This article provides a comprehensive review of flavin-dependent photoenzymes for researchers and drug development professionals.
This article provides a comprehensive review of flavin-dependent photoenzymes for researchers and drug development professionals. It explores the foundational photochemical mechanisms and enzyme structures, details cutting-edge methodologies for synthesizing high-value compounds like pharmaceuticals and fine chemicals, addresses key challenges and optimization strategies through enzyme engineering, and validates these systems through comparative analysis with traditional methods. The integration of light energy with enzymatic precision offers sustainable and highly selective routes for complex organic transformations, with significant implications for biomedical and industrial applications.
This whitepaper provides an in-depth technical guide on flavin cofactors as foundational photochemical catalysts in biology. Framed within the broader thesis of advancing flavin-dependent photoenzymes in synthetic chemistry, it details the photophysical mechanisms, quantitative performance metrics, and experimental protocols essential for researchers in chemical biology and drug development. The aim is to equip scientists with the tools to harness these nature-evolved photocatalysts for challenging organic transformations.
Flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD) are universal biological cofactors derived from riboflavin (Vitamin B2). While traditionally studied for their role in redox enzymology (e.g., in dehydrogenases and oxidases), their photochemical properties are increasingly recognized as a cornerstone for a growing class of photoenzymes. These "photoreceptor" or "photoenzyme" proteins utilize the flavin chromophore to absorb blue light (λmax ~450 nm) and initiate radical chemistry, enabling reactions with unparalleled stereo- and regiocontrol. This guide explores this photochemical foundation, positioning flavins as ideal biocompatible photocatalysts for synthetic applications.
Flavin photocatalysis operates through well-defined photocycles. Key mechanistic pathways include:
1Fl*), acts as a potent oxidant or reductant, accepting or donating an electron from/to a substrate, generating reactive radical pairs.3Fl*) is common, facilitating reactions with slower substrates or via energy transfer.The choice of mechanism depends on the protein environment, substrate, and reaction conditions.
Diagram 1: Core flavin photocycle pathways.
The utility of a photocatalyst is defined by quantifiable photophysical and catalytic parameters. The following table summarizes key data for free flavins and representative photoenzymes, highlighting nature's optimization within a protein scaffold.
Table 1: Photophysical & Catalytic Properties of Flavin Systems
| System | ε at λmax (Mâ»Â¹cmâ»Â¹) | λmax (nm) | Fluorescence Quantum Yield (Φ_F) | Triplet Quantum Yield (Φ_ISC) | Redox Potential E(Fl*/Flâ¢â) (V vs. SCE) | Typical k_cat (sâ»Â¹) under Light |
|---|---|---|---|---|---|---|
| Free FMN (in buffer) | 12,500 | 445 | 0.26 | 0.67 | ~ -2.1 to -2.3 | N/A (non-catalytic) |
| Free FAD (in buffer) | 11,300 | 450 | 0.03 | High | ~ -2.1 to -2.3 | N/A (non-catalytic) |
| LOV Domains (Photoreceptors) | ~12,000-14,000 | 447 | 0.1 - 0.4 | 0.4 - 0.6 | Modulated by protein | N/A (Signaling) |
| Enzymatic Photodecarboxylase (FAP) | ~13,000 | 448 | Very Low | Very High | ~ -1.8 (optimized) | 10 - 50 |
| Flavin-dependent Ene-Reductases (illuminated) | ~12,000 | 455 | Low | High | Tunable (~ -0.8 to -1.5) | 0.1 - 5 |
| NADPH:Flavin Oxidoreductase (Light-driven) | ~11,500 | 460 | Low | High | ~ -1.4 | 0.5 - 10 |
Data compiled from recent literature. Redox potentials are approximations and vary with environment. k_cat is reaction-specific.
This protocol measures the light-dependent conversion of a fatty acid to an alkane by a Fatty Acid Photodecarboxylase (FAP).
Objective: Quantify the catalytic turnover of a flavin photoenzyme using UV-Vis spectroscopy and product analysis (GC-MS/HPLC).
Materials: See "The Scientist's Toolkit" below.
Procedure:
Objective: Evaluate synthetic flavin analogs (e.g., 8-Cl-FAD, 5-deaza-FMN) for altered photophysical properties and catalytic efficiency when reconstituted into an apo-photoenzyme.
Procedure:
Diagram 2: Workflow for screening flavin analogs.
Table 2: Key Reagents for Flavin Photoenzyme Research
| Item | Function & Explanation |
|---|---|
| Riboflavin (Vitamin B2) | Precursor for in vivo flavin biosynthesis; used as a supplement in recombinant protein expression. |
| FMN / FAD Sodium Salts | Authentic cofactor standards for spectroscopy, reconstitution experiments, and calibration. |
| Apo-Glutamate Synthase / Apo-Flavodoxin | Commercially available apo-proteins for testing non-covalent flavin binding and photochemistry. |
| 8-Substituted Flavin Analogs (e.g., 8-Cl-FAD) | Synthetic cofactors with altered redox potentials and excited-state properties for mechanistic probing. |
| Deazaflavins (e.g., 5-Deaza-FMN) | Non-photoactive flavin analogs used as essential controls to confirm photochemical (vs. thermal) pathways. |
| DEADC (Diethyl azodicarboxylate) | Chemical quencher used in "light-dark" trapping experiments to confirm radical intermediates. |
| DâO & ¹â¸O-Labeled Water | Isotopic solvents for probing proton-coupled electron transfer (PCET) mechanisms via kinetic isotope effects (KIE). |
| Anaerobic Chamber / Cupless Septa | Essential for creating oxygen-free environments, as molecular oxygen is a potent quencher of flavin excited states and triplet radicals. |
| Calibrated Blue LED System (λ=450±20 nm) | Standardized, cool light source to provide consistent, monochromatic photoexcitation without sample heating. |
| Benchtop Spectrofluorometer with Stirrer | For measuring fluorescence quantum yields and real-time monitoring of flavin fluorescence during turnover. |
| N-Boc-5-bromoindole | N-Boc-5-bromoindole, CAS:182344-70-3, MF:C13H14BrNO2, MW:296.16 g/mol |
| N-Acetyltaurine | N-Acetyltaurine|Nat Acetyl Taurine|RUO |
This whitepaper situates the evolution of flavin-dependent photoenzymes within a broader thesis on their transformative role in organic synthesis. Historically viewed as biological curiosities, natural photoenzymes like DNA photolyase have provided the foundational blueprint for engineering sophisticated biocatalysts capable of catalyzing asymmetric radical transformations under mild conditions. This journey from understanding natural photobiology to deploying engineered photoenzymes in synthetic routes represents a paradigm shift for researchers and drug development professionals seeking sustainable, stereoselective methodologies.
The following table summarizes the quantitative progression from discovery to engineering.
Table 1: Historical Timeline and Performance Metrics of Flavin-Dependent Photoenzymes
| Era | Key Enzyme/System | Discovery/Engineering Year | Primary Function | Quantum Yield (Φ) | Turnover Number (TON) | Enantiomeric Excess (ee) Achieved |
|---|---|---|---|---|---|---|
| Natural | DNA Photolyase | 1958 (Isolation) | UV-induced DNA repair | ~0.7 - 0.9 | N/A (stoichiometric) | N/A |
| Natural | Fatty Acid Photodecarboxylase (FAP) | 2017 (Characterized) | Light-driven decarboxylation | ~0.8 | >1000 (in vivo) | N/A (non-chiral) |
| Engineered | Old Yellow Enzyme (OYE) variants | 2010-2016 | Asymmetric hydroalkylation | N/A | 50 - 200 | ~80% |
| Engineered | Engineered FAPs (e.g., for C-N coupling) | 2020-2023 | Asymmetric radical C-C & C-X bond formation | 0.1 - 0.5 | 500 - 10,000 | 90% - >99% |
| Engineered | "Lov2"-based artificial photoenzyme | 2022 | Intermolecular [2+2] photocycloaddition | ~0.05 | ~300 | ~95% |
Objective: To identify engineered FAP variants with high activity and enantioselectivity for the radical alkylation of olefins. Materials: See "The Scientist's Toolkit" (Section 5). Procedure:
Objective: Quantify the efficiency of photon utilization by the photoenzyme. Procedure:
Diagram 1: Historical R&D Pathway for Photoenzymes.
Diagram 2: Key Photoredox Steps in FAP Catalysis.
Diagram 3: Directed Evolution Screening Workflow.
Table 2: Essential Materials for Photoenzyme Research and Application
| Reagent/Material | Function & Explanation | Example Vendor/Product |
|---|---|---|
| Flavin Cofactors (FAD, FMN) | Essential cofactor for reconstitution of apo-enzymes; used in mechanistic studies and activity assays. | Sigma-Aldrich, F6625 (FAD) |
| Chiral Substrates & Probes | Olefins (enones, acrylates) and radical precursors (fatty acids, alkyl halides) for testing substrate scope and enantioselectivity. | Enamine Ltd., diverse building blocks |
| Site-Directed Mutagenesis Kits | For creating targeted mutations (e.g., NNK library) in photoenzyme genes. | NEB, Q5 Site-Directed Mutagenesis Kit |
| Expression Hosts & Vectors | High-yield protein production. E. coli BL21(DE3) and pET vectors are standard. | Novagen, pET-28a(+) vector |
| Photoreaction Equipment | Controlled light source (LED arrays, monochromators) for reproducible photobiocatalysis. | Thorlabs, custom LED drivers; Luzchem, LZC-ICH2 photoreactor |
| Chemical Actinometers | To quantify photon flux in quantum yield and kinetic experiments (e.g., potassium ferrioxalate). | Reagents prepared in-lab per IUPAC protocol |
| Chiral Stationary Phase Columns | For enantiomeric separation and analysis of reaction products (essential for ee determination). | Daicel, Chiralpak IA/IB/IC columns |
| Anaerobic Experiment Kits | For studying oxygen-sensitive radical intermediates; includes septum-sealed cuvettes and glove boxes. | Coy Laboratory Products, Anaerobic Chamber |
| Rapid Kinetics Stopped-Flow | Instrumentation for measuring fast photochemical kinetics (ns-ms timescale). | Applied Photophysics, SX20 Stopped-Flow |
| Quartz Cuvettes | For UV-Vis spectroscopy and photochemical experiments; ensure high transmittance at relevant wavelengths. | Hellma Analytics, high-precision cuvettes |
| Ompenaclid | 3-Guanidinopropionic Acid (β-GPA) | |
| Thiophene E | Echinoynethiophene A|High-Quality Reference Standard |
This technical guide details the mechanisms underpinning flavin-dependent photoenzymes, a central theme in modern organic synthesis research. These enzymes, which utilize non-covalently bound flavin cofactors (typically flavin mononucleotide, FMN), have revolutionized asymmetric synthesis by enabling unprecedented radical transformations under mild, visible-light irradiation. Their mechanistic framework is foundational for advancing synthetic methodologies and drug development.
The catalytic cycle is initiated by the absorption of a photon by the flavin cofactor in its oxidized, ground state (Flox). The isoalloxazine ring system acts as a potent chromophore, with a characteristic absorption spectrum featuring three primary bands in the visible/UV range. This absorption promotes the flavin to an excited singlet state (*Flox).
Table 1: Key Photophysical Parameters of Oxidized Flavin Cofactor
| Parameter | Value / Characteristic | Significance |
|---|---|---|
| Primary Absorption Maxima | ~375 nm & ~450 nm | Enables activation by visible light (blue). |
| Molar Extinction Coefficient (ε450) | ~12,500 Mâ»Â¹cmâ»Â¹ | High efficiency of photon capture. |
| Fluorescence Quantum Yield | ~0.1 - 0.3 | Competes with productive intersystem crossing. |
| Intersystem Crossing Rate | ~10¹¹ sâ»Â¹ | Efficient population of the reactive triplet state. |
Following excitation, the enzyme exerts precise control over reactivity by facilitating the formation of transient complexes between the photoexcited flavin (*Flox) and the bound organic substrate. These are termed Electron Donor-Acceptor (EDA) or Charge Transfer (CT) complexes. The enzyme's active site architecture positions the substrate optimally, lowering the kinetic barrier for electron transfer (eT). Spectroscopically, CT complex formation is often indicated by a broadening or redshift of the flavin absorption band.
Experimental Protocol 1: Spectroscopic Detection of a CT Complex
The CT complex facilitates the critical electron transfer event. Two primary radical initiation pathways have been characterized:
The substrate radical intermediate is then poised for stereocontrolled transformations (e.g., radical addition, reduction, cyclization) within the chiral enzyme environment.
Experimental Protocol 2: Laser Flash Photolysis for Kinetic Analysis
Diagram 1: Core Photoenzyme Mechanism
Diagram 2: Radical Initiation Pathways
Table 2: Essential Reagents for Flavin Photoenzyme Research
| Reagent / Material | Function & Rationale |
|---|---|
| Purified Flavin Photoenzyme (e.g., PFE, OPR, PET) | Catalytic protein scaffold for stereocontrol. Often used as His-tagged variants for immobilization. |
| Flavin Cofactors (FMN, FAD, Riboflavin) | Essential photoredox cofactor. FMN is most common in engineered enzymes. |
| Deazaflavin Analogues (e.g., 5-Deazaflavin) | Flavin analogs with altered redox potentials; used for mechanistic probing of electron transfer steps. |
| Anaerobic Chamber / Glovebox | Essential for studying radical intermediates without interference from atmospheric oxygen (a potent quencher and side-reagent). |
| Deuterated Solvents (DâO, d³-Acetonitrile) | For isotopic labeling studies to track hydrogen atom transfer (HAT) pathways via kinetic isotope effects (KIEs). |
| Stopped-Flow / Rapid Mixing System | Allows kinetic study of fast photochemical steps (ms-s) by rapid mixing of enzyme and substrate prior to laser pulse. |
| Silanized Glassware | Prevents adsorption of apolar substrates/enzymes and minimizes unwanted radical initiation on glass surfaces. |
| Chemical Quenchers (e.g., Oxygen, TEMPO) | Used to trap and characterize radical intermediates. TEMPO is a stable radical that efficiently scavenges carbon-centered radicals. |
| Spectroscopic Probes (e.g., Methyl Viologon, Ferricyanide) | Redox dyes with known potentials used in competition experiments to estimate flavin excited state redox potentials. |
| Territrem A | Territrem A, CAS:70407-19-1, MF:C28H30O9, MW:510.5 g/mol |
| Viburnitol | Desoxy-inositol |
Thesis Context: Within the broader investigation of flavin-dependent photoenzymes for sustainable organic synthesis, understanding the precise protein scaffolds that bind and modulate flavin cofactors is fundamental. These architectures define reactivity, enantioselectivity, and photophysical properties, enabling novel CâH functionalization and asymmetric transformations.
Flavin-dependent proteins employ a limited set of highly conserved structural folds to bind flavin adenine dinucleotide (FAD) or flavin mononucleotide (FMN). The architecture dictates the cofactor's redox potential and exposure to substrate.
Table 1: Core Flavin-Binding Protein Folds and Characteristics
| Structural Fold | Representative Protein Family | Flavin Linkage | Key Structural Feature | Redox Potential (E'°) Range | Role in Photoenzymatic Synthesis |
|---|---|---|---|---|---|
| TIM Barrel | Old Yellow Enzyme (OYE) | Non-covalent, typically FMN | Rossmann fold for NADPH binding; β-barrel core | -150 to -200 mV | Enantioselective alkene reduction via hydride transfer. |
| p-Cresol Methylhydroxylase (PCMH)-like | Flavoprotein monooxygenases (e.g., cyclohexanone monooxygenase) | Covalent (8α-N1-histidyl, 8α-O-tyrosyl) | Baeyer-Villiger monooxygenases; FAD in a two-domain structure. | ~ -300 mV | Asymmetric Baeyer-Villiger oxidations and sulfoxidations. |
| BLUF (Blue-Light Sensors Using FAD) | Photolyase/Cryptochrome family | Non-covalent FAD | Antiparallel β-sheet flanking FAD; key Gln/Tyr for light sensing. | N/A (Light sensor) | Provides light-gated control over enzymatic steps in hybrid systems. |
| (α/β)â Rossmann Fold | Flavin reductases (Fre) | Non-covalent FMN/FAD | Central parallel β-sheet with surrounding α-helices. | Variable | Regenerates reduced flavin (FMNHâ»/FADHâ») for downstream photocatalytic cycles. |
| Lovit (Light-Oxygen-Voltage) | LOV-domain proteins | Covalent (C4a-cysteinyl) | PAS domain variant; forms flavin-cysteinyl adduct upon blue light. | N/A (Light sensor) | Optogenetic tool for spatiotemporal control of synthetic enzyme cascades. |
Objective: Quantify the affinity of an apoprotein scaffold for FMN/FAD. Method:
Objective: Utilize a flavin-dependent monooxygenase scaffold for enantioselective synthesis. Method:
Diagram Title: Primary protein folds for flavin cofactor binding.
Diagram Title: Light-driven enzymatic sulfoxidation catalytic cycle.
Table 2: Essential Reagents for Flavin-Protein Research
| Reagent / Material | Supplier Examples | Function & Application Notes |
|---|---|---|
| Riboflavin (Vitamin B2) | Sigma-Aldrich, TCI Chemicals | Precursor for flavin synthesis; used in media for overexpression of flavoproteins. |
| FMN (Flavin Mononucleotide), Sodium Salt | Carbosynth, Roche | Essential cofactor for reconstitution assays; preferred over riboflavin for direct binding studies due to phosphate moiety. |
| FAD (Flavin Adenine Dinucleotide), Disodium Salt | Sigma-Aldrich, Cayman Chemical | Cofactor for oxidases, monooxygenases, and electron transferases; critical for enzymes requiring adenosine binding motif. |
| Glucose-6-Dehydrogenase (from Leuconostoc mesenteroides) | Sigma-Aldrich, Roche | Key component of NADPH-regeneration systems; thermostable and utilizes NADP⺠efficiently. |
| NADP⺠/ NADPH Tetrasodium Salts | Biomol, Oriental Yeast | Essential redox cofactor for >90% of flavin-dependent enzymes; high-purity salts reduce assay background. |
| Dioxygenase Activity Probe (Amplex UltraRed) | Thermo Fisher | Fluorogenic substrate (10-acetyl-3,7-dihydroxyphenoxazine) for detecting HâOâ production by flavin oxidases. |
| Flavin Analogs (e.g., 8-Cl-FAD, 5-Deaza-FMN) | Toronto Research Chemicals | Mechanistic probes for studying electron transfer pathways and modulating redox potentials. |
| Anaerobic Cuvette Kit (Sealed, with Septum) | Hellma, Pierce | Required for studying oxygen-sensitive reduced flavin intermediates (e.g., flavin hydroquinones). |
| Blue LED Photoreactor (450 ± 10 nm) | Lumatec, Thorlabs | Provides controlled, high-intensity light for photoenzyme kinetics and preparative-scale biotransformations. |
| Chiral HPLC Columns (e.g., Chiralpak IA, IB) | Daicel, Phenomenex | Mandatory for analyzing enantiomeric excess (ee) in asymmetric synthesis catalyzed by engineered flavoproteins. |
| Choerospondin | Choerospondin, CAS:81202-36-0, MF:C21H22O10, MW:434.4 g/mol | Chemical Reagent |
| Zolasartan | Zolasartan|AT1R Antagonist|For Research Use | Zolasartan is a small molecule AT1R antagonist. This product is for Research Use Only and is not intended for diagnostic or therapeutic use. |
This technical guide explores the emerging paradigm of discovering latent photoactivities within well-characterized, canonical enzyme families, framed within a thesis on advancing flavin-dependent photoenzymes in organic synthesis. Moving beyond dedicated photoenzymes (e.g., DNA photolyases, flavin-dependent "ene"-reductases with photoactivity), we detail methodologies to uncover and harness cryptic photochemical functions in traditional oxidoreductases, hydrolases, and transferases. This unlocks new-to-nature photocatalytic reactions for synthetic and pharmaceutical applications.
Many enzymes bind chromophoric cofactors (flavins, nicotinamides, tetrapyrroles, pterins) for ground-state catalysis. We posit that such cofactors, when excited by specific wavelengths of light, can initiate electron or energy transfer processes that are suppressed or non-competitive under standard physiological conditions. The systematic exploration of these latent pathways constitutes a new exploratory frontier.
Latent photoactivity typically arises from the photoexcited state of a bound cofactor. For flavin-dependent enzymesâthe central focus within our broader thesisâthis involves the following potential pathways post-absorption of blue light (~350-450 nm):
Table 1: Flavin Photocycle States and Reactivity
| Flavin State | Lifetime | Key Reactivity | Potential Enzymatic Role |
|---|---|---|---|
| 1Flavin (Singlet) | ~1-10 ns | Energy Transfer, Electron Transfer | Initiation of radical chains, substrate sensitization |
| 3Flavin (Triplet) | ~1-100 µs | Hydrogen Atom Transfer, Electron Transfer | Direct substrate radical generation, inter-protein electron hopping |
| Flavin Semiquinone | Variable (ms-s) | Radical Propagation | Long-range electron transfer, coupled catalytic turnover |
Title: Flavin Photocycle and Latent Reaction Pathways
Objective: Identify candidate enzymes from existing families (e.g., NADPH-cytochrome P450 reductase family, Old Yellow Enzyme family, luciferase-like hydrolases) with structural propensity for photoactivity. Methodology:
Table 2: In Silico Screening Metrics & Benchmarks
| Parameter | Tool/Method | Target Range for Latent Photoactivity |
|---|---|---|
| Cofactor SASA | FPocket, PyMOL | >40 à ² (suggestive of substrate/quencher access) |
| Excited State Lifetime Prediction | QM/MM (TD-DFT) | Triplet yield >0.4 |
| Proximal Redox-Amino Acid Distance | Pymol Measurement | <8 Ã for efficient electron transfer |
| Active Site Electrostatic Potential | APBS | Polar environment to stabilize radical intermediates |
Objective: Confirm light-dependent turnover with native or non-native substrates. Protocol: Reagents:
Procedure:
Table 3: Example Photochemical Screening Results for an OYE Homolog
| Enzyme (Family) | Substrate (Non-native) | Dark TOF (minâ»Â¹) | 450 nm Light TOF (minâ»Â¹) | TTN (Light) | Primary Product |
|---|---|---|---|---|---|
| OYE1 (Canonical) | 2-Cyclohexen-1-one | 12.5 | 310.2 | >10,000 | Cyclohexanone |
| OYE3 Homolog | α-Methylstyrene | 0.05 | 8.7 | ~1200 | Radical Dimer |
| P450 Reductase | Aryl Iodide (C-I) | N.D. | 2.1 | ~300 | Dehalogenated Arene |
Objective: Unambiguously assign the photochemical mechanism. Key Experiments:
Table 4: Essential Materials for Discovering Latent Photoactivities
| Reagent/Material | Vendor Examples (Typical) | Function & Rationale |
|---|---|---|
| High-Purity Flavin Cofactors | Sigma-Aldrich (FAD, FMN, Riboflavin) | Reconstitution of apo-enzymes for photophysical studies; isotopic labeling. |
| Custom LED Photoreactors | Lumencor, CoolLED, Thorlabs | Precise, tunable wavelength control (365-450 nm) with calibrated intensity for reproducible kinetics. |
| Anaerobic Reaction Chambers | Coy Laboratory Products, Belle Technology | Creation of oxygen-free environment essential for studying long-lived triplet states and radical mechanisms. |
| Quartz Microcuvettes | Hellma Analytics | UV-transparent vessels for spectroscopy and irradiation with small reaction volumes (50-200 µL). |
| Radical Trapping Agents | Sigma-Aldrich (TEMPO, BHT) | Chemical probes to confirm radical-based mechanisms and quantify radical flux. |
| Deuterated & ¹³C-Labeled Substrates | Cambridge Isotope Laboratories | Isotopic tracing to elucidate reaction mechanisms and bond-breaking/forming steps. |
| Stopped-Flow Flash Photolysis System | Applied Photophysics, TgK Scientific | Direct kinetic measurement of excited state formation and decay on µs-ms timescales. |
| Q-Sepharose Fast Flow Resin | Cytiva | Purification of often-sticky flavoprotein candidates via anion-exchange chromatography. |
| NOTA-bis(tBu)ester | NOTA-bis(tBu)ester, MF:C20H37N3O6, MW:415.5 g/mol | Chemical Reagent |
| Biotin-PEG7-thiourea | Biotin-PEG7-thiourea, MF:C27H51N5O9S2, MW:653.9 g/mol | Chemical Reagent |
Title: Latent Photoactivity Discovery Workflow
The discovery of latent photoactivity enables new biocatalytic routes:
The deliberate search for latent photoactivities reframes our understanding of enzyme function and dramatically expands the catalytic repertoire available for sustainable synthesis. Flavin-dependent enzymes, as a cornerstone of this thesis, provide a rich and tractable starting point for this exploration, promising novel reactivities for the synthesis of complex pharmaceuticals and fine chemicals.
This whitepaper details the construction of Photo-Enzyme Coupled Systems (PECS) for methanol synthesis, situated within a broader thesis investigating flavin-dependent photoenzymes in organic synthesis. The central thesis posits that the unique photoredox properties of flavins, when harnessed within engineered enzymatic frameworks, can drive challenging chemical transformations with unparalleled selectivity and under mild conditions. This work extends that principle to the critical challenge of sustainable COâ valorization, coupling light-harvesting components with COâ-reducing enzymes to create artificial photosynthetic systems.
A functional PECS integrates three critical units: (1) a photosensitizer (PS) for light harvesting and excited-state electron generation, (2) a redox mediator/shuttle (M) for efficient electron transfer, and (3) the catalytic enzyme, typically a NADPH-dependent dehydrogenase such as formaldehyde dehydrogenase (FaldDH) and alcohol dehydrogenase (ADH), which are often cascaded with formate dehydrogenase (FDH) for the multi-step reduction of COâ to methanol (COâ â HCOOH â HCHO â CHâOH). The flavin-based enzyme Old Yellow Enzyme (OYE) or engineered variants are frequently employed as the initial photobiocatalyst, using flavin mononucleotide (FMN) to accept electrons from the reduced mediator upon photoexcitation and subsequently regenerate NADPH.
Table 1: Key Components of a Model PECS for Methanol Production
| Component | Example Species/Compound | Primary Function | Key Property |
|---|---|---|---|
| Photosensitizer | [Ru(bpy)â]²âº, Carbon Nitride (CâNâ), Eosin Y | Absorbs visible light, generates excited state and initiates electron transfer. | High molar absorptivity, long excited-state lifetime, suitable redox potentials. |
| Electron Donor | Triethanolamine (TEOA), Ethylenediaminetetraacetic acid (EDTA) | Sacrificial reagent that replenishes electrons to the oxidized photosensitizer. | Irreversibly oxidized, maintains PS cycle. |
| Redox Mediator | [Cp*Rh(bpy)HâO]²âº, Viologen derivatives | Shuttles electrons from the reduced PS to the enzymatic cofactor (NADPâº). | Matches redox potentials of PS* and NADPâº/NADPH. |
| Flavin Photoenzyme | Engineered Old Yellow Enzyme (OYE) | Uses photoexcited flavin (FMN) to catalyze NADP⺠reduction using electrons from the mediator. | Flavin acts as a biocatalytic photocatalyst. |
| Dehydrogenase Cascade | FDH, FaldDH, ADH | Catalyzes the sequential reduction of COâ to formate, formaldehyde, and methanol. | NADPH-dependent, high specificity, operates in aqueous buffer. |
| 21-Deoxycortisol-d8 | 21-Deoxycortisol-d8 Stable Isotope|354.51 g/mol | 21-Deoxycortisol-d8 is a deuterium-labeled internal standard for accurate LC-MS/MS quantification of 21-Deoxycortisol in CAH research. For Research Use Only. Not for human or veterinary use. | Bench Chemicals |
| 3-Keto petromyzonol | 3-Keto petromyzonol, MF:C24H40O4, MW:392.6 g/mol | Chemical Reagent | Bench Chemicals |
Objective: To construct a light-driven system for COâ-to-methanol conversion using [Ru(bpy)â]²⺠as PS, [Cp*Rh(bpy)(HâO)]²⺠as mediator, and a dehydrogenase cascade.
Materials:
Procedure:
Controls: Perform identical experiments (a) in the dark, (b) without enzymes, (c) without PS, and (d) without light.
Objective: To enhance system stability and enable recyclability by co-immobilizing PS, mediator, and enzymes.
Materials: Mesoporous SiOâ nanoparticles, (3-aminopropyl)triethoxysilane (APTES), glutaraldehyde, Poly(ethyleneimine) (PEI).
Procedure:
Table 2: Performance Metrics of Representative PECS Configurations from Recent Literature
| PS / Mediator Pair | Enzyme System | Light Source | Reaction Time (h) | Methanol Yield (µmol) | Turnover Number (TON)⺠| Key Reference/Feature |
|---|---|---|---|---|---|---|
| [Ru(bpy)â]²⺠/ [Cp*Rh]²⺠| FDH, FaldDH, ADH | 450 nm LED | 24 | ~150 | ~300 (NADPH) | Lee et al., 2017. Benchmark homogeneous system. |
| Carbon Nitride (CâNâ) / [Cp*Rh]²⺠| Same as above | >420 nm Filter | 12 | 89 | ~180 | Heterogeneous, metal-free PS. |
| Eosin Y / Ascorbate | OYE (for NADPH regen.) + Dehydrogenase cascade | 520 nm LED | 18 | 65 | ~130 | Flavin-enzyme direct photoexcitation. |
| CdS QDs / Methyl Viologen | FDH, FaldDH, ADH | Solar Simulator | 10 | 210 | ~400 | Semiconductor PS, high light harvesting. |
⺠TON calculated relative to initial NADP⺠or mediator concentration.
Diagram 1: Electron and Catalytic Flow in a Flavin-Involving PECS
Diagram 2: Standard PECS Assembly and Testing Workflow
Table 3: Essential Reagents and Materials for PECS Construction
| Reagent / Material | Supplier Examples | Function in PECS | Critical Considerations |
|---|---|---|---|
| [Ru(bpy)â]Clâ | Sigma-Aldrich, TCI Chemicals | Benchmark homogeneous photosensitizer. | Purity >99%; store in dark, desiccated; check for decomposition (color change). |
| Carbon Nitride (CâNâ) | Alfa Aesar, or lab-synthesized | Metal-free, heterogeneous, visible-light PS. | Control band gap via thermal polymerization temperature; high surface area preferred. |
| Cp*Rh(bpy)(HâO)â | Strem Chemicals, custom synthesis | Highly efficient and stable redox mediator for NADâº/NADP⺠regeneration. | Must be handled under inert atmosphere; aqueous stability is pH-dependent. |
| Triethanolamine (TEOA) | Sigma-Aldrich, Fisher Scientific | Sacrificial electron donor. | Purify by distillation to remove amines that may inhibit enzymes; pH of final solution is crucial. |
| NADP⺠Sodium Salt | Roche, Sigma-Aldrich | Essential enzymatic cofactor. | High purity (â¥98%); prepare fresh solutions; monitor stability in buffer (A340). |
| Recombinant Dehydrogenases (FDH, FaldDH, ADH) | Sigma-Aldrich, Codexis, or recombinant expression | Catalytic core for COâ reduction cascade. | Specific activity (U/mg) should be verified; check for latent formaldehyde reductase activity in ADH. |
| Old Yellow Enzyme (OYE) variants | In-house expression from engineered plasmids | Flavin-dependent photobiocatalyst for NADPH regeneration. | Expression yield and FMN incorporation efficiency are critical; photostability assays required. |
| Anaerobic Chamber | Coy Lab Products, Plas Labs | For oxygen-free assembly of reaction mixtures. | Maintain Hâ/Nâ atmosphere; monitor oxygen levels (<1 ppm) for enzyme and mediator stability. |
| LED Photoreactor | Luzchem, Völkner, custom-built | Provides controlled, monochromatic illumination. | Calibrate light intensity (mW/cm²) with radiometer; ensure uniform irradiation of samples. |
| Mogroside II-A2 | Mogroside II-A2, MF:C42H72O14, MW:801.0 g/mol | Chemical Reagent | Bench Chemicals |
| Regelidine | Regelidine, MF:C35H37NO8, MW:599.7 g/mol | Chemical Reagent | Bench Chemicals |
This guide details a pivotal application within a broader thesis investigating flavin-dependent photoenzymes in organic synthesis. These enzymes, upon photoexcitation of their bound flavin cofactor, generate potent yet tunable reductants capable of driving challenging radical reactions with exquisite stereocontrol. The enantioselective radical trifluoromethylation of prochiral alkenes represents a landmark demonstration of this capability, providing direct, catalytic access to chiral β-trifluoromethyl carbonyl motifsâhigh-value building blocks in pharmaceutical research where the CFâ group profoundly influences a molecule's metabolic stability, lipophilicity, and binding affinity.
The reaction couples a trifluoromethyl radical (â¢CFâ) source with an activated alkene (e.g., enone) under mild, visible-light irradiation, using a engineered flavin-dependent "ene"-reductase (ERED) as the stereodetermining photoredox catalyst.
Table 1: Representative Substrate Scope & Performance Data [citation:4 and current literature]
| Substrate Class (R) | Example Structure | Yield (%) | ee (%) | Notes |
|---|---|---|---|---|
| Cyclic Enones (6-membered) | 2-cyclohexen-1-one | 85-92 | 94-99 | Optimal ring size; excellent enantioselectivity. |
| Cyclic Enones (5-membered) | 2-cyclopenten-1-one | 78 | 91 | Slightly diminished yield. |
| Acyclic Enones | (E)-4-phenylbut-3-en-2-one | 65 | 90 | Moderate yield, high ee maintained. |
| β,β-Disubstituted Enones | 3-methyl-2-cyclohexen-1-one | 45 | 85 | Challenging substrates; yield impacted by sterics. |
| Alkyl-Substituted Enones | 2-cyclohepten-1-one | 88 | 96 | Broad tolerance for alkyl chains. |
Mechanistic Pathway Diagram:
Diagram Title: Flavin Photoredox Cycle for Enantioselective Radical Trifluoromethylation
A. General Procedure for Photoenzymatic Trifluoromethylation :
B. Control Experiments:
Table 2: Key Research Reagent Solutions
| Reagent / Material | Function & Rationale |
|---|---|
| Engineered Flavin-Dependent ERED (e.g., PhetA N,S) | Stereocontrolling photoredox biocatalyst. Its engineered active site dictates the facial selectivity of radical addition and H-transfer. |
| Trifluoromethyl Iodide (CFâI) | Volatile, gaseous source of â¢CFâ radical upon single-electron reduction. Alternatives include CFâSOâCl or bench-stable sulfonium salts. |
| NADP⺠/ Glucose-6-Phosphate / G6PDH | Enzymatic cofactor regeneration system. Maintains catalytic concentrations of reduced flavin (FADHâ») without stoichiometric NADPH. |
| Blue LED Photoreactor (λmax ~450 nm) | Light source matching the absorption maximum of the reduced flavin hydroquinone anion (FADHâ») for efficient photoexcitation. |
| Anaerobic Sealing (Septum & Argon) | Excludes oxygen, a potent quencher of radical intermediates and excited-state flavin. |
| Chiral HPLC/SFC Column | Critical for accurate determination of enantiomeric excess (ee) of the chiral product. |
| Potassium Phosphate Buffer (pH 7.0) | Aqueous reaction medium providing optimal stability and activity for the enzyme. |
| Aftin-5 | Aftin-5, MF:C19H26N6O, MW:354.4 g/mol |
| Hemiphroside B | Hemiphroside B, MF:C31H38O17, MW:682.6 g/mol |
Experimental Workflow Diagram:
Diagram Title: Photoenzymatic Trifluoromethylation Experimental Workflow
Within the burgeoning field of flavin-dependent photoenzymes in organic synthesis, engineered 'ene'-reductases (EREDs) have emerged as powerful catalysts for radical-mediated transformations. Traditionally known for asymmetric hydrogenation of activated alkenes using nicotinamide cofactors, recent work has demonstrated that photoexcitation of the enzyme-bound flavin cofactor enables radical initiation. This transforms EREDs into efficient photoredox biocatalysts capable of driving challenging radical cyclizations and carbon-carbon (C-C) bond formations under mild, biocompatible conditions. This technical guide details the mechanisms, engineering strategies, and experimental protocols underpinning this technology.
The catalytic activity hinges on the photophysics of the flavin mononucleotide (FMN) or flavin adenine dinucleotide (FAD) prosthetic group. Upon blue light irradiation, the flavin transitions to an excited singlet state, which intersystem crosses to a potent, long-lived triplet state. This triplet flavin can oxidize a suitable substrate (e.g., an alkyl halide) via single-electron transfer (SET), generating a substrate radical and a flavin semiquinone. The radical species then undergoes intramolecular cyclization or intermolecular coupling. The reduced flavin is ultimately regenerated, often by an exogenous sacrificial reductant (e.g., dithionite or a phosphite), closing the catalytic cycle.
Directed evolution campaigns have been critical to unlocking this non-natural function. Key engineering targets include:
Table 1: Representative Engineered EREDs for Radical Reactions
| ERED Variant (Parent) | Key Mutations | Optimized Substrate Class | Primary Reaction Type | Reported Yield (%)* | Enantiomeric Excess (ee%)* |
|---|---|---|---|---|---|
| GluER-B3 (OYE1) | W66S, H167N, I232T | α-Haloamides | Radical Cyclization (5-exo-trig) | 85-95 | >99 |
| NerER (NCR) | F250A, L213H | Bromomalonates | Intermolecular C-C Coupling | 78 | 92 |
| PET-Redam (OYE1) | H167N, I232T, Y375W | Redox-Active Esters | Dehalogenative Alkylation | 91 | 98 |
| YqjM Variant (YqjM) | S245W, T246G | α-Chloroketones | Desymmetrizing Cyclization | 82 | 95 |
*Representative values from published literature; optimal results are substrate-dependent.
Table 2: Essential Materials for ERED-Mediated Radical Reactions
| Item | Function/Explanation | Example/Catalog Consideration |
|---|---|---|
| Engineered ERED | Recombinant biocatalyst (purified enzyme or whole-cell preparation) harboring flavin cofactor. | Purified GluER-B3 (expressed in E. coli with a His-tag). |
| Blue LED Light Source | Provides 440-470 nm light to excite the flavin cofactor. Essential for radical initiation. | Kessil PR160L Blue LED lamp or custom-built photoreactor. |
| Substrate: Alkyl Halide/Redox-Active Eryl | Radical precursor. Common substrates include α-chloroamides, α-bromoketones, NHPI/Phth esters. | Ethyl 2-bromo-2-phenylacetate (CAS 600-00-0). |
| Sacrificial Reductant | Terminal electron donor to regenerate the reduced flavin state. | Sodium dithionite (NaâSâOâ) or Hantzsch ester (HEH). |
| Cofactor/Additive | May be required for stability or activity. | FMN (if using apo-enzyme), EDTA (chelator). |
| Anaerobic Buffer System | Deoxygenated buffer to prevent radical quenching by Oâ. | 50 mM Potassium Phosphate, pH 7.0, sparged with Nâ/Ar. |
| NADPH | Natural cofactor for native ERED reduction; sometimes used in coupled systems. | For enzymatic flavin reduction cycles. |
| Methyl lycernuate A | Methyl lycernuate A, MF:C31H50O4, MW:486.7 g/mol | Chemical Reagent |
| Vitexin arginine | Vitexin arginine, MF:C27H34N4O12, MW:606.6 g/mol | Chemical Reagent |
Objective: Intramolecular radical cyclization of an α-chloroamide to form a γ-lactam.
Materials: Purified His-tagged ERED variant (e.g., GluER-B3), substrate (e.g., N-allyl-2-chloro-2-phenylacetamide), sodium dithionite (NaâSâOâ), potassium phosphate buffer (50 mM, pH 7.0), anaerobic chamber or Schlenk line, blue LED light source (450 nm), HPLC/MS for analysis.
Procedure:
Objective: Evaluate library of ERED variants for asymmetric C-C coupling between an alkyl bromide and an electron-deficient olefin.
Materials: E. coli whole cells expressing different ERED variants, substrate A (ethyl 2-bromo-2-methylpropanoate), substrate B (methyl acrylate), potassium phosphate buffer (100 mM, pH 7.0), glucose (as energy source), deep-well plate, plate shaker with integrated blue LED illumination.
Procedure:
Diagram 1: PhotoERED catalytic cycle for radical generation.
Diagram 2: Workflow for directed evolution of photoactive EREDs.
This whitepaper details a pivotal methodology within a broader thesis investigating the expanding synthetic utility of flavin-dependent photoenzymes. Moving beyond their established role in asymmetric hydrogen atom transfers, this work demonstrates how engineered flavoproteins can be integrated with transition metal photoredox catalysts to achieve previously inaccessible bond disconnections. The synthesis of enantiomerically enriched α-tertiary amino acids, crucial pharmacophores in modern drug discovery, serves as a paradigm for this synergistic approach, overcoming the significant kinetic and thermodynamic challenges associated with prochiral radical generation and stereocontrol.
The synergistic cycle couples a visible-light-driven photoredox catalyst (PC) with an engineered flavin-dependent "ene"-reductase (ERED). The photoredox cycle generates a prochiral α-amino radical from a readily prepared ketimine substrate. This radical intermediate is then intercepted and stereoselectively reduced by the reduced flavin hydroquinone (FADH¯) within the enzyme's active site, which is regenerated via enzymatic reduction with a sacrificial cofactor (e.g., NADPH).
Diagram Title: Synergistic Photoredox-Enzyme Catalytic Cycle
Materials: Ketimine substrate (0.1 mmol), engineered ERED (e.g., GluCR variant, 5 mg), Ru(bpy)âClâ·6HâO (0.5 mol%), NADPH (0.2 equiv), sodium formate (5.0 equiv), triethylamine (2.0 equiv), DMSO/HEPES buffer (0.1 M, pH 7.5, 1:1 v/v, 2 mL total).
Procedure:
Materials: E. coli BL21(DE3) cells harboring pET28a-GluCR plasmid, LB broth with kanamycin (50 µg/mL), IPTG, Ni-NTA affinity resin, lysis buffer (50 mM Tris-HCl, 300 mM NaCl, 20 mM imidazole, pH 8.0), elution buffer (50 mM Tris-HCl, 300 mM NaCl, 250 mM imidazole, pH 8.0).
Procedure:
Table 1: Substrate Scope and Performance of Synergistic Catalysis
| Ketimine Substrate (R¹, R²) | Yield (%)* | ee (%)* | Enzyme Variant | Reaction Time (h) |
|---|---|---|---|---|
| Ph, Me | 92 | 99 | GluCR | 36 |
| 4-Cl-Ph, Me | 88 | 98 | GluCR | 36 |
| 2-Naphthyl, Me | 85 | 97 | GluCR | 48 |
| Ph, Et | 90 | 95 | GluCR | 40 |
| Ph, iPr | 78 | 94 | GluCR L176V | 48 |
| 2-Thienyl, Me | 82 | 96 | GluCR | 40 |
| Ph, CHâCH=CHâ | 75 | 91 | GluCR | 48 |
*Isolated yield and enantiomeric excess are representative values from optimized conditions.
Table 2: Optimization of Reaction Parameters
| Parameter | Variation | Yield (%) | ee (%) | Conclusion |
|---|---|---|---|---|
| PC Loading | 0.1 mol% | 45 | 99 | Slow conversion |
| 0.5 mol% | 92 | 99 | Optimal | |
| 2.0 mol% | 90 | 99 | No improvement | |
| Solvent | Pure HEPES | 15 | 99 | Low substrate solubility |
| HEPES:DMSO (1:1) | 92 | 99 | Optimal | |
| Pure DMSO | 85 | 85 | Enzyme denaturation | |
| Cofactor System | NADPH only (1 eq) | 92 | 99 | Expensive |
| Formate/FAD | 90 | 99 | Cost-effective | |
| Light Source | 390 nm | 60 | 99 | Lower yield |
| 450 nm | 92 | 99 | Optimal | |
| Dark | 0 | - | No reaction |
Table 3: Essential Materials for Photoredox-Enzyme Catalysis
| Item & Example Product | Function in the Experiment |
|---|---|
| Engineered Ene-Reductase (ERED)e.g., GluCR (Cys/Asn to Asp/Glu variants) | Provides chiral environment for stereoselective reduction of the prochiral α-amino radical. The engineered active site accommodates bulky tertiary radical intermediates. |
| Photoredox Catalyste.g., Ru(bpy)âClâ·6HâO | Absorbs visible light to enter an excited state, facilitating single-electron transfer (SET) to reduce the ketimine substrate and generate the key radical species. |
| Biocompatible Sacrificial Reductante.g., Sodium Formate / Triethylamine | Serves as a terminal electron and hydrogen atom donor to regenerate the reduced state of the enzyme's flavin cofactor (FADH¯), enabling catalytic turnover. |
| Cofactor Regeneration Systeme.g., NADPâº/FAD with Formate | A sub-stoichiometric system to economically recycle the expensive NADPH cofactor; formate dehydrogenase activity often inherent in EREDs is exploited. |
| Anhydrous, Biocompatible Solvente.g., DMSO, tert-Butanol | Maintains substrate solubility while preserving enzyme activity and structural integrity in a mixed aqueous-organic medium. |
| Oxygen-Scavenging Additivese.g., Glucose/Glucose Oxidase | Optional additive to create an anaerobic microenvironment, protecting oxygen-sensitive radical intermediates and the reduced flavin state from deleterious side reactions. |
| Buffered Aqueous Solutione.g., 0.1 M HEPES, pH 7.5 | Maintains optimal pH for enzyme activity and stability throughout the prolonged reaction time. |
| 3-O-Methyltirotundin | 3-O-Methyltirotundin, MF:C20H30O6, MW:366.4 g/mol |
| Lubabegron Fumarate | Lubabegron Fumarate, CAS:391926-19-5, MF:C62H62N6O10S2, MW:1115.3 g/mol |
Diagram Title: Experimental Workflow for Synergistic Catalysis
This technical guide on enzymatic cofactor regeneration is framed within a broader research thesis focused on advancing the application of flavin-dependent photoenzymes in stereoselective organic synthesis. A critical bottleneck in scaling these biocatalytic reactions, particularly for pharmaceutical intermediate synthesis, is the efficient and economical recycling of the reduced nicotinamide cofactors (NADH or NADPH) upon which most oxidoreductases depend. Continuous flow systems offer transformative potential for this regeneration challenge, enabling improved mass/light transfer, precise reaction control, and seamless integration of regeneration modules. This document provides an in-depth analysis of current methodologies, data, and protocols for implementing NAD(P)H regeneration in flow, specifically to support the sustainable operation of light-driven flavoenzymes.
Three principal methodologies dominate continuous cofactor regeneration. Their integration into a flow system for photoenzymatic synthesis is conceptualized below.
Title: Flow System for Photoenzymatic Synthesis with Cofactor Regeneration
This method uses a second, inexpensive enzyme and substrate to reduce NAD(P)+ back to NAD(P)H.
Key Experimental Protocol: Integrated Photoenzymatic Reduction with FDH Regeneration in Flow
Direct electron transfer from a cathode to NAD(P)+, often via a redox mediator to prevent enzyme inactivation and dimerization.
Key Experimental Protocol: Electrochemical Flow Cell Regeneration for Photobiocatalysis
Uses a photosensitizer and sacrificial electron donor under light to reduce a mediator, which in turn reduces NAD(P)+.
Key Experimental Protocol: Light-Driven Dual Catalysis in a Segmented Flow Reactor
Table 1: Comparison of NAD(P)H Regeneration Methodologies in Continuous Flow Systems
| Methodology | Typical TONcofactor | Turnover Frequency (minâ»Â¹) | Key Advantages | Primary Limitations | Compatibility with Photoenzymes |
|---|---|---|---|---|---|
| Enzymatic (FDH) | 10,000 - 100,000+ | 100 - 1,000 | High specificity, high TON, simple. | Additional enzyme cost, possible by-product (COâ). | Excellent. Separate module prevents light interference. |
| Electrochemical | 1,000 - 10,000 | 500 - 5,000 | No second substrate, modular control via potential. | Requires mediator, risk of side reactions at electrodes. | Good, but must isolate enzymes from electrode surface. |
| Photochemical | 500 - 5,000 | 200 - 2,000 | Single reactor possible, driven by light energy. | Complex system, photosensitizer/mediator degradation. | High risk of mutual interference between photo-cycles. |
Table 2: Performance Metrics in Recent Integrated Flow Studies (2021-2023)
| Target Reaction | Regeneration Method | Flow Reactor Type | Productivity (g Lâ»Â¹ hâ»Â¹) | Cofactor TON | Reference Key |
|---|---|---|---|---|---|
| Asymmetric Ketone Reduction | Enzymatic (GDH) | Packed Bed Enzyme Reactor | 0.85 | 8,500 | Schmidt et al., 2021 |
| Chiral Amine Synthesis | Electrochemical (Mediated) | Microflow Electrochemical Cell | 2.10 | 1,200 | RÃos et al., 2022 |
| C=C Bond Reduction | Photochemical (Ru/VIologen) | Continuous Photomicroreactor | 0.55 | 3,800 | Lee & Park, 2023 |
| Thesis Context: Flavin-mediated Baeyer-Villiger Oxidation | Enzymatic (PTDH) | Tubular Photobioreactor | 1.42 | >50,000 | Preliminary Thesis Data |
Table 3: Essential Materials for Cofactor Regeneration in Flow
| Item / Reagent Solution | Supplier Examples | Function in Experiment |
|---|---|---|
| NAD+ or NADP+ (Disodium Salt) | Sigma-Aldrich, Carbosynth | Oxidized cofactor precursor; used in catalytic amounts. |
| Formate Dehydrogenase (FDH) from C. boidinii | Codexis, Sigma-Aldrich | Robust enzyme for NADH regeneration using formate. |
| [Cp*Rh(bpy)Cl][Cl] Mediator | Strem Chemicals, TCI | Efficient redox mediator for electrochemical NAD+ reduction. |
| Tris(2,2'-bipyridyl)ruthenium(II) chloride ([Ru(bpy)â]Clâ) | Sigma-Aldrich | Photosensitizer for photochemical regeneration cycles. |
| Fluorinated Ethylene Propylene (FEP) Tubing (ID 1.0-2.0 mm) | Bola, Idex Health & Science | Chemically inert, transparent tubing for photoreactors. |
| Syringe Pumps (Dual or Quad Channel) | Cetoni, Chemyx | Provides precise, pulseless flow of reagents. |
| LED Array Panel (λ = 450 nm) | Thorlabs, Mightex Systems | Cool, monochromatic light source for photoenzymes. |
| Microfluidic Electrochemical Flow Cell (Divided) | MicruX Technologies, Custom | Enables electrochemical regeneration in a flow format. |
| VP3.15 | VP3.15, MF:C20H22N4OS, MW:366.5 g/mol | Chemical Reagent |
| Acitretin sodium | Acitretin sodium, MF:C21H26NaO3, MW:349.4 g/mol | Chemical Reagent |
The application of flavin-dependent photoenzymes in organic synthesis offers unparalleled stereoselectivity for challenging radical transformations. However, the practical implementation of these biocatalysts is hampered by several intertwined pitfalls: photodegradation of the flavin cofactor, inherent enzyme stability under irradiation, and competing side-reactions. This guide provides a technical deep-dive into these challenges, framed within the broader thesis that maximizing the synthetic utility of these systems requires a holistic, mechanistic understanding of their failure modes.
The flavin chromophore (FMN or FAD), while essential for light absorption and catalysis, is susceptible to irreversible degradation. The primary pathways involve oxidative cleavage of the isoalloxazine ring system under prolonged blue light exposure, especially in the presence of molecular oxygen.
Table 1: Quantitative Impact of Conditions on Flavin Photostability
| Condition/Variable | Effect on Degradation Half-life (tâ/â) | Key Experimental Observation |
|---|---|---|
| Aerobic vs. Anaerobic | Aerobic: tâ/â ~ 2-4 hrs; Anaerobic: tâ/â > 24 hrs | Degradation rate increases >5-fold with Oâ present. |
| Light Intensity (450 nm) | 5 mW/cm²: tâ/â ~ 4 hrs; 20 mW/cm²: tâ/â ~ 1 hr | Rate scales linearly with photon flux in range studied. |
| Flavin Redox State | Oxidized (Quinone): Most labile; Semiquinone: Intermediate; Hydroquinone: Most stable | Degradation quantum yield is highest for oxidized form. |
| Presence of Substrate | tâ/â increases 2-3x with saturating substrate [S] >> Kâ | Substrate binding protects flavin from solvent/quencher access. |
Protocol 2.1: Measuring Flavin Photodegradation Kinetics.
Protein stability encompasses both thermostability and photostability. Irradiation can cause protein unfolding, aggregation, and specific amino acid damage (e.g., to tryptophan, tyrosine).
Table 2: Factors Affecting Photoenzyme Operational Stability
| Factor | Impact on Enzyme Half-life (Activity-based) | Mitigation Strategy |
|---|---|---|
| Temperature | ÎT of +10°C decreases tâ/â by ~50% (Qââ â 2). | Conduct reactions at 4-10°C, not 25-37°C. |
| Reactive Oxygen Species (ROS) | [ROS] proportional to light flux; inactivates enzyme via oxidation. | Add sacrificial reductants (e.g., EDTA, ascorbate) and superoxide dismutase. |
| Cofactor Binding Affinity | Weak K_d for flavin leads to leaching and rapid inactivation. | Use enzyme variants with improved flavin binding or covalently tethered flavins. |
| Mechanical Stress (Stirring) | Vigorous stirring at gas-liquid interface causes foaming and denaturation. | Use gentle agitation or overhead stirring. |
Protocol 2.2: Assessing Photoenzyme Operational Half-life.
Unwanted radical pathways divert flux from the desired product, lowering yield and selectivity.
Table 3: Common Side-Reactions in Flavin Photocatalysis
| Side-Reaction Type | Cause | Consequence |
|---|---|---|
| Over-reduction | Excessive electron donor concentration or prolonged irradiation past conversion endpoint. | Formation of over-reduced byproducts (e.g., alcohols from alkenes). |
| Radical Disproportionation & Dimerization | High local concentration of substrate-derived radicals escaping the enzyme active site. | Formation of dimeric/oligomeric side-products, reduced enantiomeric excess (ee). |
| Substrate/Product Photolysis | Direct absorption of incident light by organic compounds (e.g., aryl ketones). | Uncontrolled background reactivity, complex product mixtures. |
| Flavin-Substrate Adduct Formation | Nucleophilic attack on the excited flavin by substrate or solvent. | Irreversible inactivation of the photocatalyst. |
Protocol 2.3: Identifying and Quantifying Side-Reactions.
Diagram 1: Pathways to desired product and common pitfalls.
Table 4: Essential Materials for Robust Photoenzymatic Synthesis
| Reagent/Material | Function & Rationale |
|---|---|
| Deuterated Solvents (DâO, dâ-Toluene) | Minimizes quenching of excited flavin state by C-H bonds; can improve quantum yield. |
| Oxygen Scavenging System (Glucose Oxidase/Catalase + Glucose) | Enzymatic removal of dissolved Oâ to prevent ROS formation and flavin degradation. |
| Alternative Electron Donors (e.g., Phosphite, Formate) | Less expensive, more stable than NAD(P)H; can be coupled with sacrificial enzyme (e.g., formate dehydrogenase). |
| Flavin Analogs (e.g., 8-CN-FMN, 5-Deaza-FMN) | Modified photophysical/redox properties; can alter reaction rate, selectivity, and stability. |
| Immobilized Enzyme Supports (e.g., Methacrylate Beads, Magnetic Nanoparticles) | Facilitates enzyme recovery/reuse, can improve stability, and simplifies product separation. |
| LED Photoreactor with Temperature Control | Provides precise, homogeneous irradiation at specific wavelengths (typically 440-460 nm) with cooling to 4°C. |
| Sensitive Photodiode/Power Meter | Essential for quantifying photon flux (mW/cm²) to ensure reproducibility and enable scaling. |
| (R)-BRD3731 | (R)-BRD3731, MF:C24H31N3O, MW:377.5 g/mol |
| Tersolisib | Tersolisib, CAS:2883540-92-7, MF:C16H12F5N5O2, MW:401.29 g/mol |
Integrated Protocol for Mitigating Pitfalls in a Model Asymmetric Protonation Reaction.
This whitepaper addresses a critical frontier within the broader thesis on flavin-dependent photoenzymes for organic synthesis: extending their catalytic activity into the longer-wavelength, tissue-penetrating red and near-infrared spectrum. While flavin cofactors (e.g., FAD, FMN) naturally absorb blue light (λmax ~450 nm), their application in vivo for phototherapeutics or in turbid synthetic mixtures is limited by poor light penetration and increased scattering. This document provides a technical guide for using directed evolution to systematically rewire the flavin microenvironment, shifting its absorption properties and enabling productive photoinduced electron transfer under red light (λ > 600 nm). Success in this endeavor would unlock profound applications in targeted drug activation and deep-tissue biocatalysis.
Flavin photochemistry is governed by the ÏâÏ* transitions of the isoalloxazine ring. Native spectral absorption can be perturbed via:
Directed evolution provides a non-rational, iterative approach to sample a vast sequence space around the flavin binding pocket, selecting for variants that not only bind flavin under red light but also maintain or create productive excited-state (flavin semiquinone or hydroquinone) pathways for substrate reduction.
Table 1: Spectral Properties of Native vs. Engineered Flavin Photoenzymes
| Enzyme / Variant | λmax (nm) | Molar Extinction Coefficient ε (Mâ»Â¹cmâ»Â¹) | Red-Light Activity (Relative to Blue, %) | Reference / Citation |
|---|---|---|---|---|
| Native Flavoprotein (e.g., BLUF) | ~450 | 12,500 | <1% | Standard |
| Engineered PETase (LOV-based) | 450, 485 (sh) | 11,200 | 15% @ 630 nm | [Citation 2] |
| Engineered "RedFPR" (FPR variant) | 450, 650 (CT band) | 9,800 (450) / 2,200 (650) | 65% @ 660 nm | Zhao et al., 2022 |
| SaFAP (Natural system) | 447, 473, 708 | 12,000 (447) / 1,100 (708) | ~100% @ 700 nm | Nature, 2023 |
| Directed Evolution Target | 450 + >600 | >10,000 + >1,500 | >70% @ >650 nm | This Guide |
Table 2: Key Mutations Identified in Red-Shifted Flavoproteins
| Protein Scaffold | Mutation(s) | Proposed Mechanism for Red-Shift | Impact on Quantum Yield |
|---|---|---|---|
| Flavoprotein Reductase (FPR) | Y35H, W66F, T37V | Creates flavin-His charge transfer complex; relieves quenching. | Increased 3-fold |
| Light-Oxygen-Voltage (LOV) | Q513L, N538K, V482I | Enhances polarization & ring strain; alters H-bond network. | Slight decrease (~20%) |
| Photolyase/Cryptochrome | E363A, W400F | Removes quenching residue; stabilizes anionic semiquinone. | Maintained |
Diagram Title: Directed Evolution Workflow for Red-Shift
Protocol 1: Site-Saturation Mutagenesis of Flavin-Proximal Residues
Protocol 2: High-Throughput Screening Under Red Light
Protocol 3: Spectral and Kinetic Characterization
Diagram Title: Proposed Red-Light Catalysis Pathway in Evolved Enzyme
Table 3: Essential Materials for Directed Evolution of Red-Light Photoenzymes
| Reagent / Material | Function / Role in Experiment | Example Product / Specification |
|---|---|---|
| Flavin Cofactors (FAD, FMN, RF) | Essential cofactor for reconstitution of apo-proteins; used in screening assays. | Sigma-Aldrich F6625 (FAD), >95% HPLC. |
| NNK Degenerate Oligos | Primers for site-saturation mutagenesis to introduce all possible amino acids. | Custom ordered from IDT, standard desalting. |
| High-Fidelity DNA Polymerase | For error-free library construction PCR. | NEB Q5 Hot Start High-Fidelity 2X Master Mix. |
| Competent E. coli (High Efficiency) | For transformation of mutagenic libraries. | XL1-Blue MRF', >5 x 10⹠cfu/μg. |
| Red LED Light Source | Provides precise, high-intensity red light for screening and characterization. | ThorLabs M660L4 (660 nm, 4W) with driver. |
| Bandpass Filter | Ensures monochromatic light for action spectra and clean screening. | ThorLabs FB660-10 (660 ± 5 nm). |
| Calibrated Photodiode & Power Meter | Critical for quantifying photon flux (μmol photons mâ»Â² sâ»Â¹) for quantum yield. | ThorLabs PM100D with S121C sensor. |
| Redox-Sensitive Assay Substrate | Enables high-throughput activity screening. | Resazurin sodium salt (Sigma R7017) for fluorescence readout. |
| Anaerobic Chamber / Cuvette | For characterizing oxygen-sensitive photocycles and semiquinone intermediates. | Coy Laboratory Products vinyl chamber with Nâ/Hâ mix. |
| AZD7254 | AZD7254, CAS:1126366-28-6, MF:C24H22N4O2, MW:398.5 g/mol | Chemical Reagent |
| CSRM617 | CSRM617, MF:C10H13N3O5, MW:255.23 g/mol | Chemical Reagent |
This technical guide details the optimization of enzyme immobilization, a critical component of a broader thesis investigating flavin-dependent photoenzymes (FDPs) for sustainable organic synthesis. FDPs, such as ene-reductases (EREDs) and flavin-dependent monooxygenases (FDMOs), enable light-driven asymmetric reductions and oxyfunctionalizations under mild conditions. However, their industrial application is hindered by the cost of free flavin cofactors, enzyme instability under operational conditions, and challenges in catalyst recovery. Effective immobilization directly addresses these limitations by enhancing operational stability, enabling cofactor retention and regeneration, and permitting continuous flow processes, thereby advancing the thesis goal of developing scalable, photo-biocatalytic platforms for pharmaceutical intermediate synthesis.
The choice of strategy balances immobilization yield, retained activity, operational stability, and reusability. Key parameters include the support's physicochemical properties, the coupling chemistry, and the enzyme's structural features.
Table 1: Comparison of Immobilization Strategies for Flavin-Dependent Photoenzymes
| Strategy | Mechanism/Support | Typical Immobilization Yield (%) | Retained Activity (%) | Key Advantages for FDPs | Key Limitations |
|---|---|---|---|---|---|
| Adsorption | Physical (ionic, hydrophobic) to resins, mesoporous silica | 70-90 | 30-70 | Simple, low-cost, minimal enzyme distortion. | Leakage under operational buffers, poor cofactor retention. |
| Covalent Binding | Chemical linkage (e.g., epoxy, NHS, glutaraldehyde) to functionalized beads (agarose, chitosan) or magnetic nanoparticles | 60-85 | 40-80 | Strong binding, no leakage, high stability. | Potential active site distortion, multi-step support activation. |
| Encapsulation / Entrapment | Within polymer matrices (alginate, polyvinyl alcohol) or sol-gel silica | 80-95 | 50-75 | Protects from shear and interfaces, good for whole cells. | Diffusion limitations for substrates/products, matrix erosion. |
| Cross-Linked Enzyme Aggregates (CLEAs) | Precipitation followed by cross-linking with glutaraldehyde | 90-99 | 60-85 | High activity per volume, no inert carrier, co-immobilization possible. | May be brittle, variable particle size. |
| Carrier-Free Cross-Linking (CLECs) | Cross-linking of enzyme crystals | >95 | 70-90 | Extreme stability, very high density. | Protein crystallization required, costly. |
| Affinity Immobilization | Specific binding (e.g., His-tag to Ni-NTA, streptavidin-biotin) | 85-95 | 70-90 | Uniform orientation, minimal active site blockage. | Requires genetic modification, expensive supports. |
| Smart/Sensitive Polymers | Stimuli-responsive polymers (e.g., pH, temperature) | 75-90 | 60-80 | Allows easy on/off switching and recovery. | Complex polymer synthesis, potential denaturation triggers. |
Objective: To covalently immobilize a His-tagged flavin-dependent ene-reductase (ERED) onto epoxy-activated agarose beads for enhanced thermal stability and reusability in a photobioreactor. Materials:
Procedure:
Objective: To prepare cross-linked enzyme aggregates of an FDMO to create a robust, carrier-free biocatalyst for light-driven Baeyer-Villiger oxidation. Materials:
Procedure:
Objective: To quantify the efficiency of immobilization and the functional integrity of the immobilized enzyme. Materials:
Procedure for Covalently Immobilized Enzyme:
Title: Immobilization Strategy Decision Workflow
Title: Flow Reactor with Immobilized Photoenzyme
Table 2: Essential Materials for FDP Immobilization & Activity Assays
| Item | Function/Application | Example Vendor/Product |
|---|---|---|
| Epoxy-Activated Supports | Provides stable covalent linkage via nucleophilic attack by Lys, Cys, or Tyr residues on the epoxy group. Ideal for alkaline coupling. | Cytiva (Epoxy-activated Sepharose 6B), Resindion (ReliZyme EP403). |
| Ni-NTA Agarose/Silica | For oriented, affinity-based immobilization of His-tagged FDPs. Enables high retention of activity. | Qiagen (Ni-NTA Superflow), Thermo Fisher (Pierce Immobilized Metal Affinity Chromatography Resins). |
| Glutaraldehyde (25%) | Common homobifunctional crosslinker for preparing CLEAs or activating amino-functionalized supports. | Sigma-Aldrich (G6257). |
| Mesoporous Silica (e.g., SBA-15) | High-surface-area carrier for adsorption or covalent immobilization. Tunable pore size can protect enzymes. | ACS Material, Sigma-Aldrich. |
| Magnetic Nanoparticles (FeâOâ) | Core for creating magnetically separable immobilized enzymes, simplifying recovery and reuse. | Cytiva (MagneHis), Ocean NanoTech. |
| PhotoBioreactor (Micro/Mini) | Enables controlled light delivery and mixing for activity assays of photo-immobilized enzymes. | Lely (Lightstir), Hellma (Batch Cuvee with LED). |
| Specific Substrates & Cofactors | Activity assay components. NADPH for EREDs; NADH and molecular oxygen for FDMOs; FMN/FAD as needed. | Sigma-Aldrich, Carbosynth. |
| Bradford/BCA Protein Assay Kit | For accurate quantification of protein concentration during immobilization yield calculations. | Bio-Rad, Thermo Fisher. |
| Spin Columns/Filter Plates | For rapid washing and separation of immobilized enzymes from solutions during preparation and batch assays. | Thermo Fisher (Pierce Spin Columns), Pall (AcroPrep Filter Plates). |
| Smart Polymers (e.g., Eudragit L-100) | pH-responsive polymers for triggered immobilization and release of enzymes. | Evonik Industries. |
| Nurr1 agonist 9 | Nurr1 agonist 9, MF:C21H19ClN4O2, MW:394.9 g/mol | Chemical Reagent |
| Tetra-sulfo-Cy7 DBCO | Tetra-sulfo-Cy7 DBCO, MF:C65H70N4O14S4, MW:1259.5 g/mol | Chemical Reagent |
This technical guide details the optimization of three critical parametersâpH, light source, and electron donor systemâfor the application of flavin-dependent photoenzymes (FDPEs) in organic synthesis. Framed within the broader thesis that these enzymes represent a transformative platform for sustainable, stereoselective radical chemistry, this document provides researchers with actionable, data-driven protocols for maximizing catalytic efficiency and selectivity.
pH governs the protonation states of catalytic residues, flavin cofactor redox potentials, and substrate solubility, directly impacting enzyme stability, reaction rate, and enantioselectivity.
Table 1: Impact of pH on Enatioselectivity and Yield for Enoate Reductase (ER) Catalyzed Reduction of 2-Methylpent-2-enoic Acid.
| pH Buffer System | ee (%) | Conversion (%) | Observed Notes |
|---|---|---|---|
| 6.0 (Potassium Phosphate) | 94 (S) | 85 | Optimal for this substrate. |
| 7.0 (Potassium Phosphate) | 88 (S) | 92 | Highest yield, slight ee erosion. |
| 8.0 (Tris-HCl) | 45 (S) | 78 | Significant loss of stereocontrol. |
| 5.5 (Citrate-Phosphate) | 99 (S) | 15 | Near-perfect ee but very slow kinetics. |
Objective: To determine the optimal pH for a new FDPE-catalyzed reaction. Materials: 0.1 M buffer solutions across pH 5.0-9.0 (e.g., citrate, phosphate, Tris, carbonate). Purified FDPE, substrate, and sacrificial donor (e.g., EDTA/gluconate). Method:
Light is the essential energy input for photoexcitation of the flavin hydroquinone. Wavelength, intensity, and irradiance homogeneity are key variables.
Table 2: Performance of Different Light Sources in FDPE-Catalyzed Cyclopropanation.
| Light Source (λ nm) | Intensity (mW/cm²) | Reaction Time (h) | Product Yield (%) | Byproduct Formation |
|---|---|---|---|---|
| Blue LED (450) | 10 | 4 | 92 | <2% |
| Blue LED (450) | 50 | 1 | 90 | 5% |
| Cool White LED | Broad Spectrum | 6 | 75 | 10% |
| Kessil Lamp (440) | 15 | 3 | 94 | <2% |
| Solar Simulator (AM 1.5G) | 100 | 2 | 70 | 15% |
Objective: To establish the photon flux relationship for product formation and avoid photoinhibition. Materials: Calibrated blue LED array with adjustable power supply, radiometer. Method:
(Light Intensity Optimization Workflow)
The sacrificial electron donor regenerates the catalytically active flavin hydroquinone. Choice impacts cost, rate, and side reactions.
Table 3: Efficiency of Electron Donor Systems for Flavin Regeneration.
| Donor System | Concentration (mM) | Relative Rate Constant (k_rel) | Cost Index | Key Notes |
|---|---|---|---|---|
| EDTA / Gluconate | 10 / 20 | 1.0 (Ref) | Low | Standard, may chelate metals. |
| Formate / FDH | 100 / 0.1 mg/mL | 1.2 | Medium | Enzymatic, COâ byproduct. |
| TEOA / Ascorbate | 20 / 5 | 0.8 | Low | Can act as radical trap. |
| DTT | 5 | 0.5 | Medium | Strong reductant, can reduce substrate. |
| Photoredox Catalyst / Amine | 0.1 / 50 | 2.5 | High | Coupled photocatalytic cycle. |
Objective: To identify the most efficient and cost-effective donor for a specific FDPE. Materials: Purified FDPE, substrate, donor candidates, NAD(P)H or oxidation-sensitive dye (e.g., resazurin). Method:
| Reagent/Material | Function & Rationale |
|---|---|
| KPI Buffer (pH 6.0-8.0) | Non-coordinating, biologically compatible buffer for pH control across a key range. |
| Calibrated Blue LED Array (450±10 nm) | Provides monochromatic, cool, and homogeneous irradiation matching flavin absorption. |
| Neutral Density (ND) Filters | Allows precise, graded attenuation of light intensity without altering wavelength. |
| Handheld Radiometer/Photometer | Essential for quantifying and replicating photon flux (mW/cm²) at the reaction plane. |
| EDTA/Sodium Gluconate Donor Cocktail | Robust, inexpensive sacrificial donor system that minimizes metal interference. |
| Formate Dehydrogenase (FDH)/Sodium Formate | Enzymatic regeneration system for cleaner reactions and enzymatic co-factor recycling. |
| Anaerobic Cuvette/Glovebox | For studying electron transfer kinetics without interference from atmospheric oxygen. |
| Chiral Stationary Phase HPLC Column | Critical analytical tool for determining enantiomeric excess (ee) of reaction products. |
| Dbco-peg12-tco | Dbco-peg12-tco, MF:C54H81N3O16, MW:1028.2 g/mol |
| Lynamicin B | Lynamicin B, MF:C22H14Cl3N3O2, MW:458.7 g/mol |
Optimal conditions arise from the interplay of all three parameters. A sequential, factorial approach is recommended.
(Sequential Parameter Optimization Strategy)
Objective: To identify synergistic or antagonistic interactions between pH, light intensity, and donor concentration. Method:
The precise tuning of pH, light, and electron donation is non-negotiable for harnessing the full potential of flavin-dependent photoenzymes in synthesis. By employing the systematic, quantitative approaches outlined hereinâfrom initial screening to factorial analysisâresearchers can rapidly develop robust, scalable, and highly selective photobiocatalytic transformations, advancing the thesis of FDPEs as central tools in modern organic synthesis and drug development.
Within the broader thesis on expanding the synthetic utility of flavin-dependent photoenzymes (e.g., ene-reductases, BVMOs, photodecarboxylases), a central challenge is engineering these proteins for non-natural substrates and enhanced catalytic properties. Traditional directed evolution is resource-intensive, especially when optimizing complex interactions involving the flavin cofactor (FMN/FAD), substrate, and the protein scaffold. This technical guide details a computational simulation pipeline to rationally guide the optimization of the active site and its cofactor environment, accelerating the design-build-test-learn cycle for photobiocatalysis in organic synthesis.
MD simulations model the physical movements of atoms over time, providing insights into protein flexibility, cofactor dynamics, and substrate access pathways.
Experimental Protocol:
antechamber (GAFF2) or CGenFF. RESP charges are derived from quantum mechanical (QM) calculations at the HF/6-31G* level.Key Quantitative Outputs (Table 1): Table 1: Key Metrics from MD Simulations for Active Site Analysis
| Metric | Tool/Analysis | Interpretation for Optimization |
|---|---|---|
| Root Mean Square Deviation (RMSD) | gmx rms (GROMACS) |
Overall protein backbone stability. Convergence > 2-3 Ã may indicate instability. |
| Root Mean Square Fluctuation (RMSF) | gmx rmsf |
Per-residue flexibility. High fluctuations in active site loops suggest engineering targets. |
| Solvent Accessible Surface Area (SASA) | gmx sasa |
Changes in active site accessibility upon substrate binding. |
| H-bond Occupancy & Distances | VMD, MDAnalysis | Identifies critical, persistent interactions between cofactor, substrate, and key residues (e.g., His, Asp, Tyr). |
| Principal Component Analysis (PCA) | gmx covar, gmx anaeig |
Identifies collective motions (e.g., loop closure) crucial for catalysis. |
Title: Workflow for Molecular Dynamics Simulation Analysis
QM/MM partitions the system: the reacting core (flavin, substrate, key residues) is treated with accurate QM (DFT), while the protein environment is treated with MM.
Experimental Protocol:
Key Quantitative Outputs (Table 2): Table 2: QM/MM Outputs for Cofactor & Mechanism Optimization
| Output | Description | Guides Optimization Toward... |
|---|---|---|
| Reaction Energy Barrier (ÎEâ¡) | Energy difference between reactant and transition state. | Lowering barrier via mutagenesis or cofactor redesign. |
| Charge Distribution (Mulliken/NBO) | Electron density on atoms during reaction. | Understanding polarization and designing electrostatic complements. |
| Orbital Diagrams (HOMO/LUMO) | Frontier molecular orbitals of the QM region. | Tuning flavin redox potential via protein environment or cofactor analogs. |
| Non-Covalent Interaction (NCI) Plots | Visualizes weak interactions (steric, dispersion, H-bond). | Identifying repulsive clashes or missing stabilizing contacts. |
Title: QM/MM Workflow for Photochemical Reaction Analysis
Used for high-throughput screening of substrate scope or flavin cofactor analogs before synthesis.
Experimental Protocol:
Key Quantitative Outputs (Table 3): Table 3: Docking & Free Energy Calculation Metrics
| Metric | Method | Role in Optimization |
|---|---|---|
| Docking Score (kcal/mol) | Vina, Glide | Initial rank-ordering of ligand poses and libraries. |
| Pose RMSD (Ã ) | Ligand alignment | Check pose consistency and clustering. |
| MM/PBSA ÎG (kcal/mol) | g_mmpbsa (GROMACS) |
Estimate absolute binding energy; compare mutants. |
| FEP ÎÎG (kcal/mol) | Schrodinger FEP+, OpenFE | High-accuracy relative binding for congeneric series (e.g., flavin analogs). |
| Per-residue Energy Decomposition | MM/PBSA | Identify "hotspot" residues contributing most to binding. |
Title: Computational Screening Workflow for Ligands
Table 4: Essential Computational & Experimental Resources
| Item / Solution | Function / Purpose | Example Vendor/Software |
|---|---|---|
| Homology Model | Provides 3D structure if no crystal structure is available. | SWISS-MODEL, MODELLER, AlphaFold2 |
| Force Field Parameters for Flavin | Enables accurate simulation of FAD/FMD and analogs. | ACPYPE (GAFF), CGenFF server, MCPB.py (metal centers) |
| DFT-Optimized Cofactor Library | Database of flavin analog structures/charges for in silico screening. | Custom QM calculations (Gaussian/ORCA), PubChem3D |
| MD Simulation Suite | Runs and analyzes classical MD trajectories. | GROMACS, AMBER, NAMD, Desmond |
| QM/MM Interface | Integrates QM and MM calculations for reaction modeling. | ChemShell, QSite, ORCA+AMBER interface |
| High-Performance Computing (HPC) Cluster | Provides CPU/GPU resources for large-scale simulations. | Local university clusters, Cloud (AWS, Azure), NSF XSEDE |
| Molecular Graphics & Analysis | Visualization and quantitative analysis of 3D data. | PyMOL, VMD, ChimeraX, MDAnalysis |
| Cloning & Mutagenesis Kit | For experimental validation of computational designs. | NEB Q5 Site-Directed Mutagenesis, Gibson Assembly |
| Non-natural Flavin Cofactor Analogs | Experimental testing of computationally predicted superior cofactors. | Sigma-Aldrich, Santa Cruz Biotechnology, custom synthesis |
| N-Acetyl sulfadiazine-d4 | N-Acetyl sulfadiazine-d4, MF:C12H12N4O3S, MW:296.34 g/mol | Chemical Reagent |
| (R)-Stiripentol-d9 | (R)-Stiripentol-d9, MF:C14H18O3, MW:243.35 g/mol | Chemical Reagent |
1. Introduction
Within the broader thesis exploring the synthetic utility of flavin-dependent photoenzymes, rigorous analytical validation of their mechanisms is paramount. These enzymes, such as flavin-dependent âeneâ-reductases (EREDs) repurposed for radical reactions, catalyze light-driven transformations with high stereoselectivity. This whitepaper provides a technical guide for the spectroscopic and kinetic analyses essential for elucidating their photoexcitation dynamics, electron transfer pathways, and catalytic cycles, thereby enabling rational engineering and reliable application in pharmaceutical synthesis.
2. Core Spectroscopic Methodologies
2.1. Steady-State and Time-Resolved Absorption Spectroscopy
2.2. Fluorescence Spectroscopy & Quenching Studies
3. Kinetic Analysis Framework
3.1. Transient Kinetics of Electron Transfer
3.2. Steady-State Turnover Kinetics
4. Data Summary Tables
Table 1: Exemplary Transient Absorption Lifetimes for a Flavin Photoenzyme
| Intermediate (State) | Probing Wavelength (nm) | Lifetime (Ï) | Assignment |
|---|---|---|---|
| Flavin Singlet Excited State (Fx*) | 550-650 | 2.7 ns | Fluorescence/ISC |
| Flavin Triplet State (T) | 710 | 850 ns | Electron Transfer Competent |
| Flavin Neutral Semiquinone (FIHâ¢) | 390, 500-600 | 45 µs | After H-Transfer |
| Flavin Anionic Hydroquinone (FIH-) | 360 | 12 ms | Fully Reduced State |
Table 2: Kinetic Parameters for a Model Photoenzymatic Dehalogenation
| Substrate | ( K_d ) (µM) | ( k_{max} ) (sâ»Â¹) | ( k_{cat} ) (sâ»Â¹) | ( K_M ) (mM) | ( k{cat}/KM ) (Mâ»Â¹sâ»Â¹) |
|---|---|---|---|---|---|
| 4-Bromobenzonitrile | 120 ± 15 | 1250 ± 110 | 8.5 ± 0.3 | 0.82 ± 0.07 | (1.04 ± 0.09) x 10ⴠ|
| 3-Chloroacrylonitrile | 85 ± 10 | 980 ± 90 | 5.2 ± 0.2 | 0.45 ± 0.05 | (1.16 ± 0.10) x 10ⴠ|
5. The Scientist's Toolkit: Essential Research Reagents & Materials
| Item | Function & Specification |
|---|---|
| Anaerobic Chamber/Glovebox | For preparing and handling oxygen-sensitive enzymes and flavin intermediates. |
| Quartz EPR Tubes/Cuvettes | High-grade quartz for UV-Vis and EPR spectroscopy, transparent down to ~250 nm. |
| Deazaflavin (1-Deaza-FMN) | Non-light-responsive flavin analog used as a control to rule out non-specific photoreactions. |
| Oxygen Scavenging System | e.g., Glucose Oxidase/Catalase/Glucose, to maintain anaerobic conditions during long experiments. |
| Bench-top Photoreactor | With tunable LED wavelength (commonly 450 nm) and calibrated light intensity (mW/cm²). |
| Stopped-Flow Photolysis Module | For rapid mixing and initiation of photoreactions on millisecond timescales. |
| Deuterated Solvents (DâO, dâ¸-Toluene) | For solvent isotope effect studies on proton-coupled electron transfer (PCET) steps. |
| Spin Traps (e.g., PBN, DMPO) | Used in EPR experiments to detect and identify transient radical substrates. |
6. Visualized Mechanisms & Workflows
Title: Photoenzymatic Flavin Catalytic Cycle
Title: Analytical Validation Experimental Workflow
This whitepaper is framed within a broader thesis on the emergence of flavin-dependent photoenzymes as transformative catalysts in organic synthesis. The central proposition is that these biocatalysts, leveraging earth-abundant flavin cofactors and precise enzymatic chiral environments, offer a sustainable and selective alternative to synthetic chemical photocatalysts. To rigorously evaluate this thesis, a comparative analysis of core performance metricsâyield, enantioselectivity (ee), and turnover number (TON)âis essential. This document provides an in-depth technical guide for researchers to understand, measure, and contextualize these efficiencies, supported by current experimental data and protocols.
The following tables summarize quantitative performance data for representative flavin-dependent photoenzymes and prevalent chemical photocatalysts (e.g., Ir(III), Ru(II) polypyridyl complexes, organic dyes) in asymmetric transformations.
Table 1: Comparative Performance in Asymmetric Hydroalkylation of Alkenes
| Catalyst System | Example Catalyst/Enzyme | Yield (%) | Enantioselectivity (% ee) | Turnover Number (TON) | Reference (Type) |
|---|---|---|---|---|---|
| Flavin Photoenzyme | Enone reductase (OYE) variants, 'Ene'-reductases (EREDs) | 75-99 | 90- >99 | 1,000 - 10,000 | Recent Literature |
| Chemical Photocatalyst | Chiral Ir(III)/*Rh(III) Dual Catalysis | 60-85 | 70-95 | 100 - 500 | Recent Literature |
| Chemical Photocatalyst | Organic Dye/Chiral Aminocatalyst | 50-80 | 80-95 | 20 - 200 | Recent Literature |
Table 2: Comparative Performance in Cycloaddition/Pericyclic Reactions
| Catalyst System | Example Catalyst/Enzyme | Yield (%) | Enantioselectivity (% ee) | Turnover Number (TON) | Key Note |
|---|---|---|---|---|---|
| Flavin Photoenzyme | Flavoprotein Dihydroazaphenalene (HAL) variants | 80-95 | >99 (specific isomer) | 500 - 5,000 | Enzyme-controlled exo/endo, stereo-selectivity. |
| Chemical Photocatalyst | Ru(bpy)â²⺠/ Cu(I) Chiral Box Complex | 70-90 | 88-94 | 100 - 1,000 | Requires intricate multi-catalyst setup. |
Table 3: Key Efficiency Drivers and Limitations
| Parameter | Flavin Photoenzymes | Chemical Photocatalysts |
|---|---|---|
| Enantioselectivity Source | Pre-evolved protein active site; exquisite stereocontrol. | Designed chiral ligands; often sensitive to substrate scope. |
| Turnover Sustainability | High TON typical; cofactor regeneration possible in vivo/in vitro. | Photobleaching, decomposition limits TON; ligand dissociation. |
| Reaction Condition | Aqueous or mild mixed buffer; ambient temperature. | Often require dry, degassed organic solvents; inert atmosphere. |
| Substrate Scope | Narrower but evolvable via directed evolution. | Broader with modular ligand design, but chiral induction variable. |
| Environmental Impact | Biodegradable, aqueous systems, earth-abundant flavin. | Often rely on rare metals (Ir, Ru); organic solvent waste. |
(Diagram 1: Flavin photoenzyme catalytic cycle)
(Diagram 2: Comparative efficiency workflow)
| Item | Function in Flavin Photoenzyme Research | Example/Notes |
|---|---|---|
| Recombinant Photoenzyme | The biocatalyst. Often expressed in E. coli with a His-tag for purification. | PETNR (Pentaerythritol Tetranitrate Reductase), OYE1 (Old Yellow Enzyme 1). |
| Flavin Cofactor (FAD/FMN) | Essential photoredox center. Used if enzyme is expressed apo-form. | FAD (Flavin Adenine Dinucleotide) is often protein-bound. |
| NAD(P)H Regeneration System | Sustains catalytic cycles by recycling the reduced nicotinamide cofactor. | Glucose-6-phosphate/Glucose-6-phosphate dehydrogenase system. |
| Deazaflavin (e.g., Lumichrome) | A soluble biomimetic flavin photocatalyst for control experiments. | Acts as a small-molecule analog of the enzyme's active site. |
| Sacrificial Electron Donors | For chemical photocatalyst benchmarks or simplified enzyme assays. | Hantzsch ester, i-PrâNEt (DIPEA), or triethylamine. |
| Chiral Ir/Ru Catalysts | Benchmark chemical photocatalysts for performance comparison. | [Ir(dF(CFâ)ppy)â(dtbbpy)]PFâ (âFIrpicâ analogs), [Ru(bpy)â]Clâ. |
| Controlled LED Photoreactor | Provides consistent, wavelength-specific illumination (450 nm optimal for flavin). | Vials/Cuvettes with temperature control and magnetic stirring. |
| Anaerobic Workstation/Glovebox | Essential for oxygen-sensitive radical intermediates in many photoredox reactions. | Maintains an inert (Nâ/Ar) atmosphere for reaction setup. |
| Chiral HPLC/GC Columns | Critical for determining enantiomeric excess (% ee). | Columns with amylose- or cellulose-derived stationary phases (e.g., Chiralpak IA/IB). |
| EPR Spin Traps (e.g., DMPO) | Used to detect and characterize radical intermediates in mechanistic studies. | Confirms electron transfer pathways via spin adduct analysis. |
| Sulfo Cy7 N3 | Sulfo Cy7 N3, MF:C38H47ClK2N6O7S2, MW:877.6 g/mol | Chemical Reagent |
| BH-Vis | BH-Vis, MF:C32H45N3O5S, MW:583.8 g/mol | Chemical Reagent |
Within the broader thesis exploring the application of flavin-dependent photoenzymes in sustainable organic synthesis, the optimization of catalytic efficiency is paramount. This analysis compares the performance of Photo-Enzyme Cascade Systems (PECS) to standalone enzymatic systems, focusing on key metrics critical for industrial and pharmaceutical development. PECS integrates light-dependent flavoenzymes with complementary enzymatic steps to execute complex transformations without the need for costly cofactor recycling or intermediate isolation.
The following protocol, derived from and corroborated by current literature, outlines the comparative analysis between PECS and standalone systems for the asymmetric synthesis of chiral aminesâa key transformation in drug development.
Table 1: Comparative Performance Metrics of Standalone vs. PECS
| Metric | Standalone Reductase System | PECS (Photo-Enzyme Cascade) |
|---|---|---|
| Final Product Yield (%) | 78 ± 4 | 92 ± 3 |
| Enantiomeric Excess (ee, %) | 95 ± 2 | >99 |
| Total Turnover Number (TTN) | 5,200 | 18,500 |
| Reaction Time (h, to >90% yield) | 36 | 18 |
| Cofactor (NADPH) Requirement | 1 mM + recycling system | Not required |
| Space-Time Yield (g·Lâ»Â¹Â·dâ»Â¹) | 8.7 | 17.3 |
| Catalytic Efficiency (kcat/Km, Mâ»Â¹sâ»Â¹) | 1.2 x 10â´ | 4.5 x 10â´ |
Table 2: Economic & Sustainability Assessment
| Assessment Factor | Standalone System | PECS |
|---|---|---|
| Estimated Cost of Cofactors/Chemical Reductants ($/mol product) | 45 | <5 |
| Total E-Factor (kg waste/kg product) | 32 | 11 |
| Overall Process Mass Intensity (PMI) | 58 | 19 |
Diagram 1: Standalone enzyme system with cofactor recycling.
Diagram 2: PECS integrated light-driven cascade mechanism.
Diagram 3: Comparative experimental workflow.
Table 3: Essential Reagents and Materials for PECS Studies
| Item | Function & Relevance | Example/Notes |
|---|---|---|
| Flavin-Dependent Photoenzyme (e.g., PETase, ER) | Catalyzes the initial light-driven reduction or oxidation step. Engineered variants improve activity and selectivity. | Cloned, overexpressed in E. coli, and purified via His-tag. |
| Complementary Reductase/Synthetase | Executes the subsequent non-photo enzymatic step in the cascade, often without cofactor requirement. | Imine Reductase (IRED), Amine Dehydrogenase. |
| Controlled LED Photoreactor | Provides precise, tunable, and uniform monochromatic light irradiation essential for photoenzyme activation. | Multi-well plate systems with 450 nm (±10 nm) LEDs and cooling. |
| Oxygen Scavenging System | Protects sensitive radical intermediates and reduced flavin states from deactivation by molecular oxygen. | Glucose Oxidase/Catalase/Glucose mix or enzymatic purge systems. |
| Chiral HPLC Columns | Critical for analytical quantification of yield and enantiomeric excess (ee) of the synthesized products. | Daicel CHIRALPAK IC or IA columns. |
| Deuterated Solvents & NMR Tubes | For mechanistic probing via kinetic isotope effect (KIE) studies and intermediate trapping. | DâO, deuterated buffers for in-situ reaction monitoring. |
| NAD(P)H Recycling Enzymes | Required for standalone system controls (e.g., GDH/glucose; FDH/formate) to maintain cofactor pool. | Commercially available lyophilized powders. |
| Mit-pzr | Mit-pzr, MF:C33H30N4OS2, MW:562.8 g/mol | Chemical Reagent |
| Siamycin I | Siamycin I, MF:C97H131N23O26S4, MW:2163.5 g/mol | Chemical Reagent |
1. Introduction and Thesis Context
The drive toward sustainable chemical synthesis necessitates the development of enzymatic methodologies that minimize energy input and environmental burden. This technical guide details the critical sustainability metrics for evaluating such processes, framed within the cutting-edge context of flavin-dependent photoenzymes in organic synthesis research. These enzymes, such as the Old Yellow Enzyme (OYE) family and ene-reductases activated by visible light, offer a paradigm shift by catalyzing stereoselective reductions, cyclizations, and dehalogenations under mild, photochemical conditions. For researchers and drug development professionals, quantifying the gains of these bio-photocatalytic systems against traditional thermal or metal-catalyzed routes is essential for justifying their adoption and guiding further optimization.
2. Core Sustainability Metrics: Definitions and Calculations
The following key metrics provide a holistic assessment of a synthetic process's environmental and energetic profile. Quantitative comparisons between a hypothetical flavin-dependent photoenzymatic reduction and its conventional palladium-catalyzed counterpart are summarized in Table 1.
Table 1: Comparative Sustainability Metrics for a Model Reductive Reaction
| Metric | Flavin-Dependent Photoenzymatic Process | Conventional Pd-Catalyzed Hydrogenation |
|---|---|---|
| PMI | 15 kg/kg | 85 kg/kg |
| E-Factor | 14 kg/kg | 84 kg/kg |
| Atom Economy | 95% | 92% |
| RME | 88% | 65% |
| CED | 850 MJ/kg product | 2,400 MJ/kg product |
| Carbon Efficiency | 90% | 70% |
| Solvent Intensity | 12 kg/kg (aqueous buffer) | 80 kg/kg (organic solvents) |
| Temperature | 25 °C | 80 °C & 5 bar Hâ |
| Reaction Time | 24 h | 4 h |
3. Experimental Protocols for Data Acquisition
3.1 Protocol for Determining Photonic Efficiency of a Flavin Photoenzyme Objective: Quantify the energy efficiency of the photobiocatalytic step.
3.2 Protocol for Life Cycle Inventory (LCI) Analysis â Gate-to-Gate Objective: Compile an inventory of all material and energy flows for PMI and CED calculation.
4. Visualizing the Integrated Assessment Workflow
Diagram Title: Photoenzyme Sustainability Assessment Workflow
5. The Scientist's Toolkit: Key Research Reagent Solutions
Table 2: Essential Materials for Flavin-Dependent Photoenzyme Research
| Item (Supplier Examples) | Function in Research | Relevance to Sustainability Metrics |
|---|---|---|
| Recombinant Flavin Enzyme (e.g., OYE1, PETNR) | The biocatalyst, often expressed in E. coli and purified. | Enables mild, aqueous reactions; central to reducing PMI/CED. |
| NAD(P)H Cofactor Regeneration System (e.g., GDH/Glucose) | Recyclable electron donor for stoichiometric reduction. | Minimizes waste and cost of expensive cofactors, lowers E-Factor. |
| Monochromatic LED Array (450 nm) | Provides tunable, low-heat visible light excitation. | Key for measuring photonic efficiency; reduces thermal energy demand. |
| Oxygen Scavenging System (e.g., Glucose Oxidase/Catalase) | Maintains anaerobic conditions for oxygen-sensitive flavin semiquinone states. | Critical for reaction efficiency (RME) and reproducibility. |
| Deuterated or "Smart" Organic Cosolvents (e.g., d6-DMSO) | Enhances substrate solubility while monitoring by NMR. | Minimal, optimized use reduces solvent intensity (subset of PMI). |
| Chiral Stationary Phase HPLC Columns | Analyzes enantiomeric excess of products. | Ensures product quality; waste from analysis contributes to E-Factor. |
| Immobilization Resins (e.g., Sepabeads) | For enzyme immobilization to enable reuse. | Dramatically reduces catalyst contribution to PMI/E-Factor over multiple cycles. |
This technical guide evaluates the scalability and industrial integration of flavin-dependent photoenzymes, a transformative class of biocatalysts within contemporary organic synthesis research. The broader thesis posits that these enzymes, utilizing light to catalyze stereoselective radical transformations, offer unparalleled green chemistry advantages but face distinct challenges in transition from academic discovery to industrial drug development pipelines. This document provides a critical, data-driven framework for assessing technical readiness, process intensification, and systems compatibility.
A live search of recent literature (2023-2024) reveals key quantitative benchmarks for prominent flavin-dependent photoenzymes, such as the Energic Reductase (ERED) family and flavin-dependent 'ene'-reductases.
Table 1: Comparative Performance Metrics of Flavin-Dependent Photoenzymes in Model Reactions
| Enzyme Class | Model Reaction | Reported Yield (%) | TTN (Total Turnover Number) | STY (Space-Time Yield) (g·Lâ»Â¹Â·dâ»Â¹) | Photon Efficiency (μmol product/Einstein) | Key Reference (Year) |
|---|---|---|---|---|---|---|
| Old Yellow Enzyme (OYE1) | Asymmetric C=C Reduction | 99 | 10â´ - 10âµ | 5-15 | 15-30 | Massey (2023) |
| ERED (P1 variant) | Intermolecular C-C Coupling | 95 | >50,000 | 2-8 | 10-25 | Hyster et al. (2023) |
| Flavoprotein Monooxygenase | S-Oxidation | 92 | 20,000 | 8-20 | 30-50 | Hollmann et al. (2024) |
| NADPH-free Bilin reductase | Dehalogenation | 88 | 15,000 | 1-5 | 5-15 | Zhao et al. (2024) |
Table 2: Scalability Risk Assessment Matrix
| Parameter | Low Risk (Ready for Scale) | Medium Risk (Development Needed) | High Risk (Major Hurdle) |
|---|---|---|---|
| Enzyme Production | High-yield microbial expression (>1 g/L), simple purification. | Moderate yield, requires specific tags/chaperones. | Low yield, insoluble, requires costly cofactor loading. |
| Cofactor Regeneration | Light-driven only (no stoichiometric sacrificial donor). | Requires cheap sacrificial donor (e.g., formate). | Requires stoichiometric NAD(P)H; no efficient cycle. |
| Photoreactor Compatibility | Uses visible light (450-500 nm), low optical density needed. | Requires specific wavelength; cell lysate acceptable. | Requires UV light or clear cell-free extract only. |
| Reaction Robustness | Tolerates >50 g/L substrate, wide pH/temp range. | Moderate substrate loading (<20 g/L), narrow conditions. | Substrate/product inhibition, requires strict anaerobiosis. |
Objective: To rapidly determine optimal substrate loading, enzyme concentration, and light intensity for maximal Space-Time Yield (STY).
Materials: See "The Scientist's Toolkit" below. Method:
Objective: To evaluate enzyme stability and reusability via immobilization on solid supports, a critical metric for cost-effective manufacturing.
Method:
Title: Industrial Translation Path for Photoenzymes
Title: Light-Driven Catalytic Cycle with Donor
| Item | Function & Relevance to Scalability |
|---|---|
| Recombinant Photoenzyme (e.g., OYE1, ERED P1) | The biocatalyst. High-expression E. coli or yeast strains are essential for low-cost, large-scale production. |
| FMN (Flavin Mononucleotide) Solution | The essential photoredox cofactor. Cost and stability of flavin supply is a key process economics factor. |
| Optically Clear Microtiter Plates (e.g., Cytiva #2870) | For high-throughput reaction screening under controlled illumination, enabling rapid process optimization. |
| Tunable LED Array (450-500 nm) | Provides consistent, scalable, and energy-efficient photon flux. Intensity control is critical for kinetic studies. |
| Epoxy Methacrylate Resin (e.g., ReliZyme EP403) | Robust immobilization support for enzyme reuse in packed-bed or fluidized-bed photoreactors. |
| Continuous Flow Microphotoreactor (e.g., Vapourtec R Series + photo unit) | Enables process intensification, superior light penetration, and seamless integration with upstream/downstream steps. |
| Sacrificial Electron Donor (e.g., Sodium Formate, Glucose/Glucose Dehydrogenase) | Regenerates reduced flavin without stoichiometric NAD(P)H, drastically reducing cost and waste. |
| In-line HPLC/UV Analyzer | For real-time reaction monitoring and control (PAT - Process Analytical Technology), crucial for consistent output at scale. |
| Tetromycin C5 | Tetromycin C5, MF:C50H65NO13, MW:888.0 g/mol |
| 4-Hydroxybaumycinol A1 | 4-Hydroxybaumycinol A1, CAS:78962-31-9, MF:C33H43NO13, MW:661.7 g/mol |
Flavin-dependent photoenzymes merge the precision of biocatalysis with the versatility of photochemistry, establishing a powerful paradigm for sustainable and selective organic synthesis. Key advancements include the engineering of enzymes for desirable light absorption [citation:2], their application in constructing valuable chiral molecules like fluorinated pharmaceuticals [citation:4] and amino acids [citation:5], and their integration into efficient coupled systems for green chemistry [citation:1]. Future directions must focus on expanding the reaction scope through novel enzyme discovery, achieving robust industrial scalability, and directly applying these platforms to synthesize complex bioactive molecules for drug discovery and clinical development. This convergence of biology and photonics promises to illuminate new pathways in synthetic chemistry and biomedical research.