ACADSB primarily processes short/branched-chain acyl-CoA substrates, with distinct activity profiles:
Substrate | Relative Activity | Metabolic Pathway |
---|---|---|
(S)-2-methylbutyryl-CoA | High | Isoleucine degradation |
Isobutyryl-CoA | Moderate | Valine metabolism |
2-methylhexanoyl-CoA | Low | Medium-chain fatty acid oxidation |
The human enzyme exhibits reduced activity against longer-chain substrates compared to rat orthologs due to structural differences in substrate-binding residues .
Mutations in ACADSB cause short/branched-chain acyl-CoA dehydrogenase deficiency (SBCADD), characterized by:
Elevated 2-methylbutyrylglycine and 2-methylbutyrylcarnitine in blood/urine
Symptoms: hypoglycemia, muscle atrophy, developmental delays (30% of cases)
Detection: Newborn screening via tandem mass spectrometry (MS/MS) for C5-carnitine
ACADSB downregulation correlates with poor survival in multiple cancers:
Cancer Type | Hazard Ratio (Low vs. High ACADSB) | Study Cohort |
---|---|---|
Clear cell RCC | 2.1 (95% CI: 1.5–2.9) | TCGA-KIRC (n=522) |
Colon adenocarcinoma | 1.8 (95% CI: 1.2–2.7) | TCGA-COAD |
Breast cancer (Luminal) | 1.5 (95% CI: 1.1–2.1) | TCGA-BRCA |
In clear cell renal cell carcinoma (ccRCC), ACADSB expression discriminates tumor vs. normal tissue with AUC values of 0.952–0.966 across datasets .
ACADSB Human is utilized in:
Enzyme kinetics studies: Investigating substrate specificity and inhibition
Cancer biomarker research: Validating ACADSB as a prognostic indicator via IHC and RNA-seq
Metabolic disorder models: Screening therapeutics for SBCADD
A 2022 cohort study identified nine ACADSB variants across 10 patients:
Common mutations: c.1165A>G (p.Lys389Glu), c.1159G>A (p.Glu387Lys)
Novel variants: c.1102T>C (VUS/likely pathogenic), c.823C>T (p.Arg275Ter)
Diagnostic biomarkers: Urinary 2-methylbutyrylglycine levels show 100% sensitivity for SBCADD .
Therapeutic monitoring: L-carnitine supplementation (100 mg/kg/day) normalized metabolic profiles in 90% of patients .
Cancer immunotherapy: Low ACADSB expression correlates with reduced CD8+ T-cell infiltration in ccRCC .
Short/branched-chain specific acyl-CoA dehydrogenase (ACADSB) is a member of the acyl-CoA dehydrogenase enzyme family. These enzymes play a crucial role in fatty acid and branched-chain amino acid metabolism by catalyzing the dehydrogenation of acyl-CoA derivatives. ACADSB is specifically involved in the breakdown of L-isoleucine and exhibits a high affinity for substrates like (s)-2-methylbutyryl-CoA, isobutyryl-CoA, and 2-methylhexanoyl-CoA. Additionally, it may utilize valproyl-CoA as a substrate. Genetic defects in the ACADSB gene can lead to short/branched-chain acyl-CoA dehydrogenase deficiency (SBCADD), an autosomal recessive disorder. This disorder is characterized by elevated levels of 2-methylbutyrylglycine and 2-methylbutyrylcarnitine in the bloodstream and urine.
The ACADSB gene encodes for the mitochondrial short/branched-chain acyl-CoA dehydrogenase (SBCAD), a member of the acyl-CoA dehydrogenase family of enzymes. This protein catalyzes two critical reactions: (1) the third reaction in the L-isoleucine degradation pathway, specifically the conversion of 2-methylbutyryl-CoA to tiglyl-CoA, and (2) the first oxidative step of short straight-chain acyl-CoAs, including butyryl-CoA and hexanoyl-CoA . Additionally, SBCAD can utilize valproyl-CoA as a substrate, suggesting a potential role in valproate metabolism .
Research methodology: To characterize ACADSB protein function, researchers typically employ enzyme activity assays using purified recombinant protein with various acyl-CoA substrates. Spectrophotometric methods measuring the reduction of electron acceptors can quantify dehydrogenase activity, while mass spectrometry can identify reaction products and intermediates.
The ACADSB gene is located on chromosome 10, specifically within the chromosomal band 10q25-q26 . The gene has been characterized at the molecular level with a genomic reference sequence of NG_008003.1 and transcript reference of NM_001609.3 .
Research methodology: Chromosomal localization was initially determined through Southern blot analysis using human/rodent somatic cell hybrids and further refined through fluorescence in situ hybridization (FISH) . Current mapping approaches include next-generation sequencing and bioinformatic analysis of reference genomes.
SBCAD deficiency (SBCADD, OMIM# 600301/610006) is identified through metabolite analysis of blood, urine, and fibroblast samples . The primary diagnostic markers include:
Elevated C5-carnitine levels detected through newborn screening
Elevated 2-methylbutyrylglycine (2MBG) in urine
Long-term monitoring involves regular assessment of:
DBS (dried blood spot) or serum C5 concentration
Urine 2MBG concentration
Serum glucose, ammonia, and CK concentrations
Liver function tests
Blood gases
Cardiac function via heart ultrasound and electrocardiogram
Research methodology: Acylcarnitine analysis is performed using tandem-mass spectrometry (LC/MS-MS) on either DBS or serum samples, with established intra-day precision of approximately 0.7% coefficient of variation (CV) and inter-day precision of approximately 5% CV . Urine organic acid profiles are assessed via gas chromatography-mass spectrometry (GC-MS) .
Molecular characterization of ACADSB variants involves multiple steps:
DNA extraction from EDTA peripheral venous blood samples
PCR amplification of all exons and parts of flanking intron regions
Sequencing according to standard procedures
Variation reporting following the Human Genome Variation Society (HGVS) nomenclature
Annotation according to:
Research methodology: While standard PCR and Sanger sequencing remain valid, next-generation sequencing approaches including targeted gene panels, whole-exome sequencing, and whole-genome sequencing offer higher throughput for variant identification. Functional validation of variants can be performed through expression studies in cellular models or enzyme activity assays with purified recombinant proteins containing the specific mutations.
ACADSB has been found to be down-regulated in multiple cancers, with decreased expression correlating with poor prognosis in certain cancer types . Specific findings include:
ACADSB plays important roles in glioma, colorectal cancer (CRC), and hepatocellular carcinoma (HCC)
In clear cell renal cell carcinoma (ccRCC), decreased ACADSB expression predicts poor prognosis
Research methodology: To study ACADSB expression in cancer, researchers employ:
RNA-sequencing or qRT-PCR for transcriptional analysis
Western blotting or immunohistochemistry for protein expression
Cancer tissue microarrays for high-throughput expression profiling
Kaplan-Meier survival analysis to correlate expression levels with patient outcomes
Gene knockdown or overexpression experiments to assess functional consequences of altered ACADSB expression
SBCADD shows variable clinical presentation. The current understanding is:
Most individuals with SBCADD, including those identified through newborn screening, show no health problems
A small percentage develop symptoms shortly after birth or later in childhood
Initial symptoms may include poor feeding, lethargy, vomiting, and irritability
Other features can include poor growth, muscle weakness, delay in motor skills, and intellectual disability
A founder mutation exists in the Hmong Chinese population, where affected individuals have remained largely asymptomatic
Research methodology: Population studies utilize newborn screening data analysis, with biochemical confirmation of suspected cases. Case-control studies examining genotype-phenotype correlations help elucidate the clinical spectrum. Long-term follow-up studies track developmental outcomes and metabolic stability.
Research on ACADSB's metabolic roles employs multiple experimental approaches:
Metabolomic Profiling: Using LC-MS/MS or GC-MS to identify metabolic perturbations in ACADSB-deficient cells or tissues
Stable Isotope Tracing: Employing 13C-labeled isoleucine or other substrates to track metabolic flux through ACADSB-dependent pathways
CRISPR-Cas9 Gene Editing: Creating cellular models with ACADSB knockout or specific mutations
Recombinant Enzyme Assays: Assessing substrate specificity and kinetic parameters of wild-type and mutant ACADSB proteins
Mitochondrial Functional Assays: Measuring oxygen consumption rates and mitochondrial membrane potential in ACADSB-deficient models
These approaches provide complementary insights into ACADSB's role in branched-chain amino acid metabolism and fatty acid oxidation.
Determining ACADSB variant pathogenicity involves multiple lines of evidence:
Population Frequency Analysis: Evaluating variant prevalence in population databases like gnomAD
In Silico Prediction Tools: Using algorithms like SIFT, PolyPhen-2, and CADD to predict functional impact
Conservation Analysis: Assessing evolutionary conservation of affected amino acid residues
Functional Assays: Measuring enzyme activity of recombinant proteins containing variants
Clinical Correlation: Analyzing biochemical profiles of patients carrying specific variants
Segregation Analysis: Examining variant co-segregation with disease in affected families
The ACMG guidelines provide a standardized framework for integrating these evidence types to classify variants as pathogenic, likely pathogenic, variant of uncertain significance (VUS), likely benign, or benign .
Long-term monitoring of SBCADD patients employs standardized approaches:
Biomarker Trend Analysis: Tracking serum C5 and urine 2MBG trends, classified as:
Biochemical Surveillance: Regular assessment of metabolic parameters including:
Clinical Monitoring: Tracking growth parameters, developmental milestones, and system-specific assessments (cardiac, neurological)
The 10% threshold for trend classification was established based on analytical performance characteristics, specifically setting the threshold at approximately 10 times the intraday CV and twice the interday CV of the analytical methods .
Recent research has identified ACADSB as a potential biomarker in multiple cancer types:
Expression Profiling: Decreased ACADSB expression predicts poor prognosis in clear cell renal cell carcinoma (ccRCC)
Functional Studies: ACADSB plays important roles in glioma, colorectal cancer, and hepatocellular carcinoma
Metabolic Reprogramming: ACADSB alterations may contribute to cancer-specific metabolic phenotypes through effects on branched-chain amino acid metabolism
Research methodology: Biomarker validation studies typically employ multi-cohort designs with discovery and validation phases. Techniques include immunohistochemistry on tissue microarrays, transcriptomic analysis of patient samples, and correlation with clinical outcomes. Mechanistic studies investigate how ACADSB affects cancer cell proliferation, migration, and response to therapy.
Computational biology has become increasingly important for ACADSB research:
Homology Modeling: Utilizing structural information from related acyl-CoA dehydrogenases to predict ACADSB tertiary structure
Molecular Dynamics Simulations: Investigating how mutations affect protein stability and substrate binding
Systems Biology Approaches: Integrating ACADSB into metabolic network models to predict systemic effects of altered function
Machine Learning Applications: Developing improved algorithms for variant pathogenicity prediction based on multiple data types
These computational methods complement experimental approaches and can generate hypotheses for further laboratory investigation.
Acyl-CoA dehydrogenases (ACADs) are a class of mitochondrial flavoenzymes that play a crucial role in the metabolism of fatty acids and amino acids. These enzymes catalyze the initial step in each cycle of fatty acid β-oxidation, introducing a trans double-bond between the α and β carbon atoms of the acyl-CoA thioester substrate . Among the various types of ACADs, the short-chain acyl-CoA dehydrogenase (SCAD) specifically targets short-chain fatty acids.
Short-chain acyl-CoA dehydrogenase (SCAD) is responsible for the dehydrogenation of saturated short-chain acyl-CoA molecules, converting them into their corresponding enoyl-CoA derivatives. This reaction is the first and rate-limiting step in the β-oxidation pathway, which ultimately leads to the production of acetyl-CoA. Acetyl-CoA then enters the tricarboxylic acid (TCA) cycle, contributing to the production of ATP through oxidative phosphorylation .
The enzyme utilizes flavin adenine dinucleotide (FAD) as a cofactor to facilitate the transfer of electrons from the acyl-CoA substrate to the electron transfer flavoprotein (ETF). The ETF then transfers these electrons to the mitochondrial respiratory chain, where they contribute to the generation of ATP .
The gene encoding SCAD is known as ACADS. Mutations in this gene can lead to short-chain acyl-CoA dehydrogenase deficiency (SCADD), an autosomal recessive disorder characterized by impaired fatty acid oxidation. Individuals with SCADD may exhibit a range of clinical symptoms, from severe metabolic or neuromuscular disabilities to being completely asymptomatic .
Human recombinant SCAD is produced using recombinant DNA technology. This involves inserting the human ACADS gene into a suitable expression vector, which is then introduced into a host cell, such as Escherichia coli or yeast. The host cells are cultured under conditions that promote the expression of the SCAD protein. The recombinant protein is then purified using various chromatographic techniques to obtain a highly pure and active enzyme.
Recombinant SCAD is used in various research applications to study the biochemical and physiological roles of the enzyme. It is also employed in the development of diagnostic assays for detecting SCADD and other related metabolic disorders. Additionally, recombinant SCAD can be used in drug discovery and development to screen for potential therapeutic compounds that target fatty acid oxidation pathways.