Mechanism: ECH1 overexpression suppresses ferroptosis (iron-dependent cell death) by modulating the Erk signaling pathway .
Outcomes:
Adipose Browning: ECH1 promotes thermogenesis by upregulating UCP1 and PGC1α in white adipose tissue .
Energy Expenditure:
ECH1 is downregulated in calcified human aortic valves. Overexpression reduces calcium deposition by inhibiting osteogenic pathways .
Tissue Distribution: Highly expressed in liver, adipose tissue, and kidneys .
Transcriptional Networks: Associated with lipid metabolism pathways (PPARα, CPT1α) .
ECH1 (delta(3,5)-Delta(2,4)-dienoyl-CoA isomerase) is a member of the hydratase/isomerase superfamily that plays an important role in the auxiliary step of the fatty acid beta-oxidation pathway . The protein shows high sequence similarity to enoyl-CoA hydratases across multiple species, particularly within conserved domains characteristic of these enzymes. Its primary function is to isomerize 3-trans,5-cis-dienoyl-CoA to 2-trans,4-trans-dienoyl-CoA, a critical step in the metabolism of unsaturated fatty acids with odd-numbered double bonds .
For researchers investigating ECH1 function, enzyme activity assays measuring isomerization rates provide direct functional evidence. These assays typically employ spectrophotometric measurements or HPLC analysis of substrate-product conversion. Complementary approaches include metabolomic profiling of cells with modulated ECH1 expression to identify changes in fatty acid intermediates, providing insight into the specific metabolic pathways affected by ECH1 activity.
ECH1 contains a C-terminal peroxisomal targeting sequence and primarily localizes to peroxisomes . Interestingly, studies of the rat ortholog indicate that the protein can also localize to the matrix of both peroxisomes and mitochondria, suggesting potential dual compartmentalization depending on cellular conditions . This dual localization may reflect the importance of ECH1 in fatty acid metabolism occurring in both organelles.
To methodologically investigate ECH1 localization, researchers should employ a multi-faceted approach including:
Subcellular fractionation followed by Western blotting
Immunofluorescence microscopy with organelle-specific markers
Expression of fluorescently-tagged ECH1 constructs
Proximity labeling approaches such as BioID or APEX
For quantitative assessment, high-content imaging systems with automated image analysis can measure colocalization coefficients with known peroxisomal and mitochondrial markers, providing statistical robustness to localization studies.
Recombinant ECH1 protein requires specific storage and handling conditions to maintain structural integrity and enzymatic activity. Based on manufacturer specifications, the optimal formulation includes 20 mM Tris-HCl buffer (pH 8.0), 10% glycerol, 1 mM DTT, and 50 mM NaCl . This formulation provides pH stability, prevents protein denaturation, maintains reducing conditions, and provides ionic strength.
Storage Parameter | Recommendation | Rationale |
---|---|---|
Short-term storage | 4°C | Minimizes degradation while maintaining accessibility |
Long-term storage | -20°C | Prevents proteolytic degradation and denaturation |
Working concentration | 1 mg/ml | Standard for most enzymatic assays |
Avoid | Freeze-thaw cycles | Prevents protein aggregation and activity loss |
Researchers should aliquot the protein upon receipt to minimize freeze-thaw cycles and validate protein integrity by SDS-PAGE before experimental use . For enzymatic assays, activity controls should be included to ensure the protein maintains its functional properties throughout experimental procedures.
Recombinant human ECH1 protein for research applications is typically produced in E. coli expression systems . This bacterial expression platform offers advantages for producing non-glycosylated human proteins like ECH1, including high yield, cost-effectiveness, and established purification protocols. The recombinant protein is typically tagged with a polyhistidine sequence (His-tag) at the N-terminus to facilitate purification by metal affinity chromatography .
For researchers producing their own ECH1 protein, optimization of expression conditions is critical. This includes selection of appropriate E. coli strains (BL21(DE3) derivatives are common), induction parameters (IPTG concentration, temperature, duration), and purification strategies. Following purification, validation of protein identity and purity should be performed using techniques such as SDS-PAGE (targeting >90% purity), Western blotting with anti-ECH1 antibodies, and mass spectrometry .
For rigorous biochemical characterization of ECH1 activity, researchers should establish well-controlled reaction conditions that mimic physiological environments while enabling precise measurements. Based on biochemical principles and the protein's characteristics, the following parameters are recommended:
Parameter | Optimal Condition | Notes |
---|---|---|
Buffer system | 50 mM phosphate or HEPES | Maintains physiological pH stability |
pH | 7.2-7.4 | Reflects physiological conditions |
Temperature | 37°C | Human physiological temperature |
Protein concentration | 0.1-1.0 μM | Adjust based on specific activity |
Substrate concentration | 5-100 μM | Range for kinetic parameter determination |
Cofactors | None required | Unlike other β-oxidation enzymes |
Detection method | HPLC-UV or LC-MS/MS | For direct measurement of substrate/product |
Activity assays should include appropriate controls such as heat-inactivated enzyme preparations and reaction mixtures lacking substrate. For kinetic analysis, researchers should collect time-course data with multiple substrate concentrations to determine Km, Vmax, and kcat values. These parameters provide quantitative measures of enzyme efficiency and can be compared across experimental conditions or between wild-type and mutant proteins.
Investigation of ECH1's protein-protein interactions requires a multi-method approach to capture both stable and transient interactions within metabolic pathways. Given ECH1's role in fatty acid metabolism and its dual localization, interaction studies should focus on both peroxisomal and mitochondrial protein networks.
Methodologically, researchers should begin with affinity purification approaches using tagged ECH1 as bait, followed by mass spectrometry (AP-MS) to identify potential interacting partners. For validation and detailed characterization, techniques including:
Bimolecular fluorescence complementation (BiFC) for visualizing interactions in living cells
Förster resonance energy transfer (FRET) for measuring interaction distances
Co-immunoprecipitation with endogenous proteins for confirming physiological relevance
Proximity-dependent labeling methods (BioID, APEX2) for capturing transient interactions
When analyzing interaction data, researchers should apply stringent statistical filtering (typically fold change >2, p<0.01) and validate key interactions using at least two independent methods. Functional validation through co-expression, co-depletion, or enzymatic coupling assays provides evidence for the biological significance of identified interactions.
To investigate the functional consequences of ECH1 mutations or deficiency, researchers should employ complementary genetic and biochemical approaches. CRISPR-Cas9 gene editing provides a precise method for creating cellular models with ECH1 knockout or specific mutations. Lentiviral shRNA systems offer an alternative approach for temporary knockdown when complete gene deletion is not desired.
For phenotypic characterization, a comprehensive analysis should include:
Analysis Level | Techniques | Expected Outcomes |
---|---|---|
Molecular | RNA-seq, proteomics | Changes in related metabolic enzymes |
Biochemical | Fatty acid oxidation assays | Altered oxidation of specific fatty acids |
Metabolomic | Targeted LC-MS/MS | Accumulation of specific intermediates |
Cellular | Mitochondrial function, peroxisome morphology | Organelle dysfunction |
Physiological | Energy expenditure, stress resistance | Metabolic adaptation |
For clinical relevance, patient-derived cell lines or induced pluripotent stem cells (iPSCs) carrying ECH1 mutations can be differentiated into relevant cell types to study tissue-specific effects. Rescue experiments with wild-type ECH1 and various mutant constructs help distinguish between loss-of-function and gain-of-function effects.
Understanding ECH1's role within the context of integrated metabolism requires systems biology approaches that capture pathway interactions and regulatory mechanisms. Metabolic flux analysis using stable isotope-labeled substrates (13C-labeled fatty acids) enables quantitative measurement of carbon flow through ECH1-dependent and alternative pathways.
Researchers should combine targeted experimental manipulations with computational modeling:
Experimental approaches:
Metabolic tracing using labeled substrates and mass spectrometry
Pathway inhibition studies using specific inhibitors of related enzymes
Time-course analyses of metabolite changes after ECH1 modulation
Multi-omics integration (transcriptomics, proteomics, metabolomics)
Computational approaches:
Constraint-based metabolic modeling (e.g., flux balance analysis)
Kinetic modeling of fatty acid oxidation pathways
Network analysis of metabolic interactions
Machine learning for pattern detection in complex datasets
These integrated approaches help identify compensatory mechanisms, regulatory feedback loops, and pathway crosstalk that may not be apparent from isolated studies of ECH1 function.
ECH1 expression and function may vary significantly across tissue types, reflecting tissue-specific metabolic requirements. The Human Protein Atlas and other tissue expression databases provide insight into expression patterns across tissues , but functional studies require tissue-specific methodological adaptations.
For tissue-specific investigations, researchers should consider:
Expression analysis:
Single-cell RNA sequencing to resolve cell type-specific expression
Immunohistochemistry with validated antibodies for protein localization
Tissue microarrays for comparative expression profiling
Functional studies:
Primary cell isolation from relevant tissues
Organoid cultures maintaining tissue architecture
Tissue-specific conditional knockout animal models
Metabolism studies under tissue-relevant substrate conditions
Pathological relevance:
Expression analysis in disease-affected tissues
Correlation with tissue-specific biomarkers
Patient-derived samples for validation studies
When interpreting results, researchers should account for tissue-specific factors including metabolic specialization, environmental conditions, and developmental stage. These considerations ensure that findings accurately reflect the biological role of ECH1 in specific physiological contexts.
The ECH1 gene is located on chromosome 19q13.2 . The gene encodes a protein that consists of 329 amino acids and contains a C-terminal peroxisomal targeting signal (SKL), which is crucial for its localization to the peroxisomes . The protein shares a high degree of sequence similarity with enoyl-CoA hydratases from various species, indicating its evolutionary conservation and importance .
ECH1 is involved in the auxiliary step of the fatty acid beta-oxidation pathway. Specifically, it catalyzes the isomerization of 3-trans, 5-cis-dienoyl-CoA to 2-trans, 4-trans-dienoyl-CoA . This reaction is essential for the metabolism of unsaturated fatty acids, allowing them to be further processed by other enzymes in the beta-oxidation pathway .
Interestingly, ECH1 is not only localized to the peroxisomes but also to the mitochondria . This dual localization suggests that ECH1 plays a role in both peroxisomal and mitochondrial fatty acid metabolism, highlighting its versatility and importance in cellular energy production .
ECH1 is ubiquitously expressed in various human tissues, with the highest levels observed in skeletal muscle . The expression of ECH1 can be induced by peroxisome proliferators, which are compounds that promote the proliferation of peroxisomes and enhance the expression of genes involved in peroxisomal beta-oxidation . This regulation is mediated through peroxisome proliferator-activated receptors (PPARs), which bind to specific response elements in the promoter regions of target genes .
Mutations or dysregulation of the ECH1 gene can have significant clinical implications. For instance, defects in the beta-oxidation pathway can lead to the accumulation of fatty acids and their derivatives, which can be toxic to cells. This can result in various metabolic disorders, including adrenoleukodystrophy and central nervous system leiomyoma . Understanding the function and regulation of ECH1 is therefore crucial for developing therapeutic strategies for these conditions.
Recombinant ECH1 refers to the enzyme produced through recombinant DNA technology, which involves inserting the ECH1 gene into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. This recombinant protein can be used for various research and therapeutic purposes, including studying the enzyme’s function, screening for potential inhibitors, and developing treatments for metabolic disorders.