Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Faraday Discuss ; 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847587

ABSTRACT

Genetic code expansion has emerged as a powerful tool in enzyme design and engineering, providing new insights into sophisticated catalytic mechanisms and enabling the development of enzymes with new catalytic functions. In this regard, the non-canonical histidine analogue Nδ-methylhistidine (MeHis) has proven especially versatile due to its ability to serve as a metal coordinating ligand or a catalytic nucleophile with a similar mode of reactivity to small molecule catalysts such as 4-dimethylaminopyridine (DMAP). Here we report the development of a highly efficient aminoacyl tRNA synthetase (G1PylRSMIFAF) for encoding MeHis into proteins, by transplanting five known active site mutations from Methanomethylophilus alvus (MaPylRS) into the single domain PylRS from Methanogenic archaeon ISO4-G1. In contrast to the high concentrations of MeHis (5-10 mM) needed with the Ma system, G1PylRSMIFAF can operate efficiently using MeHis concentrations of ∼0.1 mM, allowing more economical production of a range of MeHis-containing enzymes in high titres. Interestingly G1PylRSMIFAF is also a 'polyspecific' aminoacyl tRNA synthetase (aaRS), enabling incorporation of five different non-canonical amino acids (ncAAs) including 3-pyridylalanine and 2-fluorophenylalanine. This study provides an important step towards scalable production of engineered enzymes that contain non-canonical amino acids such as MeHis as key catalytic elements.

2.
Protein Sci ; 32(5): e4640, 2023 05.
Article in English | MEDLINE | ID: mdl-37051694

ABSTRACT

The availability of an expanded genetic code opens exciting new opportunities in enzyme design and engineering. In this regard histidine analogues have proven particularly versatile, serving as ligands to augment metalloenzyme function and as catalytic nucleophiles in designed enzymes. The ability to genetically encode multiple functional residues could greatly expand the range of chemistry accessible within enzyme active sites. Here, we develop mutually orthogonal translation components to selectively encode two structurally similar histidine analogues. Transplanting known mutations from a promiscuous Methanosarcina mazei pyrrolysyl-tRNA synthetase (MmPylRSIFGFF ) into a single domain PylRS from Methanomethylophilus alvus (MaPylRSIFGFF ) provided a variant with improved efficiency and specificity for 3-methyl-L-histidine (MeHis) incorporation. The MaPylRSIFGFF clone was further characterized using in vitro biochemical assays and x-ray crystallography. We subsequently engineered the orthogonal MmPylRS for activity and selectivity for 3-(3-pyridyl)-L-alanine (3-Pyr), which was used in combination with MaPylRSIFGFF to produce proteins containing both 3-Pyr and MeHis. Given the versatile roles played by histidine in enzyme mechanisms, we anticipate that the tools developed within this study will underpin the development of enzymes with new and enhanced functions.


Subject(s)
Amino Acyl-tRNA Synthetases , Histidine , Histidine/genetics , Lysine/chemistry , RNA, Transfer/genetics , RNA, Transfer/metabolism , Amino Acyl-tRNA Synthetases/chemistry , Methanosarcina/genetics , Methanosarcina/metabolism
3.
JACS Au ; 3(1): 13-24, 2023 Jan 23.
Article in English | MEDLINE | ID: mdl-36711092

ABSTRACT

Nucleosides, nucleotides, and oligonucleotides modulate diverse cellular processes ranging from protein production to cell signaling. It is therefore unsurprising that synthetic analogues of nucleosides and their derivatives have emerged as a versatile class of drug molecules for the treatment of a wide range of disease areas. Despite their great therapeutic potential, the dense arrangements of functional groups and stereogenic centers present in nucleic acid analogues pose a considerable synthetic challenge, especially in the context of large-scale manufacturing. Commonly employed synthetic methods rely on extensive protecting group manipulations, which compromise step-economy and result in high process mass intensities. Biocatalytic approaches have the potential to address these limitations, enabling the development of more streamlined, selective, and sustainable synthetic routes. Here we review recent achievements in the biocatalytic manufacturing of nucleosides and cyclic dinucleotides along with progress in developing enzymatic strategies to produce oligonucleotide therapies. We also highlight opportunities for innovations that are needed to facilitate widespread adoption of these biocatalytic methods across the pharmaceutical industry.

4.
Nature ; 606(7912): 49-58, 2022 06.
Article in English | MEDLINE | ID: mdl-35650353

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.


Subject(s)
Biocatalysis , Biotechnology , Protein Engineering , Proteins , Biotechnology/methods , Biotechnology/trends , Protein Engineering/methods , Protein Engineering/trends , Proteins/chemistry , Proteins/metabolism
5.
J Am Chem Soc ; 144(9): 3761-3765, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35224970

ABSTRACT

The Covid-19 pandemic highlights the urgent need for cost-effective processes to rapidly manufacture antiviral drugs at scale. Here we report a concise biocatalytic process for Molnupiravir, a nucleoside analogue recently approved as an orally available treatment for SARS-CoV-2. Key to the success of this process was the development of an efficient biocatalyst for the production of N-hydroxy-cytidine through evolutionary adaption of the hydrolytic enzyme cytidine deaminase. This engineered biocatalyst performs >85 000 turnovers in less than 3 h, operates at 180 g/L substrate loading, and benefits from in situ crystallization of the N-hydroxy-cytidine product (85% yield), which can be converted to Molnupiravir by a selective 5'-acylation using Novozym 435.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , Cytidine Deaminase/metabolism , Cytidine/analogs & derivatives , SARS-CoV-2 , Biocatalysis , Cytidine/biosynthesis , Cytidine/metabolism , Cytidine Deaminase/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Hydroxylamines , Metabolic Engineering , Protein Engineering , Uridine/metabolism
6.
Nat Chem ; 14(3): 313-320, 2022 03.
Article in English | MEDLINE | ID: mdl-34916595

ABSTRACT

The combination of computational design and directed evolution could offer a general strategy to create enzymes with new functions. So far, this approach has delivered enzymes for a handful of model reactions. Here we show that new catalytic mechanisms can be engineered into proteins to accelerate more challenging chemical transformations. Evolutionary optimization of a primitive design afforded an efficient and enantioselective enzyme (BH32.14) for the Morita-Baylis-Hillman (MBH) reaction. BH32.14 is suitable for preparative-scale transformations, accepts a broad range of aldehyde and enone coupling partners and is able to promote selective monofunctionalizations of dialdehydes. Crystallographic, biochemical and computational studies reveal that BH32.14 operates via a sophisticated catalytic mechanism comprising a His23 nucleophile paired with a judiciously positioned Arg124. This catalytic arginine shuttles between conformational states to stabilize multiple oxyanion intermediates and serves as a genetically encoded surrogate of privileged bidentate hydrogen-bonding catalysts (for example, thioureas). This study demonstrates that elaborate catalytic devices can be built from scratch to promote demanding multi-step processes not observed in nature.


Subject(s)
Proteins , Catalysis , Hydrogen Bonding , Molecular Conformation , Stereoisomerism
7.
ACS Catal ; 10(4): 2735-2746, 2020 Feb 21.
Article in English | MEDLINE | ID: mdl-32550044

ABSTRACT

Nature employs a limited number of genetically encoded axial ligands to control diverse heme enzyme activities. Deciphering the functional significance of these ligands requires a quantitative understanding of how their electron-donating capabilities modulate the structures and reactivities of the iconic ferryl intermediates compounds I and II. However, probing these relationships experimentally has proven to be challenging as ligand substitutions accessible via conventional mutagenesis do not allow fine tuning of electron donation and typically abolish catalytic function. Here, we exploit engineered translation components to replace the histidine ligand of cytochrome c peroxidase (CcP) by a less electron-donating N δ-methyl histidine (Me-His) with little effect on the enzyme structure. The rate of formation (k 1) and the reactivity (k 2) of compound I are unaffected by ligand substitution. In contrast, proton-coupled electron transfer to compound II (k 3) is 10-fold slower in CcP Me-His, providing a direct link between electron donation and compound II reactivity, which can be explained by weaker electron donation from the Me-His ligand ("the push") affording an electron-deficient ferryl oxygen with reduced proton affinity ("the pull"). The deleterious effects of the Me-His ligand can be fully compensated by introducing a W51F mutation designed to increase "the pull" by removing a hydrogen bond to the ferryl oxygen. Analogous substitutions in ascorbate peroxidase lead to similar activity trends to those observed in CcP, suggesting that a common mechanistic strategy is employed by enzymes using distinct electron transfer pathways. Our study highlights how noncanonical active site substitutions can be used to directly probe and deconstruct highly evolved bioinorganic mechanisms.

8.
Nature ; 570(7760): 219-223, 2019 06.
Article in English | MEDLINE | ID: mdl-31132786

ABSTRACT

The combination of computational design and laboratory evolution is a powerful and potentially versatile strategy for the development of enzymes with new functions1-4. However, the limited functionality presented by the genetic code restricts the range of catalytic mechanisms that are accessible in designed active sites. Inspired by mechanistic strategies from small-molecule organocatalysis5, here we report the generation of a hydrolytic enzyme that uses Nδ-methylhistidine as a non-canonical catalytic nucleophile. Histidine methylation is essential for catalytic function because it prevents the formation of unreactive acyl-enzyme intermediates, which has been a long-standing challenge when using canonical nucleophiles in enzyme design6-10. Enzyme performance was optimized using directed evolution protocols adapted to an expanded genetic code, affording a biocatalyst capable of accelerating ester hydrolysis with greater than 9,000-fold increased efficiency over free Nδ-methylhistidine in solution. Crystallographic snapshots along the evolutionary trajectory highlight the catalytic devices that are responsible for this increase in efficiency. Nδ-methylhistidine can be considered to be a genetically encodable surrogate of the widely employed nucleophilic catalyst dimethylaminopyridine11, and its use will create opportunities to design and engineer enzymes for a wealth of valuable chemical transformations.


Subject(s)
Directed Molecular Evolution , Hydrolases/genetics , Hydrolases/metabolism , Protein Engineering , 4-Aminopyridine/analogs & derivatives , 4-Aminopyridine/metabolism , Biocatalysis , Catalytic Domain/genetics , Crystallography, X-Ray , Esters/metabolism , Genetic Code , Hydrolases/chemistry , Hydrolysis , Methylhistidines/metabolism , Models, Molecular , Mutagenesis , Mutation , Pyrococcus horikoshii/enzymology , Pyrococcus horikoshii/genetics , Substrate Specificity/genetics
9.
J Biotechnol ; 293: 56-65, 2019 Mar 10.
Article in English | MEDLINE | ID: mdl-30690098

ABSTRACT

N-Alkylated-α-amino acids are useful building blocks for the pharmaceutical and fine chemical industries. Enantioselective methods of N-alkylated-α-amino acid synthesis are therefore highly valuable and widely investigated. While there are a variety of chemical methods for their synthesis, they often employ stoichiometric quantities of hazardous reagents such as pyrophoric metal hydrides or genotoxic alkylating agents, whereas biocatalytic routes can provide a greener and cleaner alternative to existing methods. This review highlights the occurrence of the N-alkyl-α-amino acid motif and its role in nature, important applications towards human health and biocatalytic methods of preparation. Several enzyme classes that can be used to access chiral N-alkylated-α-amino acids and their substrate selectivities are detailed.


Subject(s)
Amino Acids/biosynthesis , Amino Acids/chemistry , Animals , Biocatalysis , Humans , Nature
10.
Angew Chem Int Ed Engl ; 57(42): 13821-13824, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30138551

ABSTRACT

N-Functionalized amino acids are important building blocks for the preparation of diverse bioactive molecules, including peptides. The development of sustainable manufacturing routes to chiral N-alkylated amino acids remains a significant challenge in the pharmaceutical and fine-chemical industries. Herein we report the discovery of a structurally diverse panel of biocatalysts which catalyze the asymmetric synthesis of N-alkyl amino acids through the reductive coupling of ketones and amines. Reactions have been performed on a gram scale to yield optically pure N-alkyl-functionalized products in high yields.


Subject(s)
Amino Acids/chemistry , Biocatalysis , Alkylation , Amino Acids/metabolism , Animals , Chromatography, High Pressure Liquid , Humans , Ketones/chemistry , Pseudomonas/enzymology , Stereoisomerism
11.
Angew Chem Int Ed Engl ; 54(15): 4608-11, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25728350

ABSTRACT

The synthesis of substituted D-phenylalanines in high yield and excellent optical purity, starting from inexpensive cinnamic acids, has been achieved with a novel one-pot approach by coupling phenylalanine ammonia lyase (PAL) amination with a chemoenzymatic deracemization (based on stereoselective oxidation and nonselective reduction). A simple high-throughput solid-phase screening method has also been developed to identify PALs with higher rates of formation of non-natural D-phenylalanines. The best variants were exploited in the chemoenzymatic cascade, thus increasing the yield and ee value of the D-configured product. Furthermore, the system was extended to the preparation of those L-phenylalanines which are obtained with a low ee value using PAL amination.


Subject(s)
Anabaena variabilis/enzymology , Phenylalanine Ammonia-Lyase/metabolism , Phenylalanine/analogs & derivatives , Amination , Chemistry Techniques, Synthetic , Oxidation-Reduction , Phenylalanine/chemical synthesis , Phenylalanine/metabolism , Stereoisomerism
12.
Angew Chem Weinheim Bergstr Ger ; 127(15): 4691-4694, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-27478261

ABSTRACT

The synthesis of substituted d-phenylalanines in high yield and excellent optical purity, starting from inexpensive cinnamic acids, has been achieved with a novel one-pot approach by coupling phenylalanine ammonia lyase (PAL) amination with a chemoenzymatic deracemization (based on stereoselective oxidation and nonselective reduction). A simple high-throughput solid-phase screening method has also been developed to identify PALs with higher rates of formation of non-natural d-phenylalanines. The best variants were exploited in the chemoenzymatic cascade, thus increasing the yield and ee value of the d-configured product. Furthermore, the system was extended to the preparation of those l-phenylalanines which are obtained with a low ee value using PAL amination.

13.
Bioorg Med Chem ; 22(20): 5555-7, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25037641

ABSTRACT

Phenylalanine ammonia lyases (PALs) catalyse the regio- and stereoselective hydroamination of cinnamic acid analogues to yield optically enriched α-amino acids. Herein, we demonstrate that a bacterial PAL from Anabaena variabilis (AvPAL) displays significantly higher activity towards a series of non-natural substrates than previously described eukaryotic PALs. Biotransformations performed on a preparative scale led to the synthesis of the 2-chloro- and 4-trifluoromethyl-phenylalanine derivatives in excellent ee, highlighting the enormous potential of bacterial PALs as biocatalysts for the synthesis of high value, non-natural amino acids.


Subject(s)
Anabaena variabilis/enzymology , Biocatalysis , Phenylalanine Ammonia-Lyase/metabolism , Kinetics , Models, Molecular , Phenylalanine Ammonia-Lyase/chemistry , Stereoisomerism , Substrate Specificity
14.
Angew Chem Int Ed Engl ; 53(18): 4652-6, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24692092

ABSTRACT

Phenylalanine ammonia lyases (PALs) belong to a family of 4-methylideneimidazole-5-one (MIO) cofactor dependent enzymes which are responsible for the conversion of L-phenylalanine into trans-cinnamic acid in eukaryotic and prokaryotic organisms. Under conditions of high ammonia concentration, this deamination reaction is reversible and hence there is considerable interest in the development of PALs as biocatalysts for the enantioselective synthesis of non-natural amino acids. Herein the discovery of a previously unobserved competing MIO-independent reaction pathway, which proceeds in a non-stereoselective manner and results in the generation of both L- and D-phenylalanine derivatives, is described. The mechanism of the MIO-independent pathway is explored through isotopic-labeling studies and mutagenesis of key active-site residues. The results obtained are consistent with amino acid deamination occurring by a stepwise E1 cB elimination mechanism.


Subject(s)
Amino Acids/chemical synthesis , Phenylalanine Ammonia-Lyase/metabolism , Phenylalanine/chemical synthesis , Ammonia/metabolism , Catalysis , Catalytic Domain , Cinnamates/metabolism , Imidazoles/metabolism , Mutation/genetics , Phenylalanine Ammonia-Lyase/genetics , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...