Far1 catalyzes the reduction of saturated and unsaturated C16–C18 fatty acyl-CoA to fatty alcohols, a reaction essential for ether lipid biosynthesis. Its activity is tightly linked to plasmalogen production, as fatty alcohols are incorporated into the sn-1 position of 1-alkyl-dihydroxyacetone phosphate (DHAP) intermediates via alkyl-DHAP synthase .
Parameter | Recombinant Rat Far1 | Human Far1 |
---|---|---|
Substrate Preference | C16–C18 fatty acyl-CoAs | C16–C18 (saturated/unsaturated) |
Reaction Product | Fatty alcohols | Fatty alcohols |
Biological Role | Ether lipid synthesis | Plasmalogen biosynthesis |
Key Pathway | Peroxisomal ether lipid pathway | Peroxisomal ether lipid pathway |
Far1 stability is dynamically regulated by cellular plasmalogen levels, ensuring feedback control of ether lipid synthesis. In plasmalogen-deficient cells, Far1 protein levels rise (~4-fold), while plasmalogen supplementation restores them to baseline . This regulation occurs post-translationally, as FAR1 mRNA levels remain unchanged .
Plasmalogen-Dependent Degradation:
Protein Stability Modulation:
Property | Value |
---|---|
Source | E. coli expression system |
Expression System | In vitro recombinant production |
Molecular Weight | Not explicitly stated |
Purity | High (as per supplier standards) |
Applications | Enzymatic assays, lipid studies |
Plasmalogen Deficiency and Far1 Upregulation:
Role in Ether Lipid Synthesis:
Disease Implications:
While Far1 and Far2 share catalytic activity, their subcellular roles and regulation differ significantly.
Aspect | Far1 | Far2 |
---|---|---|
Catalytic Output | Fatty alcohols | Fatty alcohols |
Localization | Peroxisomes | Cytosol/ER |
Substrate Scope | Broad (C16–C18) | Narrow (saturated C16–C18) |
Regulation | Plasmalogen-dependent degradation | None reported |
Tissue-Specific Expression | Widespread | Testis, retina |
Fatty acyl-CoA reductase 1 (FAR1) is an enzyme that catalyzes the reduction of fatty acyl-CoA to fatty alcohols, playing a crucial role in lipid metabolism. In rats, as in humans, FAR1 is primarily involved in the biosynthesis of ether lipids and waxes. The enzyme demonstrates both fatty-acyl-CoA reductase (alcohol-forming) activity and long-chain-fatty-acyl-CoA reductase activity, as confirmed by functional studies. FAR1 participates in the peroxisome pathway, where it contributes to the metabolism of very long-chain fatty acids and interacts with multiple peroxisomal proteins including PEX14 . This enzyme represents an important component in cellular lipid homeostasis, with implications for membrane structure and function across mammalian species.
Several expression systems can be employed for producing recombinant rat FAR1, each with distinct advantages:
Bacterial expression (E. coli): The pGEX vector system, which creates GST-fusion proteins, offers a robust platform for initial expression attempts. This approach allows for inducible expression using IPTG (typically 0.1 mM) with optimal induction periods of approximately 6 hours . The primary advantage is high yield, though proper folding may be compromised.
Yeast expression systems: These provide a eukaryotic environment with some post-translational processing capabilities while maintaining relatively high yields.
Insect cell expression: Baculovirus-infected insect cells offer improved folding and post-translational modifications compared to prokaryotic systems.
Mammalian cell expression: For the most physiologically relevant form of rat FAR1, mammalian expression systems such as HEK293 cells provide appropriate folding and post-translational modifications . This approach is particularly relevant when investigating protein-protein interactions or when enzymatic activity requires specific modifications.
Selection of an appropriate expression system should be guided by the specific experimental requirements, balancing protein yield with functional considerations.
Optimizing purification of recombinant rat FAR1 requires careful attention to several critical factors:
Lysis conditions: Use gentle lysis methods such as sonication (3-5 cycles of 30 seconds on/off) in buffers containing protease inhibitors and reducing agents (DTT or β-mercaptoethanol) . This prevents protein degradation and oxidation of cysteine residues.
Affinity purification: For GST-tagged FAR1, glutathione-Sepharose affinity chromatography represents an effective first purification step. After binding, wash extensively with PBS to remove non-specifically bound proteins before eluting with 10 mM reduced glutathione in buffer .
Tag removal: If a tag-free protein is desired, include a protease cleavage site between the tag and FAR1. For GST-tagged proteins, PreScission Protease can efficiently cleave the tag under mild conditions while maintaining enzyme activity .
Buffer optimization: Test different buffers (pH 7.0-8.0) containing stabilizing agents such as glycerol (10-20%) and reducing agents. Include cofactor (NADPH) at low concentrations to stabilize the active site.
Storage conditions: Aliquot purified FAR1 into small volumes to avoid freeze-thaw cycles and store at -80°C in buffer containing 10-20% glycerol and reducing agent .
The purification process should be monitored at each step using activity assays to ensure the final product retains catalytic function.
Multiple complementary techniques should be employed to verify the purity and identity of recombinant rat FAR1:
SDS-PAGE: Assess protein purity and approximate molecular weight. Expect a band at approximately 60 kDa for rat FAR1 (or higher if fusion tags are present) .
Western blotting: Confirm identity using antibodies specific to FAR1 or to the fusion tag (e.g., anti-GST) . This provides specificity that SDS-PAGE alone cannot offer.
Mass spectrometry: MALDI-QqTOF mass spectrometry can provide precise molecular weight determination and, following protease digestion, peptide mapping for sequence confirmation . This approach can identify post-translational modifications and verify sequence integrity.
Size exclusion chromatography: Analyze the oligomeric state and detect potential aggregates or degradation products.
Circular dichroism: Assess secondary structure to confirm proper folding, particularly when comparing different expression systems or purification methods.
Enzymatic activity assays: Measure the conversion of fatty acyl-CoA substrates to fatty alcohols using spectrophotometric methods tracking NADPH consumption as the definitive test of functional integrity.
These methods collectively provide a comprehensive characterization profile for recombinant rat FAR1 preparations.
Inconsistent activity between recombinant rat FAR1 batches represents a common challenge requiring systematic troubleshooting:
Standardization of expression conditions: Maintain consistent cell density before induction (OD600 of 0.6-0.8), IPTG concentration (0.1 mM has been demonstrated effective for recombinant proteins), and induction time (6 hours optimum for many GST-fusion proteins) . Document growth curves for each production batch.
Protein characterization matrix:
Parameter | Method | Acceptance Criteria |
---|---|---|
Purity | SDS-PAGE | >95% single band |
Identity | Western blot | Single band at expected MW |
Oligomeric state | Size exclusion | Predominantly monomeric |
Activity | NADPH consumption | >80% of reference standard |
Stability | Thermal shift assay | Tm within ±2°C of reference |
Reference standard implementation: Maintain a well-characterized reference standard preparation and include it in each activity assay as an internal control.
Cofactor and substrate quality: Use fresh NADPH preparations and verify the quality of fatty acyl-CoA substrates, as these can degrade and significantly impact measured activity.
Environmental variables: Control temperature precisely during activity assays, as enzymatic rates are highly temperature-dependent. Standardize all buffer components, including salt concentration and pH.
Statistical validation: Perform at least triplicate measurements for each activity determination and apply appropriate statistical analyses to determine if observed differences are significant.
Implementing these approaches creates a robust framework for identifying sources of variability and establishing reliable quality control parameters.
Investigating substrate specificity of rat FAR1 requires a systematic experimental design:
Substrate panel preparation: Assemble a diverse panel of fatty acyl-CoA substrates including:
Kinetic characterization protocol:
Measure initial reaction rates across a range of substrate concentrations (typically 1-100 μM)
Maintain constant enzyme concentration and reaction conditions
Determine Km, Vmax, and catalytic efficiency (kcat/Km) for each substrate
Plot structure-activity relationships based on chain length, degree of unsaturation, etc.
Competition experiments:
Perform assays with multiple substrates simultaneously
Analyze product formation using GC-MS or LC-MS to determine preference ratios
Use varying ratios of competing substrates to establish preference profiles
Structure-function analysis:
Design and express FAR1 variants with mutations in the predicted substrate binding pocket
Compare substrate profiles of wild-type and mutant proteins
Correlate changes in specificity with structural features
Computational modeling:
Develop homology models of rat FAR1 based on related enzymes
Perform molecular docking with various substrates
Identify key residues involved in substrate recognition
This comprehensive approach provides both qualitative and quantitative insights into the substrate preference landscape of rat FAR1.
Investigating rat FAR1's role in peroxisomal lipid metabolism requires a multi-faceted approach:
Subcellular localization studies:
Protein-protein interaction network:
Metabolic impact assessment:
Lipidomic analysis comparing wild-type and FAR1-deficient cells
Focus on ether lipid precursors and derivatives
Stable isotope labeling to track metabolic flux through FAR1-dependent pathways
Analysis of peroxisomal β-oxidation efficiency
Genetic manipulation strategies:
siRNA knockdown of FAR1 in rat hepatocytes or other relevant cell types
CRISPR-Cas9 genome editing to create FAR1-null cells
Rescue experiments with wild-type and mutant FAR1 constructs
Conditional knockout models to study tissue-specific effects
Disease model correlations:
These approaches collectively provide a comprehensive understanding of FAR1's contribution to peroxisomal metabolism and potential implications for metabolic disorders.
Systematic investigation of structure-function relationships in rat FAR1 requires a coordinated experimental approach:
Structural analysis foundations:
Domain mapping strategy:
Create truncation mutants to identify minimal functional units
Design chimeric proteins with domains from related enzymes (e.g., FAR2)
Systematically replace regions with the corresponding sequences from other species
Site-directed mutagenesis design:
Target predicted catalytic residues (typically involving S, Y, K catalytic triad in SDR enzymes)
Modify residues in the NAD(P)H-binding region (typically a Rossmann fold)
Alter potential membrane-association domains
Create charge-reversal mutations at potential protein-protein interaction interfaces
Functional characterization matrix:
Construct | Enzymatic Activity | Substrate Specificity | Protein Interactions | Cellular Localization |
---|---|---|---|---|
Wild-type | Baseline | Baseline | Baseline | Peroxisomal |
Catalytic mutants | Reduced/eliminated | Altered | Maintained | Maintained |
Binding pocket mutants | Maintained | Altered | Maintained | Maintained |
Localization mutants | Maintained | Maintained | Altered | Cytosolic/mislocalized |
Biophysical analysis:
Circular dichroism to assess secondary structure changes
Thermal stability measurements to identify destabilizing mutations
Intrinsic fluorescence to monitor conformational changes upon substrate/cofactor binding
This systematic approach correlates structural elements with specific functional properties, providing mechanistic insights into rat FAR1's catalytic activity and biological role.
Establishing optimal assay conditions for rat FAR1 requires careful optimization of multiple parameters:
Basic assay components:
Buffer: 100 mM phosphate buffer or 50 mM Tris-HCl, pH 7.4-7.8
Reducing environment: 1-5 mM DTT or β-mercaptoethanol
Cofactor: 0.1-0.5 mM NADPH (freshly prepared)
Substrate: 10-100 μM fatty acyl-CoA (chain length appropriate for experiment)
Enzyme: 0.1-1.0 μg purified recombinant FAR1
Spectrophotometric measurement protocol:
Monitor NADPH consumption at 340 nm (ε = 6,220 M⁻¹cm⁻¹)
Maintain temperature at 25°C or 37°C (consistent between experiments)
Measure initial rates (first 1-5 minutes) to ensure linearity
Include appropriate controls (no enzyme, no substrate, heat-inactivated enzyme)
Alternative product detection methods:
Gas chromatography analysis of fatty alcohols following extraction
HPLC separation with UV or fluorescence detection (may require derivatization)
LC-MS/MS for highest sensitivity and specificity
Optimization considerations:
Temperature dependence: Test activity at 25°C, 30°C, and 37°C
pH profile: Test activity across pH range 6.5-8.5
Salt sensitivity: Test NaCl concentrations from 0-500 mM
Divalent cation effects: Test impact of Mg²⁺, Mn²⁺, Ca²⁺ (0-5 mM)
Data analysis:
Calculate specific activity as nmol NADPH consumed/min/mg protein
For kinetic studies, fit data to Michaelis-Menten equation to determine Km and Vmax
For inhibition studies, determine IC₅₀ values and inhibition mechanisms
These optimized conditions ensure reproducible and physiologically relevant measurement of rat FAR1 activity.
Post-translational modifications (PTMs) of recombinant rat FAR1 can be analyzed using multiple complementary techniques:
Mass spectrometry-based identification:
Analyze peptides by MALDI-QqTOF MS in positive ionization mode
Compare observed masses with theoretical peptide masses
Identify mass shifts corresponding to potential modifications
Confirm PTM identity and location using MS/MS fragmentation patterns
Modification-specific enrichment strategies:
Phosphorylation: Immobilized metal affinity chromatography (IMAC) or phospho-specific antibodies
Glycosylation: Lectin affinity chromatography
Ubiquitination: Affinity purification with anti-ubiquitin antibodies
Acetylation: Anti-acetyllysine antibodies for enrichment
Site-directed mutagenesis approach:
Identify potential modification sites by in silico prediction
Create site-specific mutants (e.g., S→A to prevent phosphorylation, K→R to prevent acetylation)
Compare properties of wild-type and mutant proteins
Generate phosphomimetic mutants (S→D or S→E) to simulate phosphorylation
Expression system comparison:
Express rat FAR1 in different systems (E. coli, yeast, insect cells, mammalian cells)
Compare PTM patterns between expression systems
Correlate modifications with functional properties
Enzymatic demodification:
Treat purified FAR1 with phosphatases, deglycosylases, or deacetylases
Monitor changes in electrophoretic mobility and activity
Perform peptide mass fingerprinting before and after treatment
These approaches provide comprehensive characterization of FAR1 PTMs and their functional significance.
Multiple complementary approaches can effectively detect and characterize protein-protein interactions involving rat FAR1:
Affinity-based isolation techniques:
Proximity-based detection methods:
Bimolecular fluorescence complementation (BiFC) for direct visualization in cells
Förster resonance energy transfer (FRET) to measure proximity between fluorescently tagged proteins
Proximity ligation assay (PLA) for detection of endogenous protein interactions
BioID or APEX2 proximity labeling to identify proteins in the vicinity of FAR1
Genetic interaction screens:
Yeast two-hybrid screening using rat FAR1 as bait
Mammalian two-hybrid assays for verification in more relevant cell types
Synthetic lethality screens to identify functional interactions
Quantitative interaction characterization:
Surface plasmon resonance (SPR) to measure binding kinetics and affinity
Isothermal titration calorimetry (ITC) for thermodynamic parameters
Microscale thermophoresis (MST) for interaction studies in solution
Interaction mapping:
Domain mapping through truncation mutants
Alanine scanning of predicted interaction surfaces
Crosslinking mass spectrometry to identify interaction interfaces
Functional validation:
Co-localization studies in cells using immunofluorescence
Mutagenesis of interaction interfaces and functional testing
Competition assays with peptides derived from interaction regions
These approaches collectively provide robust identification and characterization of the FAR1 interactome, including interactions with peroxisomal proteins such as PEX14 and metabolic enzymes like GSTK1 .
Designing rigorous controls for rat FAR1 knockdown or overexpression experiments requires addressing several potential sources of experimental variability:
Knockdown experiment controls:
Non-targeting siRNA/shRNA with similar GC content to FAR1-targeting sequences
Multiple independent siRNA/shRNA sequences targeting different regions of FAR1 mRNA
Rescue experiments with siRNA-resistant FAR1 cDNA (containing silent mutations)
Quantification of knockdown efficiency by qRT-PCR and Western blot
Assessment of potential off-target effects using transcriptome analysis
Evaluation of compensatory upregulation of FAR2 or other related enzymes
Overexpression experiment controls:
Empty vector transfection maintaining identical promoter and regulatory elements
Expression of unrelated protein of similar size (e.g., GFP) from the same vector
Catalytically inactive mutant FAR1 (for separating enzymatic from structural functions)
Titration of expression levels to avoid non-physiological effects
Subcellular localization verification to ensure proper targeting
Time-course experiments to distinguish acute from adaptive responses
Experimental design considerations:
Cell-type specific controls (primary cells vs. cell lines)
Appropriate timepoints based on protein half-life and cellular processes
Paired biological replicates to minimize batch effects
Randomization of sample processing order
Blinded analysis of experimental outcomes where possible
Phenotypic validation:
Multiple independent methods to assess the same endpoint
Complementary approaches (e.g., genetic and pharmacological)
Dose-dependency of observed effects
Correlation between degree of knockdown/overexpression and phenotype magnitude
These comprehensive controls ensure that observed phenotypes can be confidently attributed to specific alterations in FAR1 expression or function.
Overcoming expression and solubility challenges with recombinant rat FAR1 requires systematic optimization at multiple levels:
Vector design optimization:
Expression host selection:
Expression condition optimization matrix:
Parameter | Variables to Test | Expected Impact |
---|---|---|
Temperature | 16°C, 25°C, 30°C, 37°C | Lower temperatures reduce inclusion body formation |
Induction | 0.01-1.0 mM IPTG | Lower inducer concentrations can improve solubility |
Media | LB, TB, 2xYT, minimal | Rich media can support proper folding |
Additives | Glycerol, sucrose, sorbitol | Osmolytes can stabilize folding intermediates |
Duration | 3-24 hours | Optimal time balances yield vs. degradation |
Protein extraction and purification strategies:
Buffer optimization:
Screen buffer compositions (HEPES, Tris, phosphate)
Vary pH conditions (pH 6.0-9.0)
Test salt concentrations (0-500 mM NaCl)
Include stabilizing additives (glycerol, arginine, trehalose)
Add reducing agents (DTT, β-mercaptoethanol) to prevent oxidation
These strategies have successfully addressed similar challenges in recombinant protein expression as documented for other challenging enzymes .