Chromosome: 2p23.3
Protein:
HADHB catalyzes the final step of β-oxidation: cleavage of 3-ketoacyl-CoA into acetyl-CoA and a shortened acyl-CoA . The MTP complex includes:
HADHB also regulates RNA stability and interacts with estrogen receptor α (ERα) to modulate lipid metabolism .
Over 102 pathogenic variants in HADHB are documented, causing mitochondrial trifunctional protein deficiency (MTPD) .
c.1175C>T (p.A392V): Associated with adult-onset neuropathy and cerebral demyelination .
Exons 6–9 deletion: Causes severe neonatal cardiomyopathy and rhabdomyolysis .
Deep intronic mutations: Disrupt splicing, leading to frameshifts (e.g., p.Pro270Profs*14) .
Respiratory chain defects: Compound-heterozygous HADHB variants (e.g., FB854) reduce maximal mitochondrial respiration by 30–40% .
Oxidative stress: Impaired β-oxidation elevates reactive oxygen species (ROS), contributing to tissue damage .
Case report (2023): A 61-year-old patient with HADHB c.1175C>T mutation exhibited peripheral neuropathy, cognitive decline, and gadolinium-enhanced brain lesions .
HELLP syndrome/AFLP: Mothers carrying fetuses with biallelic HADHB mutations risk severe liver disease .
Mutation Type | Frequency (%) | Examples |
---|---|---|
Missense | 45 | p.A392V, p.Pro270Profs*14 |
Frameshift | 30 | Exons 6–9 deletion |
Splice-site | 15 | c.33627A>G |
Nonsense | 10 | p.Arg283Ter |
HADHB (Hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoA hydratase, beta subunit) encodes the beta subunit of the mitochondrial trifunctional protein (MTP). This 51.2 kDa protein composed of 474 amino acids catalyzes the final thiolytic cleavage step in mitochondrial beta-oxidation of long-chain fatty acids. Specifically, HADHB provides the 3-ketoacyl-CoA thiolase activity, where the thiol group of Coenzyme A cleaves 3-ketoacyl CoA between C-2 and C-3, yielding an acetyl CoA molecule and an acyl CoA molecule that is two carbons shorter .
The MTP complex forms an α₂β₂ heterotetrameric structure with two HADHB (β) subunits forming a homodimer in the center, flanked by one HADHA (α) subunit on each side. Recent cryo-electron microscopy studies have elucidated this quaternary structure, revealing that proper complex assembly is essential for efficient substrate channeling through the beta-oxidation pathway .
For experimental investigation of HADHB structure:
X-ray crystallography or cryo-EM can resolve detailed protein structure
Protein-protein interaction studies using co-immunoprecipitation can examine HADHB-HADHA interactions
Site-directed mutagenesis of key domains can identify critical residues for enzyme function
HADHB functions within the mitochondrial trifunctional protein, which catalyzes three consecutive reactions in the long-chain fatty acid beta-oxidation pathway. This metabolic pathway represents the major energy-producing process in tissues, especially during fasting states . Within this pathway:
HADHA (α-subunit) performs two enzymatic activities:
2,3-enoyl-CoA hydratase activity
3-hydroxyacyl-CoA dehydrogenase activity
HADHB (β-subunit) performs the final step:
3-ketoacyl-CoA thiolase activity
When studying HADHB's role in this pathway, researchers should consider:
Measuring specific intermediates of beta-oxidation using LC-MS/MS
Analyzing fatty acid utilization in cells with wild-type versus mutant HADHB
Examining compensatory mechanisms when HADHB function is compromised
Beyond its canonical role in fatty acid metabolism, HADHB demonstrates additional functions that expand its biological significance. The protein can bind RNA and decrease the stability of some mRNAs, suggesting a role in post-transcriptional regulation . This RNA-binding capacity may represent a mechanism by which metabolic status is communicated to gene expression regulation.
Methodological approaches to investigate these non-canonical functions include:
RNA immunoprecipitation (RIP) to identify HADHB-bound transcripts
RNA stability assays comparing wild-type and HADHB-deficient cells
Polysome profiling to assess effects on translation
Protein-protein interaction networks to identify non-metabolic binding partners
Researchers should design experiments that can differentiate direct effects of HADHB from indirect consequences of altered fatty acid metabolism when investigating these alternative functions.
The HADHB gene is located on chromosome 2p23 and contains 17 exons encoding a protein of 474 amino acids . The gene is positioned in a head-to-head orientation with HADHA, suggesting coordinated regulation of these functionally related genes.
For comprehensive genetic analysis of HADHB:
Analysis Approach | Methodology | Applications |
---|---|---|
Exome sequencing | NGS of coding regions | Identification of coding variants |
Whole genome sequencing | Complete genomic analysis | Detection of intronic and regulatory variants |
mRNA analysis | RT-PCR and sequencing | Assessment of splicing defects |
Copy number variation | MLPA or array CGH | Detection of large deletions/duplications |
Promoter analysis | Reporter assays | Evaluation of regulatory region function |
When interpreting sequencing results, researchers should consider:
Evolutionary conservation of affected amino acids
Structural impact using in silico prediction tools
Population frequency in databases like gnomAD
Functional consequences on protein stability and enzyme activity
Potential effects on HADHA-HADHB interaction
Various types of mutations in the HADHB gene have been identified in patients with MTP deficiency:
Mutation Type | Example from Literature | Predicted Effect | Detection Method |
---|---|---|---|
Missense | c.694G>A p.(Ala232Thr) | Amino acid substitution affecting protein function | Sanger sequencing, NGS |
Splice site | c.255-1G>A | Likely exon 6 skipping resulting in frameshift | mRNA analysis, RT-PCR |
Nonsense | Various | Premature stop codon leading to truncated protein | Protein truncation testing |
Frameshift | Various | Altered reading frame and nonfunctional protein | Fragment analysis |
For functional characterization of HADHB variants:
Express recombinant wild-type and mutant proteins to assess stability
Perform enzyme activity assays to measure catalytic function
Analyze complex formation with HADHA using co-immunoprecipitation
Conduct in silico structural analysis to predict impacts on protein folding
Assess cellular phenotypes in patient-derived cells or model systems
Structural analysis based on cryo-electron microscopy of the human MTP reveals that mutations can affect the homodimerization of HADHB subunits or disrupt the interaction with HADHA subunits, compromising the integrity of the entire complex .
MTP deficiency presents with a broad spectrum of clinical phenotypes correlating with specific HADHB mutations. This genotype-phenotype relationship can be stratified into several categories:
Phenotype | Clinical Manifestations | Associated Mutation Characteristics | Diagnostic Approach |
---|---|---|---|
Severe infantile | Lethal cardiomyopathy, liver failure, coma | Often null mutations with no residual activity | Acylcarnitine profile, enzyme activity in fibroblasts |
Intermediate | Recurrent rhabdomyolysis, hypoglycemia | Mutations with residual enzyme activity | CK levels, urine organic acids, acylcarnitine profile |
Mild adult-onset | Axonal neuropathy, episodic weakness | Missense mutations with significant residual activity | Neurophysiology studies, enzyme activity in lymphocytes |
A particularly noteworthy finding is the identification of a mild phenotype in three young adults with HADHB mutations (c.694G>A p.(Ala232Thr) and c.255-1G>A) characterized by axonal neuropathy and frequent intermittent weakness episodes without myoglobinuria. These cases highlight that MTP deficiency should be considered in the differential diagnosis of patients with milder fluctuating neuromuscular symptoms, even without elevated CK or rhabdomyolysis .
Research approaches to investigate genotype-phenotype correlations include:
Systematic collection of clinical data from patients with defined mutations
Biochemical characterization of enzyme activity for different variants
Development of cellular and animal models expressing specific mutations
Longitudinal studies to assess disease progression with different genotypes
Accurate measurement of HADHB enzyme activity is crucial for both research and diagnostic applications. Multiple methodological approaches can be employed:
Sample Type | Measurement Technique | Advantages | Limitations |
---|---|---|---|
Lymphocytes | Spectrophotometric assay | Easily accessible sample, standardized protocols | Lower enzyme activity than in affected tissues |
Fibroblasts | Radioisotope-based assay | Stable cell line, reproducible results | Requires skin biopsy, lengthy culture period |
Muscle tissue | LC-MS/MS | Direct assessment of affected tissue | Invasive sampling, tissue heterogeneity |
Liver tissue | Combined enzyme activity assay | High enzyme content, physiologically relevant | Highly invasive sampling, ethical limitations |
For research applications, the specific protocol should be selected based on:
Multiple validated antibodies and detection methods are available for HADHB research:
Antibody | Type | Applications | Dilution/Concentration | Species Reactivity |
---|---|---|---|---|
ab230667 | Rabbit Polyclonal | WB, IHC-P, ICC/IF | 1/500 for WB | Human, Mouse, Rat |
ab110302 | Mouse Monoclonal | IHC-P, ICC, IP, Flow Cyt | 1μg/ml for flow cytometry | Human |
Methodological considerations for different applications:
Western Blot:
Immunohistochemistry:
Flow Cytometry:
Immunoprecipitation:
Developing appropriate models is essential for studying HADHB function, disease mechanisms, and potential therapeutic approaches:
Model Type | Development Method | Applications | Limitations |
---|---|---|---|
Cell lines with HADHB knockout | CRISPR-Cas9 gene editing | Basic functional studies, high-throughput screening | Limited physiological context |
Patient-derived fibroblasts | Skin biopsy from affected individuals | Disease mechanism studies with actual mutations | Variable expression of phenotype in culture |
iPSC-derived cell types | Reprogramming patient cells and differentiation | Tissue-specific effects in relevant cell types | Complex differentiation protocols, variability |
HADHB knockout mice | Germline or conditional gene targeting | Systemic effects, tissue interactions, in vivo dynamics | Species differences in metabolism |
Zebrafish models | Morpholino knockdown or CRISPR editing | Developmental effects, high-throughput screening | Evolutionary distance from humans |
Methodological considerations for model development:
Validate models by confirming HADHB expression levels and enzyme activity
Characterize metabolic profiles using targeted metabolomics
Assess mitochondrial function using respirometry, membrane potential, and ROS production
Challenge models with metabolic stressors to reveal phenotypes (fasting, exercise, temperature)
Use dietary interventions to modulate phenotype severity
For phenotypic characterization, researchers should examine:
Fatty acid oxidation capacity using labeled substrate oxidation assays
Accumulation of intermediate metabolites by mass spectrometry
Tissue-specific manifestations, particularly in muscle and nerve
Compensatory mechanisms that may mask phenotypes
Stress-induced decompensation that mimics clinical triggers
Researchers face several challenges when reconciling contradictory findings regarding HADHB mutations and their clinical manifestations:
Challenge | Methodological Approach | Implementation Strategy |
---|---|---|
Phenotypic heterogeneity | Standardized clinical assessment protocols | Develop and implement consistent evaluation tools across research centers |
Variable tissue expression | Multi-tissue sampling and analysis | Compare enzyme activity and metabolite profiles across affected tissues |
Genetic modifiers | Whole exome/genome sequencing | Identify additional variants that may influence phenotype |
Environmental factors | Detailed patient history documentation | Record dietary patterns, exercise, infections, and other triggers |
Methodological differences | Interlaboratory standardization | Establish reference standards and protocols for enzyme assays |
To systematically address these challenges:
Establish international patient registries with standardized data collection
Develop consensus guidelines for phenotypic classification
Implement multi-omics approaches (genomics, transcriptomics, proteomics, metabolomics)
Create in vitro systems to test variant combinations and environmental conditions
Design longitudinal studies to track phenotypic evolution over time
The case of three patients with similar mild phenotypes despite different HADHB mutations illustrates the complexity of genotype-phenotype correlations. These patients presented with axonal neuropathy and intermittent weakness episodes without myoglobinuria, challenging the traditional association of MTP deficiency with severe manifestations or rhabdomyolysis .
Several cutting-edge methodological approaches can advance HADHB research:
Emerging Technique | Application to HADHB Research | Methodological Advantages |
---|---|---|
Single-cell metabolomics | Heterogeneity in metabolic responses | Reveals cell-to-cell variation in HADHB function |
CRISPR screens | Genetic modifiers of HADHB function | Systematic identification of interacting genes |
Proximity labeling proteomics | HADHB interactome in different conditions | Maps dynamic protein-protein interactions |
Cryo-electron tomography | In situ structural analysis | Visualizes HADHB within native mitochondrial environment |
Stable isotope tracing | Dynamic flux through beta-oxidation | Quantifies pathway activity in living systems |
Organoid models | Tissue-specific HADHB function | Recapitulates complex tissue architecture and function |
Implementation strategies:
Combine multiple approaches to triangulate findings
Establish interdisciplinary collaborations between metabolic specialists, neurologists, and basic scientists
Develop computational models to integrate diverse datasets
Apply systems biology approaches to understand network-level effects of HADHB dysfunction
Translate fundamental findings into potential therapeutic strategies
Current and emerging therapeutic strategies for HADHB deficiency require rigorous evaluation methodologies:
Therapeutic Approach | Mechanism | Evaluation Methods | Outcome Measures |
---|---|---|---|
Dietary management | Restriction of long-chain fats, MCT supplementation | Controlled dietary trials | Acylcarnitine profiles, frequency of metabolic decompensations |
Triheptanoin | Anaplerotic therapy providing alternative energy | Randomized controlled trials | Muscle strength, exercise tolerance, quality of life |
Chaperone therapy | Stabilization of mutant HADHB protein | In vitro folding assays, cellular models | Enzyme activity, protein stability, complex formation |
Gene therapy | Delivery of functional HADHB gene | Animal models, ex vivo cell correction | Tissue-specific enzyme activity, phenotypic correction |
mRNA therapy | Transient expression of functional HADHB | Lipid nanoparticle delivery systems | Duration of expression, mitochondrial targeting efficiency |
Mitochondrial transplantation | Replacement of defective mitochondria | Cell-based assays, animal models | Engraftment efficiency, functional improvement |
For clinical trial design, researchers should consider:
Appropriate control groups and randomization
Selection of clinically meaningful endpoints
Biomarkers that reflect disease activity and treatment response
Long-term follow-up to assess durability of effects
Patient-reported outcomes to capture quality of life impacts
Stratification based on genotype and baseline disease severity
Several critical knowledge gaps persist in HADHB research that warrant focused investigation:
Research Gap | Priority Questions | Methodological Approach |
---|---|---|
Genotype-phenotype correlation | Why do some mutations cause severe disease while others present with mild symptoms? | Large cohort studies correlating genetic variants with standardized phenotyping |
Tissue specificity | Why does HADHB deficiency particularly affect cardiac, muscle, and nerve tissues? | Tissue-specific metabolomics and single-cell transcriptomics |
Neuropathy mechanism | How does HADHB deficiency lead to axonal degeneration? | Nerve-specific cellular models, molecular imaging of axonal transport |
Metabolic adaptation | What compensatory mechanisms emerge in HADHB deficiency? | Flux analysis using stable isotope tracers in different nutritional states |
Non-canonical functions | What is the full spectrum of HADHB functions beyond metabolism? | Interactome analysis, RNA-binding studies, subcellular localization |
Methodological considerations for addressing these questions:
Develop collaborative networks to increase patient numbers
Standardize phenotypic assessments across centers
Implement multiple complementary approaches in parallel
Create biorepositories of patient samples for future analyses
Utilize advanced computational methods to integrate diverse datasets
Integrating multi-omics approaches provides a comprehensive view of HADHB biology:
Omics Approach | Application to HADHB Research | Integration Strategy |
---|---|---|
Genomics | Identify HADHB variants and potential modifiers | Correlate with functional outcomes |
Transcriptomics | Assess expression changes in HADHB deficiency | Identify compensatory pathways |
Proteomics | Map protein interactions and post-translational modifications | Connect to metabolic alterations |
Metabolomics | Profile metabolite changes in various tissues | Link to clinical manifestations |
Lipidomics | Characterize lipid composition changes | Associate with membrane integrity |
Epigenomics | Identify regulatory mechanisms affecting HADHB | Correlate with expression patterns |
Methodological framework for integration:
Collect matched samples for multiple omics analyses
Apply standardized protocols for sample preparation and analysis
Develop computational pipelines for data integration
Use network analysis to identify key nodes and pathways
Validate findings in multiple model systems
Translate integrated insights into testable therapeutic hypotheses
To facilitate meaningful international collaboration, standardized protocols are essential:
Research Area | Proposed Standard Protocol | Implementation Strategy |
---|---|---|
Clinical assessment | Unified phenotyping tool for MTP deficiency | Web-based secure database with standardized forms |
Biochemical diagnosis | Reference methods for enzyme activity measurement | Round-robin testing between reference laboratories |
Genetic analysis | Variant classification framework specific to HADHB | Expert consortium to evaluate novel variants |
Functional validation | Standardized assays for determining pathogenicity | Distribution of reference materials and controls |
Treatment protocols | Consensus guidelines for dietary and supportive care | Multi-center agreement on intervention parameters |
Outcome measures | Validated tools for assessing disease progression | Training programs to ensure consistent application |
For successful implementation:
Establish an international HADHB/MTP deficiency consortium
Secure funding for infrastructure to support collaborative efforts
Develop data sharing agreements that respect privacy regulations
Create repositories for reference materials and biological samples
Organize regular workshops for protocol harmonization and training
Implement quality control programs to ensure consistent results across sites
Collaborative research studies are particularly needed to determine benefit from early treatment identified through newborn screening, as individual studies have had small sample sizes and inconclusive results regarding complications such as neuropathy and retinopathy .
2-Enoyl-Coenzyme A (CoA) Hydratase, Beta, also known as HADHB, is an enzyme that plays a crucial role in the metabolism of fatty acids. This enzyme is part of the mitochondrial trifunctional protein complex, which is involved in the beta-oxidation pathway of fatty acid metabolism. The human recombinant form of this enzyme is produced using recombinant DNA technology, which allows for the production of large quantities of the enzyme for research and therapeutic purposes.
HADHB is a non-glycosylated polypeptide chain consisting of 464 amino acids and has a molecular mass of approximately 49.9 kDa . The enzyme is fused to a 23 amino acid His-tag at the N-terminus, which facilitates its purification through chromatographic techniques .
The primary function of HADHB is to catalyze the hydration of 2-trans-enoyl-CoA to L-3-hydroxyacyl-CoA, a critical step in the beta-oxidation of fatty acids . This reaction involves the addition of a hydroxyl group and a proton to the unsaturated beta-carbon on a fatty-acyl CoA, converting it into a beta-hydroxy acyl-CoA . The enzyme’s efficiency is attributed to its hexameric structure, which provides six active sites for catalysis .
HADHB is essential for the efficient metabolism of fatty acids, which are a significant source of energy for the human body. The beta-oxidation pathway, in which HADHB is involved, breaks down long-chain fatty acids into acetyl-CoA units. These units then enter the citric acid cycle (Krebs cycle) to produce ATP, the primary energy currency of the cell .
In addition to its role in energy production, HADHB is also involved in the metabolism of branched-chain amino acids, such as leucine . This enzyme’s activity is crucial for maintaining energy homeostasis, especially during periods of fasting or prolonged exercise when fatty acids become the primary energy source.
Mutations in the HADHB gene can lead to metabolic disorders, such as mitochondrial trifunctional protein deficiency (MTPD) and long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHAD). These disorders are characterized by the accumulation of fatty acids and their derivatives, leading to symptoms such as hypoglycemia, muscle weakness, and cardiomyopathy .
Recombinant HADHB is used in research to study these metabolic disorders and to develop potential therapeutic interventions. The availability of human recombinant HADHB allows for detailed biochemical and structural studies, which are essential for understanding the enzyme’s function and its role in disease.