SORD Human, His adopts a homotetrameric structure (four identical subunits), similar to its native form. Each subunit contains:
Catalytic Sites: Serine and histidine residues coordinated with zinc, enabling NAD⁺-dependent oxidation-reduction reactions .
Subunit Interactions: Stabilized by hydrophobic effects, hydrogen bonds, and electrostatic interactions between subunits .
SORD belongs to the medium-chain dehydrogenase/reductase (MDR) superfamily, sharing structural homology with alcohol dehydrogenases (ADHs). Unlike dimeric ADHs, SORD’s tetrameric structure is conserved across species, from bacteria to humans .
The enzyme facilitates the reversible conversion:
This reaction bypasses ATP-dependent pathways, enabling efficient fructose production in tissues like the liver and kidney .
Biallelic mutations in SORD (e.g., c.753delG or c.757delG) cause autosomal recessive neuropathies (e.g., Charcot-Marie-Tooth disease type 2), characterized by:
Elevated Sorbitol Levels: >100× higher in patients vs. controls, leading to osmotic stress and neuronal damage .
Disease Severity Correlation: Higher sorbitol levels correlate with worse motor/upper limb symptoms and disability .
ARIs (e.g., AT-007) target the upstream polyol pathway enzyme aldose reductase, reducing sorbitol accumulation and mitigating neuropathy. Preclinical models show:
Sorbitol Reduction: AT-007 lowers intracellular sorbitol to near-normal levels in patient-derived cells .
Motor Recovery: Improved motor neuron function in Drosophila models and human cell cultures .
Phase III Trials: Ongoing for AT-007 in SORD-deficiency neuropathy, with a focus on serum sorbitol as a biomarker .
Diagnostic Challenges: SORD mutations are often misattributed to its pseudogene SORD2P, complicating genetic testing .
Feature | SORD Human, His (ENZ-1151) | SORD Human, His (ENZ-520) |
---|---|---|
Amino Acid Sequence | 357 residues + His tag | 357 residues + His tag |
Molecular Mass | 38.3 kDa | 40.4 kDa |
Purity | >90% (SDS-PAGE) | >90% (SDS-PAGE) |
Storage Conditions | -20°C (long-term), 4°C (short-term) | -20°C (long-term), 4°C (short-term) |
Activity | >15 units/mg | Not specified |
Allele Frequency: The c.757delG variant has an allele frequency of ~0.5% in healthy controls, making it a common pathogenic allele .
Geographic Distribution: SORD mutations are identified in patients from Europe, Asia, the Middle East, and the Americas .
Biomarkers: Fasting serum sorbitol levels serve as a reliable diagnostic marker for SORD neuropathy .
Sorbitol dehydrogenase (SORD) is a critical enzyme in the polyol pathway that catalyzes the second step of this metabolic pathway, converting sorbitol to fructose. In humans, SORD functions as a NAD+-dependent enzyme that plays an essential role in glucose metabolism. When this pathway is disrupted due to SORD deficiency, sorbitol accumulates in tissues and blood, with levels over 100 times higher than normal in affected patients . The polyol pathway becomes particularly important under hyperglycemic conditions, making SORD relevant to both normal physiology and pathological states.
Methodologically, researchers studying SORD function typically employ enzyme activity assays using purified protein or tissue homogenates, with standardized spectrophotometric measurement of NADH production as sorbitol is converted to fructose.
The human SORD gene (NM_003104.6) encodes the sorbitol dehydrogenase protein . A particular challenge in SORD research is the presence of a pseudogene, SORD2P, which has significant sequence similarity to SORD and has historically complicated genetic analysis . SORD is expressed in multiple tissues, with particularly notable expression in peripheral nerves, which explains the neuropathic phenotype when the gene is defective.
To study SORD expression patterns, researchers should employ quantitative PCR with careful primer design to distinguish between SORD and SORD2P, complemented by Western blotting and immunohistochemistry using validated antibodies.
Histidine residues play critical roles in SORD structure and catalytic function. The His135 residue in particular appears to be essential for proper protein folding and stability, as the His135Arg mutation results in aggregate formation and decreased protein solubility . Histidine residues often participate in metal coordination, hydrogen bonding networks, and catalytic mechanisms in dehydrogenases.
Researchers investigating histidine functions in SORD should consider site-directed mutagenesis studies accompanied by circular dichroism spectroscopy to assess structural changes, thermal shift assays to measure protein stability, and activity assays to determine functional impacts.
The His135Arg mutation causes significant structural disruption to SORD. Research shows that while wild-type SORD displays diffuse intracellular staining, the His135Arg mutant forms distinct granular aggregates throughout the cytosol . This aggregation correlates with reduced solubility, as demonstrated by significantly decreased levels of the mutant protein in soluble fractions and increased presence in insoluble fractions during biochemical fractionation experiments .
Table 1: Solubility Comparison Between Wild-Type and Mutant SORD
SORD Variant | Soluble Fraction (% of total) | Insoluble Fraction (% of total) | Aggregate Formation |
---|---|---|---|
Wild-Type | 85-95% | 5-15% | Minimal |
His135Arg | 30-40% | 60-70% | Extensive |
Ala253GlnfsTer27 | <10% | >90% | Complete |
For researchers studying the structural impacts of SORD mutations, a comprehensive approach should include:
Immunofluorescence microscopy with appropriate controls
Biochemical fractionation to quantify soluble vs. insoluble protein
Dynamic light scattering to characterize aggregate size distribution
In silico molecular dynamics simulations to predict structural perturbations
SORD deficiency manifests as a slowly progressive hereditary motor neuropathy with distinct clinical features. Patients typically present with muscle weakness in the lower limbs during their teenage years (mean age of onset: 12.5 ± 3.5 years) . The disease follows a distal-to-proximal progression pattern, beginning with atrophy in the feet and legs, later affecting the intrinsic muscles of the hands .
Electrophysiological examination reveals length-dependent axonal motor neuropathy with:
Decreased compound muscle action potential amplitudes
Mild reduction in motor nerve conduction velocities in lower limb nerves
Preserved sensory nerve conduction velocities and action potentials
A definitive diagnosis requires genetic testing showing biallelic pathogenic variants in SORD, supported by elevated blood sorbitol levels (>100x normal) .
SORD deficiency represents a significant portion of previously undiagnosed hereditary neuropathies. Research indicates it accounts for approximately 10% of undiagnosed Charcot-Marie-Tooth type 2 (CMT2) cases . The c.753delG (p.Ala253GlnfsTer27) variant has an allele frequency of ~0.5% in healthy control populations, making it one of the most common pathogenic alleles in humans .
Unlike other forms of CMT that may present with significant sensory involvement, SORD deficiency predominantly affects motor neurons with minimal sensory disruption. This clinical distinction, combined with elevated sorbitol levels as a biomarker, provides a means to differentiate SORD-related neuropathy from other genetic causes.
A significant challenge in SORD genetic research is the presence of the pseudogene SORD2P, which shares high sequence homology with functional SORD. The most common pathogenic variant (c.753delG) is constitutively present in SORD2P, which had previously led to misalignment of whole-exome sequencing reads and missed diagnoses .
When designing genetic studies for SORD, researchers should implement:
Long-read sequencing technologies that can span ambiguous regions
Custom capture methods specifically designed to distinguish SORD from SORD2P
Validation with Sanger sequencing using unique primers that discriminate between the gene and pseudogene
Functional validation using ex vivo cDNA PCR and protein expression studies
For novel SORD variants, comprehensive functional validation is essential. Based on published research, an effective validation pipeline includes:
Ex vivo cDNA PCR to confirm splicing defects for intronic or potential splice-affecting variants (as demonstrated with the c.908+1 G>C variant)
In vitro expression studies using tagged constructs to assess protein localization, solubility, and aggregation propensity
Biochemical fractionation to quantify soluble vs. insoluble protein fractions
Enzyme activity assays to measure the catalytic function of mutant proteins
Structural modeling to predict the impact of amino acid substitutions on protein folding and stability
When reporting novel variants, researchers should provide evidence across multiple assays rather than relying on a single functional test.
Selecting appropriate model systems is crucial for advancing SORD research. Based on current literature, researchers should consider:
Cellular Models:
HeLa cells for transfection studies and protein localization (as used in the His135Arg characterization)
Primary fibroblasts from patients for physiological studies of endogenous SORD
iPSC-derived motor neurons to model the cell type most affected in the disease
Animal Models:
SORD knockout mice to study systemic effects of SORD deficiency
Drosophila models for high-throughput screening of genetic modifiers and potential therapeutics
C. elegans for studying conserved aspects of sorbitol metabolism
Each model system offers distinct advantages, and researchers should select based on their specific research questions while acknowledging the limitations of each system in their experimental design and interpretation.
Sorbitol accumulation serves as a valuable biomarker for SORD deficiency, with patient serum levels reported to be over 100 times higher than normal . Accurate measurement requires:
Liquid chromatography-mass spectrometry (LC-MS/MS) for precise quantification in biological samples
Enzymatic assays using sorbitol dehydrogenase from alternative sources
Sample preparation protocols that minimize sorbitol degradation or metabolism during processing
Appropriate control samples matched for age, sex, and other relevant variables
When reporting sorbitol levels, researchers should include detailed methodology, reference ranges, and statistical analysis to facilitate cross-study comparisons.
Aldose reductase inhibitors (ARIs) represent a promising therapeutic approach for SORD deficiency based on their mechanism of action within the polyol pathway. Aldose reductase catalyzes the first step in the pathway, converting glucose to sorbitol, which is then normally converted to fructose by SORD . In SORD deficiency, sorbitol accumulates to toxic levels.
ARIs block the production of sorbitol at the source, potentially preventing its pathological accumulation. This approach addresses the biochemical defect upstream of the genetic mutation, offering a mechanistically sound therapeutic strategy that doesn't require correcting the underlying genetic defect.
Researchers investigating ARIs should assess:
Dose-dependent effects on sorbitol levels in patient-derived cells
Potential off-target effects through comprehensive metabolomic profiling
Comparative efficacy of different ARI compounds
Bioavailability in target tissues, particularly peripheral nerves
Gene therapy approaches for SORD deficiency remain in early research stages but hold significant potential. Key considerations for researchers in this area include:
Delivery challenges: Research should focus on developing vectors capable of efficiently targeting peripheral nerves where SORD function is critical
Expression regulation: Design of promoter elements that recapitulate physiological SORD expression patterns
Gene editing approaches: CRISPR-Cas9 strategies to correct common mutations like c.753delG
Alternative splicing modulation: For splice-affecting variants like c.908+1 G>C, antisense oligonucleotides might redirect splicing to restore normal transcript processing
Gene therapy studies should incorporate long-term safety assessments and functional outcome measures relevant to the progressive neuropathic phenotype in SORD deficiency.
Sorbitol Dehydrogenase (SORD), also known as L-iditol 2-dehydrogenase or SORD1, is an enzyme that plays a crucial role in the polyol pathway. This enzyme is responsible for the zinc-dependent interconversion of polyols, such as sorbitol and xylitol, to their respective ketoses . The recombinant form of this enzyme, tagged with a His-tag, is widely used in research to study its structure, function, and potential therapeutic applications.
The recombinant human Sorbitol Dehydrogenase is typically expressed in Escherichia coli (E. coli) and purified using conventional chromatography techniques . The protein consists of 357 amino acids and has a molecular weight of approximately 40.4 kDa . The His-tag, which is fused to the N-terminus of the protein, facilitates its purification and detection.
Sorbitol Dehydrogenase catalyzes the conversion of sorbitol to fructose, a reaction that is essential for the metabolism of glucose and fructose . This enzyme is widely expressed in various tissues, with the highest expression observed in the kidney and the lens of the eye . The specific activity of the recombinant enzyme is greater than 20,000 pmol/min/µg, indicating its high catalytic efficiency .
The recombinant form of Sorbitol Dehydrogenase is used extensively in biochemical and physiological studies. Researchers utilize this enzyme to investigate its role in diabetic complications, such as diabetic retinopathy and neuropathy, where the accumulation of sorbitol is implicated . Additionally, the enzyme is used in studies related to fructose metabolism and its impact on various metabolic disorders.