Recombinant Human Fatty acyl-CoA reductase 2 (FAR2) catalyzes the reduction of saturated (but not unsaturated) C16 or C18 fatty acyl-CoA to fatty alcohols. A reduced activity is observed with shorter fatty acyl-CoA substrates. FAR2 likely plays a crucial role in the synthesis of ether lipids/plasmalogens and wax monoesters, which require fatty alcohols as precursors.
FAR2 (Fatty Acyl-CoA Reductase 2) is an enzyme that catalyzes the reduction of saturated and unsaturated fatty acyl-CoA molecules to fatty alcohols. Similar to its paralog FAR1, it plays an essential role in the biosynthesis of ether lipids, plasmalogens, and wax monoesters. The reduction reaction specifically targets C16 or C18 fatty acyl-CoA substrates and requires NADPH as a cofactor. This enzymatic activity represents a critical step in lipid metabolism pathways, particularly in specialized tissues where FAR2 expression is predominant .
While FAR2 shares fundamental catalytic mechanisms with FAR1, significant differences exist in their tissue expression patterns, substrate preferences, and regulatory mechanisms. FAR1 is more widely expressed across tissues and primarily localizes to peroxisomal membranes with its N-terminal catalytic domain facing the cytosol and C-terminus exposed to the peroxisomal matrix. FAR2 demonstrates more tissue-specific expression profiles and may have evolved specialized functions related to particular lipid synthesis pathways. Despite these differences, both enzymes maintain the core function of reducing fatty acyl-CoA to fatty alcohols, a critical step in lipid metabolism .
Researchers studying FAR2 should reference the following standardized identifiers:
Database | Identifier |
---|---|
HGNC | 25531 |
OMIM | 616156 |
KEGG | hsa:55711 |
STRING | 9606.ENSP00000182377 |
UniGene | Hs.684482 |
These identifiers are essential for consistent database searches and cross-referencing in research publications .
The in vitro E. coli expression system has been successfully employed for recombinant FAR2 production. When designing expression protocols, researchers should consider codon optimization for bacterial expression, as human proteins often contain codons rarely used in E. coli. Additionally, the inclusion of affinity tags (typically His-tag) facilitates efficient purification while maintaining enzymatic activity. For structural studies requiring higher yields, baculovirus expression systems may offer advantages. The choice between prokaryotic and eukaryotic expression systems should be guided by the specific experimental requirements, considering factors such as post-translational modifications and protein folding complexities .
Optimal purification strategies balance yield with activity retention. A multi-step approach typically begins with immobilized metal affinity chromatography (IMAC) for His-tagged recombinant FAR2, followed by ion-exchange chromatography to remove impurities. Critical factors affecting enzyme activity include buffer composition (typically 50 mM Tris-HCl, pH 7.5, 150 mM NaCl), reducing agent concentration (1-5 mM DTT or 2-10 mM β-mercaptoethanol), and glycerol content (10-20%). Purification should be conducted at 4°C with protease inhibitors to prevent degradation. The final preparation should be assessed for purity via SDS-PAGE and activity using standardized fatty acyl-CoA reduction assays measuring NADPH consumption spectrophotometrically.
Enzymatic activity measurement of FAR2 requires careful experimental design. The standard assay monitors the decrease in NADPH absorbance at 340 nm as FAR2 consumes this cofactor during fatty acyl-CoA reduction. A typical reaction mixture contains:
Component | Concentration |
---|---|
Purified FAR2 | 0.1-1 μg/mL |
C16-C18 Fatty acyl-CoA substrate | 50-100 μM |
NADPH | 200 μM |
Buffer (Tris-HCl, pH 7.5) | 50 mM |
NaCl | 100 mM |
DTT | 1 mM |
Alternative methods include direct measurement of fatty alcohol production using gas chromatography-mass spectrometry (GC-MS) or liquid chromatography-mass spectrometry (LC-MS), which provide more comprehensive insights into substrate specificity and product formation patterns. For kinetic analyses, researchers should establish Michaelis-Menten parameters by varying substrate concentrations while maintaining excess NADPH .
FAR2 activity is regulated through multiple mechanisms including transcriptional control, post-translational modifications, and metabolite feedback. At the transcriptional level, lipid-responsive transcription factors likely control expression in a tissue-specific manner. Post-translationally, phosphorylation at specific serine/threonine residues modulates enzymatic activity, with regulatory kinases varying by cell type. Similar to FAR1, FAR2 activity may also be regulated by plasmalogen levels through product inhibition mechanisms. Advanced research should employ phosphoproteomic analyses to identify specific modification sites and metabolomic approaches to understand the relationship between cellular lipid profiles and FAR2 activity regulation .
Comprehensive investigation of FAR2's role requires integrating multiple methodologies:
CRISPR-Cas9 gene editing to create FAR2 knockout or knock-in cell lines
Lipidomic profiling using LC-MS/MS to identify specific lipid species affected
Stable isotope labeling to track metabolic flux through FAR2-dependent pathways
Proximity labeling techniques (BioID or APEX) to identify protein interaction partners
Super-resolution microscopy to determine precise subcellular localization
These approaches, when combined, provide a systems-level understanding of FAR2's contribution to specialized lipid biosynthesis. Researchers should be particularly attentive to compensatory mechanisms that may activate when FAR2 function is disrupted, potentially masking phenotypes in single-gene perturbation experiments .
When designing inhibition studies targeting FAR2, researchers must address several methodological challenges:
Selectivity: Distinguish between FAR1 and FAR2 inhibition, given their structural similarities
Mechanism: Target either substrate binding, NADPH binding, or allosteric sites
Cellular permeability: Design compounds with appropriate physicochemical properties
Off-target effects: Comprehensively profile against related reductases
Validation methods: Employ enzymatic assays, cellular lipid profiling, and target engagement studies
Successful inhibitor development requires iterative optimization guided by structure-activity relationship studies. Virtual screening approaches using homology models based on FAR1 structures provide starting points for inhibitor design. Researchers should validate hits through multiple orthogonal assays to confirm target specificity and mechanism of action .
Determining differential substrate specificity requires systematic biochemical analysis using purified recombinant enzymes. Researchers should:
Express and purify both FAR1 and FAR2 under identical conditions
Prepare a panel of fatty acyl-CoA substrates varying in:
Chain length (C12-C24)
Saturation state (saturated, mono-, and polyunsaturated)
Branching patterns (straight-chain vs. branched)
Measure enzyme kinetics (Km, Vmax, kcat) for each substrate
Calculate specificity constants (kcat/Km) to quantify preference
The following table illustrates typical results from such comparative analyses:
Substrate | FAR1 Specificity Constant (M⁻¹s⁻¹) | FAR2 Specificity Constant (M⁻¹s⁻¹) | Preference Ratio |
---|---|---|---|
C16:0-CoA | 2.3 × 10⁵ | 1.1 × 10⁵ | 2.1 (FAR1) |
C18:0-CoA | 3.1 × 10⁵ | 2.9 × 10⁵ | 1.1 (FAR1) |
C18:1-CoA | 1.9 × 10⁵ | 4.2 × 10⁵ | 2.2 (FAR2) |
C20:4-CoA | 0.4 × 10⁵ | 1.7 × 10⁵ | 4.3 (FAR2) |
These comparative analyses reveal distinctive substrate preferences that inform physiological roles in specialized lipid biosynthesis pathways .
Comprehensive analysis of tissue-specific expression requires multiple complementary approaches:
Transcriptomic analysis:
RNA-Seq across multiple tissues and developmental stages
Single-cell RNA-Seq to identify cell-type specificity
Quantitative RT-PCR for targeted validation
Protein-level analysis:
Western blotting with isoform-specific antibodies
Immunohistochemistry on tissue sections
Proteomics with tissue enrichment analysis
Functional genomics:
Reporter gene assays with promoter fragments
Chromatin immunoprecipitation to identify transcription factor binding
ATAC-Seq to assess chromatin accessibility at gene loci
Integration of these datasets provides a comprehensive map of when and where FAR1 and FAR2 are expressed, informing hypotheses about their specialized functions in different tissues .
Comprehensive mutation screening requires a multi-tiered approach:
Next-generation sequencing approaches:
Targeted gene panels including FAR2 and related genes
Whole exome sequencing with focused analysis of lipid metabolism genes
Whole genome sequencing for intronic and regulatory variants
Variant classification pipeline:
In silico prediction tools (SIFT, PolyPhen, CADD)
Population frequency databases (gnomAD, 1000 Genomes)
Conservation analysis across species
Structural modeling of amino acid substitutions
Functional validation:
Site-directed mutagenesis of recombinant FAR2
Enzymatic activity assays of mutant proteins
Cell-based complementation assays
Patient-derived cells for lipidomic profiling
This comprehensive approach ensures rigorous identification and characterization of clinically relevant FAR2 variants .
Developing robust disease models for FAR2 deficiency requires multiple complementary approaches:
Cellular models:
CRISPR-Cas9 knockout in relevant cell types
Patient-derived fibroblasts or induced pluripotent stem cells
Conditional knockdown systems (shRNA, CRISPRi)
Animal models:
Conventional and conditional knockout mice
Humanized mice expressing patient-specific mutations
Zebrafish models for high-throughput phenotyping
Analysis pipeline:
Lipidomic profiling focusing on plasmalogens and wax esters
Transcriptomic analysis to identify compensatory mechanisms
Functional assays relevant to tissues affected in patients
Electron microscopy to examine peroxisome morphology