SULT2A1 is a cytosolic sulfotransferase encoded by the SULT2A1 gene (UniProt ID: Q06520) that catalyzes the transfer of sulfate groups to hydroxysteroids, bile acids, and xenobiotics, enhancing their solubility for excretion . The SULT2A1 antibody is designed to specifically bind to this enzyme, enabling its detection in tissues like the liver, adrenal glands, and epididymis . Antibodies are available in monoclonal and polyclonal forms, with applications ranging from Western blotting (WB) to immunohistochemistry (IHC) .
SULT2A1 antibodies are pivotal in diverse experimental workflows:
Western Blotting: Detects SULT2A1 at ~34–35 kDa in liver and adrenal tissues .
Immunohistochemistry (IHC): Localizes SULT2A1 in adrenal zona reticularis and hepatic parenchyma .
Flow Cytometry: Identifies intracellular SULT2A1 in cell lines like HepG2 .
Enzyme Activity Studies: Validates functional changes in SULT2A1 during inflammatory responses .
Lipopolysaccharide (LPS) administration in mice reduced hepatic SULT2A1 mRNA by 50% within 4 hours and decreased enzyme activity by 60%, correlating with reduced serum DHEA-sulfate (DHEA-S) levels .
Proinflammatory cytokines TNF-α and IL-1β suppressed SULT2A1 expression in human hepatoma cells .
Activation of dopamine receptor D1 (DRD1) in HepG2 cells increased SULT2A1 mRNA, protein expression, and enzyme activity via cAMP signaling .
DRD1 knockdown reduced SULT2A1 activity by 20%, highlighting a novel regulatory pathway .
SULT2A1 is highly expressed in Leydig cells of the testis and the epididymal body in pigs, suggesting roles in steroid metabolism beyond the liver .
Specificity: Antibodies like ab194113 (Abcam) show no cross-reactivity with SULT2B1 isoforms .
Band Confirmation: Western blot validation in human liver (35 kDa) and adrenal gland lysates .
Functional Assays: Activity assays correlate with antibody-detected protein levels, as seen in LPS-induced SULT2A1 suppression .
SULT2A1 antibodies are indispensable for studying:
Sulfotransferase family cytosolic 2A dehydroepiandrosterone (DHEA)-preferring member 1, DHEA-ST, STD, HST, ST2, ST2A1, ST2A3, Alcohol/hydroxysteroid sulfotransferase, bile-salt sulfotranasferase 2A1, EC 2.8.2.14.
SULT2A1 antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
PAT13E10AT.
Anti-human SULT2A1 mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human SULT2A1 amino acids 1-285 purified from E. coli.
Mouse IgG2a heavy chain and κ light chain.
SULT2A1, also known as alcohol/hydroxysteroid sulfotransferase, belongs to the SULT2 family and catalyzes the sulfonation of hydroxysteroids and various endogenous steroids including androgens . This enzyme is highly expressed in adrenal and liver tissues and plays a critical role in hepatic cholesterol homeostasis . SULT2A1's significance in biomedical research stems from its involvement in multiple physiological processes, including steroid hormone metabolism, bile acid homeostasis, and detoxification of xenobiotics. The enzyme's activity affects drug bioavailability, hormone regulation, and toxicant metabolism, making it relevant for research in endocrinology, hepatology, pharmacology, and toxicology . Additionally, altered SULT2A1 expression and activity have been associated with various liver diseases, including nonalcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma, highlighting its potential as both a biomarker and therapeutic target .
SULT2A1 antibodies serve multiple essential applications in biomedical research. Western blot analysis using anti-SULT2A1 antibodies allows for protein quantification in tissue lysates, with validated applications in human liver and adrenal gland tissues . Immunohistochemistry applications enable the visualization of SULT2A1 expression patterns in tissue sections, particularly in human kidney tissue using antigen retrieval with TE buffer pH 9.0 or citrate buffer pH 6.0 . Immunofluorescence and immunocytochemistry with SULT2A1 antibodies have been validated for cellular localization studies, demonstrating cytoplasmic staining in cell lines such as HepG2 human hepatocellular carcinoma cells . Flow cytometry applications allow for quantitative assessment of SULT2A1 expression at the single-cell level . These antibodies are also valuable in mechanistic studies, as demonstrated in research examining dopamine D₁ receptor activation's effects on SULT2A1 expression and activity . Additionally, SULT2A1 antibodies can validate gene silencing experiments, as seen in studies where SULT2A1-specific siRNAs reduced protein expression by 40% and enzyme activity by 20% .
SULT2A1 has a calculated molecular weight of approximately 34 kDa based on its 285 amino acid sequence, and this is consistently observed in experimental applications . In Western blot analyses, SULT2A1 typically appears as a specific band at approximately 34-35 kDa under reducing conditions, as demonstrated in human liver tissue lysates . To verify antibody specificity, researchers should implement multiple validation strategies. First, testing the antibody in known positive control tissues with high SULT2A1 expression (liver and adrenal gland) is essential . Negative controls should include tissues or cell types with minimal SULT2A1 expression. Peptide competition assays, where pre-incubation of the antibody with the immunizing peptide blocks specific staining, provide strong evidence of specificity . Knockdown/knockout validation using SULT2A1-specific siRNAs, as demonstrated in HepG2 cells where 40% reduction in SULT2A1 protein expression was observed following siRNA treatment, represents a gold standard for antibody validation . Cross-reactivity assessment with related sulfotransferases should be performed, especially given the structural similarities within the SULT family. Lastly, confirming consistent results across multiple detection methods (WB, IHC, IF) with the same antibody increases confidence in specificity .
Optimization of antibody dilutions for SULT2A1 detection requires systematic testing across application-specific ranges. For Western blot applications, the recommended starting dilution range is 1:500-1:2000, with researchers advised to begin at 1:1000 and adjust based on signal intensity and background levels . For immunohistochemistry, a broader dilution range of 1:2000-1:8000 is suggested, with an initial dilution of 1:4000 followed by serial adjustments to achieve optimal signal-to-noise ratio . For immunofluorescence and immunocytochemistry applications, studies have successfully used SULT2A1 antibodies at 10 μg/mL for detection in HepG2 cells . For flow cytometry, antibody titration experiments should be performed starting from the manufacturer's recommendations, typically using a similar range as for immunofluorescence applications . The optimization process should include positive controls (tissues with known high SULT2A1 expression like liver and adrenal gland) and negative controls (omission of primary antibody) . Sample-specific factors may necessitate further adjustments; for instance, reduced SULT2A1 expression in diseased liver samples might require lower antibody dilutions for detection . Researchers should document optimization procedures thoroughly and maintain consistent protocols across experiments to ensure reproducibility, particularly when comparing expression levels between different experimental conditions .
Comprehensive validation of SULT2A1 antibodies in new experimental systems requires a multi-faceted approach. First, researchers should perform Western blot analysis on control samples known to express SULT2A1 (such as human liver tissue) alongside their experimental system, confirming detection of a single band at the expected molecular weight of 34-35 kDa . Immunoprecipitation followed by mass spectrometry can provide definitive identification of the protein recognized by the antibody. For genetic validation, siRNA-mediated knockdown experiments should demonstrate corresponding reduction in antibody signal, as shown in studies where SULT2A1-specific siRNAs decreased protein expression by 40% . Peptide competition assays, where pre-incubation with the immunizing peptide blocks specific staining, provide evidence of epitope specificity . When transitioning to new species or cell types, cross-reactivity should be systematically assessed, noting that certain SULT2A1 antibodies have been validated for human, mouse, and rat samples . Orthogonal validation using multiple antibodies targeting different epitopes of SULT2A1 increases confidence in specificity. For imaging applications, colocalization studies with established subcellular markers help confirm the expected cytoplasmic distribution of SULT2A1 . Quantitative validation should include standard curves with recombinant SULT2A1 protein to establish detection limits and linear range. Lastly, functional correlation between antibody-detected SULT2A1 levels and enzyme activity measurements provides validation of biological relevance .
Integration of SULT2A1 antibodies into multiplex detection systems requires careful consideration of antibody compatibility, spectral overlap, and epitope accessibility. For multiplex immunofluorescence, SULT2A1 antibodies from different host species (sheep, rabbit) are available, facilitating co-detection with other markers of interest . When designing multiplex panels, researchers should consider SULT2A1's cytoplasmic localization, enabling simultaneous detection with nuclear, membrane, or organelle-specific markers without spatial interference . For flow cytometry applications, careful selection of fluorophores with minimal spectral overlap is essential when combining SULT2A1 detection with other cellular markers. Optimization of staining protocols should include: sequential versus simultaneous antibody incubation comparison, determination of optimal fixation and permeabilization conditions for all targets, and thorough blocking to minimize non-specific binding . Multiplex immunohistochemistry requires testing of antibody combinations to ensure no cross-reactivity or steric hindrance, with tyramide signal amplification potentially enabling detection of low-abundance SULT2A1. For multiplex Western blotting, selecting SULT2A1 antibodies from different host species than other target antibodies allows simultaneous detection of multiple proteins with species-specific secondary antibodies. Mass cytometry (CyTOF) represents an advanced multiplex approach, using metal-conjugated antibodies including anti-SULT2A1 to simultaneously detect dozens of proteins without spectral overlap concerns. Regardless of the multiplex platform, comprehensive validation studies must confirm that antibody performance in multiplex settings matches that observed in single-target applications .
SULT2A1 expression demonstrates significant disease-specific alterations across liver pathologies, with antibody-based detection revealing important pathophysiological insights. In nonalcoholic fatty liver disease progression, immunohistochemical and Western blot analyses reveal a progressive decline in SULT2A1 expression and activity with increasing disease severity . Specifically, studies have shown that sulfation activity was substantially reduced in liver tissue from subjects with steatosis (23%), diabetic cirrhosis (16%), and alcoholic cirrhosis (18%) compared to healthy controls (100%) . This decreased expression correlates with reduced capacity to metabolize endogenous substrates and environmental toxicants such as bisphenol A, potentially contributing to disease progression through prolonged exposure to these compounds . In hepatocellular carcinoma, antibody-based studies have demonstrated variable SULT2A1 expression patterns, with potential connections to the Arg213His polymorphism found in SULT1A1, suggesting shared regulatory mechanisms within the sulfotransferase family . Immunolocalization studies in cholestatic liver diseases show alterations in zonal distribution of SULT2A1, potentially affecting bile acid metabolism and detoxification capacity. The cytoplasmic localization of SULT2A1 observed in HepG2 hepatocellular carcinoma cells suggests retained functionality in some liver cancer models, making these cells valuable for in vitro studies of SULT2A1 regulation . These findings highlight SULT2A1's potential as both a biomarker for liver disease progression and a therapeutic target, with antibody-based detection providing crucial spatial and quantitative information about expression patterns across different hepatic pathologies .
SULT2A1 plays a crucial role in phase II hepatic drug metabolism through catalyzing the sulfonation of various xenobiotics, thereby modifying their bioavailability, activity, and elimination. Antibody-based approaches offer multiple avenues to advance understanding of SULT2A1's role in drug metabolism. Immunohistochemistry and immunofluorescence with anti-SULT2A1 antibodies enable visualization of zonal expression patterns within the liver, helping identify hepatic regions with high sulfonation capacity for specific drugs . Quantitative Western blot analysis allows monitoring of SULT2A1 expression changes in response to drug inducers or inhibitors, providing insights into regulatory mechanisms . The application of SULT2A1 antibodies in flow cytometry enables assessment of expression heterogeneity across hepatocyte populations, potentially identifying subpopulations with different drug-metabolizing capacities . Co-immunoprecipitation studies using SULT2A1 antibodies can identify protein-protein interactions that influence enzyme activity or substrate specificity. Research has demonstrated that dopamine D₁ receptor activation upregulates both SULT2A1 expression and activity in HepG2 cells, revealing a novel regulatory pathway that could influence drug metabolism . This finding suggests that the dopamine D₁ receptor subtype may be involved in modulating drug metabolism through SULT2A1 regulation, highlighting potential drug-drug interactions between dopaminergic agents and SULT2A1 substrates . Antibody-based monitoring of SULT2A1 expression in patient-derived liver organoids or primary hepatocytes provides a platform for personalized prediction of drug metabolism capacity and potential adverse reactions, advancing precision medicine approaches in pharmacotherapy .
SULT2A1 antibodies offer valuable tools for investigating hormone-dependent diseases due to the enzyme's critical role in steroid hormone metabolism. SULT2A1 specifically catalyzes the sulfonation of dehydroepiandrosterone (DHEA) and various endogenous steroids including testosterone and estrogen, affecting their bioavailability and activity . In hormone-dependent cancer research, immunohistochemical detection of SULT2A1 in tumor samples can reveal alterations in steroid metabolism pathways that may contribute to cancer progression or resistance to hormone therapies. Western blot analysis using SULT2A1 antibodies enables quantitative assessment of expression changes in response to hormone-targeting therapies, potentially identifying mechanisms of treatment resistance . Co-localization studies combining SULT2A1 antibodies with markers for hormone receptors can uncover spatial relationships between hormone metabolism and signaling pathways within target tissues. Research has demonstrated that dopamine D₁ receptor activation induces both SULT2A1 expression and activity in HepG2 cells, revealing a novel neuroendocrine regulatory pathway with potential implications for hormone-dependent conditions . Specifically, treatment with the D₁ receptor agonists SKF82958 or SKF38393 significantly increased SULT2A1 mRNA expression, protein levels, and enzyme activity in a manner partially blocked by the D₁ antagonist SCH23390 . This unexpected connection between dopaminergic signaling and steroid sulfonation opens new avenues for investigating neuroendocrine influences on hormone-dependent diseases. SULT2A1 antibodies also enable mechanistic studies in models of endocrine disorders involving altered steroid metabolism, such as polycystic ovary syndrome, congenital adrenal hyperplasia, and adrenal insufficiency, potentially revealing disease-specific alterations in SULT2A1 expression or localization .
Researchers frequently encounter several challenges when performing SULT2A1 immunodetection, but these can be addressed with appropriate technical modifications. Cross-reactivity with other sulfotransferase family members represents a significant concern due to sequence homology. This can be overcome by selecting antibodies validated specifically for SULT2A1, such as those raised against unique epitopes or recombinant full-length human SULT2A1 protein . Western blot detection issues often include weak signal intensity; researchers can enhance detection by increasing protein loading (40-50 μg), optimizing transfer conditions for proteins around 34-35 kDa, and using high-sensitivity detection systems . Background staining in immunohistochemistry and immunofluorescence applications can be minimized through extended blocking steps (5% BSA or 10% normal serum), thorough washing, and careful antibody titration . Antigen retrieval challenges are common in formalin-fixed tissues; optimal SULT2A1 detection typically requires TE buffer at pH 9.0, though citrate buffer at pH 6.0 can serve as an alternative . Variability between lots of SULT2A1 antibodies may occur; researchers should validate each new lot against previous standards and maintain consistent protocols . Reduced SULT2A1 detection in diseased liver tissues compared to healthy controls may reflect biological downregulation rather than technical limitations, as studies have demonstrated decreased SULT2A1 activity with increasing severity of liver disease . Sample preparation issues can be addressed by including protease inhibitors in lysis buffers and maintaining cold temperatures throughout processing . Finally, discrepancies between protein detection and enzyme activity measurements may reflect post-translational modifications affecting function without altering antibody recognition; complementary approaches measuring both protein levels and enzymatic activity provide more comprehensive assessment .
When confronted with discrepancies between SULT2A1 protein levels detected by antibody-based methods and measured enzyme activity, researchers should consider multiple physiological and methodological factors. Post-translational modifications may significantly affect SULT2A1 activity without altering antibody recognition sites, necessitating phospho-specific or other modification-specific antibodies to resolve these discrepancies . Competitive inhibition by endogenous compounds or experimental reagents can suppress enzyme activity without affecting protein levels; activity assays with sample dilution series or in the presence of potential inhibitors may identify such effects . Cofactor availability is critical for SULT2A1 function, particularly 3'-phosphoadenosine-5'-phosphosulfate (PAPS); supplementation of this cofactor in activity assays can determine if limited availability explains reduced activity despite normal protein levels . Genetic polymorphisms may result in normal expression of functionally impaired enzyme variants; complementary genotyping for known functional polymorphisms can clarify such cases . Antibody epitope accessibility might be affected by protein-protein interactions or conformational changes that don't impact catalytic activity; varying extraction conditions can help resolve these issues. Methodological differences in sensitivity and dynamic range between antibody detection and activity assays should be considered; calibration with recombinant SULT2A1 standards across a concentration range can align these measurements . The study on dopamine D₁ receptor activation provides an informative example where both SULT2A1 protein expression and enzyme activity increased following receptor stimulation, though not to identical degrees (protein expression increased more substantially than activity), highlighting the complex relationship between these parameters . When persistent discrepancies remain unexplained, researchers should consider subcellular compartmentalization or microenvironmental factors that might differentially affect antibody binding versus enzymatic function .
Optimal storage and handling practices for SULT2A1 antibodies are essential for maintaining functionality and ensuring experimental reproducibility. Long-term storage recommendations include keeping antibodies at -20°C to -70°C for up to 12 months from the date of receipt, with -20°C adequate for most research applications . Antibodies should be stored in appropriate buffers, typically PBS containing 0.02% sodium azide and 50% glycerol at pH 7.3, which provides stability and prevents microbial growth . Aliquoting is crucial to avoid repeated freeze-thaw cycles; dividing stock solutions into single-use volumes (20-50 μL) prevents degradation from multiple freeze-thaw events . For short-term storage during ongoing experiments, antibodies can be maintained at 2-8°C for up to one month under sterile conditions after reconstitution . Working dilutions should be prepared fresh for each experiment rather than stored for extended periods, as diluted antibodies are more susceptible to degradation and microbial contamination . During experimental procedures, antibodies should be kept on ice when in use and returned to appropriate storage immediately afterward . Centrifuging antibody vials briefly before opening helps collect solution at the bottom and prevent loss. Proper documentation of storage conditions, freeze-thaw cycles, and lot numbers enables tracking of antibody performance over time . Performance validation through regular testing with positive controls (such as human liver tissue for SULT2A1) helps identify potential degradation before it affects experimental results . Some SULT2A1 antibody preparations may contain carriers like BSA (0.1%) or stabilizers that should be considered when designing blocking strategies for immunoassays . Following these comprehensive storage and handling practices will maximize antibody shelf life and maintain consistent performance in various applications including Western blot, immunohistochemistry, immunofluorescence, and flow cytometry .
SULT2A1 antibodies offer valuable tools for investigating xenobiotic metabolism in environmental exposure research through multiple innovative approaches. Immunohistochemical mapping of SULT2A1 distribution in exposed tissues can reveal site-specific metabolism capacity and potentially identify susceptible cell populations . Studies have demonstrated that liver diseases significantly impact SULT2A1-mediated metabolism of environmental compounds; for example, the sulfonation of bisphenol A was substantially reduced in liver samples from patients with steatosis (23%), diabetic cirrhosis (16%), and alcoholic cirrhosis (18%) compared to healthy controls . This reduced detoxification capacity could result in prolonged exposure to environmental toxicants, potentially exacerbating liver disease. Antibody-based protein quantification via Western blot or ELISA enables correlation of SULT2A1 expression levels with biomarkers of environmental exposure or toxicity endpoints . Flow cytometry applications with SULT2A1 antibodies allow assessment of cell-specific responses to xenobiotic exposure across heterogeneous cell populations . Co-localization studies combining SULT2A1 antibodies with markers of cellular stress or damage can establish spatial relationships between metabolism capacity and toxicity. Mechanistic studies can leverage SULT2A1 antibodies to investigate how environmental compounds might alter the enzyme's expression or localization; for instance, research has shown that dopaminergic signaling regulates SULT2A1 expression, suggesting potential neuroendocrine influence on xenobiotic metabolism . Time-course experiments using antibody detection can track adaptation of SULT2A1 expression following acute versus chronic exposure scenarios. Population studies may employ antibody-based SULT2A1 quantification in accessible samples (such as peripheral blood mononuclear cells) as potential biomarkers of metabolic capacity, especially when combined with genotyping for relevant polymorphisms . These applications collectively provide crucial insights into how environmental exposures interact with metabolic capabilities across diverse populations and disease states .
SULT2A1 antibodies hold significant potential for developing innovative diagnostic and prognostic tools for liver diseases based on their ability to detect disease-specific alterations in expression patterns. Immunohistochemical assessment of SULT2A1 in liver biopsies could serve as a functional biomarker, as studies have demonstrated progressive decline in SULT2A1 activity with increasing severity of liver disease from simple steatosis to cirrhosis . This pattern suggests that SULT2A1 expression levels might predict disease progression risk or treatment response. Multiplex immunoassays incorporating SULT2A1 alongside other markers of liver function could create comprehensive tissue fingerprints for more precise disease stratification . In hepatocellular carcinoma research, SULT2A1 antibody-based detection may have prognostic value, particularly when integrated with analysis of the Arg213His polymorphism, which has been associated with HCC susceptibility . For noninvasive diagnostics, developing sensitive immunoassays detecting circulating SULT2A1 released from damaged hepatocytes could provide a minimally invasive assessment of liver-specific injury. Digital pathology approaches combining SULT2A1 immunostaining with artificial intelligence analysis could identify subtle pattern changes not apparent to the human eye. Microfluidic platforms utilizing immobilized SULT2A1 antibodies could enable point-of-care testing of enzymatic activity in small tissue samples or bodily fluids. Liquid biopsy applications might detect SULT2A1 in extracellular vesicles released from liver cells, potentially providing tissue-specific information without invasive procedures. Significant validation work remains necessary, but research has established that SULT2A1 expression varies meaningfully across different liver diseases, with functional consequences for both endogenous metabolism and xenobiotic detoxification . The demonstration that SULT2A1 activity significantly decreases with disease progression suggests that quantitative assessment of this enzyme could provide valuable prognostic information beyond current standard liver function tests .
SULT2A1 antibodies offer multiple pathways to advance personalized medicine approaches for optimizing drug therapy based on individual metabolic profiles. Patient stratification strategies can be developed using immunohistochemical analysis of SULT2A1 in liver biopsies to predict sulfonation capacity for specific medications, potentially identifying individuals at risk for adverse reactions or treatment failure . Ex vivo drug metabolism assays combining SULT2A1 antibody detection with functional activity measurements in patient-derived samples can provide personalized predictions of drug clearance rates . High-throughput screening platforms utilizing SULT2A1 antibodies can identify novel drug candidates or repurpose existing medications based on their interactions with this enzyme, facilitating development of personalized therapeutic regimens . Development of companion diagnostics incorporating SULT2A1 detection could guide prescription decisions for medications primarily metabolized through sulfonation pathways. The discovery that dopamine D₁ receptor activation upregulates SULT2A1 expression and activity suggests potential regulatory mechanisms that could be therapeutically exploited; for example, patients receiving dopaminergic medications might experience altered metabolism of drugs that are SULT2A1 substrates . Specifically, research has demonstrated that D₁ receptor agonists SKF82958 (2.5 μmol/L) and SKF38393 (5-50 μmol/L) significantly increased SULT2A1 mRNA expression, protein levels, and enzymatic activity in HepG2 cells, effects that were partially blocked by the D₁ antagonist SCH23390 . This finding has significant implications for drug interaction predictions and personalized dosing regimens. Longitudinal monitoring of SULT2A1 expression using antibody-based methods could track changes in metabolic capacity during disease progression or treatment, enabling dynamic adjustment of medication dosing . Point-of-care tests incorporating SULT2A1 antibodies might eventually provide rapid assessment of metabolic capacity in clinical settings, facilitating real-time personalization of drug therapy . These applications collectively contribute to the evolving paradigm of precision medicine by accounting for individual variations in drug metabolism capacity .
Current limitations in SULT2A1 antibody research encompass both technical challenges and knowledge gaps that require targeted development efforts. Epitope specificity remains a primary concern, as antibodies may cross-react with other sulfotransferase family members due to sequence homology; development of highly specific monoclonal antibodies against unique SULT2A1 epitopes would address this limitation . Standardization across laboratories represents another challenge, with variations in protocols, antibody sources, and detection methods complicating cross-study comparisons; establishment of standardized procedures and reference materials would enhance reproducibility . Limited ability to distinguish between functionally distinct SULT2A1 variants or post-translationally modified forms restricts mechanistic insights; next-generation antibodies specifically recognizing these variants would advance functional studies . Most current antibodies primarily detect total protein rather than active enzyme; development of conformation-specific antibodies that differentiate active versus inactive forms would better correlate with functional capacity . Species cross-reactivity limitations constrain translational research; while some antibodies react with human, mouse, and rat SULT2A1, broader cross-species reactivity would facilitate comparative studies . Temporal resolution is inadequate with current methodologies; development of biosensor approaches using SULT2A1 antibody-derived fragments could enable real-time monitoring of expression changes . Limited correlation between immunodetection and functional activity measurements creates interpretation challenges; integrated platforms simultaneously measuring both parameters would provide more comprehensive assessment . Accessibility of SULT2A1 antibody-based tools in clinical settings remains limited; translation into robust point-of-care formats would expand research applications into clinical practice . Addressing these limitations through targeted research and development efforts would significantly advance SULT2A1 research and its applications in both basic science and clinical medicine.
Designing comprehensive validation strategies for SULT2A1 antibodies in translational research requires a multi-dimensional approach addressing analytical validity, clinical validity, and practical utility. Initial analytical validation should verify antibody specificity through Western blot analysis confirming detection of a single band at the expected molecular weight (34-35 kDa) in well-characterized positive control tissues such as human liver and adrenal gland . Orthogonal method comparison can strengthen validation by correlating antibody-based detection with mass spectrometry-based proteomics and mRNA expression analyses. Genetic validation using SULT2A1 knockdown/knockout models provides critical evidence of specificity; studies have shown that siRNA-mediated reduction of SULT2A1 mRNA by 40% resulted in corresponding 40% reduction in protein expression detectable by antibodies . Cross-reactivity assessment should include testing against recombinant sulfotransferase family members and in tissues known to express related enzymes. For translational applications, antibody performance must be evaluated across diverse sample types including fresh tissues, FFPE samples, blood fractions, and clinically relevant biofluids . Reproducibility assessment should include inter-laboratory comparison studies using standardized samples and protocols. Functional correlation between antibody-detected SULT2A1 levels and enzyme activity measurements is essential, as demonstrated in studies where D₁ receptor activation increased both protein expression and activity . Clinical validation requires demonstrating association between antibody-detected SULT2A1 expression and relevant clinical parameters, such as drug metabolism capacity or disease severity . Practical considerations include determination of pre-analytical variables (sample collection, processing, storage), analytical factors (detection limits, linear range, precision), and post-analytical aspects (reference ranges, clinical decision thresholds). Implementing this comprehensive validation strategy ensures that SULT2A1 antibodies perform reliably across the research-to-clinic continuum, supporting robust translational outcomes .
The intersection of SULT2A1 biology and antibody technology creates fertile ground for interdisciplinary research spanning multiple scientific domains. Biomarker development represents a promising area where SULT2A1 antibodies could enable liquid biopsy approaches for liver diseases, potentially detecting enzyme released from damaged hepatocytes or in extracellular vesicles . In cancer research, the connection between SULT2A1, hormone metabolism, and tumor growth creates opportunities for antibody-based investigations of metabolic rewiring in hormone-dependent cancers . The discovery that dopamine D₁ receptor activation upregulates SULT2A1 expression and activity opens interdisciplinary research at the neuroendocrine-metabolism interface, potentially revealing novel regulatory pathways connecting central nervous system signaling with peripheral metabolic functions . Nanomedicine applications could leverage SULT2A1 antibodies in targeted drug delivery systems directed at tissues with high enzyme expression, combining pharmaceutical science with immunotechnology . In toxicology, antibody-based detection of SULT2A1 alterations following environmental exposures could elucidate mechanisms of toxicity and individual susceptibility differences, as suggested by studies showing reduced bisphenol A sulfation in diseased liver tissues . Imaging technology integration, combining SULT2A1 antibodies with advanced microscopy techniques, could reveal subcellular localization dynamics and co-localization with metabolic partners . Synthetic biology approaches might utilize antibody-derived binding domains to create artificial regulatory circuits modulating SULT2A1 activity. In pharmacogenomics, correlating antibody-detected SULT2A1 protein variants with genetic polymorphisms could improve prediction of drug metabolism phenotypes beyond genotyping alone . Digital pathology combined with artificial intelligence could extract complex pattern information from SULT2A1 immunostaining that exceeds human visual processing capabilities. These interdisciplinary opportunities highlight how the convergence of SULT2A1 biology and antibody technology can catalyze innovative research spanning basic science, technological development, and clinical applications .
The Sulfotransferase Family, Cytosolic, 2A, Member 1 (SULT2A1) is a crucial enzyme involved in the metabolism of various endogenous and exogenous compounds. This enzyme is part of the larger sulfotransferase (SULT) family, which plays a significant role in the sulfonation process, aiding in the detoxification and excretion of numerous substances.
SULT2A1 is a protein-coding gene that encodes for the enzyme responsible for the sulfonation of steroids and bile acids. The gene is located on chromosome 19 in humans and has several aliases, including DHEA-ST, STD, and HST . The enzyme utilizes 3’-phospho-5’-adenylyl sulfate (PAPS) as a sulfonate donor to catalyze the sulfonation process .
The primary function of SULT2A1 is to catalyze the sulfonation of a wide range of steroids and sterols, including pregnenolone, androsterone, dehydroepiandrosterone (DHEA), bile acids, and cholesterol . This process increases the water solubility of these compounds, facilitating their renal excretion. Additionally, SULT2A1 can sulfonate various xenobiotics containing alcohol and phenol functional groups .
SULT2A1 is predominantly expressed in the liver and adrenal glands, where it plays a vital role in steroid metabolism and bile acid regulation . The enzyme’s activity is crucial for maintaining hormonal balance and detoxifying potentially harmful substances. In women with polycystic ovary syndrome (PCOS), SULT2A1 may contribute to inherited adrenal androgen excess .
Research on SULT2A1 has expanded our understanding of its role in chemical metabolism and detoxification. Studies have shown that the enzyme’s activity can be influenced by genetic polymorphisms, which may affect an individual’s response to drugs and environmental toxins . Additionally, SULT2A1 is used in various biochemical assays to study steroid metabolism and drug interactions.