SUOX Human

Sulfite Oxidase Human Recombinant
Shipped with Ice Packs
In Stock

Description

Definition and Biochemical Properties

SUOX Human refers to the recombinant human sulfite oxidase protein, a mitochondrial enzyme critical for sulfur amino acid metabolism. Produced in E. coli, SUOX Human is a non-glycosylated polypeptide with a molecular mass of 53.9 kDa, comprising 489 amino acids (residues 80–545) . It is a homodimeric protein with two distinct domains:

  • Heme-binding domain: Anchors a heme group (cytochrome b5) for electron transfer.

  • Molybdopterin-binding domain: Contains a molybdenum cofactor (MoCo) essential for catalysis .

Table 1: Key Biochemical Features of SUOX Human

PropertyDescriptionSource
Molecular Mass53.9 kDa (monomer)
StructureHomodimer with heme and molybdopterin domains
CofactorsMolybdenum (Mo), iron (heme)
Expression SystemRecombinant E. coli
Purity>90% (SDS-PAGE)

Functional Role in Sulfur Amino Acid Metabolism

SUOX Human catalyzes the oxidation of sulfite (SO₃²⁻) to sulfate (SO₄²⁻), the terminal step in cysteine and methionine degradation . This reaction prevents toxic sulfite accumulation and maintains cellular redox balance.

Mechanism:

  1. Sulfite → Sulfate: Transfer of an oxygen atom from water to sulfite, reducing molybdenum (MoVI → MoIV).

  2. Electron Transfer: Electrons from MoIV are relayed via heme to cytochrome c, linking the enzyme to mitochondrial oxidative phosphorylation .

Clinical Implications of SUOX Deficiency

Mutations in the SUOX gene cause isolated sulfite oxidase deficiency (ISOD), a severe neurometabolic disorder.

Key Features of ISOD:

  • Neurological Symptoms: Seizures, developmental delay, and brain damage due to sulfite toxicity .

  • Ocular Manifestations: Ectopia lentis (lens dislocation) in 50–60% of cases .

  • Biochemical Markers: Elevated urine sulfite and S-sulfocysteine; low homocysteine in blood spots .

Table 2: SUOX Mutations and Associated Pathology

Mutation TypeExample VariantsFunctional ImpactSource
Nonsensep.Glu159*Truncated protein, loss of molybdopterin domain
Missensep.Lys401GluReduced molybdopterin binding, enzyme instability
Compound Heterozygousp.A69P + p.Y400*Mild symptoms, later onset, and regression

Laboratory Use:

  • Enzymatic Assays: SUOX Human is used to study sulfite oxidation kinetics and cofactor interactions .

  • Genetic Testing: Identified via next-generation sequencing (NGS) in ISOD cases .

Genotype-Phenotype Correlations:

  • Missense Mutations: Associated with milder symptoms (e.g., isolated ectopia lentis) and delayed onset .

  • Nonsense Mutations: Lead to severe early-onset ISOD with brain damage .

Table 3: SUOX Mutation Spectrum

Mutations IdentifiedFrequencyPopulation
Missense44/59Global
Nonsense10/59Global
Deletions/Insertions5/59Global

Data from LOVD SUOX database .

Novel Variants and Their Effects:

  • p.Lys401Glu: Disrupts molybdopterin binding, reducing catalytic efficiency .

  • p.Glu159*: Truncates the protein, eliminating the molybdopterin domain .

Molecular Modeling:

  • p.A69P Mutation: Causes structural instability but preserves sulfite affinity .

  • p.Y400*: Impairs heme binding, reducing electron transfer capacity .

Product Specs

Introduction
Sulfite oxidase (SUOX) is a mitochondrial enzyme that catalyzes the oxidation of sulfite to sulfate. This reaction is the final step in the degradation of sulfur-containing amino acids. SUOX deficiency is a rare genetic disorder that can cause severe neurological problems.
Description
Recombinant human SUOX protein was expressed in E. coli and purified to greater than 90% purity. The protein is a single, non-glycosylated polypeptide chain with a molecular mass of 53.9 kDa. The recombinant protein includes a 23 amino acid His-tag at the N-terminus.
Physical Appearance
Clear, colorless liquid.
Formulation
The SUOX protein is supplied as a 1 mg/mL solution in phosphate-buffered saline (pH 7.4), 30% glycerol, and 1 mM DTT.
Stability
The protein is stable for 2-4 weeks at 4°C. For long-term storage, the protein should be stored at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid multiple freeze-thaw cycles.
Purity
Greater than 90% pure as determined by SDS-PAGE analysis.
Synonyms
Sulfite Oxidase, EC 1.8.3.1, Sulfite oxidase, mitochondrial.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSESTHIYT KEEVSSHTSP ETGIWVTLGS EVFDVTEFVD LHPGGPSKLM LAAGGPLEPF WALYAVHNQS HVRELLAQYK IGELNPEDKV APTVETSDPY ADDPVRHPAL KVNSQRPFNA EPPPELLTEN YITPNPIFFT RNHLPVPNLD PDTYRLHVVG APGGQSLSLS LDDLHNFPRY EITVTLQCAG NRRSEMTQVK EVKGLEWRTG AISTARWAGA RLCDVLAQAG HQLCETEAHV CFEGLDSDPT GTAYGASIPL ARAMDPEAEV LLAYEMNGQP LPRDHGFPVR VVVPGVVGAR HVKWLGRVSV QPEESYSHWQ RRDYKGFSPS VDWETVDFDS APSIQELPVQ SAITEPRDGE TVESGEVTIK GYAWSGGGRA VIRVDVSLDG GLTWQVAKLD GEEQRPRKAW AWRLWQLKAP VPAGQKELNI VCKAVDDGYN VQPDTVAPIW NLRGVLSNAW HRVHVYVSP.

Q&A

How is the SUOX protein structured and what cofactors does it require?

The SUOX enzyme is a homodimeric protein located in the mitochondrial intermembrane space. Each subunit has a distinct molecular architecture featuring two primary domains: a heme domain and a molybdopterin-binding domain . The complete functional enzyme has a molecular mass of approximately 53.9 kDa per monomer, as indicated by recombinant protein analysis .
SUOX requires two essential cofactors for catalytic activity:

  • Molybdenum: Incorporated into the molybdopterin cofactor within the molybdopterin-binding domain, which is critical for the redox reaction catalyzed by the enzyme .

  • Iron: Present in the heme domain and also crucial for electron transfer during the oxidation reaction .
    The functional dependence on these cofactors is evidenced by the existence of molybdenum cofactor deficiency (MoCD), which presents with symptoms similar to ISOD but also affects other molybdenum-dependent enzymes such as xanthine dehydrogenase and aldehyde oxidase . The proper assembly of both domains with their respective cofactors is essential for SUOX to effectively catalyze the conversion of toxic sulfite to excretable sulfate .

What methods are used to measure SUOX activity in clinical and research settings?

Measuring SUOX activity involves several complementary approaches:
Biochemical Analysis:

  • Quantification of sulfite, thiosulfate, and S-sulfocysteine levels in urine, which are typically elevated in SUOX deficiency .

  • Analysis of homocysteine levels, which are often lower than normal in ISOD patients. This is an important diagnostic consideration as many laboratories focus primarily on elevated homocysteine levels for other conditions, potentially overlooking ISOD when levels are decreased .

  • Measurement of uric acid levels to distinguish ISOD from molybdenum cofactor deficiency (MoCD), as MoCD also affects xanthine dehydrogenase, resulting in reduced uric acid levels and elevated xanthine and hypoxanthine in urine .
    Enzyme Assays:

  • ELISA-based methods can quantitatively measure SUOX protein levels in human serum, plasma, and tissue samples using sandwich enzyme immunoassay techniques with antibodies specific for human SUOX .

  • Direct enzyme activity assays measuring the rate of sulfite to sulfate conversion, typically using spectrophotometric methods.
    Genetic Analysis:

  • Next-generation sequencing (NGS) to identify mutations in the SUOX gene .

  • Sanger sequencing for confirmation of candidate variants identified through NGS .
    These methodologies provide complementary data, with biochemical tests offering immediate clinical relevance and genetic analyses providing definitive molecular diagnosis and potential for genetic counseling .

How do specific mutations in the SUOX gene correlate with clinical phenotypes of ISOD?

The correlation between SUOX gene mutations and ISOD clinical phenotypes demonstrates significant genotype-phenotype relationships that impact disease severity and progression . Research indicates that:
Different types of mutations produce varying effects:

  • Nonsense mutations (e.g., c.1200C>G resulting in p.Y400*) typically cause complete loss of enzyme function, resulting in severe early-onset disease with profound neurological symptoms .

  • Frameshift mutations (e.g., c.1549_1574dup resulting in p.I525Mfs*102) alter the reading frame, generally leading to non-functional protein and severe disease manifestation .

  • Missense mutations may permit residual enzyme activity, potentially resulting in attenuated phenotypes with later onset and slower progression, depending on the specific amino acid substitution and its location within functional domains .
    Clinical manifestation patterns commonly include:

  • Early-onset cases (neonatal or early infantile) typically presenting with seizures resistant to anticonvulsant medications

  • Rapidly progressive encephalopathy resembling neonatal hypoxic-ischemic injury

  • Development of microcephaly and feeding difficulties

  • Dislocated ocular lenses in some patients

  • Less commonly reported features including elevated creatine kinase, creatine kinase isoenzyme, and lactate dehydrogenase, as documented in recent case reports
    Recent research has expanded the mutation spectrum through identification of novel variants in diverse populations, including compound heterozygous cases in non-consanguineous Chinese families . These findings suggest that geographic and ethnic factors may influence the distribution of specific mutations. Comprehensive genotype-phenotype studies require international collaboration and detailed clinical phenotyping to better understand the full spectrum of ISOD manifestations and their relationship to specific SUOX mutations .

What are the current challenges in developing therapeutic approaches for SUOX deficiency?

Developing effective therapeutics for SUOX deficiency presents several significant challenges:
Targeting the Root Cause:

  • SUOX deficiency results from genetic mutations affecting enzyme expression or function, requiring gene therapy or protein replacement approaches that can effectively target mitochondria .

  • The blood-brain barrier presents a significant obstacle for delivering therapeutic agents to the central nervous system, where the most devastating effects of ISOD occur .
    Managing Disease Progression:

  • ISOD typically manifests early with rapid progression of neurological damage, creating a narrow therapeutic window before irreversible damage occurs .

  • Currently, there is "no long-term effective treatment, and patients often die in infancy" , highlighting the urgent need for early intervention strategies.
    Research Limitations:

  • The rarity of ISOD (fewer than 50 reported cases worldwide) complicates clinical trials and treatment development .

  • Limited availability of appropriate animal models that accurately recapitulate human ISOD pathophysiology hampers preclinical testing.

  • The complexity of the enzyme structure, requiring both molybdenum and iron cofactors, complicates enzyme replacement approaches .
    Potential therapeutic strategies under investigation include:

  • Dietary modification to reduce intake of sulfur-containing amino acids

  • Molybdenum supplementation in cases where cofactor binding is affected

  • Novel approaches to reduce sulfite accumulation through alternative metabolic pathways

  • Gene therapy approaches targeting SUOX expression
    The development of effective treatments will likely require multidisciplinary approaches combining genetic diagnostics, biochemical intervention, and possibly gene therapy strategies tailored to specific mutations .

How can recombinant SUOX protein be effectively produced and purified for structural and functional studies?

Production and purification of recombinant SUOX protein for research applications involves several critical considerations:
Expression Systems:

  • E. coli expression systems have been successfully employed to produce recombinant human SUOX, as evidenced by commercial preparations yielding single, non-glycosylated polypeptide chains containing 489 amino acids (residues 80-545) with a molecular mass of 53.9kDa .

  • Fusion tags, particularly N-terminal His-tags (e.g., 23 amino acid His-tag), facilitate purification while maintaining enzyme function .

  • Alternative expression systems, including yeast or mammalian cells, may be considered for producing protein with post-translational modifications more closely resembling native human SUOX.
    Purification Strategy:

  • Multi-step chromatographic techniques are essential for obtaining high-purity SUOX (>90% as determined by SDS-PAGE) .

  • Affinity chromatography utilizing His-tag affinity is typically employed as an initial capture step.

  • Further purification may involve ion exchange, size exclusion, or other specialized chromatographic methods.
    Cofactor Integration:

  • Ensuring proper incorporation of molybdopterin and heme cofactors is critical for producing functionally active enzyme .

  • Supplementation of growth media or post-purification reconstitution with molybdenum and iron may be necessary.
    Stability Considerations:

  • Optimal buffer formulation (e.g., phosphate-buffered saline at pH 7.4 with 30% glycerol and 1mM DTT) helps maintain protein stability .

  • For long-term storage, addition of carrier proteins (0.1% HSA or BSA) can enhance stability .

  • Storage recommendations include keeping the protein at 4°C for short-term use (2-4 weeks) or frozen at -20°C for longer periods, avoiding multiple freeze-thaw cycles .
    Functional Verification:

  • Activity assays measuring sulfite to sulfate conversion are essential for confirming that the recombinant protein retains catalytic function.

  • Structural studies using X-ray crystallography or cryo-electron microscopy may require specific modifications to purification protocols to yield protein suitable for crystallization.
    These methodological approaches provide a foundation for producing high-quality recombinant SUOX for various research applications, from biochemical characterization to structural studies and therapeutic development .

What are the optimal methods for genetic testing and diagnosis of ISOD?

Establishing an effective genetic testing and diagnostic strategy for ISOD requires a comprehensive approach integrating multiple techniques:
Next-Generation Sequencing (NGS):

  • Targeted gene panel sequencing focusing on sulfur metabolism genes, particularly SUOX and molybdenum cofactor synthesis genes (MOCS1, MOCS2, MOCS3, and GEPH), provides efficient first-line screening .

  • Whole exome sequencing (WES) may be preferred when clinical suspicion is high but targeted panels yield negative results, as it can identify novel variants or genes potentially associated with similar phenotypes .

  • NGS data analysis should include thorough coverage analysis to ensure complete assessment of the SUOX gene, as some regions may be poorly captured .
    Confirmation and Validation:

  • Sanger sequencing should be employed to verify candidate variants identified through NGS, particularly novel variants that haven't been previously reported .

  • When novel variants are identified, parental testing is essential to confirm their inheritance pattern and phase (compound heterozygosity versus homozygosity) .

  • According to ACMG/AMP guidelines, comprehensive pathogenicity analysis of novel variants should be performed, including in silico prediction tools, conservation analysis, and functional impact assessment .
    Biochemical Correlation:

  • Diagnostic genetic findings should be correlated with biochemical markers, including urinary sulfite, thiosulfate, and S-sulfocysteine levels .

  • Assessment of plasma homocysteine (typically lower than normal in ISOD) and uric acid levels (normal in ISOD but decreased in MoCD) helps distinguish between sulfite oxidase deficiency and molybdenum cofactor deficiency .

  • Whenever possible, direct enzyme activity measurement in accessible tissues provides valuable confirmation of genetic findings.
    Time-Sensitive Considerations:

  • Given the rapid progression and severe consequences of ISOD, establishing a "comprehensive genetic diagnosis strategy... within a limited time" is crucial .

  • Rapid turnaround protocols for NGS and confirmatory testing should be implemented when ISOD is clinically suspected.

  • Prenatal and preimplantation genetic diagnosis options should be made available to affected families for future pregnancies .
    This multi-faceted approach enables accurate diagnosis of ISOD and provides the foundation for genetic counseling, family planning, and potential future targeted therapies .

How can researchers develop effective animal models for studying SUOX deficiency?

Developing animal models that accurately recapitulate human SUOX deficiency requires strategic consideration of several factors:
Selection of Appropriate Species:

  • Mice models offer advantages of genetic manipulability and relatively fast reproduction cycles, making them suitable for studying basic mechanisms of SUOX deficiency.

  • Larger animal models (e.g., pigs) may better approximate human neurological development and could provide more translatable insights for therapeutic testing.

  • Multiple model systems may be necessary to address different aspects of the disease, from molecular mechanisms to therapeutic development.
    Genetic Modification Strategies:

  • Knockout models: Complete SUOX gene knockout may be lethal during embryonic development, necessitating conditional knockout approaches.

  • Knockin models incorporating human SUOX mutations: These can more accurately reflect the specific molecular defects observed in patients, particularly for studying missense mutations with residual enzyme activity .

  • Conditional expression systems allow for temporal control of SUOX deficiency, enabling study of both developmental and acute effects.
    Evaluation Parameters:

  • Biochemical analysis: Measurement of sulfite, thiosulfate, and S-sulfocysteine levels to confirm biochemical phenotype .

  • Neurological assessment: Comprehensive evaluation of seizure activity, neuronal loss, brain development, and behavioral changes.

  • Histopathological examination: Assessment of tissue damage patterns, particularly in the central nervous system.

  • Molecular characterization: Analysis of compensatory mechanisms and downstream effects on related metabolic pathways.
    Translational Considerations:

  • Models should incorporate biomarkers that can be monitored non-invasively to facilitate translation to clinical applications.

  • Developmental timing differences between humans and model organisms must be carefully considered when interpreting results.

  • Dose-response studies for potential therapeutics should account for species-specific differences in drug metabolism and distribution.
    Ethical and Practical Challenges:

  • Animal welfare concerns are particularly relevant for models with severe neurological phenotypes.

  • Development of less severe models that still capture key disease aspects may be preferable for long-term studies and therapeutic testing.

  • Collaborative approaches between multiple research groups may help maximize knowledge gained while minimizing animal usage.
    Effective animal models must balance physiological relevance with practical considerations to advance understanding of SUOX deficiency pathophysiology and facilitate development of therapeutic interventions .

What techniques are most effective for studying the structural and functional impacts of SUOX mutations?

Investigating the structural and functional consequences of SUOX mutations requires an integrated approach combining several advanced methodologies:
Structural Analysis Techniques:

  • X-ray crystallography provides high-resolution structural data of wild-type and mutant SUOX proteins, revealing how specific mutations affect protein folding, cofactor binding, and active site geometry .

  • Cryo-electron microscopy (cryo-EM) offers advantages for examining large protein complexes and capturing alternative conformational states that may be affected by mutations.

  • Computational modeling and molecular dynamics simulations can predict structural perturbations caused by specific mutations, particularly useful when experimental structures are challenging to obtain .

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS) can reveal regional flexibility and solvent accessibility changes induced by mutations.
    Functional Characterization Methods:

  • Enzyme kinetics assays measuring sulfite oxidation rates provide direct quantification of how mutations affect catalytic efficiency, substrate binding, and product formation .

  • Thermostability assessments using differential scanning fluorimetry or calorimetry can determine if mutations destabilize the protein structure.

  • Cofactor binding studies utilizing isothermal titration calorimetry or surface plasmon resonance can reveal changes in molybdenum or heme binding affinity and kinetics .

  • Cellular localization studies using fluorescence microscopy with tagged proteins can determine if mutations disrupt mitochondrial targeting or inter-membrane space localization.
    Expression Systems for Mutant Analysis:

  • Site-directed mutagenesis in bacterial expression systems allows production of specific mutant proteins for in vitro characterization .

  • Mammalian cell models expressing mutant SUOX variants can reveal cellular consequences of mutations, including effects on mitochondrial function and cellular metabolism.

  • Patient-derived cell lines (such as fibroblasts or induced pluripotent stem cells) provide physiologically relevant contexts for studying naturally occurring mutations .
    Integration of Clinical Data:

  • Correlation of biochemical phenotypes (sulfite levels, enzyme activity) with specific structural and functional changes provides insights into structure-function relationships .

  • Analysis of mutation clusters in the three-dimensional structure can reveal particularly important functional regions of the protein.
    These complementary approaches collectively enable comprehensive characterization of how SUOX mutations disrupt enzyme function at molecular, cellular, and organismal levels, potentially identifying mutation-specific therapeutic strategies .

How do SUOX deficiency biomarkers correlate with disease severity and progression?

The relationship between biochemical markers and clinical outcomes in SUOX deficiency demonstrates important correlations that can guide both diagnosis and prognosis:
Primary Biomarkers and Their Clinical Significance:

  • Urinary sulfite, thiosulfate, and S-sulfocysteine levels serve as direct indicators of SUOX dysfunction, with higher concentrations generally correlating with more severe enzyme deficiency .

  • Plasma homocysteine levels are typically reduced in ISOD patients, with the degree of reduction potentially reflecting disease severity. This has been observed in at least eight examined patients with "obviously lower homocysteine level than normal controls" .

  • Neuroimaging findings resembling hypoxic-ischemic injury provide another quantifiable marker that correlates with clinical severity, particularly regarding central nervous system damage .
    Emerging Biomarkers:

  • Recent research has identified additional biochemical abnormalities in ISOD patients, including "increase in creatine kinase, creatine kinase isoenzyme, and lactate dehydrogenase," which may represent novel biomarkers for disease monitoring .

  • These markers potentially reflect secondary effects of sulfite toxicity on energy metabolism and tissue damage.
    Temporal Patterns:

  • Early and rapidly elevated toxic metabolites generally correlate with neonatal or early infantile onset and more severe clinical course .

  • The rate of increase in sulfite and related metabolites may serve as a predictor of disease progression speed.

  • Longitudinal monitoring of these biomarkers can potentially track therapeutic efficacy if interventions are attempted.
    Genotype-Biomarker-Phenotype Relationships:

  • Different SUOX mutations may produce varying levels of residual enzyme activity, resulting in distinct biochemical profiles that correlate with phenotypic severity .

  • Complete loss-of-function mutations typically lead to higher toxic metabolite levels and more severe clinical manifestations compared to mutations that permit some residual activity .

  • Compound heterozygous patients with two different mutations may show intermediate biochemical profiles reflecting the combined effect of both variants .
    Understanding these correlations can improve clinical management by:

  • Providing prognostic information based on initial biomarker levels

  • Enabling personalized monitoring protocols based on specific genetic variants

  • Identifying potential therapeutic windows for intervention

  • Establishing objective measures for assessing treatment efficacy in future clinical trials

What are the differential diagnostic considerations when evaluating suspected SUOX deficiency?

Accurate differential diagnosis of SUOX deficiency requires systematic evaluation to distinguish it from conditions with overlapping clinical presentations:
Primary Differential Diagnosis:

  • Molybdenum Cofactor Deficiency (MoCD): Presents with similar neurological manifestations but can be distinguished by laboratory findings including reduced uric acid levels and elevated xanthine and hypoxanthine in urine due to concurrent xanthine dehydrogenase deficiency . SUOX deficiency maintains normal uric acid levels.

  • Hypoxic-Ischemic Encephalopathy: Neuroimaging findings in ISOD often resemble those of hypoxic-ischemic injury, creating potential for misdiagnosis, particularly in neonatal presentations . Detailed birth history and biochemical testing are crucial for differentiation.

  • Other Early-Onset Epileptic Encephalopathies: Various genetic epilepsies can present with similar seizure patterns and developmental regression, requiring comprehensive genetic and metabolic evaluation.
    Diagnostic Approach:

  • Clinical Assessment:

    • Evaluate for characteristic features: seizures resistant to anticonvulsant medications, rapidly progressive encephalopathy, microcephaly, feeding difficulties, and dislocated ocular lenses .

    • Document timing of symptom onset, as ISOD typically manifests in the neonatal or early infantile period.

  • First-Line Biochemical Testing:

    • Urinary sulfite, thiosulfate, and S-sulfocysteine levels (elevated in ISOD)

    • Plasma homocysteine levels (typically lower than normal in ISOD)

    • Serum uric acid levels (normal in ISOD, decreased in MoCD)

  • Neuroimaging:

    • MRI typically shows cortical atrophy, white matter changes, and cystic lesions resembling hypoxic-ischemic injury

    • Sequential imaging may reveal progressive nature of changes consistent with ongoing metabolic damage

  • Genetic Testing:

    • NGS-based targeted gene panel including SUOX and molybdenum cofactor synthesis genes (MOCS1, MOCS2, MOCS3, and GEPH)

    • Whole exome sequencing when targeted testing is negative despite strong clinical suspicion

    • Confirmation of variants by Sanger sequencing and parental testing

  • Additional Considerations:

    • Enzyme activity testing in appropriate tissues when available

    • Evaluation for recently identified associated features like recurrent pneumonia, umbilical hernia, and inguinal oblique femoral hernia
      This systematic approach enables clinicians to accurately diagnose ISOD, distinguish it from similar conditions, and provide appropriate genetic counseling to affected families .

How does SUOX interact with other metabolic pathways in cellular homeostasis?

SUOX occupies a critical position within cellular metabolism, with connections to multiple pathways that collectively maintain cellular homeostasis:
Sulfur Amino Acid Metabolism:

  • SUOX catalyzes the final step in the oxidative degradation of sulfur-containing amino acids cysteine and methionine, converting toxic sulfite to excretable sulfate .

  • This reaction represents a critical junction between amino acid catabolism and cellular detoxification systems.

  • Impaired SUOX function leads to accumulation of upstream metabolites including sulfite, thiosulfate, and S-sulfocysteine, which have downstream toxic effects .
    Redox Homeostasis:

  • The SUOX-catalyzed reaction involves electron transfer, with electrons ultimately transferred to cytochrome c in the respiratory chain, connecting sulfur metabolism to mitochondrial energetics .

  • Sulfite is a potent reductant that can disrupt cellular redox balance when accumulated, potentially explaining the oxidative stress observed in SUOX deficiency.

  • The enzyme's dependence on molybdenum and iron cofactors links it to cellular metal homeostasis and distribution systems .
    Mitochondrial Function:

  • Localized to the mitochondrial intermembrane space, SUOX is integrated with mitochondrial protein import and folding systems .

  • Dysfunction in SUOX may trigger mitochondrial stress responses and potentially impact energy production.

  • Recent findings of elevated creatine kinase, creatine kinase isoenzyme, and lactate dehydrogenase in ISOD patients suggest secondary effects on energy metabolism .
    Developmental Processes:

  • The severe neurological impact of SUOX deficiency indicates its particular importance in neural development and function .

  • The temporal pattern of symptom emergence in ISOD suggests critical periods where SUOX activity is especially vital for normal brain development.

  • Potential interactions with developmental signaling pathways may explain the particular vulnerability of the central nervous system to SUOX deficiency.
    Systemic Integration:

  • Beyond neurological effects, SUOX deficiency has been associated with systemic manifestations including recurrent pneumonia, umbilical hernia, and inguinal oblique femoral hernia .

  • These diverse effects suggest broader interactions between sulfur metabolism and connective tissue development or maintenance.

  • Recent research has also linked SUOX genetic variations to conditions like polycystic ovarian syndrome (PCOS), indicating potential roles in endocrine regulation .
    Understanding these metabolic interconnections provides insight into the multisystemic effects of SUOX deficiency and highlights potential points for therapeutic intervention beyond direct enzyme replacement .

What emerging technologies could advance early detection and treatment of SUOX deficiency?

Several cutting-edge technologies show promise for revolutionizing the diagnosis and management of SUOX deficiency:
Advanced Diagnostic Technologies:

  • Rapid whole genome sequencing (rWGS) in neonatal intensive care settings could enable diagnosis within 24-48 hours, critical for conditions like ISOD where early intervention may improve outcomes .

  • Development of newborn screening methods targeting S-sulfocysteine or other ISOD biomarkers could enable pre-symptomatic identification and intervention.

  • Metabolomic profiling using high-resolution mass spectrometry may identify novel biomarkers that predict disease severity or monitor treatment response with greater sensitivity.

  • Non-invasive imaging techniques such as functional MRI or PET with specialized tracers might detect early biochemical changes before structural damage occurs.
    Therapeutic Approaches:

  • Gene therapy using adeno-associated viral vectors (AAVs) with specific tropism for affected tissues could potentially deliver functional SUOX genes to target cells .

  • mRNA therapeutics delivering transient SUOX expression might provide a renewable source of functional enzyme without permanent genetic modification.

  • CRISPR-Cas9 gene editing technologies could potentially correct specific SUOX mutations in patient cells, though delivery to affected tissues remains challenging.

  • Enzyme replacement therapy using engineered variants with enhanced stability, blood-brain barrier penetration, and mitochondrial targeting could address the central pathophysiology .
    Supportive Technologies:

  • Bioengineered alternative metabolic pathways that bypass the need for sulfite oxidase activity might reduce toxic metabolite accumulation.

  • Development of small molecule chaperones that stabilize partially functional mutant SUOX proteins could enhance residual enzyme activity in patients with specific missense mutations.

  • Advanced biomarker monitoring systems for home use could enable more personalized management by detecting metabolic decompensation before clinical deterioration.
    Translational Research Infrastructure:

  • International patient registries with standardized data collection would facilitate natural history studies and identification of genotype-phenotype correlations in this rare disorder .

  • Biobanking of patient samples with comprehensive clinical data would accelerate research into disease mechanisms and biomarker discovery.

  • Development of validated cellular and animal models that accurately recapitulate human ISOD would enhance preclinical testing of novel therapeutics .
    These emerging technologies collectively offer hope for transforming ISOD from a devastating, untreatable condition to one where early detection and effective intervention become possible, potentially changing the natural history of the disease .

How might genetic variations in SUOX contribute to other human diseases beyond ISOD?

The potential role of SUOX genetic variations in conditions beyond classical ISOD represents an emerging area of research with significant implications:
Metabolic and Endocrine Disorders:

  • Genetic variations in SUOX have been linked to polycystic ovarian syndrome (PCOS), suggesting that altered sulfur metabolism may contribute to endocrine dysfunction .

  • Subclinical sulfite oxidase insufficiency might potentially contribute to metabolic syndromes through impaired handling of dietary sulfur compounds or altered redox homeostasis.

  • The connection between SUOX function and hormonal regulation merits further investigation to understand potential mechanistic links.
    Neurological Conditions:

  • Milder SUOX variants might contribute to less severe but more common neurological conditions, particularly those involving oxidative stress.

  • The spectrum of SUOX-related neurological manifestations likely extends beyond the severe phenotype of classical ISOD, potentially including migraine, certain epilepsy syndromes, or neurodegenerative conditions.

  • Carrier status for recessive SUOX mutations might confer subtle neurological effects or susceptibility to environmental stressors.
    Oxidative Stress-Related Pathologies:

  • Given SUOX's role in preventing sulfite-induced oxidative damage, genetic variations affecting enzyme efficiency could modify risk for conditions where oxidative stress plays a pathogenic role .

  • Cardiovascular diseases, neurodegeneration, and aging-related disorders might be influenced by SUOX functional variation.

  • The interaction between SUOX variants and environmental sulfite exposure (from food preservatives, industrial pollution, etc.) could represent an important gene-environment interaction.
    Population-Specific Considerations:

  • Recent identification of novel SUOX variants in different ethnic populations, including Chinese Han families, suggests population-specific genetic contributions that may influence disease susceptibility .

  • Comprehensive population genetic studies of SUOX variation could reveal selection pressures and adaptive significance of different variants.
    Research Methodologies to Explore These Associations:

  • Large-scale genomic association studies incorporating SUOX variants

  • Metabolomic profiling in carriers of SUOX variants to identify subclinical biochemical signatures

  • Cellular models examining how different SUOX variants affect response to oxidative stressors

  • Population studies examining interaction between dietary sulfite exposure and SUOX genotypes
    The expanding understanding of SUOX's contributions to human health and disease beyond classical ISOD highlights the importance of this enzyme in broader physiological processes and suggests potential for targeted interventions in multiple conditions .

What are the critical methodological challenges in researching rare metabolic disorders like SUOX deficiency?

Researching rare metabolic disorders such as SUOX deficiency presents unique methodological challenges that require innovative approaches:
Patient Recruitment and Cohort Assembly:

  • With fewer than 50 reported ISOD cases worldwide , assembling cohorts of sufficient size for statistically meaningful analysis is extremely difficult.

  • Geographic dispersion of cases necessitates international collaboration and standardized data collection protocols.

  • Phenotypic heterogeneity, even within patients sharing similar genetic variants, complicates group analyses and requires careful stratification.
    Diagnostic Standardization:

  • Biochemical testing for sulfite metabolites is not routinely available in many clinical laboratories, leading to potential underdiagnosis .

  • Lack of standardized reference ranges and testing methodologies across different centers complicates data comparison and meta-analysis.

  • Evolving diagnostic criteria and improved genetic testing capabilities create temporal biases in patient identification.
    Research Design Considerations:

  • Traditional randomized controlled trials are often infeasible due to small patient numbers and ethical considerations.

  • Alternative designs such as N-of-1 trials, natural history studies, and crossover designs may be more appropriate but present their own methodological challenges.

  • Control selection is particularly challenging given the unique metabolic profile of ISOD patients.
    Biospecimen Collection and Analysis:

  • Tissue-specific effects of SUOX deficiency require appropriate sampling strategies, yet affected neural tissue is rarely accessible.

  • Long-term biobanking is essential but requires careful consideration of sample stability and preparation methods.

  • Development of surrogate biomarkers that can be measured in accessible tissues or fluids remains a significant challenge.
    Translational Research Barriers:

  • Animal models may not fully recapitulate human ISOD due to species differences in sulfur metabolism and neurodevelopment.

  • In vitro systems must account for the mitochondrial localization of SUOX and its dependence on specific cofactors .

  • Therapeutic development faces significant challenges, including delivery across the blood-brain barrier and achieving sufficient enzyme replacement in affected tissues.
    Knowledge Synthesis and Dissemination:

  • Publication bias may limit reporting of negative findings, particularly important in rare disorders where every case provides valuable information.

  • Balancing patient privacy with the need for detailed case reporting requires careful ethical consideration.

  • Funding limitations for rare disease research necessitate creative approaches to study design and resource utilization.
    Addressing these methodological challenges requires interdisciplinary collaboration, innovative research designs, and development of specialized infrastructures supporting rare disease research. International patient registries, biobanks with standardized protocols, and data sharing initiatives represent essential foundations for advancing SUOX deficiency research .

Product Science Overview

Introduction

Sulfite oxidase (SOX) is a crucial enzyme in the oxidative degradation of sulfur-containing amino acids, such as cysteine and methionine. This enzyme catalyzes the oxidation of sulfite to sulfate, a vital reaction in sulfur metabolism. The human recombinant form of sulfite oxidase is produced using recombinant DNA technology, allowing for its study and application in various research and clinical settings.

Structure and Classification

Sulfite oxidase is a homodimeric protein localized to the intermembrane space of mitochondria. Each subunit of the enzyme contains two key domains:

  1. Heme Domain: This domain is responsible for electron transfer during the oxidation process.
  2. Molybdopterin-binding Domain: This domain binds the molybdenum cofactor (Moco), which is essential for the enzyme’s catalytic activity .
Biological Properties and Functions

Sulfite oxidase plays a critical role in the final step of the oxidative degradation of sulfur amino acids. The enzyme’s primary function is to catalyze the conversion of sulfite to sulfate, which is then excreted from the body. This reaction is vital for maintaining sulfur homeostasis and preventing the accumulation of toxic sulfite levels .

Mode of Action

The catalytic mechanism of sulfite oxidase involves the transfer of electrons from sulfite to the molybdenum center in the molybdopterin-binding domain. The electrons are then transferred to the heme domain, where they are ultimately passed to cytochrome c, a component of the mitochondrial electron transport chain. This process results in the oxidation of sulfite to sulfate .

Regulatory Mechanisms

The activity of sulfite oxidase is regulated by the availability of its substrates and cofactors. The enzyme requires the molybdenum cofactor (Moco) for its catalytic activity. Deficiencies in Moco or mutations in the SUOX gene, which encodes sulfite oxidase, can lead to sulfite oxidase deficiency. This condition is characterized by severe neurological abnormalities and is often fatal at an early age .

Applications of Human Recombinant Sulfite Oxidase

Recombinant human sulfite oxidase is produced using E. coli expression systems and is purified using conventional chromatography techniques. This recombinant form is used in various research applications, including studies on sulfur metabolism, enzyme kinetics, and the development of therapeutic interventions for sulfite oxidase deficiency .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.