Supporting Information

Similar documents
CypExpress 3A4 Catalyzed Conversion of Testosterone (TE) to 6β- Hydroxytestosterone (HT)

SI for: Photobiocatalytic alcohol oxidation using LED light sources

Selective Formation of Benzo[c]cinnoline by Photocatalytic Reduction of 2,2 Dinitrobiphenyl with TiO 2 and UV light irradiation

Amine dehydrogenases: Efficient biocatalysts for the stereoselective reductive amination of carbonyl compounds

SUPPORTING INFORMATION

A biphasic oxidation of alcohols to aldehydes and ketones using a simplified packed-bed microreactor

CypExpress 2C9 Catalyzed Conversion of Diclofenac (DN) to to 4- Hydroxydiclofenac (HDN)

Supporting information

Exploring the biocatalytic scope of a bacterial flavin-containing monooxygenase

Codex KRED Screening Kit

Encapsulation of enzyme in metal ion-surfactant nanocomposites for

Electronic Supplementary Information

Access from the University of Nottingham repository:

ADVANTAME (TENTATIVE)

Automated Sample Preparation of Headspace Standards Using the Agilent 7696 WorkBench

Appendix II- Bioanalytical Method Development and Validation

Enantioselective desymmetrisation of citric acid catalysed by the substratetolerant petrobactin biosynthetic enzyme AsbA

*Correspondence to:

H-Bonding Promotion of Peptide Solubility and Cyclization by Fluorinated Alcohols

KINETIC ANALYSIS OF THE HBTM-CATALYZED ESTERIFICATION OF AN ENANTIOPURE SECONDARY ALCOHOL

Ondansetron Hydrochloride Tablets

Substrate Specificity of Alcohol Dehydrogenase

ADVANTAME. Not less than 97.0% and not more than 102.0% on the anhydrous basis. Sweetener, flavour enhancer

Supplemental Information

Backbone modification of a parathyroid hormone receptor-1 antagonist/inverse agonist

ADH Activity Assay Kit

A Fluorescence Turn-On Sensor for the Detection of Palladium Ions that Operates Through In-Situ Generation of Palladium Nanoparticles

Analysis of Residual Solvents in Pharmaceuticals (USP<467>) with Shimadzu GC-2010 Plus and HS-10 Headspace Sampler

Nicotinamide Adenine Dinucleotide-Dependent Redox-Neutral Convergent Cascade for Lactonizations with Type II Flavin-Containing Monooxygenase

Ionic liquid-supported Pt nanoparticles as catalysts for enantioselective hydrogenation

Supporting Information. A fluorogenic assay for screening Sirt6 modulators

RAFT /MADIX polymerization of N-vinylcaprolactam in water-ethanol solvent mixtures

Convenient Synthesis of Nucleoside 5 -Triphosphates for RNA Transcription. Supplemental Materials

guanidine bisurea bifunctional organocatalyst

Synergistic Cu/Ir Catalysis. Table of Contents

in reaction buffer (40 mm Tris-HCl, ph 8.0, 100 mm NaCl and 10 mm MgCl 2 ). After

Supporting Information. Copyright Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim, 2007

Data sheet. UV-Tracer TM Biotin-Maleimide. For Labeling of Thiol-groups with UV-detectable Biotin CLK-B Description

Supplementary Material (ESI) for Chemical Communications This journal is (c) The Royal Society of Chemistry 2010

SUPPORTING INFORMATION. Elucidation of the role of betaine hydrochloride in glycerol esterification: towards bio-based ionic building blocks

Supplementary Materials for

Supporting information. Direct Enantioselective Aldol Reactions catalyzed by a Proline-Thiourea Host- Guest Complex

Supporting Information for. PNA FRET Pair Miniprobes for Quantitative. Fluorescent in Situ Hybridization to Telomeric DNA in Cells and Tissue

Supporting Information for: Using a Lipase as a High Throughput Screening Method for Measuring the Enantiomeric. Excess of Allylic Acetates

7. Stability indicating analytical method development and validation of Ramipril and Amlodipine in capsule dosage form by HPLC.

1,1,3,3-Tetramethylguanidine-Promoted Ring-Opening Polymerization of N-Butyl N-Carboxyanhydride Using Alcohol Initiators

Electronic Supplementary Information

Electronic supplementary information

Recyclable Enamine Catalysts for Asymmetric Direct Cross-Aldol

Supplementary Materials for

Reduction-free synthesis of stable acetylide cobalamins. Table of Contents. General information. Preparation of compound 1

Supporting Information

CIPAC. CIPAC Free relevant impurities methods:

Sulfotepp impurities in Chlorpyrifos EC formulations

Dissolution Test 5 was validated using a Zodiac C18 brand of L1 column. The typical retention time for atorvastatin is about min.

Photochemical synthesis of 3-azabicyclo[3.2.0]heptanes: advanced building blocks for drug discovery

Molecular Weight Distribution of Living Chains in Polystyrene Pre-pared by Atom Transfer Radical Polymerization

Acrylates via Metathesis of Crotonates

In vivo monitoring of hydrogen sulfide using a cresyl violet-based ratiometric fluorescence probe

A contribution from the Department of Chemistry, Washington University, Campus Box 1134, One Brookings Drive, Saint Louis, Missouri 63130

Evolution of Solid Phase Homochirality for a. Proteinogenic Amino Acid

Electronic Supplementary Information

Supporting Information

Supporting Information for

Optical Isomer Separation Columns and Packing Materials

NAD + /NADH Assay [Colorimetric]

Supporting Information. for. Angew. Chem. Int. Ed. Z Wiley-VCH 2003

Supplementary Figure 1. Structures of substrates tested with 1. Only one enantiomer is shown.

A New Model for Asymmetric Amplification in Amino Acid Catalysis - Supporting information

Discovery of Decamidine as a New and Potent PRMT1 Inhibitor

Encapsulated enzymes with integrated fluorescence-control of enzymatic activity. Supporting Information

Supporting Information. Design and construction of artificial photoresponsive protocells capable of converting day light to chemical energy

A RP-HPLC METHOD DEVELOPMENT AND VALIDATION OF PARA- PHENYLENEDIAMINE IN PURE FORM AND IN MARKETED PRODUCTS

Specifically colorimetric recognition of calcium, strontium, barium. ions using 2-mercaptosuccinic acid-functionalized gold nanoparticles

Selective Reduction of Carboxylic acids to Aldehydes Catalyzed by B(C 6 F 5 ) 3

Dual Use of a Chemical Auxiliary: Molecularly Imprinted Polymers for the Selective Recovery of Products from Biocatalytic Reaction Mixtures

HYDROXYPROLINE IN URINE by UV/VIS Code Z01010

Determination of Carbonyl Compounds In Water by Dinitrophenylhydrazine Derivatization and HPLC/UV*

Electronic Supplementary Information for:

Alcohol Dehydrogenase Detection Kit

Dynamic covalent assembly of stimuli responsive vesicle gels

GAFTI Analytical method for ISO/TS 16179:2012 Detection and Determination of Organotin Compounds in Footwear and Apparel Materials by GC-MS

Enzymatic Assay of PHOSPHOLIPASE D (EC )

Supporting Information

Supporting Information

Green Approaches to Late-stage Fluorination: Radiosyntheses of 18 F-Labelled Radiopharmaceuticals in Ethanol and Water

Analysis of BTEX in Natural Water with SPME

Supplemental data. Supplemental Figure 1: Alignment of potential ERRE1 and 2 in human, mouse and rat. PEPCK promoter.

A. 50 mm Sodium Lauryl Sulfate Solution (SDS) (Prepare 10 ml in deionized water using Lauryl Sulfate, Sodium Salt, Sigma Prod. No. L-5750.

SUPPORTING INFORMATION. Photoswitchable catalysis by a nanozyme mediated by a light-sensitive cofactor

Supporting Information

Uncontrolled Copy. SOP 109 Ethylene Glycol Screen by Gas Chromatography/Mass Spectrometry. Table of Contents. 1. Principle of Assay...

Supporting Information for: In vitro biocatalytic pathway design: orthogonal network for the quantitative and stereospecific amination of alcohols

Anethole. Gas chromatograhpy determination of trans-anethole in Spirit srinks of viti-vinicultural origin

Impact factor: 3.958/ICV: 4.10 ISSN:

Rapid Microwave-Assisted CNBr Cleavage of Bead-Bound Peptides

Towards the use of Alcohol dehydrogenases as biocatalysts for stereoselective isotope labeling of aromatic alcohols.

A fluorinated dendritic TsDPEN-Ru(II) catalyst for asymmetric transfer hydrogenation of prochiral ketones in aqueous media

BUTYL ALCOHOL in urine by GC-FID Code GC05510

CYCLOSERINE Final text for addition to The International Pharmacopoeia. (November 2008) CYCLOSERINUM CYCLOSERINE

Transcription:

Supporting Information More efficient redox biocatalysis by utilising 1,4-butanediol as smart cosubstrate Selin Kara, a Dominik Spickermann, b Joerg H. Schrittwieser, a Christan Leggewie, b Willem J. H. van Berkel, c Isabel W. C. E. Arends, a and Frank Hollmann* a a b c Department of Biotechnology, Delft University of Technology, Julianalaan 136, 2628BL Delft (The Netherlands) evocatal GmbH Merowingerplatz 1A, 40225 Düsseldorf (Germany) Laboratory of Biochemistry, Wageningen University, Dreijenlaan 3, 6703 HA Wageningen (The Netherlands) CONTENTS 1 Materials... 2 2 Experimental details and supporting information... 2 2.1 Kinetic Analysis of HLADH-Catalysed Oxidation of 1,4-Butanediol... 2 2.2 HLADH-Catalysed Reductions with Cofactor Regeneration... 3 2.3 Analysis of Product Inhibition for the Reduction of Trans-Cinnamaldehyde... 4 2.4 HLADH-Catalysed Reduction of 2-Phenyl-1-Propanal... 4 2.5 TsER-Catalysed Reduction of Ketoisophorone... 5 2.6 3HB6H-Catalysed Hydroxylation of 3-Hydroxybenzoate... 5 2.7 Hydroxylation of 3-Hydroxybenzoate at 50 mm-scale... 5 3 Analytics... 6 S1

1 Materials HLADH, isoform E recombinantly expressed in Escherichia coli is commercially available from evocatal GmbH (Duesseldorf, Germany). TsER was produced according to literature procedure (Opperman et al., 2008) by recombinant expression in E. coli followed by heat-purification. 3HB6H was produced by recombinant expression in E. coli following a literature procedure (Montersino and van Berkel, 2012). All chemicals were purchased from Sigma Aldrich (Zwijndrecht, The Netherlands) in the highest purity available and used as received. 2 Experimental details and supporting information 2.1 Estimation of the the conversion achievable as a function of molar cosubstrate surplus The reversible MPV-reduction can be described by the law of mass action (Equation 1). Using isopropanol as cosubstrate for the reduction of ketones, it is justified to assume an equilibrium constant of 1. ( ) ( ) ( ) ( ) (Equation 1, equilibrium concentrations) From this Eq.2 can be derived, which relates the conversion to the initial concentrations of substrate and cosubstrate. [ ] [ ) [ ] (Equation 2) All calculations shown in Scheme 1 were done according to these eqations. 2.2 Kinetic Analysis of HLADH-Catalysed Oxidation of 1,4-Butanediol Stock solutions of 1,4-butanediol (1,4-BD, 5 M), NAD + (25 mm) and HLADH (12 gl 1 ) were freshly prepared in 50 mm Tris-HCl buffer at ph 7. Reaction mixture containing reaction buffer (V µl), 1,4-butanediol stock (970 µl V µl) and NAD + stock (20 µl) was incubated at 30 C for 3 min. Reactions were started by the addition of the HLADH stock (10 µl). Formation of NADH was followed by UV/Vis spectroscopy and quantified using an absorption coefficient of 6.22 mm 1 cm 1. The starting concentrations of NAD + and HLADH were 0.5 mm and 0.12 gl 1, respectively. The following graph (Fig. S1) shows the kinetic behavior of HLADH for 1,4-butanediol oxidation. The simulation was performed using MATLAB 2010 based on Michaelis-Menten double substrate kinetics. In case of excess substrate inhibition the following equation was used: (( ( ) ( ( ) ( ) ( )) ( ( )) S2

Fig. S1 Kinetic analysis of oxidation of 1,4-butanediol catalysed by HLADH. Left: Reaction conditions: 0 4200 mm 1,4-butanediol, 0.5 mm NAD +, 0.12 gl 1 enyzme, 50 mm Tris-HCl at ph 7 and 30 C. K M, 1,4-BD= 23.3 ± 8.8 mm, V max = 0.34 ± 0.01 Umg 1, K i/k M = 57. Right: Reaction conditions: 0 1.5 mm NAD +, 150 mm 1,4-BD, 0.12 gl 1 enyzme, 50 mm Tris-HCl at ph 7 and 30 C. K M,NAD+ = 0.16 ± 0.04 mm, V max = 0.35 ± 0.03 Umg 1. Similarly the kinetic properties for ethanol (EtOH) and isopropanol ( i PrOH) were determined (Table S1). Table S1 Summary of the kinetic parameters of some selected cosubstrates with HLADH. Cosubstrate V max [Umg 1 ] K M [mm] K i /K M [-] 1,4-BD 0.34 ± 0.001 23.3 ± 8.8 57 EtOH 0.26 ± 0.03 5.8 ± 0.5 n.d. i PrOH 0.03 ± 0.001 261.1 ± 51.0 n.d. 2.3 HLADH-Catalysed Reductions with Cofactor Regeneration Due to the poor solubility of some of the applied substrates (1c, 1e and 1f), stock solutions (0.5 M) were prepared in acetonitrile (MeCN). The final MeCN concentration was kept at maximum 1% (v/v). There was no negative effect on the enzyme activity with this amount of MeCN (data not shown). Cosubstrate stocks (30 mm each), NAD + stock (15 mm) and HLADH stock (5 gl 1 ) were freshly prepared in 50 mm Tris-HCl buffer at ph 7. The mixture of substrate stock (15 µl), cosubstrate stock (125 µl), NAD + stock (10 µl), buffer (1200 µl) was incubated at 30 C for 3 min. Biotransformations were initiated with the HLADH solution (150 µl) and the reaction mixtures (1.5 ml) were efficiently shaken (700 rpm) in screw-capped glass vials at 30 C. Aliquots (50 µl) were removed at intervals and mixed with 200 µl of ethyl acetate (EtOAc). The mixture was vortexed for 20 sec, followed by centrifugation (16,100 g, 0.5 min). The clear organic phase was removed and dried over anhydrous MgSO 4. Dodecane (5 mm) was used as an external standard in EtOAc and the product formation was assessed using GC (see 3. Analytics). For all substrates we ran negative control experiments whereby no HLADH was added. No formation of reduction product or γ-butyrolactone (GBL) was detected in the negative controls. Following table summarises the maximum conversions achieved using either isopropanol ( i PrOH), ethanol (EtOH) or 1,4- butanediol (1,4-BD) as cosubstrate. Table S2 Evaluation of isopropanol ( i PrOH), ethanol (EtOH) and 1,4-butanediol (1,4-BD) as cosubstrates for the HLADH-catalysed reductions. Entry Product Cosubstrate Conversion [%] i PrOH 17.8 ± 0.5 1 i PrOH [a] 38.6 ± 1.6 EtOH 24.4 ± 1.9 2a 1,4-BD 58.6 ± 2.8 i PrOH 0 2 EtOH 5.3 ± 0.5 2b 1,4-BD 44.2 ± 2.6 i PrOH 3.5 ± 0.2 3 EtOH 6.0 ± 0.3 2c BDO 48.8 ± 2.3 i PrOH 30.1 ± 2.1 4 EtOH 44.6 ± 1.7 S3

2d 1,4-BD 73.0 ± 2.3 i PrOH 25.9 ± 2.2 5 EtOH 47.5 ± 0.2 2e 1,4-BD 97.7 ± 1.7 EtOH 12.9 ± 1.1 6 [b] EtOH [c] 17.0 ± 0.8 2f 1,4-BD 44.1 ± 0.6 Conditions: c(substrate) = 5 mm, c(cosubstrate) = 2.5 mm, c(nad + ) = 0.1 mm, c(hladh) = 0.5 gl 1, buffer: Tris-HCl (50 mm, ph 7.0), T = 30 o C, reaction time 96 h. In case of poorly water-soluble substrates 1% (v/v) acetonitrile was added. Entry 1 c(hladh) = 1 gl 1. [a] 5 eq of isopropanol, [b] ee>98% (R), [c] 1 eq of ethanol. 2.4 Analysis of Product Inhibition for the Reduction of Trans-Cinnamaldehyde Since in most of cases full conversion to the reduction product could not be observed, we examined a potential product inhibition. Stock solutions of trans-cinnamaldehyde (0.5 M) and trans-cinnamyl alcohol (0.5 M) were prepared in DMSO. Stocks of NADH (5 mm) and HLADH (12 gl 1 ) were freshly prepared in 50 mm Tris-HCl buffer at ph 7. The mixture containing reaction buffer (960-V µl), trans-cinnamaldehyde stock (10 µl), NADH stock (20 µl) and trans-cinnamyl alcohol (V µl) was incubated at 30 C for 3 min. Reactions were started by the addition of the HLADH stock (10 µl). Depletion of NADH was followed by UV/Vis spectroscopy and quantified using an absorption coefficient of 6.22 mm 1 cm 1. Maximum 2% (v/v) of DMSO was applied whereby no negative effect of DMSO at this concentration was detected (data not shown). The following graph (Fig. S2) shows decrease in the reduction activity of HLADH towards transcinnamaldehyde in the presence of trans-cinnamyl alcohol. Our experiments showed a severe product inhibition: loss of 75% of activity in the presence in the presence of 0.2 eq alcohol (approx. corresponding to 20% conversion). Fig. S2 Reduction of trans-cinnamaldehyde in the presence of trans-cinnamyl alcohol (0 5 mm). Reaction conditions: 5 mm trans-cinnamaldehyde, 0.1 mm NADH, 0.12 gl 1 enyzme, 50 mm Tris-HCl at ph 7 and 30 C. 2.5 HLADH-Catalysed Reduction of 2-Phenyl-1-Propanal 2-Phenyl-1-propanal stock (0.5 M) was prepared in MeCN. Cosubstrate stocks (30 mm each), NAD + stock (15 mm) and HLADH stock (0.1 gl 1 ) were freshly prepared in 50 mm Tris-HCl buffer at ph 7. The mixture of 2-phenyl-1-propanal stock (15 µl), cosubstrate stock (125 µl), NAD + stock (10 µl), reaction buffer (1200 µl) was incubated at 30 C for 3 min. Reactions were started by the addition of HLADH solution (150 µl, 0.1 gl 1 ) and were incubated at 30 C in screw-capped glass vials by effectively shaking (700 rpm). Aliquot samples (150 µl) were removed at intervals and mixed with 50 µl of TFA (10%) and 400 µl of n-heptane. The mixture was vortexed for 45 sec, followed by centrifugation (16,100 g, 1 min). The clear organic phase was separated and dried over anhydrous MgSO 4. Conversion of 2-phenyl-1-propanal and the formation of (R/S)-2-phenyl-1-propanol were monitored by HPLC. Formation of GBL was followed by GC analysis (see 3. Analytics, Table S2). S4

2.6 TsER-Catalysed Reduction of Ketoisophorone A ketoisophorone stock (KIP, 1M) stock was prepared in MeCN, whereas NAD + stock (15 mm), HLADH stock (10 gl 1 ) and cosubstrate stocks (EtOH or 1,4-BD, 50 mm) were freshly prepared in 50 mm MOPS buffer, 5 mm CaCl 2, at ph 7. The mixture of KIP stock (15 µl), cosubstrate stock (150 µl), NAD + stock (100 µl) and buffer (710 µl) was incubated at 30 C for 3 min. Reactions were started by the addition of TsER (375 µl, 1 gl 1 ) and HLADH solution (150 µl, 10 gl 1 ) and were incubated at 30 C in screw-capped glass vials by shaking (800 rpm). The reaction conditions were: 10 mm KIP, 5 mm cosubstrate, 1 mm NAD +, 0.25 gl 1 TsER and 1 gl 1 HLADH in 1.5 ml. Aliquot samples (50 µl) were removed at intervals and mixed with 200 µl of EtOAc containing 5 mm dodecane. The mixture was vortexed for 20 sec, followed by centrifugation (16,100 g, 1 min). The clear organic phase was separated and dried over anhydrous MgSO 4. The consumption of KIP and the formation of (R/S)-levodione were monitored by GC (see 3. Analytics, Table S3). 2.7 3HB6H-Catalysed Hydroxylation of 3-Hydroxybenzoate The stocks 3-hydroxybenzoate (3HB, 10 mm), cosubstrates (EtOH, i PrOH or 1,4-BD, 30 mm), NAD + (15 mm), HLADH (10 gl 1 ) were prepared in 20 mm Tris-SO 4 buffer at ph 8.0. The mixture containing 3HB stock (750 µl), cosubstrate stock (125 µl), NAD + stock (100 µl) and the reaction buffer (350 µl) were incubated at 30 C for 3 min. The reactions were started with the addition of 3HB6H (25 µl) and HLADH (150 µl) and incubated at 30 C and 800 rpm in screw-capped vials. The reaction conditions were: 5 mm 3HB, 2.5 mm cosubstrate, 1 mm NAD +, 0.25 gl 1 3HB6H and 1 gl 1 HLADH in 1.5 ml. Aliquot samples (100 µl) were removed at intervals and mixed with 100 µl of MeCN. The mixture was vortexed for 30 sec, followed by centrifugation (16,100 g, 1 min). The consumption of 3HB and the formation of 2,5-dihydroxybenzoate (2,5- DHB) were monitored by HPLC (see 3. Analytics, Table S3). 2.8 Hydroxylation of 3-Hydroxybenzoate at 50 mm-scale The stocks of 1,4-BD (250 mm), NAD + (20 mm) and HLADH (12.5 gl 1 ) were freshly prepared in 100 mm Tris-SO 4 at ph 8. First, 550 µl of reaction buffer (100 mm Tris-SO 4, ph 8) was given to 3HB (6.9 mg, 50 µmol) and ph was readjusted to 8 using 4M of NaOH (~10 µl was needed). After that, 100 µl of NAD + was added and reaction mixture was incubated at 30 C for 3 min. The reactions were started with the addition of 3HB6H (50 µl, 15 gl 1 ) and HLADH (200 µl, 12.5 gl 1 ) and incubated at 30 C and 800 rpm in screw-capped vials. The reaction conditions were: 50 mm 3HB, 25 mm 1,4-BD, 2 mm NAD +, 0.75 gl 1 3HB6H and 2.5 gl 1 HLADH in 1.0 ml. Aliquot samples (10 µl) were removed at intervals and mixed with 190 µl of MeCN. The mixture was vortexed for 30 sec, followed by centrifugation (16,100 g, 3 min). The consumption of 3HB and the formation of 2,5-DHB were monitored by HPLC (see 3. Analytics, Table S4). The formation of 2,5-DHB is shown in the following graph. Fig. S3 Hydroxylation of 3-hydroxybenzoate (3HB) to 2,5- dihydroxybenzoate (2,5-DHB). Reaction conditions: c(3- hydroxybenzoate) = 50 mm, c(1,4-bd) = 25 mm, c(nad + ) = 2 mm, c(3hb6h) = 0.75 gl 1, c(hladh) = 2.5 gl 1, buffer: Tris- SO 4 (100 mm, ph 8), T = 30 o C. S5

3 Analytics The reaction progress and the optical purity of the products were determined using GC or HPLC. Table S3 Details for the analytics used in ADH-catalysed reductions. Entry Product Analysis Column Temperature Profile t R [min] 1 2 3 4 5 6 7 2a 2b 2c 2d 2e 2f GC [b] GC [c] CP-Chiralsil dex CB, 25 m 0.32 mm 0.25 µm - 60 1.0 50 150 2.2 50 160 1.5 50 190 2.0 50 250 1.0-50 5.0 40 85 3.0 40 250 1.0-80 3.0 40 120 6.0 40 250 1.0-80 3.0 40 120 4.0 40 250 1.0-55 3.0 25 60 7.0 20 160 2.0 25 250 3.0-100 5.0 20 130 3.0 20 140 9.0 50 220 4.0 Substrate = 9.9 Product = 11.1 γ-butyrolactone = 6.1 1,4-Butanediol = 8.4 Substrate = 5.6 Product = 8.6 γ-butyrolactone = 12.0 1,4-Butanediol = 12.8 Substrate = 4.3 Product = 6.3 γ-butyrolactone = 11.2 1,4-Butanediol = 12.8 Substrate = 5.1 Product = 6.3 γ-butyrolactone = 9.7 1,4-Butanediol = 10.9 Substrate = 15.3 Product = 18.5 γ-butyrolactone = 16.3 1,4-Butanediol = 18.8 Substrate = 11.2 (R)-product = 15.1 (S)-product = 15.4 γ-butyrolactone = 7.1 1,4-Butanediol = 12.9 Derivatisation of the chiral hydroxy-ester product (2f): Reaction sample was extracted with EtOAc (700 µl) and incubated with 4-dimethylaminopyridine (3 gl 1 in EtOAc, 300 µl) and acetic anhydride (50 µl) for 90 min. After incubation, 300 µl NaOH (4 M) was added and vigorously mixed. The organic layer was then removed and 300 µl HCl (6 M) was given to this seperated phase. The mixture was vortexed; the organic phase was separated and dried over anhydrous MgSO 4 for GC analysis. 2g HPLC [d] Chiralpak AD-H, 0.46 cm 25 cm GC [e] CB-Sil 5 CB, 50 m 0.53 mm 1 µm - 40 25-80 3.0 40 105 3.0 40 160 3.0 40 335 1.0 Substrate = 6.3 (S)-product = 19.5 (R)-product = 22.2 Substrate = 5.5 Product = 7.0 γ-butyrolactone = 2.6 1,4-Butanediol = 3.2 [a] Entry 1 to 4: Total flow = 18 mlmin 1, N 2 as carrier gas. [b] Entry 5: Total flow = 20 mlmin 1, N 2 as carrier. [c] Entry 6: Pressure = 39.7 kpa, total flow = 24.4 mlmin 1, column flow = 1.02 mlmin 1, linear velocity = 22 cms 1, split = 20, He as carrier gas. [d] Entry 7: (HPLC analysis) Column flow = 1.0 mlmin 1, detection at 210 nm. Eluent = heptane: i PrOH:TFA (99:1:0.1), TFA: trifluoroacetic acid. [e] Entry 7: (GC Analysis) Total flow = 20 mlmin 1, N 2 as carrier. H: hold time (min), R: temperature ramp ( C/min). Entry 1 to 5: GC equipment: Shimadzu GC-2014 Entry 6: GC equipment: Shimadzu GC-2010 Plus Entry 7: HPLC equipment: Waters 515 Pump, Waters 717 Plus Autosampler, Shimadzu SPD-10A VP Detector, GC Equipment: Shimadzu GC-2014 S6

Following chromatograms (Fig. S4) show the conversion of substrate trans-cinnamaldehyde and the synthesis of product cinnamyl alcohol. Fig. S4 Left: The GC chromatograms of the reduction of trans-cinnamaldehyde (t R = 9.9 min) and the formation of trans-cinnamyl alcohol (t R = 11.1 min) catalysed by HLADH using 1,4-butanediol as cosubstrate. Right: The GC chromatograms of the oxidation of 1,4-butanediol (t R = 8.4 min) to γ-butyrolactone (t R = 6.1 min) catalysed by HLADH. S7

Table S4 Details for the analytics used in ER- and 3HB6H-catalysed reactions, respectively. Entry Product Analysis Column Temperature Profile t R [min] 1 Chrialdex GTA, 50 m 0.25 mm 0.12 µm - 110 25.0 40 170 3.0 Substrate = 10.7 (S)-Product = 13.9 (R)-Product = 14.4 γ-butyrolactone = 6.4 2h 1,4-Butanediol = 6.8 2 HPLC [b] SpeedROD RP-18E, 4.6 mm 50 mm - 35 7.5 Substrate = 5.6 Product = 4.6 2i [a] Entry 1: Pressure = 128 kpa, total flow = 27.9 mlmin 1, column flow = 0.8 mlmin 1, linear velocity = 20.5 cms 1, split = 30, He as carrier gas. [b] Entry 2: (HPLC analysis) Column flow = 0.8 mlmin 1, detection at 240 nm. Eluent = MilliQ:MeCN:TFA (95:5:0.1), TFA: trifluoroacetic acid. H: hold time (min), R: temperature ramp ( C/min). Entry 1: GC equipment: Shimadzu GC 2010 Entry 2: HPLC Equipment: Shimadzu LC-20AT with SDP-M20A IVDD Detector The following chromatograms (Fig. S5) show the conversion of ketoisophorone and the formation of (S/R)-levodione. Fig. S5 The GC chromatogram from the course of reaction at time 0, 1h, 3h and 24 h. The hydrogenation of KIP (t R = 10.7 min) catalyzed by TsER using 1,4-BD in HLADH-catalyzed NADH regeneration. Similarly following chromatograms (Fig. S6) represent the conversion of 3HB to 2,5-DHB catalysed by 3HB6H followed by HPLC. Fig. S6 The HPLC chromatogram from the course of reaction at time 0, 1h, 2h and 24 h. The hydroxylation of 3HB (t R = 5.6 min) to 2,5-DHB (t R = 4.6 min) catalyzed by 3HB6H using 1,4-BD in HLADH-catalyzed NADH regeneration. S8