AS3MT Antibody

Shipped with Ice Packs
In Stock

Description

AS3MT Antibody Overview

AS3MT antibodies are immunodetection tools targeting the AS3MT enzyme, which mediates arsenic methylation in humans and model organisms. These antibodies are pivotal for:

  • Western Blot (WB): Detecting AS3MT protein levels in tissues (e.g., liver, kidney) .

  • Immunohistochemistry (IHC): Localizing AS3MT in cellular compartments .

  • ELISA: Quantifying AS3MT expression in biological samples .

Studies using AS3MT-deficient mice (As3mt⁻/⁻) highlight its necessity for arsenic metabolism, as knockout models retain 10× more arsenic than wild-type mice . Polymorphisms in AS3MT are linked to differential arsenic methylation efficiency and cardiovascular risks in humans .

Mechanistic Insights into Arsenic Toxicity

  • Atherosclerosis Models: AS3MT antibodies identified elevated AS3MT expression in murine aortic plaques after arsenic exposure, linking methylation to plaque formation .

  • Knockout Studies: As3mt⁻/⁻ mice retained 20% of arsenic doses after 96 hours (vs. <2% in wild-type), confirmed via WB using AS3MT antibodies .

  • Polymorphism Effects: In Argentinian and Bangladeshi cohorts, AS3MT haplotypes correlated with DNA methylation patterns and gene expression, analyzed using IHC and WB .

Inhibitor Development

Small-molecule inhibitors of AS3MT (e.g., TPI-1 to TPI-10) were evaluated using AS3MT antibodies to monitor enzyme activity. Inhibitors reduced methylation efficacy by binding allosteric sites distant from catalytic domains .

Technical Considerations

  • Cross-Reactivity: Antibodies like ABIN2783536 show broad reactivity (92–100% across 10 species) .

  • Controls: Use GAPDH or Lamin A for normalization in WB .

  • Limitations: Some antibodies lack specificity for methylated AS3MT isoforms, requiring validation via knockout controls .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery times may vary depending on the mode of purchase and location. For specific delivery details, please contact your local distributor.
Synonyms
2310045H08Rik antibody; Arsenic (+3 oxidation state) methyltransferase antibody; Arsenite methyltransferase antibody; As3mt antibody; AS3MT_HUMAN antibody; C10ORF32 antibody; CYT19 antibody; Cyt19 protein antibody; Hypothetical protein C10orf32 antibody; Methylarsonite methyltransferase antibody; Methyltransferase cyt19 antibody; OTTHUMP00000020384 antibody; RP11-753C18.6 antibody; S adenosylmethionine arsenic (III) methyltransferase antibody; S-adenosyl-L-methionine:arsenic(III) methyltransferase antibody
Target Names
AS3MT
Uniprot No.

Target Background

Function
This antibody targets the AS3MT protein, which catalyzes the transfer of a methyl group from AdoMet to trivalent arsenicals, resulting in methylated and dimethylated arsenicals. Specifically, it methylates arsenite to form methylarsonate, Me-AsO(3)H(2). Subsequently, methylarsonate reductase reduces methylarsonate to methylarsonite, Me-As(OH)2. Importantly, methylarsonite also serves as a substrate for AS3MT and is further converted into the less toxic compound dimethylarsinate (cacodylate), Me(2)As(O)-OH.
Gene References Into Functions

Gene References and Functional Implications:

  1. A combined effect of N6AMT1 haplotype 2_GGCCAT and As3MT haplotype 2_GCAC was observed, consistent with an additive impact of each haplotype on % iAs: the mean was 5.47% for carriers with both haplotypes and 9.36% for carriers with null haplotypes. [PMID: 27637898]
  2. Maternal alleles for five of the seven SNPs (rs7085104, rs3740400, rs3740393, rs3740390, and rs1046778) were associated with urinary concentrations of iAs metabolites. Notably, alleles for one SNP (rs3740393) were associated with birth outcomes/measures. These associations were strongly dependent upon the male sex of the fetus but independent of fetal genotype for AS3MT. [PMID: 26928318]
  3. A combined analysis confirmed genome-wide significant associations in the Han Chinese population for three loci: 2p16.1 (rs1051061, in an exon of VRK2, P=1.14 x 10-12, odds ratio (OR)=1.17), 6p22.1 (rs115070292 in an intron of GABBR1, P=4.96 x 10-10, OR=0.77), and 10q24.32 (rs10883795 in an intron of AS3MT, P=7.94 x 10-10, OR=0.87; rs10883765 at an intron of ARL3, P=3.06 x 10-9, OR=0.87). [PMID: 27922604]
  4. This study examined AS3MT and N6AMT1 polymorphisms and urinary arsenic metabolites (%iAs, %MMA, %DMA) in 722 subjects from an arsenic-cancer case-control study in a uniquely exposed area in northern Chile. [PMID: 28640505]
  5. Risk alleles spanning multiple genes across the 10q24.32 schizophrenia-related locus are associated in the human brain selectively with an increase in the expression of both BLOC-1 related complex subunit 7 (BORCS7) and a previously uncharacterized, human-specific arsenite methyltransferase (AS3MT) isoform (AS3MTd2d3). [PMID: 27158905]
  6. A joint dose-response effect was identified for a AS3MT high-risk haplotype and inefficient arsenic methylation capacity on developmental delay. [PMID: 28235556]
  7. A genome-wide association study in a population in the United States (AZ, OK, SD, ND) suggests that SNPs in AS3MT (rs17878846, rs10509760) partly explain inter-individual variability in susceptibility to arsenic poisoning (and urine arsenic species patterns). [PMID: 26209557]
  8. Genotype-dependent differences in arsenic metabolism are noted. [PMID: 26411936]
  9. This study found that rs3740400 in As3MT and rs156697 in GSTO2 were strongly associated with the arsenic species (%iAs, %MMA and MMA/iAs and DMA/MMA ratios) in urine of occupationally exposed workers, suggesting these SNPs may influence methylation capacity. [PMID: 25491248]
  10. The study analyzed the functional roles of GSH and the crucial Cys residues in iAs3+ methylation catalyzed by hAS3MT. [PMID: 25349987]
  11. A strong signal of positive selection was observed in the main arsenic methyltransferase AS3MT gene, which has been previously associated with lower concentrations of the most toxic product of arsenic metabolism, monomethylarsonic acid. [PMID: 26366667]
  12. This study provides preliminary evidence for the over-transmission of the A allele at the AS3MT rs11191454 polymorphism in Attention Deficit Hyperactivity Disorder. [PMID: 25461954]
  13. The study investigated the influence of the level of arsenic exposure and the presence of T860C polymorphism in human As urinary metabolic profile. [PMID: 24845704]
  14. The functions of arsenic (III) methyltransferase S-adenosylmethionine binding site were evaluated. [PMID: 25447140]
  15. Genetic variation in the AS3MT gene is associated with arsenic metabolism and risk of basal cell carcinoma. [PMID: 25156000]
  16. Two SNPs, AS3MT 12390 (rs3740393) and 14458 (rs11191439), were consistently related to arsenic methylation regardless of the populations examined for the analysis. [PMID: 21731446]
  17. AS3MT SNPs were associated with arsenic metabolism efficiency and arsenic toxicity in exposed individuals. [PMID: 24536095]
  18. Low-level arsenic exposure is associated with cognitive functioning, but this association is modified by an AS3MT gene. [PMID: 24621105]
  19. A hypomethylated region in the AS3MT promoter was associated with higher arsenic exposure, indicating that arsenic exposure influences the epigenetic regulation of AS3MT. [PMID: 24154821]
  20. AS3MT polymorphisms may impact disease susceptibility. [PMID: 24792412]
  21. When workers were classified according to their AS3MT Met287Thr genotypes, significantly higher micronucleus values were observed for those carrying the variant allele [odds ratio (OR), 3.4 (1.6-5.2); P=0.0003)]. [PMID: 24361376]
  22. Genetic polymorphisms in the AS3MT gene in a Western Chinese population influenced urinary arsenic metabolism from arsenic in drinking water. [PMID: 24239724]
  23. Five N6AMT1 single nucleotide polymorphisms and two N6AMT1 haplotypes were significantly associated with the percentage of methylarsonic acid in urine, even after adjusting for arsenic (+3 oxidation state) methyltransferase haplotype. [PMID: 23665909]
  24. Residues Asp76, 84, 102, and 150 greatly influence hAS3MT catalytic activity, affecting S-adenosylmethionine-binding or methyl transfer. [PMID: 23742935]
  25. Individuals with M287T and G4965C AS3MT polymorphisms had higher levels of urinary DMAs(III) and were more frequently diabetic than the respective wild-type carriers, although the excess was not statistically significant. [PMID: 23093101]
  26. Data suggest that during a few thousand years, natural selection for tolerance to the environmental stressor arsenic may have increased the frequency of protective variants of AS3MT. [PMID: 23070617]
  27. The AS3MT haplotype status strongly predicted DNA methylation and gene expression of AS3MT as well as several genes in 10q2. [PMID: 23341986]
  28. Single nucleotide polymorphisms (SNPs) in arsenic methyltransferase and methylene-tetrahydrofolate reductase are associated with bladder cancer in those exposed to low concentrations of inorganic arsenic. SNPs in glutathione S-transferase omega-1 are not. [PMID: 22747749]
  29. The study analyzed rapid equilibrium kinetics of arsenite methylation catalyzed by recombinant human arsenic (+3 oxidation state) methyltransferase (hAS3MT). [PMID: 22955273]
  30. Structural insights into the mechanism of substrate binding and catalysis were provided. [PMID: 22712827]
  31. Pregnancy and the AS3MT genotype independently influence the arsenic methylation phenotype. [PMID: 22547080]
  32. Genetic polymorphisms GSTO and As3MT modify arsenic metabolism as evidenced by altered urinary arsenic excretion. [PMID: 22339537]
  33. Coronary heart disease and its main risk factors were associated with low-level arsenic exposure, AS3MT polymorphism, or both. Hypertension was associated with higher arsenic exposure, while hyperlipidemia was associated with AS3MT polymorphism. [PMID: 22341486]
  34. Data suggest that the expression pattern of splicing variants of the As3MT gene may affect the capacity for arsenic methylation in cells. [PMID: 22005461]
  35. The study investigated the roles of AS3MT in the effects of arsenicals, carcinogens found ubiquitously in nature: metabolism, toxicity, and accumulation of arsenicals in hepatocytes. [PMID: 21537954]
  36. Polymorphisms in AS3MT significantly predicted As metabolism across two very different populations, suggesting that AS3MT may have an impact on As metabolite patterns in populations worldwide. [PMID: 21247820]
  37. The distribution of 18 SNPs in AS3MT was assessed in healthy individuals in Mexico and Germany: 38 Mestizo, 69 Nahuas, 50 Huicholes, and 32 Germans. [PMID: 20571777]
  38. The study analyzed the enzymatic activity, kinetics, thermal stability, and secondary structures of the mutant enzyme. [PMID: 20971157]
  39. The study determined an ordered sequence for the binding of SAM and arsenite to the hAS3MT, while no interactions between GSH and the hAS3MT were detected. [PMID: 20621156]
  40. AS3MT effects organic arsenic metabolism. [PMID: 20532380]
  41. Genetic association analysis with arsenic metabolism confirmed the previously observed association between AS3MT variants, including this large cluster of linked polymorphisms, and arsenic methylation efficiency. [PMID: 20014157]
  42. Data suggest that the inhibitory effect of the ions (Co(2+), Mn(2+), and Zn(2+)) or selenite on AS3MT activity might be via interactions with free Cys residues in human AS3MT to form inactive protein adducts. [PMID: 20129672]
  43. This study is the first to demonstrate the existence of genetic heterogeneity in a worldwide distribution of 18 single nucleotide polymorphisms in AS3MT. [PMID: 19932709]
  44. Heterologous expression of rat AS3MT in human urothelial cells confers the capacity to methylate inorganic arsenic and alters its cytotoxicity and that of methylated arsenicals. [PMID: 15808521]
  45. A strong genetic association between polymorphisms of CYT19 and D:M ratio (urinary dimethylarsenic(V) to monomethylarsenic(V)) was observed in drinking water exposed Mexican children, but not adults. [PMID: 15929903]
  46. Inherited variation in AS3MT may contribute to variation in arsenic metabolism. [PMID: 16407288]
  47. AS3MT is strongly associated with the ratio of urinary dimethylarsinic acid to monomethylarsonic acid (D/M) in children (7-11 years) but not in adults (18-79 years). [PMID: 17306849]
  48. Results indicate that genetic polymorphisms in AS3MT contribute to inter-individual variation in arsenic biotransformation and, therefore, may contribute to inter-individual variations in risk of arsenic toxicity and arsenic carcinogenesis. [PMID: 17850829]
  49. Mutation frequencies of AS3MT in Asian populations were relatively lower than those of African and Caucasian populations: frequencies of mutation in the Mongolian, Korean, and Japanese populations were 0.040, 0.010, and 0.010, respectively. [PMID: 17889916]
  50. Individuals with the AS3MT(287)Thr variant display increased arsenic methylation and thus are associated with an increased risk for the toxic and genotoxic effects of arsenic exposure. [PMID: 18334919]

Show More

Hide All

Database Links

HGNC: 17452

OMIM: 611806

KEGG: hsa:57412

STRING: 9606.ENSP00000358896

UniGene: Hs.720370

Protein Families
Methyltransferase superfamily, Arsenite methyltransferase family
Subcellular Location
Cytoplasm, cytosol.

Q&A

What is AS3MT and why is it important in arsenic metabolism research?

AS3MT (Arsenic (+3 Oxidation State) Methyltransferase) is an enzyme that catalyzes the biotransformation of inorganic arsenic through methylation reactions. This 375-amino acid protein (376 in mice) plays a critical role in arsenic metabolism, which directly impacts arsenic toxicity and related pathologies. The enzyme transfers methyl groups from S-adenosylmethionine (SAM) to arsenic species through nucleophilic attacks, with concomitant release of S-adenosyl-homocysteine (SAH) . AS3MT is expressed in various tissues including liver, brain, adrenal gland, and peripheral blood mononuclear cells, but notably absent in human keratinocytes, urothelial cells, and brain microvascular endothelial cells . Understanding AS3MT function is crucial for arsenic toxicology research as it represents a key determinant in individual susceptibility to arsenic-related diseases.

What are the conserved functional domains in the AS3MT protein important for antibody targeting?

AS3MT contains several highly conserved and functionally significant regions that serve as important targets for antibody development. The protein contains five fully conserved cysteine residues that are essential for arsenic methylation activity. In human AS3MT, these cysteines are positioned at C32, C61, C85, C156, and C206, corresponding to C33, C62, C86, C157, and C207 in the mouse homolog . These cysteine residues play crucial roles in the catalytic mechanism and represent critical epitopes for functional antibodies. Commercial antibodies target various regions including the N-terminus, middle region (with sequences such as "GIKNESHDIV VSNCVINLVP DKQQVLQEAY RVLKHGGELY FSDVYTSLEL"), and C-terminus, each offering different advantages depending on the experimental application . The selection of antibodies targeting specific domains should be guided by whether structural or functional investigations are being conducted.

How is AS3MT expression regulated at the transcriptional level?

The transcriptional regulation of AS3MT occurs in a tissue/cell type-specific manner through multiple mechanisms. Recent research has identified the core promoter region of the human AS3MT gene, revealing a GC-rich region containing a GC box to which the stress-related transcription factor Sp1 binds . This finding indicates the involvement of stress-responsive regulatory elements in AS3MT gene expression. The promoter region contains several potential binding sites for transcription factors, suggesting complex regulation mechanisms. Methylation of the promoter may also play a role in regulating expression, as studies have employed unmethylated AS3MT-specific primers (unmethAS3MT-F and unmethAS3MT-R) to investigate this aspect . This multi-layered regulation explains the tissue-specific expression pattern observed, with notable expression in liver and brain tissues but absence in keratinocytes and certain endothelial cells.

What are the optimal conditions for using AS3MT antibodies in Western blotting experiments?

The effective use of AS3MT antibodies in Western blotting requires careful optimization of several parameters. Based on research protocols, the following conditions are recommended:

  • Sample preparation: Cell lysates should be prepared in buffer containing protease inhibitors to prevent degradation of AS3MT.

  • Antibody selection: Choose antibodies targeting regions relevant to your research question. For general detection, middle region antibodies (such as ABIN2783536) show broad species cross-reactivity with human, mouse, rat, and other mammals .

  • Dilution optimization: Experimental determination of optimal working dilutions is critical as this varies between antibody batches. Starting dilutions of 1:500 to 1:2000 are typically recommended for Western blotting applications .

  • Detection systems: Enhanced chemiluminescence (ECL) systems provide adequate sensitivity for detecting AS3MT in most tissue extracts where the protein is expressed.

  • Controls: Include positive controls such as liver cell lysates where AS3MT is abundantly expressed, and negative controls from tissues known to lack AS3MT expression (e.g., keratinocytes) .

When validating antibody specificity, compare results with recombinant AS3MT proteins or lysates from cells with confirmed AS3MT expression levels through techniques like qRT-PCR.

How can AS3MT automethylation be detected and measured in experimental settings?

Detection and measurement of AS3MT automethylation requires specialized techniques that capture this post-translational modification. Based on published methodologies, the following approaches are recommended:

  • Radioactive methylation assays: Using [3H]-labeled SAM (S-adenosylmethionine) as methyl donor, AS3MT automethylation can be visualized by fluorography after SDS-PAGE separation. Typical reaction conditions include 0.4 μM [3H]-SAM (15 Ci/mmol), purified AS3MT protein, and cofactors including TRR (0.2 μM), TRX (10 μM), NADPH (300 μM), and GSH (1 mM) .

  • Mass spectrometry: Flag-AS3MT immunoprecipitation coupled to MS/MS analysis has successfully identified specific cysteine residues (C33, C62) as acceptors of methyl groups in vivo . This approach requires stable expression of tagged AS3MT in appropriate cell lines followed by immunoprecipitation.

  • Site-directed mutagenesis: Comparing wild-type AS3MT with cysteine-to-alanine mutants (particularly at positions C33, C62, C86, C157, and C207 in mouse AS3MT) in automethylation assays can determine which residues are critical for this process .

  • Cofactor requirements: Include reducing agents such as glutathione (GSH) and dithiothreitol (DTT) which enhance automethylation by maintaining cysteines in a reduced state necessary for accepting methyl groups .

Interpretation should consider that automethylation decreases following addition of inorganic arsenic (iAs), suggesting competition between automethylation and substrate methylation.

What approaches can be used to assess AS3MT antibody cross-reactivity across different species?

Determining cross-reactivity of AS3MT antibodies across species is essential for comparative studies. Several methodological approaches can be employed:

MethodDescriptionAdvantagesLimitations
Sequence alignmentCompare AS3MT epitope sequences across target speciesPredicts potential cross-reactivityDoes not confirm actual binding
Western blottingTest antibody against lysates from multiple speciesDirect evidence of bindingRequires tissue/cell samples from each species
Recombinant protein testingUse purified recombinant AS3MT from different speciesControlled comparison of binding affinityMay not reflect native protein conformation
ImmunohistochemistryTest tissue sections from different speciesEvaluates binding in native contextTissue processing may affect epitope availability

Commercial antibodies often provide predicted reactivity data. For example, antibody ABIN2783536 shows high predicted cross-reactivity (92-100%) across mammals including human, mouse, rat, cow, dog, goat, horse, pig, rabbit, and even zebrafish . When experimental validation is required, Western blot analysis using equal amounts of protein from comparable tissues across species is recommended, with sequence analysis of the targeted epitope region to explain any observed differences in reactivity.

How should experiments be designed to study AS3MT-mediated arsenic metabolism?

Designing robust experiments to study AS3MT-mediated arsenic metabolism requires careful consideration of multiple factors:

  • Expression systems: Both prokaryotic (E. coli) and eukaryotic expression systems can be used for producing recombinant AS3MT. For human AS3MT, MBP-tagged fusion proteins expressed in E. coli provide good solubility and activity. Cultures should be grown to OD 0.6, induced with 0.3 mM IPTG for 4 hours, and purified using appropriate affinity chromatography (e.g., amylose magnetic beads for MBP-tagged proteins) .

  • Methylation reaction conditions: Standard in vitro reactions should include:

    • Purified AS3MT protein (1-5 μg)

    • SAM (0.4-1 μM if radiolabeled; higher if unlabeled)

    • Reducing system (GSH 1 mM, NADPH 300 μM, TRX 10 μM, TRR 0.2 μM)

    • Arsenic substrate (typically 0.1-10 μM iAs)

    • Buffer (50 mM MOPS pH 7.4 with 10% glycerol and 0.3 M NaCl)

    • Incubation at 37°C for optimal activity

  • Analysis of methylation products: Hydride generation atomic absorption spectrometry coupled with cryotrapping (HG-CT-AAS) provides sensitive detection of arsenic species (iAs, MAs, DMAs) with detection limits of 8-20 pg . Sample preparation should include L-cysteine treatment to reduce pentavalent arsenic species to their trivalent counterparts.

  • Controls: Include enzyme-free controls, heat-inactivated enzyme controls, and reactions without SAM to distinguish enzyme-dependent methylation from non-enzymatic processes.

For cellular studies, comparison between AS3MT-expressing and AS3MT-deficient cells (through CRISPR knockout or siRNA) provides valuable insights into the contribution of AS3MT to cellular arsenic metabolism.

What are common pitfalls when using AS3MT antibodies in immunofluorescence studies?

Immunofluorescence studies with AS3MT antibodies present several technical challenges that investigators should anticipate and address:

  • Specificity concerns: AS3MT antibodies may show cross-reactivity with other methyltransferases. Always validate specificity using AS3MT-knockout or knockdown samples as negative controls and recombinant AS3MT as a positive control.

  • Fixation sensitivity: The detection of AS3MT epitopes can be significantly affected by fixation methods. Compare paraformaldehyde (4%) with methanol fixation to determine optimal epitope preservation. Some antibodies may work better with specific fixation methods.

  • Subcellular localization artifacts: AS3MT is primarily cytosolic but may show nuclear localization under certain conditions. Differentiate true localization from artifacts by using multiple antibodies targeting different AS3MT regions and complementary techniques like cell fractionation.

  • Background fluorescence: Tissues with high levels of natural fluorophores (particularly liver tissue, where AS3MT is highly expressed) may show interfering background. Use appropriate quenching steps and careful selection of fluorophores with emission spectra distinct from autofluorescence.

  • Antibody concentration optimization: Titrate antibodies carefully, as both insufficient and excessive concentrations can lead to false negative or high background signals, respectively. Start with dilutions recommended by manufacturers (typically 1:100 to 1:500) and optimize empirically.

When performing co-localization studies, sequential staining protocols may be preferable to simultaneous application of multiple primary antibodies to minimize cross-reactivity issues.

What controls should be included when studying AS3MT promoter activity?

Investigation of AS3MT promoter activity requires rigorous controls to ensure reliable and interpretable results:

  • Positive and negative genomic controls: Include well-characterized active promoters (e.g., ACTB) as positive controls and promoter-less constructs as negative controls in reporter assays . Additionally, include regions known to lack promoter activity from the same genomic vicinity as AS3MT.

  • Deletion series controls: Generate a series of promoter fragments with progressive deletions to map the minimal promoter region and identify key regulatory elements. The AS3MT promoter has been analyzed using fragments ranging from positions -1226 to +630 relative to the transcription start site .

  • Mutational controls: Include constructs with site-directed mutations in putative transcription factor binding sites. For the AS3MT promoter, mutations in the GC box that binds Sp1 are particularly informative . Compare these with wild-type constructs to assess the contribution of specific elements.

  • Cell type controls: Test promoter activity in both AS3MT-expressing (e.g., liver-derived) and non-expressing (e.g., keratinocyte) cell lines to confirm tissue-specific regulation mechanisms.

  • Transcription factor binding controls: For EMSA or ChIP experiments investigating transcription factor binding, include competition assays with excess unlabeled probe and supershift assays with antibodies against putative binding factors (particularly Sp1 for the AS3MT promoter) .

  • Methylation controls: When investigating epigenetic regulation, include methylated and unmethylated versions of the promoter constructs, and validate with methylation-specific PCR using primers such as unmethAS3MT-F and unmethAS3MT-R .

How should discrepancies in AS3MT antibody detection across different tissues be interpreted?

Discrepancies in AS3MT antibody detection across tissues require careful interpretation and may reflect biological realities or technical limitations:

  • Tissue-specific expression levels: AS3MT is differentially expressed across tissues, with high expression in liver, kidney, and adrenal glands but low or absent expression in keratinocytes and certain endothelial cells . Low detection may reflect genuine biological variation rather than antibody failure.

  • Isoform specificity: Multiple AS3MT splice variants exist, and antibodies targeting different epitopes may detect distinct isoforms. Compare results from antibodies targeting different regions (N-terminal, central, C-terminal) to identify potential isoform-specific expression patterns .

  • Post-translational modifications: AS3MT undergoes automethylation at cysteine residues and potentially other modifications that may mask epitopes in a tissue-specific manner . Antibodies targeting modified regions may show variable detection effectiveness.

  • Technical considerations: Tissue-specific matrix effects, including different lipid compositions and endogenous peroxidases, can affect antibody performance. Optimize extraction and detection protocols for each tissue type independently.

  • Antibody validation approach: When faced with discrepancies, employ orthogonal methods such as RNA expression analysis (RT-qPCR), mass spectrometry, or functional assays to confirm protein presence independently of antibody detection.

When publishing research using AS3MT antibodies, clearly report which antibody was used (including catalog number), the specific protocols employed for each tissue type, and acknowledge potential limitations in detection sensitivity.

What is the significance of AS3MT automethylation in experimental studies of arsenic metabolism?

AS3MT automethylation represents a significant phenomenon with implications for experimental design and data interpretation:

  • Mechanistic significance: Automethylation occurs at specific cysteine residues (C33 and C62 in the mouse protein) and appears to be part of the catalytic mechanism . This self-modification may represent an intermediate state in the enzyme's catalytic cycle rather than an inactivation mechanism.

  • Experimental implications: In vitro methylation assays must account for automethylation, which occurs even in the absence of arsenical substrates. The observation that automethylated AS3MT can still methylate inorganic arsenic suggests that this modification does not abolish catalytic activity .

  • Redox sensitivity: Automethylation is enhanced by reducing agents like glutathione (GSH) and dithiothreitol (DTT), indicating that reduced cysteines are necessary for accepting methyl groups . This redox dependence suggests experimental conditions, particularly the redox environment, will significantly impact observed methylation patterns.

  • Competition with substrate methylation: The decrease in automethylation observed after adding inorganic arsenic indicates competition between protein and substrate methylation . This phenomenon may explain some of the complex kinetics observed in AS3MT-catalyzed reactions.

  • Detection strategies: When studying automethylation, radioactive labeling with [3H]-SAM provides the most direct evidence, though mass spectrometry approaches offer site-specific information about modified residues .

Understanding automethylation is essential for correctly interpreting arsenic metabolism data and designing experiments that account for this intrinsic activity of AS3MT.

How can contradictory findings regarding AS3MT protein structure-function relationships be reconciled?

Reconciling contradictory findings about AS3MT structure-function relationships requires systematic analysis of methodological differences:

  • Species differences: Human and rodent AS3MT proteins show important structural differences despite high sequence similarity. Human AS3MT contains 375 amino acids while mouse AS3MT has 376 . Critical comparison of results should account for these interspecies variations, particularly regarding conserved cysteine positions.

  • Experimental conditions: Different reducing systems used across studies (GSH/TRX/TRR vs. TCEP vs. DTT) significantly affect AS3MT activity and may lead to seemingly contradictory results. Standardization of reaction conditions or side-by-side comparisons are essential to resolve such discrepancies.

  • Oligomeric state considerations: While AS3MT has been described as monomeric in solution , other methyltransferases often function as dimers. Experimental conditions affecting protein concentration, buffer composition, or tag systems may influence oligomerization and consequently activity.

  • Mutation effects interpretation: Site-directed mutagenesis studies targeting conserved cysteines have yielded variable results. Consider whether mutations affect protein folding, stability, or catalytic mechanism specifically. Complementary approaches like circular dichroism spectroscopy can help distinguish structural from functional effects.

  • Reconciliation approaches: When faced with contradictory findings, consider:

    • Direct replication studies using multiple methodologies in parallel

    • Meta-analysis of published data with attention to methodological differences

    • Collaboration between laboratories reporting discrepant results

    • Application of emerging structural biology techniques like cryo-EM to resolve structural ambiguities

What novel applications of AS3MT antibodies are emerging in arsenic toxicology research?

Several innovative applications of AS3MT antibodies are advancing arsenic toxicology research:

  • Biomarker development: AS3MT antibodies are being employed to assess protein expression levels in peripheral blood mononuclear cells as potential biomarkers for individual differences in arsenic metabolism capacity and susceptibility to arsenic-related diseases.

  • Single-cell analysis: Integration of AS3MT immunostaining with single-cell technologies is enabling researchers to identify cell-specific responses to arsenic exposure within heterogeneous tissues, particularly in liver and kidney where cellular responses may vary dramatically.

  • Proximity labeling approaches: Antibodies conjugated to enzymes like APEX2 or BioID are being used to identify the AS3MT interactome under different arsenic exposure conditions, revealing context-specific protein-protein interactions that regulate arsenic metabolism.

  • In situ activity visualization: Development of activity-based probes coupled with anti-AS3MT antibodies allows simultaneous visualization of enzyme localization and activity in tissue sections, providing spatial information about arsenic biotransformation.

  • Conformational state-specific antibodies: Emerging research aims to develop antibodies that specifically recognize AS3MT in different conformational states (e.g., reduced vs. oxidized, automethylated vs. unmethylated), offering new insights into the dynamic regulation of enzyme activity in vivo.

These applications are expanding beyond traditional detection methods to provide mechanistic insights into how AS3MT functions within the complex cellular environment during arsenic exposure.

How can genetic variations in AS3MT be effectively studied using antibody-based approaches?

Investigating genetic variations in AS3MT using antibody-based approaches requires sophisticated methodologies:

  • Variant-specific antibodies: Development of antibodies that specifically recognize common AS3MT variants (such as those in the 10q24.32 region associated with arsenic metabolism efficiency) enables direct assessment of variant protein levels and cellular localization.

  • Allele-specific expression analysis: Combining AS3MT immunoprecipitation with mass spectrometry or RNA sequencing allows quantification of allele-specific expression in heterozygous individuals, revealing potential regulatory effects of promoter or enhancer variants.

  • Functional impact assessment: Antibodies targeting specific AS3MT domains can be used to compare wild-type and variant proteins in terms of:

    • Substrate binding capacity through co-immunoprecipitation

    • Protein stability through pulse-chase experiments followed by immunoblotting

    • Subcellular localization through immunofluorescence microscopy

    • Post-translational modifications through modification-specific antibodies

  • Tissue-specific expression patterns: Immunohistochemistry with validated AS3MT antibodies can reveal how genetic variants affect tissue-specific expression patterns, potentially explaining differential susceptibility to arsenic toxicity.

  • Methodological considerations: When studying variants, researchers should:

    • Verify antibody recognition of variant forms using recombinant proteins

    • Include appropriate controls expressing different variants at known levels

    • Consider haplotype effects rather than focusing on single nucleotide polymorphisms in isolation

    • Integrate antibody-based findings with functional assays measuring enzymatic activity

What techniques can be combined with AS3MT antibodies to advance understanding of arsenic biotransformation pathways?

Integrating AS3MT antibodies with complementary techniques creates powerful research approaches:

  • ChIP-seq analysis: Combining chromatin immunoprecipitation with sequencing and AS3MT promoter antibodies can identify transcription factors and epigenetic modifications that regulate AS3MT expression across different cellular states, particularly in response to environmental stressors .

  • Proximity-dependent labeling: BioID or APEX2 fusions with AS3MT followed by streptavidin pulldown and mass spectrometry can map the dynamic protein interaction network of AS3MT under various arsenical exposures, revealing cofactors and regulatory proteins.

  • Live-cell imaging: Development of function-blocking AS3MT antibody fragments (Fabs) or intrabodies allows visualization of enzyme dynamics in living cells during arsenic exposure, providing temporal information about localization changes and potential degradation.

  • Multi-omics integration: Correlation of AS3MT protein levels (quantified by immunoassays) with:

    • Transcriptomics data to identify co-regulated genes

    • Metabolomics profiles to map arsenical species and other affected metabolites

    • Epigenetic modifications to understand regulatory mechanisms

  • Single-molecule techniques: Combining fluorescently labeled AS3MT antibodies with techniques like FRET or super-resolution microscopy can reveal conformational changes and interactions at the molecular level.

  • In vivo imaging: Development of radiolabeled AS3MT antibodies or fragments for PET imaging in animal models could allow non-invasive tracking of AS3MT expression changes during chronic arsenic exposure, providing translational insights into human arsenic metabolism.

These integrated approaches promise to advance understanding beyond what conventional antibody applications can achieve alone, addressing complex questions about the regulation and function of AS3MT in arsenic biotransformation.

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 2025 TheBiotek. All Rights Reserved.