495 publications

495 publications

Assembly and Evolution of Artificial Metalloenzymes within E. coli Nissle 1917 for Enantioselective and Site-Selective Functionalization of C─H and C═C Bonds

Hartwig, J.F.

J. Am. Chem. Soc. 2022, 144, 883-890, 10.1021/jacs.1c10975

The potential applications afforded by the generation and reactivity of artificial metalloenzymes (ArMs) in microorganisms are vast. We show that a non-pathogenic E. coli strain, Nissle 1917 (EcN), is a suitable host for the creation of ArMs from cytochrome P450s and artificial heme cofactors. An outer-membrane receptor in EcN transports an iridium porphyrin into the cell, and the Ir-CYP119 (CYP119 containing iridium porphyrin) assembled in vivo catalyzes carbene insertions into benzylic C–H bonds enantioselectively and site-selectively. The application of EcN as a whole-cell screening platform eliminates the need for laborious processing procedures, drastically increases the ease and throughput of screening, and accelerates the development of Ir-CYP119 with improved catalytic properties. Studies to identify the transport machinery suggest that a transporter different from the previously assumed ChuA receptor serves to usher the iridium porphyrin into the cytoplasm.


Metal: Ir
Ligand type: Porphyrin
Host protein: CYP119
Anchoring strategy: Dative
Optimization: Genetic
Reaction: C-H activation
Max TON: 1314
ee: 84
PDB: ---
Notes: In vivo

Computationally Driven Design of an Artificial Metalloenzyme Using Supramolecular Anchoring Strategies of Iridium Complexes to Alcohol Dehydrogenase

Jäger, C.M.; Pordea, A.

Faraday Discuss. 2022, 10.1039/d1fd00070e

Artificial metalloenzymes (ArMs) confer non-biological reactivities to biomolecules, whilst taking advantage of the biomolecular architecture in terms of their selectivity and renewable origin. In particular, the design of ArMs by the supramolecular anchoring of metal catalysts to protein hosts provides flexible and easy to optimise systems. The use of cofactor dependent enzymes as hosts gives the advantage of both a (hydrophobic) binding site for the substrate and a cofactor pocket to accommodate the catalyst. Here, we present a computationally driven design approach of ArMs for the transfer hydrogenation reaction of cyclic imines, starting from the NADP+-dependent alcohol dehydrogenase from Thermoanaerobacter brockii (TbADH). We tested and developed a molecular docking workflow to define and optimize iridium catalysts with high affinity for the cofactor binding site of TbADH. The workflow uses high throughput docking of compound libraries to identify key structural motifs for high affinity, followed by higher accuracy docking methods on smaller, focused ligand and catalyst libraries. Iridium sulfonamide catalysts were selected and synthesised, containing either a triol, a furane, or a carboxylic acid to provide the interaction with the cofactor binding pocket. IC50 values of the resulting complexes during TbADH-catalysed alcohol oxidation were determined by competition experiments and were between 4.410 mM and 0.052 mM, demonstrating the affinity of the iridium complexes for either the substrate or the cofactor binding pocket of TbADH. The catalytic activity of the free iridium complexes in solution showed a maximal turnover number (TON) of 90 for the reduction of salsolidine by the triol-functionalised iridium catalyst, whilst in the presence of TbADH, only the iridium catalyst with the triol anchoring functionality showed activity for the same reaction (TON of 36 after 24 h). The observation that the artificial metalloenzymes developed here lacked stereoselectivity demonstrates the need for the further investigation and optimisation of the ArM. Our results serve as a starting point for the design of robust artificial metalloenzymes, exploiting supramolecular anchoring to natural NAD(P)H binding pockets.


Metal: Ir
Ligand type: Amino-sulfonamide; Cp*
Host protein: Alcohol dehydrogenase
Anchoring strategy: Supramolecular
Optimization: Chemical & genetic
Max TON: 81±0.80
ee: ---
PDB: 1YKF
Notes: ---

De Novo Metalloprotein Design

Review

DeGrado, W.F.

Nat. Rev. Chem. 2022, 6, 31-50, 10.1038/s41570-021-00339-5

Natural metalloproteins perform many functions — ranging from sensing to electron transfer and catalysis — in which the position and property of each ligand and metal are 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/multielectron reductases have been created. In addition, proteins can now be designed using xenobiological 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.


Notes: ---

Binding of Vanadium Ions and Complexes to Proteins and Enzymes in Aqueous Solution

Review

Garribba, E.

Coord. Chem. Rev. 2021, 449, 214192, 10.1016/j.ccr.2021.214192

The understanding of the role of vanadium enzymes and of vanadium compounds (VCs) in biology, as well as the design of new vanadium-based species for catalysis, materials science and medicinal chemistry has exponentially increased during the last decades. In biological systems, VCs may rapidly interconvert under physiological conditions and several V-containing moieties may be formed and bind to proteins. These interactions play key roles in the form transported in blood, in the uptake by cells, in inhibition properties and mechanism of action of essential and pharmacologically active V species. In this review, we focus on the recent advances made, namely in the application of the theoretical methodologies that allowed the description of the coordinative and non-covalent VC–protein interactions. The text is organized in six main topics: a general overview of the most important experimental and computational techniques useful to study these systems, a discussion on the nature of binding process, the recent advances on the comprehension of the V-containing natural and artificial enzymes, the interaction of mononuclear VCs with blood and other physiologically relevant proteins, the binding of polyoxidovanadates(V) to proteins and, finally, the biological and therapeutic implications of the interaction of pharmacologically relevant VCs with proteins and enzymes. Recent developments on vanadium-containing nitrogenases, haloperoxidases and nitrate reductases, and binding of VCs to transferrin, albumins, immunoglobulins, hemoglobin, lysozyme, myoglobin, ubiquitin and cytochrome c are discussed. Challenges and ideas about desirable features and potential drawbacks of VCs in biology and medicine and future directions to explore this chemistry area are also presented. The deeper understanding of the interactions of V-species with proteins, and the discussed data may provide the basis to undertake the investigation, design and development of new potentially active VCs with a more solid knowledge to predict their binding to biological receptors at a molecular point of view.


Notes: ---

Nitrene Transfers Mediated by Natural and Artificial Iron Enzymes

Review

Latour, J.-M.

J. Inorg. Biochem. 2021, 225, 111613, 10.1016/j.jinorgbio.2021.111613

Amines are ubiquitous in biology and pharmacy. As a consequence, introducing N functionalities in organic molecules is attracting strong continuous interest. The past decade has witnessed the emergence of very efficient and selective catalytic systems achieving this goal thanks to engineered hemoproteins. In this review, we examine how these enzymes have been engineered focusing rather on the rationale behind it than the methodology employed. These studies are put in perspective with respect to in vitro and in vivo nitrene transfer processes performed by cytochromes P450. An emphasis is put on mechanistic aspects which are confronted to current molecular knowledge of these reactions. Forthcoming developments are delineated.


Notes: ---

Semi-Synthetic Hydrogenases—In Vitro and In Vivo Applications

Review

Berggren, G.

Curr. Opin. Green Sustain. Chem. 2021, 32, 100521, 10.1016/j.cogsc.2021.100521

Hydrogenases are gas processing redox enzymes central in hydrogen metabolism. The interdisciplinary nature of hydrogenase research is underscored by the development of “artificial maturation”, enabling the preparation of semi-synthetic hydrogenases through the incorporation of synthetic cofactors into a range of apo-hydrogenase hosts under in vitro and in vivo conditions. Herein, we discuss how the preparation of such semi-synthetic [FeFe]-hydrogenases has elucidated structural elements of the cofactor critical for catalysis and reactivity towards known inhibitors. It has also provided a convenient method for exploring the biodiversity of this enzyme family and thereby facilitated investigation of the role of the outer-coordination sphere in tuning the reactivity of the H-cluster. In parallel, hijacking the assembly line of the [FeFe]-hydrogenase through incorporation of synthetic precursors has provided detailed insight into the biosynthesis of the H-cluster. Moreover, it has allowed the preparation of Mn analogs of [Fe] hydrogenase.


Notes: ---

Unnatural Biosynthesis by an Engineered Microorganism with Heterologously Expressed Natural Enzymes and an Artificial Metalloenzyme

Clark, D.S.; Hartwig, J.F.; Keasling, J.D.; Mukhopadhyay, A.

Nat. Chem. 2021, 13, 1186-1191, 10.1038/s41557-021-00801-3

Synthetic biology enables microbial hosts to produce complex molecules from organisms that are rare or difficult to cultivate, but the structures of these molecules are limited to those formed by reactions of natural enzymes. The integration of artificial metalloenzymes (ArMs) that catalyse unnatural reactions into metabolic networks could broaden the cache of molecules produced biosynthetically. Here we report an engineered microbial cell expressing a heterologous biosynthetic pathway, containing both natural enzymes and ArMs, that produces an unnatural product with high diastereoselectivity. We engineered Escherichia coli with a heterologous terpene biosynthetic pathway and an ArM containing an iridium–porphyrin complex that was transported into the cell with a heterologous transport system. We improved the diastereoselectivity and product titre of the unnatural product by evolving the ArM and selecting the appropriate gene induction and cultivation conditions. This work shows that synthetic biology and synthetic chemistry can produce, by combining natural and artificial enzymes in whole cells, molecules that were previously inaccessible to nature.


Metal: Ir
Ligand type: Methyl; Porphyrin
Host protein: CYP119
Anchoring strategy: Metal substitution
Optimization: Genetic
Reaction: Cyclopropanation
Max TON: 2130
ee: ---
PDB: ---
Notes: TON in vivo of (-)-carvone, WITHOUT limonene biosynthetic genes

Engineering Dirhodium Artificial Metalloenzymes for Diazo Coupling Cascade Reactions

Lewis, J.C.; Roux, B.

Angew. Chem. Int. Ed. 2021, 60, 23672-23677, 10.1002/anie.202107982

Artificial metalloenzymes (ArMs) are commonly used to control the stereoselectivity of catalytic reactions, but controlling chemoselectivity remains challenging. In this study, we engineer a dirhodium ArM to catalyze diazo cross-coupling to form an alkene that, in a one-pot cascade reaction, is reduced to an alkane with high enantioselectivity (typically >99 % ee) by an alkene reductase. The numerous protein and small molecule components required for the cascade reaction had minimal effect on ArM catalysis. Directed evolution of the ArM led to improved yields and E/Z selectivities for a variety of substrates, which translated to cascade reaction yields. MD simulations of ArM variants were used to understand the structural role of the cofactor on ArM conformational dynamics. These results highlight the ability of ArMs to control both catalyst stereoselectivity and chemoselectivity to enable reactions in complex media that would otherwise lead to undesired side reactions.


Metal: Rh
Ligand type: Dirhodium
Anchoring strategy: Covalent
Optimization: ---
Max TON: ---
ee: >99
PDB: ---
Notes: 61% max combined yield for cascade reactions

Design of Artificial Metalloenzymes with Multiple Inorganic Elements: The More the Merrier

Review

Song, W.J.

J. Inorg. Biochem. 2021, 223, 111552, 10.1016/j.jinorgbio.2021.111552

A large fraction of metalloenzymes harbors multiple metal-centers that are electronically and/or functionally arranged within their proteinaceous environments. To explore the orchestration of inorganic and biochemical components and to develop bioinorganic catalysts and materials, we have described selected examples of artificial metalloproteins having multiple metallocofactors that were grouped depending on their initial protein scaffolds, the nature of introduced inorganic moieties, and the method used to propagate the number of metal ions within a protein. They demonstrated that diverse inorganic moieties can be selectively grafted and modulated in protein environments, providing a retrosynthetic bottom-up approach in the design of versatile and proficient biocatalysts and biomimetic model systems to explore fundamental questions in bioinorganic chemistry.


Notes: ---

Engineered and Artificial Metalloenzymes for Selective C–H Functionalization

Review

Fasan, R.

Curr. Opin. Green Sustain. Chem. 2021, 31, 100494, 10.1016/j.cogsc.2021.100494

The direct functionalization of C–H bonds constitutes a powerful strategy to construct and diversify organic molecules. However, controlling the chemo- and site-selectivity of this transformation, particularly in complex molecular settings, represents a significant challenge. Metalloenzymes are ideal platforms for achieving catalyst-controlled selective C–H bond functionalization as their reactivities can be tuned by protein engineering and/or redesign of their cofactor environment. In this review, we highlight recent progress in the development of engineered and artificial metalloenzymes for C–H functionalization, with a focus on biocatalytic strategies for selective C–H oxyfunctionalization and halogenation as well as C–H amination and C–H carbene insertion via abiological nitrene and carbene transfer chemistries. Engineered heme and nonheme iron dependent enzymes have emerged as promising scaffolds for executing these transformations with high chemo-, regio-, and stereocontrol as well as tunable selectivity. These emerging systems and methodologies have expanded the toolbox of sustainable strategies for organic synthesis and created new opportunities for the generation of chiral building blocks, the late-stage C–H functionalization of complex molecules, and the total synthesis of natural products.


Notes: ---

Engineering and Emerging Applications of Artificial Metalloenzymes with Whole Cells

Review

Sauer, D.F.; Schwaneberg, U.

Nat. Catal. 2021, 4, 814-827, 10.1038/s41929-021-00673-3

The field of artificial metalloenzymes (ArMs) is rapidly growing and ArMs are attracting increasing attention, for example, in the fields of biosensing and drug therapy. Protein-engineering methods that are commonly used to tailor the properties of natural enzymes are more frequently included in the design of ArMs. In particular, directed evolution allows the fine-tuning of ArMs, ultimately assisting in the development of their enormous potential. The integration of ArMs in whole cells enables their in vivo application and facilitates high-throughput directed-evolution methodologies. In this Review, we highlight the recent progress of whole-cell conversions and applications of ArMs and critically discuss their limitations and prospects. To focus on ArMs and their specific properties, advantages and challenges, the evolution of natural enzymes for non-natural reactions will not be covered.


Notes: ---

Design of Artificial Metalloenzymes for the Reduction of Nicotinamide Cofactors

Pordea, A.

J. Inorg. Biochem. 2021, 220, 111446, 10.1016/j.jinorgbio.2021.111446

Artificial metalloenzymes result from the insertion of a catalytically active metal complex into a biological scaffold, generally a protein devoid of other catalytic functionalities. As such, their design requires efforts to engineer substrate binding, in addition to accommodating the artificial catalyst. Here we constructed and characterised artificial metalloenzymes using alcohol dehydrogenase as starting point, an enzyme which has both a cofactor and a substrate binding pocket. A docking approach was used to determine suitable positions for catalyst anchoring to single cysteine mutants, leading to an artificial metalloenzyme capable to reduce both natural cofactors and the hydrophobic 1-benzylnicotinamide mimic. Kinetic studies revealed that the new construct displayed a Michaelis-Menten behaviour with the native nicotinamide cofactors, which were suggested by docking to bind at a surface exposed site, different compared to their native binding position. On the other hand, the kinetic and docking data suggested that a typical enzyme behaviour was not observed with the hydrophobic 1-benzylnicotinamide mimic, with which binding events were plausible both inside and outside the protein. This work demonstrates an extended substrate scope of the artificial metalloenzymes and provides information about the binding sites of the nicotinamide substrates, which can be exploited to further engineer artificial metalloenzymes for cofactor regeneration.


Metal: Rh
Host protein: Alcohol dehydrogenase
Anchoring strategy: Covalent
Optimization: Chemical & genetic
Max TON: ---
ee: ---
PDB: 1YKF
Notes: ---

Noncanonical Heme Ligands Steer Carbene Transfer Reactivity in an Artificial Metalloenzyme

Hilvert, D.

Angew. Chem. Int. Ed. 2021, 60, 15063-15068, 10.1002/anie.202103437

Changing the primary metal coordination sphere is a powerful strategy for tuning metalloprotein properties. Here we used amber stop codon suppression with engineered pyrrolysyl-tRNA synthetases, including two newly evolved enzymes, to replace the proximal histidine in myoglobin with Nδ-methylhistidine, 5-thiazoylalanine, 4-thiazoylalanine and 3-(3-thienyl)alanine. In addition to tuning the heme redox potential over a >200 mV range, these noncanonical ligands modulate the protein's carbene transfer activity with ethyl diazoacetate. Variants with increased reduction potential proved superior for cyclopropanation and N–H insertion, whereas variants with reduced Eo values gave higher S–H insertion activity. Given the functional importance of histidine in many enzymes, these genetically encoded analogues could be valuable tools for probing mechanism and enabling new chemistries.


Metal: Fe
Ligand type: Histidine residues
Host protein: Myoglobin (Mb)
Anchoring strategy: Heme
Optimization: Genetic
Reaction: Cyclopropanation
Max TON: ---
ee: >99
PDB: ---
Notes: yield: styrene cyclopropanation 71% max, cf free heme <5%

Metal: Fe
Ligand type: Histidine residues
Host protein: Myoglobin (Mb)
Anchoring strategy: Heme
Optimization: Genetic
Reaction: N-H Insertion
Max TON: ---
ee: ---
PDB: ---
Notes: Yield: aniline insertion 74-93%

Metal: Fe
Ligand type: Histidine residues
Host protein: Myoglobin (Mb)
Anchoring strategy: Heme
Optimization: Genetic
Reaction: S-H insertion
Max TON: ---
ee: ---
PDB: ---
Notes: Yield: thiophenol insertion 18-36% but still outperforms heme

Diversifying Metal–Ligand Cooperative Catalysis in Semi‐Synthetic [Mn]‐Hydrogenases

Hu, X.; Shima, S.

Angew. Chem. Int. Ed. 2021, 60, 13350-13357, 10.1002/anie.202100443

The reconstitution of [Mn]-hydrogenases using a series of MnI complexes is described. These complexes are designed to have an internal base or pro-base that may participate in metal–ligand cooperative catalysis or have no internal base or pro-base. Only MnI complexes with an internal base or pro-base are active for H2 activation; only [Mn]-hydrogenases incorporating such complexes are active for hydrogenase reactions. These results confirm the essential role of metal–ligand cooperation for H2 activation by the MnI complexes alone and by [Mn]-hydrogenases. Owing to the nature and position of the internal base or pro-base, the mode of metal–ligand cooperation in two active [Mn]-hydrogenases is different from that of the native [Fe]-hydrogenase. One [Mn]-hydrogenase has the highest specific activity of semi-synthetic [Mn]- and [Fe]-hydrogenases. This work demonstrates reconstitution of active artificial hydrogenases using synthetic complexes differing greatly from the native active site.


Metal: Mn
Ligand type: CO; Pyridone
Anchoring strategy: Reconstitution
Optimization: Chemical
Reaction: Hydrogenation
Max TON: ---
ee: ---
PDB: ---
Notes: ---

Repurposing Metalloproteins as Mimics of Natural Metalloenzymes for Small-Molecule Activation

Review

Holland, P.L.

J. Inorg. Biochem. 2021, 219, 111430, 10.1016/j.jinorgbio.2021.111430

Artificial metalloenzymes (ArMs) consist of an unnatural metal or cofactor embedded in a protein scaffold, and are an excellent platform for applying the concepts of protein engineering to catalysis. In this Focused Review, we describe the application of ArMs as simple, tunable artificial models of the active sites of complex natural metalloenzymes for small-molecule activation. In this sense, ArMs expand the strategies of synthetic model chemistry to protein-based supporting ligands with potential for participation from the second coordination sphere. We focus specifically on ArMs that are structural, spectroscopic, and functional models of enzymes for activation of small molecules like CO, CO2, O2, N2, and NO, as well as production/consumption of H2. These ArMs give insight into the identities and roles of metalloenzyme structural features within and near the cofactor. We give examples of ArM work relevant to hydrogenases, acetyl-coenzyme A synthase, superoxide dismutase, heme oxygenases, nitric oxide reductase, methyl-coenzyme M reductase, copper-O2 enzymes, and nitrogenases.


Notes: ---

A De Novo‐Designed Artificial Metallopeptide Hydrogenase: Insights into Photochemical Processes and the Role of Protonated Cys

Chakraborty, S.

ChemSusChem 2021, 14, 2237-2246, 10.1002/cssc.202100122

Hydrogenase enzymes produce H2 gas, which can be a potential source of alternative energy. Inspired by the [NiFe] hydrogenases, we report the construction of a de novo-designed artificial hydrogenase (ArH). The ArH is a dimeric coiled coil where two cysteine (Cys) residues are introduced at tandem a/d positions of a heptad to create a tetrathiolato Ni binding site. Spectroscopic studies show that Ni binding significantly stabilizes the peptide producing electronic transitions characteristic of Ni-thiolate proteins. The ArH produces H2 photocatalytically, demonstrating a bell-shaped pH-dependence on activity. Fluorescence lifetimes and transient absorption spectroscopic studies are undertaken to elucidate the nature of pH-dependence, and to monitor the reaction kinetics of the photochemical processes. pH titrations are employed to determine the role of protonated Cys on reactivity. Through combining these results, a fine balance is found between solution acidity and the electron transfer steps. This balance is critical to maximize the production of NiI-peptide and protonation of the NiII−H− intermediate (Ni−R) by a Cys (pKa≈6.4) to produce H2.


Metal: Ni
Ligand type: Amino acid
Host protein: Synthetic peptide
Anchoring strategy: Dative
Optimization: Chemical
Reaction: H2 evolution
Max TON: 44
ee: ---
PDB: ---
Notes: ---

Chemogenetic Evolution of a Peroxidase-like Artificial Metalloenzyme

Okuda, J.; Schwaneberg, U.

ACS Catal. 2021, 11, 5079-5087, 10.1021/acscatal.1c00134

Directed evolution has helped enzyme engineering to remarkable successes in the past. A main challenge in directed evolution is to find the most suitable starting point, that is, an enzyme that allows maximum “evolvability”. Consisting of a synthetic cofactor embedded in a protein scaffold, artificial metalloenzymes (ArMs) are reminiscent of rough-hewn ancestral metalloproteins and thus could provide an evolutionarily clean slate. Here, we report the design and directed evolution of an ArM with peroxidase-like properties based on the nitrobindin variant, NB4. After identifying a suitable artificial metal cofactor, two rounds of directed evolution were sufficient to elevate the ArM’s activity to levels akin to those of some natural peroxidases (up to kcat = 14.1 s–1 and kcat/Km = 52,800 M–1 s–1). A substitution to arginine in the distal cofactor environment (position 76) was the key to boost the peroxidase activity. Molecular dynamics simulations reveal a remarkable flexibility in the distal site of the NB4 scaffold that is absent in the nitrobindin wildtype and which allows the unrestricted movement of the catalytically important Arg76. In addition to the oxidation of the common redox mediators (ABTS, syringaldehyde, and 2,6-dimethoxyphenol), the ArM proved efficient in the decolorization of three recalcitrant dyes (indigo carmine, reactive blue 19, and reactive black 5) and was amenable to several rounds of ArM recycling.


Metal: Mn
Ligand type: Porphyrin
Host protein: Nitrobindin (Nb)
Anchoring strategy: Supramolecular
Optimization: Chemical & genetic
Reaction: Oxidation
Max TON: ---
ee: ---
PDB: ---
Notes: kcat = 14.1 s−1 and kcat/Km = 52,800 M−1 s −1

Rational Design of a Miniature Photocatalytic CO2-Reducing Enzyme

Liu, X.; Tian, C.; Wang, J.

ACS Catal. 2021, 11, 5628-5635, 10.1021/acscatal.1c00287

Photosystem I (PSI) is a very large membrane protein complex (∼1000 kDa) harboring P700*, the strongest reductant known in biological systems, which is responsible for driving NAD(P)+ and ultimately for CO2 reduction. Although PSI is one of the most important components in the photosynthesis machinery, it has remained difficult to enhance PSI functions through genetic engineering due to its enormous complexity. Inspired by PSI’s ability to undergo multiple-step photo-induced electron hopping from P700* to iron–sulfur [Fe4S4] clusters, we designed a 33 kDa miniature photocatalytic CO2-reducing enzyme (mPCE) harboring a chromophore (BpC) and two [Fe4S4] clusters (FeA/FeB). Through reduction potential fine-tuning, we optimized the multiple-step electron hopping from BpC to FeA/FeB, culminating in a CO2/HCOOH conversion quantum efficiency of 1.43%. As mPCE can be overexpressed with a high yield in Escherichia coli cells without requiring synthetic cofactors, further development along this route may result in rapid photo-enzyme quantum yield improvement and functional expansion through an efficient directed evolution process.


Metal: Fe
Ligand type: Amino acid
Host protein: Ferredoxin (Fd)
Anchoring strategy: Dative
Optimization: Genetic
Reaction: CO2 reduction
Max TON: 35
ee: ---
PDB: ---
Notes: ---

An Artificial Cofactor Catalyzing the Baylis‐Hillman Reaction with Designed Streptavidin as Protein Host

Höcker, B.; Lechner, H.

ChemBioChem 2021, 22, 1573-1577, 10.1002/cbic.202000880

An artificial cofactor based on an organocatalyst embedded in a protein has been used to conduct the Baylis-Hillman reaction in a buffered system. As protein host, we chose streptavidin, as it can be easily crystallized and thereby supports the design process. The protein host around the cofactor was rationally designed on the basis of high-resolution crystal structures obtained after each variation of the amino acid sequence. Additionally, DFT-calculated intermediates and transition states were used to rationalize the observed activity. Finally, repeated cycles of structure determination and redesign led to a system with an up to one order of magnitude increase in activity over the bare cofactor and to the most active proteinogenic catalyst for the Baylis-Hillman reaction known today.


Metal: ---
Ligand type: ---
Host protein: Streptavidin (Sav)
Anchoring strategy: Supramolecular
Max TON: ---
ee: ---
PDB: 6T1E
Notes: Organocatalyst

Controlled Ligand Exchange Between Ruthenium Organometallic Cofactor Precursors and a Naïve Protein Scaffold Generates Artificial Metalloenzymes Catalysing Transfer Hydrogenation

Barker, P.D.; Boss, S.R.

Angew. Chem. Int. Ed. 2021, 60, 10919-10927, 10.1002/anie.202015834

Many natural metalloenzymes assemble from proteins and biosynthesised complexes, generating potent catalysts by changing metal coordination. Here we adopt the same strategy to generate artificial metalloenzymes (ArMs) using ligand exchange to unmask catalytic activity. By systematically testing RuII(η6-arene)(bipyridine) complexes designed to facilitate the displacement of functionalised bipyridines, we develop a fast and robust procedure for generating new enzymes via ligand exchange in a protein that has not evolved to bind such a complex. The resulting metal cofactors form peptidic coordination bonds but also retain a non-biological ligand. Tandem mass spectrometry and 19F NMR spectroscopy were used to characterise the organometallic cofactors and identify the protein-derived ligands. By introduction of ruthenium cofactors into a 4-helical bundle, transfer hydrogenation catalysts were generated that displayed a 35-fold rate increase when compared to the respective small molecule reaction in solution.


Metal: Ru
Ligand type: Arene; Bipyridine
Host protein: Cytochrome b562
Anchoring strategy: Dative
Optimization: ---
Max TON: ---
ee: ---
PDB: ---
Notes: 35 fold rate increase

Metal: Ru
Ligand type: Arene; Bipyridine
Host protein: Ubiquitin
Anchoring strategy: Dative
Optimization: ---
Max TON: ---
ee: ---
PDB: ---
Notes: 35 fold rate increase

Abiotic reduction of ketones with silanes catalysed by carbonic anhydrase through an enzymatic zinc hydride

Hartwig, J.F.

Nat. Chem. 2021, 13, 312-318, 10.1038/s41557-020-00633-7

Enzymatic reactions through mononuclear metal hydrides are unknown in nature, despite the prevalence of such intermediates in the reactions of synthetic transition-metal catalysts. If metalloenzymes could react through abiotic intermediates like these, then the scope of enzyme-catalysed reactions would expand. Here we show that zinc-containing carbonic anhydrase enzymes catalyse hydride transfers from silanes to ketones with high enantioselectivity. We report mechanistic data providing strong evidence that the process involves a mononuclear zinc hydride. This work shows that abiotic silanes can act as reducing equivalents in an enzyme-catalysed process and that monomeric hydrides of electropositive metals, which are typically unstable in protic environments, can be catalytic intermediates in enzymatic processes. Overall, this work bridges a gap between the types of transformation in molecular catalysis and biocatalysis.


Metal: Zn
Ligand type: Histidine residues
Anchoring strategy: Native
Optimization: Chemical
Max TON: 500
ee: >99
PDB: ---
Notes: ---

Substrate Promiscuity of a De Novo Designed Peroxidase

Anderson, J.L.R.

J. Inorg. Biochem. 2021, 217, 111370, 10.1016/j.jinorgbio.2021.111370

The design and construction of de novo enzymes offer potentially facile routes to exploiting powerful chemistries in robust, expressible and customisable protein frameworks, while providing insight into natural enzyme function. To this end, we have recently demonstrated extensive catalytic promiscuity in a heme-containing de novo protein, C45. The diverse transformations that C45 catalyses include substrate oxidation, dehalogenation and carbon‑carbon bond formation. Here we explore the substrate promiscuity of C45's peroxidase activity, screening the de novo enzyme against a panel of peroxidase and dehaloperoxidase substrates. Consistent with the function of natural peroxidases, C45 exhibits a broad spectrum of substrate activities with selectivity dictated primarily by the redox potential of the substrate, and by extension, the active oxidising species in peroxidase chemistry, compounds I and II. Though the comparison of these redox potentials provides a threshold for determining activity for a given substrate, substrate:protein interactions are also likely to play a significant role in determining electron transfer rates from substrate to heme, affecting the kinetic parameters of the enzyme. We also used biomolecular simulation to screen substrates against a computational model of C45 to identify potential interactions and binding sites. Several sites of interest in close proximity to the heme cofactor were discovered, providing insight into the catalytic workings of C45.


Metal: Fe
Ligand type: Porphyrin
Anchoring strategy: Covalent
Optimization: ---
Reaction: Peroxidation
Max TON: ---
ee: ---
PDB: ---
Notes: ---

Engineering Thermostability in Artificial Metalloenzymes to Increase Catalytic Activity

Jarvis, A.G.

ACS Catal. 2021, 11, 3620-3627, 10.1021/acscatal.0c05413

Protein engineering has shown widespread use in improving the industrial application of enzymes and broadening the conditions they are able to operate under by increasing their thermostability and solvent tolerance. Here, we show that protein engineering can be used to increase the thermostability of an artificial metalloenzyme. Thermostable variants of the human steroid carrier protein 2L, modified to bind a metal catalyst, were created by rational design using structural data and a 3DM database. These variants were tested to identify mutations that enhanced the stability of the protein scaffold, and a significant increase in melting temperature was observed with a number of modified metalloenzymes. The ability to withstand higher reaction temperatures resulted in an increased activity in the hydroformylation of 1-octene, with more than fivefold improvement in turnover number, whereas the selectivity for linear aldehyde remained high up to 80%.


Metal: Rh
Ligand type: Phosphine
Anchoring strategy: Covalent
Optimization: Genetic
Reaction: Hydroformylation
Max TON: 415
ee: ---
PDB: 1IKT
Notes: ---

Modular Design of G-Quadruplex MetalloDNAzymes for Catalytic C–C Bond Formations with Switchable Enantioselectivity

Clever, G.H.

J. Am. Chem. Soc. 2021, 143, 3555-3561, 10.1021/jacs.0c13251

Metal-binding DNA structures with catalytic function are receiving increasing interest. Although a number of metalloDNAzymes have been reported to be highly efficient, the exact coordination/position of their catalytic metal center is often unknown. Here, we present a new approach to rationally develop metalloDNAzymes for Lewis acid-catalyzed reactions such as enantioselective Michael additions. Our strategy relies on the predictable folding patterns of unimolecular DNA G-quadruplexes, combined with the concept of metal-mediated base-pairing. Transition-metal coordination environments were created in G-quadruplex loop regions, accessible by substrates. Therefore, protein-inspired imidazole ligandoside L was covalently incorporated into a series of G-rich DNA strands by solid-phase synthesis. Iterative rounds of DNA sequence design and catalytic assays allowed us to select tailored metalloDNAzymes giving high conversions and excellent enantioselectivities (≥99%). Based on their primary sequence, folding pattern, and metal coordination mode, valuable information on structure–activity relationships could be extracted. Variation of the number and position of ligand L within the sequence allowed us to control the formation of (S) and (R) enantiomeric reaction products, respectively.


Metal: Cu
Ligand type: DNA (G quadruplex)
Host protein: metalloDNAzyme
Anchoring strategy: Imidazole ligandoside
Optimization: Genetic
Reaction: Michael addition
Max TON: ---
ee: >99
PDB: ---
Notes: Km 35.2 uM, vmax-8.2 nM min-1

In Vivo Assembly of Artificial Metalloenzymes and Application in Whole‐Cell Biocatalysis

Roelfes, G.

Angew. Chem. Int. Ed. 2021, 60, 5913-5920, 10.1002/anie.202014771

We report the supramolecular assembly of artificial metalloenzymes (ArMs), based on the Lactococcal multidrug resistance regulator (LmrR) and an exogeneous copper(II)–phenanthroline complex, in the cytoplasm of E. coli cells. A combination of catalysis, cell-fractionation, and inhibitor experiments, supplemented with in-cell solid-state NMR spectroscopy, confirmed the in-cell assembly. The ArM-containing whole cells were active in the catalysis of the enantioselective Friedel–Crafts alkylation of indoles and the Diels–Alder reaction of azachalcone with cyclopentadiene. Directed evolution resulted in two different improved mutants for both reactions, LmrR_A92E_M8D and LmrR_A92E_V15A, respectively. The whole-cell ArM system required no engineering of the microbial host, the protein scaffold, or the cofactor to achieve ArM assembly and catalysis. We consider this a key step towards integrating abiological catalysis with biosynthesis to generate a hybrid metabolism.


Metal: Cu
Ligand type: Phenanthroline
Anchoring strategy: Supramolecular
Optimization: Genetic
Max TON: ---
ee: 98
PDB: 3F8F
Notes: ---

Metal: Cu
Ligand type: Phenanthroline
Anchoring strategy: Supramolecular
Optimization: Genetic
Max TON: ---
ee: 84
PDB: 3F8F
Notes: ---

Alternative Strategy to Obtain Artificial Imine Reductase by Exploiting Vancomycin/D-Ala-D-Ala Interactions with an Iridium Metal Complex

Pellegrino, S.; Rimoldi, I.

Inorg. Chem. 2021, 60, 2976-2982, 10.1021/acs.inorgchem.0c02969

Based on the supramolecular interaction between vancomycin (Van), an antibiotic glycopeptide, and D-Ala-D-Ala (DADA) dipeptides, a novel class of artificial metalloenzymes was synthesized and characterized. The presence of an iridium(III) ligand at the N-terminus of DADA allowed the use of the metalloenzyme as a catalyst in the asymmetric transfer hydrogenation of cyclic imines. In particular, the type of link between DADA and the metal-chelating moiety was found to be fundamental for inducing asymmetry in the reaction outcome, as highlighted by both computational studies and catalytic results. Using the [IrCp*(m-I)Cl]Cl ⊂ Van complex in 0.1 M CH3COONa buffer at pH 5, a significant 70% (S) e.e. was obtained in the reduction of quinaldine B.


Metal: Ir
Ligand type: Cp*; Diamine
Host protein: DADA dipeptide
Anchoring strategy: Supramolecular
Optimization: Chemical
Max TON: 50
ee: 70
PDB: ---
Notes: ---

Chemical Modifications of Proteins and Their Applications in Metalloenzyme Studies

Review

Liu, P.

Synth. Syst. Biotechnol. 2021, 6, 32-49, 10.1016/j.synbio.2021.01.001

Protein chemical modifications are important tools for elucidating chemical and biological functions of proteins. Several strategies have been developed to implement these modifications, including enzymatic tailoring reactions, unnatural amino acid incorporation using the expanded genetic codes, and recognition-driven transformations. These technologies have been applied in metalloenzyme studies, specifically in dissecting their mechanisms, improving their enzymatic activities, and creating artificial enzymes with non-natural activities. Herein, we summarize some of the recent efforts in these areas with an emphasis on a few metalloenzyme case studies.


Notes: ---

Efficient Lewis Acid Catalysis of an Abiological Reaction in a De Novo Protein Scaffold

Hilvert, D.; Jiménez-Osés, G.

Nat. Chem. 2021, 13, 231-235, 10.1038/s41557-020-00628-4

New enzyme catalysts are usually engineered by repurposing the active sites of natural proteins. Here we show that design and directed evolution can be used to transform a non-natural, functionally naive zinc-binding protein into a highly active catalyst for an abiological hetero-Diels–Alder reaction. The artificial metalloenzyme achieves >104 turnovers per active site, exerts absolute control over reaction pathway and product stereochemistry, and displays a catalytic proficiency (1/KTS = 2.9 × 1010 M−1) that exceeds all previously characterized Diels–Alderases. These properties capitalize on effective Lewis acid catalysis, a chemical strategy for accelerating Diels–Alder reactions common in the laboratory but so far unknown in nature. Extension of this approach to other metal ions and other de novo scaffolds may propel the design field in exciting new directions.


Metal: Zn
Ligand type: Amino acid
Host protein: De novo-designed protein
Anchoring strategy: Dative
Optimization: Genetic
Max TON: >10000
ee: 99
PDB: ---
Notes: PDB: 3V1C, 7BWW

Design and Engineering of Artificial Metalloproteins: From De Novo Metal Coordination to Catalysis

Review

Zeymer, C.

Protein Eng. Des. Sel. 2021, 34, 10.1093/protein/gzab003

Metalloproteins are essential to sustain life. Natural evolution optimized them for intricate structural, regulatory and catalytic functions that cannot be fulfilled by either a protein or a metal ion alone. In order to understand this synergy and the complex design principles behind the natural systems, simpler mimics were engineered from the bottom up by installing de novo metal sites in either natural or fully designed, artificial protein scaffolds. This review focuses on key challenges associated with this approach. We discuss how proteins can be equipped with binding sites that provide an optimal coordination environment for a metal cofactor of choice, which can be a single metal ion or a complex multinuclear cluster. Furthermore, we highlight recent studies in which artificial metalloproteins were engineered towards new functions, including electron transfer and catalysis. In this context, the powerful combination of de novo protein design and directed evolution is emphasized for metalloenzyme development.


Notes: ---

Directed Evolution of a Cp*RhIII‐Linked Biohybrid Catalyst Based on a Screening Platform with Affinity Purification

Hayashi, T; Onoda, A.

ChemBioChem 2021, 22, 679-685, 10.1002/cbic.202000681

Directed evolution of Cp*RhIII-linked nitrobindin (NB), a biohybrid catalyst, was performed based on an in vitro screening approach. A key aspect of this effort was the establishment of a high-throughput screening (HTS) platform that involves an affinity purification step employing a starch-agarose resin for a maltose binding protein (MBP) tag. The HTS platform enables efficient preparation of the purified MBP-tagged biohybrid catalysts in a 96-well format and eliminates background influence of the host E. coli cells. Three rounds of directed evolution and screening of more than 4000 clones yielded a Cp*RhIII-linked NB(T98H/L100K/K127E) variant with a 4.9-fold enhanced activity for the cycloaddition of acetophenone oximes with alkynes. It is confirmed that this HTS platform for directed evolution provides an efficient strategy for generating highly active biohybrid catalysts incorporating a synthetic metal cofactor.


Metal: Rh
Ligand type: Cp
Host protein: Nitrobindin (Nb)
Anchoring strategy: Covalent
Optimization: Genetic
Reaction: Cycloaddition
Max TON: ---
ee: ---
PDB: ---
Notes: ---