ACADL, also called Long-Chain Acyl-CoA Dehydrogenase (LCAD), catalyzes the initial step of long-chain fatty acid oxidation by converting acyl-CoA to trans-Δ²-enoyl-CoA. Key biochemical characteristics include:
ACADL deficiency in mice causes hypothermia, hepatic steatosis, and cardiomyopathy due to impaired fatty acid oxidation .
ACADL is essential for energy production in cardiac and skeletal muscles by breaking down fatty acids. Deficiency leads to:
ACADL inhibits cancer progression through two primary pathways:
Hippo/YAP Signaling: Restoring ACADL in hepatocellular carcinoma (HCC) cells reduces YAP nuclear translocation, downregulating oncogenes like CTGF and CYR61 .
Matrix Metalloproteinase 14 (MMP14): ACADL suppresses HCC metastasis by inhibiting MMP14, a promoter of extracellular matrix degradation .
Prognostic Marker: Low ACADL correlates with aggressive tumor features (vascular invasion, recurrence) and poor survival (Table 1) .
Therapeutic Target: Verteporfin (YAP inhibitor) suppresses growth in ACADL-low HCC organoids .
ACADL prevents immune evasion by suppressing PD-L1 expression via Hippo/YAP signaling .
While ACADL acts as a tumor suppressor in HCC and lung cancer, studies report conflicting roles:
Prostate Cancer: ACADL enhances malignant phenotypes and progression .
Breast Cancer: Hypermethylation of ACADL correlates with poor survival .
Recombinant ACADL (ENZ-190) is used to study fatty acid metabolism and cancer mechanisms. Key applications include:
In vitro assays: Assessing enzyme activity in mitochondrial extracts .
Xenograft models: Demonstrating tumor growth suppression in nude mice .
ACADL (Acyl-CoA Dehydrogenase Long Chain) is a critical enzyme in the mitochondrial fatty acid β-oxidation pathway, catalyzing the initial dehydrogenation step of long-chain fatty acyl-CoA substrates. This enzyme specifically handles fatty acids with carbon chain lengths of C14-C20, converting them to their corresponding trans-2-enoyl-CoAs.
Methodological approach for studying ACADL function:
Spectrophotometric enzyme assays using ferricenium ion as electron acceptor
Acylcarnitine profiling via tandem mass spectrometry
Oxygen consumption measurements in isolated mitochondria
Metabolic flux analysis using isotope-labeled substrates
Accurate measurement of ACADL activity is essential for both basic research and clinical investigations. Several complementary approaches are commonly employed:
Method | Principle | Advantages | Limitations |
---|---|---|---|
Spectrophotometric assay | Measures electron transfer via artificial acceptors | Rapid, high-throughput | Potential interference from other dehydrogenases |
Radioisotope-based assay | Tracks conversion of 14C-labeled substrates | High sensitivity | Requires radioactive materials handling |
LC-MS/MS analysis | Quantifies product formation directly | Specific identification of products | Equipment-intensive, lower throughput |
Oxygen consumption | Measures coupled respiratory activity | Physiologically relevant | Indirect measurement |
For optimal results, researchers should:
Include appropriate controls (positive, negative, and enzyme blanks)
Validate assay linearity across the expected activity range
Normalize to protein content or mitochondrial markers
Consider tissue-specific differences in activity levels
ACADL shows distinct tissue-specific expression patterns that align with metabolic requirements:
Tissue | Relative Expression | Functional Significance | Research Techniques |
---|---|---|---|
Heart | Very high | Critical for cardiac energy production | Immunohistochemistry, Western blotting |
Liver | High | Important for ketogenesis during fasting | RT-qPCR, proteomics |
Skeletal muscle | Moderate to high | Varies by fiber type; higher in oxidative fibers | Single-cell RNA-seq, tissue fractionation |
Brain | Low to moderate | Region-specific variation | In situ hybridization, spatial transcriptomics |
Adipose tissue | Low | Primarily expresses other FAO enzymes | RNA-seq, cell sorting approaches |
Methodological considerations when studying tissue-specific expression:
Use multiple detection methods (mRNA and protein)
Account for mitochondrial content differences between tissues
Consider developmental and nutritional status effects
Validate with human samples when possible, as animal models may show different patterns
ACADL shows preferential activity toward long-chain fatty acyl-CoAs (C14-C20) compared to shorter or very-long-chain substrates. This specificity is determined by:
Active site architecture: The substrate-binding pocket contains a hydrophobic channel that accommodates the fatty acid chain
Key residues: Specific amino acids that interact with the substrate's acyl chain through hydrophobic interactions
FAD cofactor positioning: The positioning of the flavin group relative to the substrate's α-carbon determines the efficiency of electron transfer
Quaternary structure: ACADL functions as a homotetramer, with interactions between subunits affecting substrate access
Experimental approaches to investigate substrate specificity include:
Site-directed mutagenesis of conserved residues
X-ray crystallography with substrate analogs
Molecular dynamics simulations
Enzyme kinetics with varied chain-length substrates
ACADL undergoes several post-translational modifications that modulate its activity, stability, and interactions:
Modification | Sites | Effect on Activity | Detection Methods |
---|---|---|---|
Phosphorylation | Ser/Thr residues | Can increase or decrease depending on site | Phospho-specific antibodies, mass spectrometry |
Acetylation | Lys residues | Generally decreases activity | Acetyl-lysine antibodies, HDAC inhibitors |
Glutathionylation | Cys residues | Protective during oxidative stress | Redox proteomics, biotin-switch technique |
Ubiquitination | Lys residues | Targets for degradation | Ubiquitin pull-down, proteasome inhibitors |
Research approaches should include:
Mass spectrometry-based proteomic identification of modifications
Site-directed mutagenesis to create non-modifiable variants
In vitro modification assays to determine direct effects
Identification of the enzymes responsible for adding/removing modifications
The transcriptional regulation of ACADL involves complex epigenetic control mechanisms that respond to metabolic conditions:
Epigenetic Mechanism | Effect on ACADL | Experimental Approaches | Metabolic Context |
---|---|---|---|
DNA methylation | Typically repressive | Bisulfite sequencing, methylation arrays | Development, aging |
Histone modifications | H3K27ac activates; H3K9me3 represses | ChIP-seq, ChIP-qPCR | Nutritional transitions |
Chromatin remodeling | Accessibility changes | ATAC-seq, DNase-seq | Fasting/feeding cycles |
Non-coding RNAs | miRNA repression, lncRNA scaffolding | RNA-seq, RNA pulldown | Metabolic disease states |
Methodological considerations:
Temporal resolution is critical—capture rapid transitions
Cell type-specific analyses avoid dilution of signals
Integration with transcription factor binding data (especially PPARα)
Validation in physiologically relevant models
Selecting the optimal model system is crucial for addressing specific research questions about ACADL:
Model | Advantages | Limitations | Best Applications |
---|---|---|---|
HepG2/Huh7 cells | Easy maintenance, reproducible | Lower ACADL expression than primary cells | Gene regulation studies |
Primary hepatocytes | Physiologically relevant | Short lifespan, donor variability | Acute metabolic responses |
iPSC-derived cardiomyocytes | Human cardiac-specific phenotypes | Complex differentiation, maturation issues | Cardiac metabolism studies |
ACADL knockout mice | Complete gene deletion | Compensatory mechanisms may mask phenotypes | In vivo physiological studies |
Patient-derived fibroblasts | Disease-relevant mutations | Limited metabolic capacity | Personalized medicine approaches |
Experimental design considerations:
Match model to research question (regulation vs. function vs. pathology)
Validate ACADL expression levels in your specific model
Consider tissue-specific metabolic differences
Use multiple complementary models when possible
Metabolic flux analysis requires careful experimental design to isolate ACADL's specific contribution:
Substrate selection considerations:
Use palmitate (C16:0) as a prototypical ACADL substrate
Include control substrates (e.g., octanoate for MCAD)
Consider both labeled and unlabeled substrate mixtures
Tracer experiment design:
[U-13C] palmitate for complete pathway tracing
[1-13C] palmitate for specific first-round β-oxidation
[ω-13C] labeling to track terminal oxidation
Analytical approaches:
GC-MS or LC-MS/MS for isotopologue distribution
Computational modeling to estimate flux parameters
Integration with oxygen consumption measurements
Controls and validation:
ACADL inhibition (genetic or pharmacological)
Complementary assays (acylcarnitine profiles)
Concentration dependency assessment
CRISPR/Cas9 gene editing provides powerful tools for ACADL research but requires careful planning:
Editing Strategy | Research Application | Key Design Considerations | Validation Methods |
---|---|---|---|
Complete knockout | Loss-of-function studies | Potential compensatory mechanisms | Western blot, enzyme activity assays |
Point mutations | Structure-function analysis | PAM site proximity, HDR efficiency | Sanger sequencing, functional assays |
Knock-in reporters | Expression monitoring | Maintenance of endogenous regulation | Fluorescence microscopy, flow cytometry |
CRISPRi/CRISPRa | Tunable expression | Target site accessibility, guide RNA design | qRT-PCR, Western blot |
Methodological workflow should include:
Comprehensive guide RNA design (minimum 3-4 guides per target)
Appropriate delivery method selection (transfection vs. viral)
Efficient screening strategy for edited cells
Careful clone selection and validation
Phenotypic characterization including rescue experiments
Researchers frequently encounter conflicting results when studying ACADL. A systematic approach to resolving these contradictions includes:
Methodological differences assessment:
Assay conditions (pH, temperature, substrate concentration)
Detection methods sensitivity and specificity
Sample preparation variations
Biological variables consideration:
Species differences in ACADL function
Nutritional or metabolic status of samples
Age, sex, and genetic background effects
Compensatory mechanisms in chronic models
Resolution strategies:
Direct method comparison using identical samples
Collaborative cross-validation between laboratories
Meta-analysis of published data with attention to methodological details
Development of standardized protocols
Reporting recommendations:
Transparent documentation of all experimental conditions
Inclusion of all relevant controls
Publication of negative or contradictory results
Data sharing in accessible repositories
Analysis of ACADL genetic variants requires specialized statistical methods:
Analysis Type | Application | Statistical Approach | Sample Size Considerations |
---|---|---|---|
Common variant association | Population studies | Logistic/linear regression, GWAS | Large cohorts (n>1000) |
Rare variant analysis | Clinical cohorts | Burden tests, SKAT, SKAT-O | Moderate size (n=100-500) |
Functional prediction | Variant classification | Machine learning, evolutionary conservation | Dependent on algorithm |
Genotype-phenotype correlation | Clinical translation | Multinomial models, Bayesian approaches | Variable based on effect size |
Methodological best practices include:
Power calculations before study initiation
Population stratification adjustment
Multiple testing correction
Replication in independent cohorts
Functional validation of significant variants
Integration of multiple omics datasets provides comprehensive insights into ACADL function:
Omics Layer | Contribution to ACADL Research | Integration Challenges | Analytical Approaches |
---|---|---|---|
Genomics | Genetic variants affecting function | Linking variants to mechanisms | eQTL analysis, functional genomics |
Transcriptomics | Expression regulation | Transcript-protein correlation | Weighted gene co-expression networks |
Proteomics | Protein levels and PTMs | Sample preparation compatibility | Protein interaction networks |
Metabolomics | Substrate/product profiling | Metabolite identification | Pathway enrichment analysis |
Lipidomics | Specific lipid alterations | Complex lipid annotations | Lipid pathway mapping |
Successful integration strategies include:
Consistent experimental design across platforms
Appropriate normalization methods for each data type
Dimensionality reduction techniques (PCA, t-SNE)
Network-based integration approaches
Validation of key nodes through targeted experiments
ACADL deficiency presents with heterogeneous clinical manifestations that correlate with specific genetic variants:
Variant Category | Clinical Presentation | Biochemical Profile | Research Methodology |
---|---|---|---|
Null variants | Severe, early-onset phenotype | Very low/absent enzyme activity | Next-generation sequencing, functional studies |
Missense variants | Variable severity | Residual enzyme activity (0-25%) | Structure-function analysis, stability assessment |
Regulatory variants | Exercise/fasting-triggered | Normal basal but impaired inducible expression | Promoter analysis, expression studies |
Splice variants | Tissue-specific manifestations | Aberrant protein isoforms | RNA analysis, minigene assays |
Research approaches should include:
Comprehensive clinical phenotyping with standardized protocols
Functional characterization of variants (expression systems, biochemical assays)
Computational prediction validated by experimental data
Long-term natural history studies for prognostic markers
Several therapeutic strategies for ACADL-related disorders are under investigation:
Therapeutic Approach | Mechanism | Research Methods | Development Stage |
---|---|---|---|
Small molecule activators | Direct enzyme enhancement | High-throughput screening, structure-based design | Preclinical |
Pharmacological chaperones | Stabilization of mutant protein | Thermal shift assays, proteostasis modulation | Early clinical trials |
Gene therapy | Expression restoration | AAV-mediated delivery, CRISPR-based correction | Preclinical |
Metabolic bypass inducers | Alternative pathway activation | Metabolic flux analysis, transcriptome profiling | Clinical trials |
Methodological considerations include:
Target engagement confirmation (cellular and in vivo)
Biomarker development for efficacy monitoring
Safety assessment with attention to off-target effects
Combination therapy approaches for synergistic benefits
Personalized approaches based on variant-specific responses
Biomarker selection is critical for both diagnosis and therapeutic monitoring:
Biomarker Category | Examples | Analytical Methods | Clinical Utility |
---|---|---|---|
Acylcarnitine profiles | C14:1, C14, C16 acylcarnitines | Tandem mass spectrometry | Diagnostic, treatment monitoring |
Organic acids | Dicarboxylic aciduria | GC-MS | Acute decompensation marker |
Enzyme activity | Direct ACADL activity | Fibroblasts, lymphocytes assays | Functional assessment |
Genetic markers | ACADL variants | Next-generation sequencing | Diagnosis, family screening |
Novel markers | miRNAs, metabolite ratios | Specialized assays | Early detection, prognosis |
Research considerations for biomarker development:
Validation in diverse patient populations
Establishment of age-specific reference ranges
Correlation with clinical outcomes
Development of point-of-care testing when possible
Integration into comprehensive metabolic profiles
ACADL is a mitochondrial flavoenzyme that requires flavin adenine dinucleotide (FAD) as a cofactor . It catalyzes the dehydrogenation of long-chain acyl-CoA substrates, introducing a trans double-bond between the C2 (α) and C3 (β) positions of the acyl-CoA thioester substrate . This reaction is the first step in the β-oxidation cycle, which ultimately breaks down fatty acids into acetyl-CoA, a key molecule in energy production .
The enzyme is specific for long-chain fatty acids, typically those with 8 to 18 carbon atoms . The human recombinant form of ACADL is produced using recombinant DNA technology, which allows for the expression of the human enzyme in a host organism, such as bacteria or yeast, for research and therapeutic purposes.
The gene encoding ACADL is located on chromosome 2 and is known by several aliases, including LCAD (Long-Chain Acyl-CoA Dehydrogenase) and ACAD4 . Mutations in the ACADL gene can lead to long-chain acyl-CoA dehydrogenase deficiency (LCADD), a metabolic disorder characterized by nonketotic hypoglycemia, muscle weakness, and cardiomyopathy . This condition results from the inability to properly oxidize long-chain fatty acids, leading to an accumulation of fatty acid intermediates and a deficiency in energy production.
Human recombinant ACADL is used extensively in biochemical research to study the mechanisms of fatty acid metabolism and the effects of genetic mutations on enzyme function. It is also employed in the development of diagnostic assays and potential therapeutic interventions for metabolic disorders related to fatty acid oxidation .