, 80126 Napoli, Italy
| |
Collapse
|
45
|
Abstract
Natural metalloproteins perform many functions - ranging from sensing to electron transfer and catalysis - in which the position and property of each ligand and metal, is dictated by protein structure. De novo protein design aims to define an amino acid sequence that encodes a specific structure and function, providing a critical test of the hypothetical inner workings of (metallo)proteins. To date, de novo metalloproteins have used simple, symmetric tertiary structures - uncomplicated by the large size and evolutionary marks of natural proteins - to interrogate structure-function hypotheses. In this Review, we discuss de novo design applications, such as proteins that induce complex, increasingly asymmetric ligand geometries to achieve function, as well as the use of more canonical ligand geometries to achieve stability. De novo design has been used to explore how proteins fine-tune redox potentials and catalyse both oxidative and hydrolytic reactions. With an increased understanding of structure-function relationships, functional proteins including O2-dependent oxidases, fast hydrolases, and multi-proton/multi-electron reductases, have been created. In addition, proteins can now be designed using xeno-biological metals or cofactors and principles from inorganic chemistry to derive new-to-nature functions. These results and the advances in computational protein design suggest a bright future for the de novo design of diverse, functional metalloproteins.
Collapse
Affiliation(s)
- Matthew J. Chalkley
- Department of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California at San Francisco, San Francisco, (CA), USA
| | - Samuel I. Mann
- Department of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California at San Francisco, San Francisco, (CA), USA
| | - William F. DeGrado
- Department of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California at San Francisco, San Francisco, (CA), USA
| |
Collapse
|
46
|
A bound iron porphyrin is redox active in hybrid bacterial reaction centers modified to possess a four-helix bundle domain. Photochem Photobiol Sci 2021; 21:91-99. [PMID: 34850374 DOI: 10.1007/s43630-021-00142-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/16/2021] [Indexed: 10/19/2022]
Abstract
In this paper we report the design of hybrid reaction centers with a novel redox-active cofactor. Reaction centers perform the primary photochemistry of photosynthesis, namely the light-induced transfer of an electron from the bacteriochlorophyll dimer to a series of electron acceptors. Hybrid complexes were created by the fusion of an artificial four-helix bundle to the M-subunit of the reaction center. Despite the large modification, optical spectra show that the purified hybrid reaction centers assemble as active complexes that retain the characteristic cofactor absorption peaks and are capable of light-induced charge separation. The four-helix bundle could bind iron-protoporphyrin in either a reduced and oxidized state. After binding iron-protoporphyrin to the hybrid reaction centers, light excitation results in a new derivative signal with a maximum at 402 nm and minimum at 429 nm. This signal increases in amplitude with longer light durations and persists in the dark. No signal is observed when iron-protoporphyrin is added to reaction centers without the four-helix bundle domain or when a redox-inactive zinc-protoporphyrin is bound. The results are consistent with the signal arising from a new redox reaction, electron transfer from the iron-protoporphyrin to the oxidized bacteriochlorophyll dimer. These outcomes demonstrate the feasibility of binding porphyrins to the hybrid reaction centers to gain new light-driven functions.
Collapse
|
47
|
Ebensperger P, Jessen-Trefzer C. Artificial metalloenzymes in a nutshell: the quartet for efficient catalysis. Biol Chem 2021; 403:403-412. [PMID: 34653321 DOI: 10.1515/hsz-2021-0329] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/05/2021] [Indexed: 12/22/2022]
Abstract
Artificial metalloenzymes combine the inherent reactivity of transition metal catalysis with the sophisticated reaction control of natural enzymes. By providing new opportunities in bioorthogonal chemistry and biocatalysis, artificial metalloenzymes have the potential to overcome certain limitations in both drug discovery and green chemistry or related research fields. Ongoing advances in organometallic catalysis, directed evolution, and bioinformatics are enabling the design of increasingly powerful systems that outperform conventional catalysis in a growing number of cases. Therefore, this review article collects challenges and opportunities in designing artificial metalloenzymes described in recent review articles. This will provide an equitable insight for those new to and interested in the field.
Collapse
Affiliation(s)
- Paul Ebensperger
- Institute of Organic Chemistry, University of Freiburg, Albertstrasse 21, D-79104Freiburg i. Br., Germany
| | - Claudia Jessen-Trefzer
- Institute of Organic Chemistry, University of Freiburg, Albertstrasse 21, D-79104Freiburg i. Br., Germany
| |
Collapse
|
48
|
Lee JL, Ross DL, Barman SK, Ziller JW, Borovik AS. C-H Bond Cleavage by Bioinspired Nonheme Metal Complexes. Inorg Chem 2021; 60:13759-13783. [PMID: 34491738 DOI: 10.1021/acs.inorgchem.1c01754] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The functionalization of C-H bonds is one of the most challenging transformations in synthetic chemistry. In biology, these processes are well-known and are achieved with a variety of metalloenzymes, many of which contain a single metal center within their active sites. The most well studied are those with Fe centers, and the emerging experimental data show that high-valent iron oxido species are the intermediates responsible for cleaving the C-H bond. This Forum Article describes the state of this field with an emphasis on nonheme Fe enzymes and current experimental results that provide insights into the properties that make these species capable of C-H bond cleavage. These parameters are also briefly considered in regard to manganese oxido complexes and Cu-containing metalloenzymes. Synthetic iron oxido complexes are discussed to highlight their utility as spectroscopic and mechanistic probes and reagents for C-H bond functionalization. Avenues for future research are also examined.
Collapse
Affiliation(s)
- Justin L Lee
- Department of Chemistry, University of California-Irvine, 1102 Natural Sciences II, Irvine, California 92697, United States
| | - Dolores L Ross
- Department of Chemistry, University of California-Irvine, 1102 Natural Sciences II, Irvine, California 92697, United States
| | - Suman K Barman
- Department of Chemistry, University of California-Irvine, 1102 Natural Sciences II, Irvine, California 92697, United States
| | - Joseph W Ziller
- Department of Chemistry, University of California-Irvine, 1102 Natural Sciences II, Irvine, California 92697, United States
| | - A S Borovik
- Department of Chemistry, University of California-Irvine, 1102 Natural Sciences II, Irvine, California 92697, United States
| |
Collapse
|
49
|
Unravelling the Structure of the Tetrahedral Metal-Binding Site in METP3 through an Experimental and Computational Approach. Molecules 2021; 26:molecules26175221. [PMID: 34500655 PMCID: PMC8434281 DOI: 10.3390/molecules26175221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 11/17/2022] Open
Abstract
Understanding the structural determinants for metal ion coordination in metalloproteins is a fundamental issue for designing metal binding sites with predetermined geometry and activity. In order to achieve this, we report in this paper the design, synthesis and metal binding properties of METP3, a homodimer made up of a small peptide, which self assembles in the presence of tetrahedrally coordinating metal ions. METP3 was obtained through a redesign approach, starting from the previously developed METP molecule. The undecapeptide sequence of METP, which dimerizes to house a Cys4 tetrahedral binding site, was redesigned in order to accommodate a Cys2His2 site. The binding properties of METP3 were determined toward different metal ions. Successful assembly of METP3 with Co(II), Zn(II) and Cd(II), in the expected 2:1 stoichiometry and tetrahedral geometry was proven by UV-visible spectroscopy. CD measurements on both the free and metal-bound forms revealed that the metal coordination drives the peptide chain to fold into a turned conformation. Finally, NMR data of the Zn(II)-METP3 complex, together with a retrostructural analysis of the Cys-X-X-His motif in metalloproteins, allowed us to define the model structure. All the results establish the suitability of the short METP sequence for accommodating tetrahedral metal binding sites, regardless of the first coordination ligands.
Collapse
|
50
|
|