AHCY catalyzes the reversible hydrolysis of SAH, a byproduct of methylation reactions, into adenosine and L-homocysteine. This reaction is NAD+-dependent and regulates intracellular SAH levels, which competitively inhibit methyltransferases . Key functional insights:
Catalytic Mechanism: AHCY employs a redox-driven nucleophilic cascade involving transient oxidation at the C3′ position of adenosine .
Regulation: Post-translational modifications (e.g., lysine acetylation at K401/408) reduce catalytic efficiency by altering hydrogen bonding near active sites . Copper ions non-competitively inhibit AHCY by disrupting NAD+ binding .
AHCY Human, Sf9 is produced via baculovirus-mediated expression in Sf9 cells, followed by proprietary chromatographic purification. Advantages of this system include:
High-Yield Glycosylation: Mimics human post-translational modifications more accurately than bacterial systems .
Stability: Maintains activity for 2–4 weeks at 4°C or longer at -20°C with carrier proteins (e.g., 0.1% HSA/BSA) .
Hypermethioninemia: AHCY deficiency elevates SAH and methionine levels, causing this metabolic disorder . Three pathogenic AHCY mutations (e.g., K188R, K389R) reduce enzyme activity by 30–50% .
Copper Metabolism: AHCY binds copper at levels comparable to albumin, suggesting a role in Wilson disease pathology .
Inhibitor Studies: AHCY inhibitors are explored for antiviral and anticancer therapies due to their impact on methylation-dependent viral replication and oncogene silencing .
EC 3.3.1.1, SAHH, AdoHcyase, S-adenosyl-L-homocysteine hydrolase, AHCY, Adenosylhomocysteinase.
Sf9, Baculovirus cells.
ADLMSDKLPY KVADIGLAAW GRKALDIAEN EMPGLMRMRE RYSASKPLKG ARIAGCLHMT VETAVLIETL VTLGAEVQWS SCNIFSTQDH AAAAIAKAGI PVYAWKGETD EEYLWCIEQT LYFKDGPLNM ILDDGGDLTN LIHTKYPQLL PGIRGISEET TTGVHNLYKM MANGILKVPA INVNDSVTKS KFDNLYGCRE SLIDGIKRAT DVMIAGKVAV VAGYGDVGKG CAQALRGFGA RVIITEIDPI NALQAAMEGY EVTTMDEACQ EGNIFVTTTG CIDIILGRHF EQMKDDAIVC NIGHFDVEID VKWLNENAVE KVNIKPQVDR YRLKNGRRII LLAEGRLVNL GCAMGHPSFV MSNSFTNQVM AQIELWTHPD KYPVGVHFLP KKLDEAVAEA HLGKLNVKLT KLTEKQAQYL GMSCDGPFKP DHYRYHHHHH H.
AHCY (Adenosylhomocysteinase) is an enzyme that catalyzes the reversible hydrolysis of S-adenosylhomocysteine (AdoHcy) to adenosine (Ado) and L-homocysteine (Hcy). The human recombinant version produced in Sf9 baculovirus cells is a valuable research tool because it controls intracellular S-adenosylhomocysteine (SAH) concentration that is crucial for transmethylation reactions .
AHCY deficiency causes hypermethioninemia and represents a natural model system for investigating processes related to the methylome. This enzyme plays a critical role in methylation research, which impacts gene expression, imprinting, signaling, protein synthesis, and lipid metabolism .
Methodologically, AHCY Human from Sf9 provides researchers with a consistent source of this enzyme that avoids the limitations of extracting the protein from human tissue samples, offering reproducible activity and suitable quantities for experimental work.
The recombinant AHCY Human produced in Sf9 cells is a single, glycosylated polypeptide chain containing 441 amino acids (comprising amino acids 1-432 of the native sequence) with a 6 amino acid His-Tag at the C-terminus . The protein has a molecular mass of 48.8 kDa but migrates at 40-57 kDa on SDS-PAGE under reducing conditions, likely due to its glycosylation pattern .
The protein is purified through proprietary chromatographic techniques to greater than 90% purity as determined by SDS-PAGE analysis . It is typically supplied in a solution containing Phosphate Buffered Saline (pH 7.4) and 10% glycerol .
For experimental validation, researchers should confirm:
Band pattern on SDS-PAGE (40-57 kDa range)
Enzymatic activity through substrate conversion assays
Protein concentration using standard methods (Bradford, BCA)
Glycosylation status if relevant to experimental goals
Proper storage and handling are critical for maintaining AHCY enzymatic activity. Based on manufacturer protocols and research practices, follow these evidence-based recommendations:
Storage Condition | Duration | Procedure | Notes |
---|---|---|---|
Short-term (2-4 weeks) | 4°C | Store in original buffer | Avoid contamination |
Long-term | -20°C | Frozen storage | Add carrier protein for stability |
Extended preservation | -20°C | Add 0.1% HSA or BSA | Protects against denaturation |
For optimal handling:
Avoid multiple freeze-thaw cycles as they significantly reduce enzyme activity
When working with the enzyme, keep it on ice to minimize degradation
Consider preparing single-use aliquots to prevent repeated freezing and thawing
If dilution is necessary, use the same buffer formulation as the stock solution
These storage conditions help preserve the native conformation and activity of the enzyme, ensuring reliable experimental results.
Optimizing assay conditions is crucial for obtaining reliable enzymatic activity measurements of AHCY Human, Sf9. Based on biochemical principles and research practices, consider the following parameters:
Parameter | Optimal Range | Methodological Considerations |
---|---|---|
Buffer | 50-100 mM Phosphate, pH 7.2-7.4 | Maintains physiological environment |
Reducing Agent | 1-5 mM DTT or β-mercaptoethanol | Protects critical cysteine residues |
Cofactor | 0.1-0.5 mM NAD+ | Essential for catalytic activity |
Temperature | 37°C | Reflects human physiological temperature |
Reaction direction | Hydrolysis vs. Synthesis | Use adenosine deaminase to drive hydrolysis |
Substrate concentration | 10-100 μM S-adenosylhomocysteine | Balance between sensitivity and substrate inhibition |
For the reversible reaction, note that:
The equilibrium naturally favors SAH synthesis rather than hydrolysis under physiological conditions
For hydrolysis direction assays, addition of adenosine deaminase removes adenosine, driving the reaction forward
For synthesis direction, use excess homocysteine to drive the reaction towards SAH formation
Researchers should optimize these conditions specifically for their experimental goals, conducting preliminary kinetic analyses to determine the linear range of the assay.
The choice of expression system significantly impacts post-translational modifications (PTMs) of recombinant proteins. For AHCY expressed in Sf9 cells versus mammalian systems:
Glycosylation patterns:
Sf9 cells produce primarily high-mannose type N-glycans rather than the complex glycans found in mammalian cells
This contributes to the heterogeneous migration pattern (40-57 kDa) observed on SDS-PAGE
While glycosylation differences generally don't significantly impact catalytic activity, they may affect protein stability and recognition by glycan-binding proteins
Phosphorylation profiles:
Insect cells possess different kinase specificities compared to human cells
Studies with other proteins show that phosphorylation patterns may differ substantially between Sf9 and mammalian expression
In the case of muscarinic cholinergic receptors expressed in Sf9 cells, agonist-induced phosphorylation was observed for the hm2 but not the hm1 receptor, demonstrating the system-specific nature of phosphorylation events
For research requiring native human PTM profiles, consider:
Comparing key experimental results with mammalian-expressed AHCY when feasible
Using mass spectrometry to characterize PTM differences when critical to experimental outcomes
Assessing whether the observed PTMs affect the specific protein properties under investigation
Investigating protein-protein interactions involving AHCY requires careful selection of complementary techniques. Based on published approaches and biochemical principles:
In vitro approaches:
Pull-down assays using His-tagged AHCY as bait
Surface Plasmon Resonance (SPR) for quantitative binding kinetics
Size Exclusion Chromatography with Multi-Angle Light Scattering (SEC-MALS) to characterize complex formation
Cell-based methods:
Co-immunoprecipitation: Demonstrated successful detection of AHCY-interacting proteins in cell lysates
Proximity Ligation Assay (PLA) for visualizing interactions with spatial resolution
FRET-based approaches for real-time interaction monitoring
Control experiments are critical:
Include competition experiments with untagged proteins
Perform reciprocal pull-downs where feasible
Use structurally unrelated proteins with similar tags as negative controls
Consider potential artifacts from the His-tag (see section 2.4)
A study examining tau protein interactions demonstrated successful co-immunoprecipitation methodology: When HA-tagged tau was overexpressed in HEK293T cells and exposed to a reversible crosslinker, immunoblot analysis revealed endogenous AHCYL1/IRBIT in the co-IP cell extracts. Reciprocal co-IP with AHCYL1/IRBIT antibody successfully pulled down HA-tagged tau, confirming the interaction .
The C-terminal 6× His-tag in AHCY Human, Sf9 enables efficient purification but may introduce artifacts in interaction studies. Evidence-based strategies to address this include:
Validation through multiple approaches:
Compare interactions using differently tagged versions (FLAG, GST) of AHCY
Employ tag-removal strategies using engineered protease sites when critical
Conduct competition experiments with imidazole or free histidine to identify tag-dependent interactions
Use alternative tag positions (N-terminal vs. C-terminal) to distinguish genuine from artifactual interactions
Common His-tag artifacts to consider:
Non-specific binding to metal-binding proteins
Interactions with negatively charged surfaces
Potential oligomerization mediated by the tag rather than the native protein
Structural considerations:
Assess the location of the tag relative to known binding interfaces
Consider the impact of tag position on protein folding and conformation
For crystallography studies, evaluate whether the tag affects crystal packing
When validating AHCY interactions with potential partners, combining orthogonal methods that do not rely solely on the His-tag provides the most robust evidence for physiologically relevant interactions.
Accurate quantification of AHCY in biological matrices requires sensitive and specific analytical approaches. Several validated methods include:
Mass spectrometry-based approaches:
iTRAQ (isobaric tags for relative and absolute quantitation) has successfully quantified AHCY in colorectal cancer tissue specimens with high accuracy and reproducibility
Use of double references increases reliability of quantification results
Absolute quantification can be achieved using stable isotope-labeled peptide standards
Immunoassay methods:
ELISA-based detection using antibodies specific to human AHCY
Western blotting with densitometry for semi-quantitative analysis
Capillary immunoelectrophoresis for higher sensitivity
Sample preparation considerations:
Protein extraction methods should be optimized to effectively solubilize membrane-associated pools of AHCY
Protease inhibitors must be included to prevent degradation during processing
Normalization to housekeeping proteins or total protein is essential for cross-sample comparison
Research by Herbrich et al. demonstrated that statistical inference from multiple iTRAQ experiments without using common reference standards can provide more precise estimates of protein relative abundance, which is applicable to AHCY quantification in complex samples .
AHCY deficiency causes hypermethioninemia, making the recombinant protein valuable for investigating methylation disorders. Methodological approaches include:
In vitro disease modeling:
Compare wild-type AHCY activity with known disease-causing mutations
Reconstitute the methionine cycle in vitro to assess pathway perturbations
Screen for small molecules that rescue mutant AHCY activity
Translational applications:
Develop enzyme activity assays that correlate with clinical phenotypes
Assess the impact of S-adenosylmethionine/S-adenosylhomocysteine ratio perturbations
Evaluate potential therapeutic strategies targeting the methylation pathway
Experimental design considerations:
Include appropriate disease-relevant mutations as controls
Assess both hydrolysis and synthesis directions of the reaction
Consider the physiological context of AHCY function in the methionine cycle
AHCY deficiency represents a natural model system for methylation research, linking genomics, proteomics, cellomics, lipidomics, and metabolomics in what has been termed "AHCYdomics" . This integrated approach enables exploration of the full impact of this human methylation disorder and contributes to better understanding of the human methylome.
Developing high-throughput screening (HTS) assays for AHCY inhibitors requires careful assay design and validation. Based on successful approaches in enzyme inhibitor discovery:
Assay development considerations:
Establish Z' factor >0.5 to ensure assay robustness
Determine optimal substrate concentration (typically at or below Km)
Select an assay readout compatible with automated systems (fluorescence, luminescence)
Validate with known inhibitors (adenosine dialdehyde, D-eritadenine)
Detection methods:
Mass spectrometry: The RapidFire high-throughput mass spectrometry (RF-MS) system has been successfully applied to enzyme inhibitor screening with approximately 7 seconds per sample analysis time
Coupled enzyme assays: Monitor NAD(P)H production/consumption spectrophotometrically
Direct measurement of adenosine production
Control experiments:
Include vehicle controls (matching DMSO concentrations)
Test for assay interference using counter-screens
Evaluate compound aggregation potential with detergent controls
Vary enzyme concentration to ensure linear assay response
A directed screen of 78,000 compounds for human Kynurenine 3-Monooxygenase used similar methodology with recombinant protein expressed in Sf9 cells, demonstrating the feasibility of this approach for membrane-associated enzymes .
The choice of expression system significantly impacts recombinant protein yield and activity. Comparative studies provide valuable insights:
Expression efficiency comparison:
Sf9 cells typically yield significantly higher amounts of active protein compared to mammalian systems for many human proteins
For similar recombinant proteins, studies show approximately 60-fold higher activity in Sf9-derived preparations compared to HEK293-derived preparations
Active site titration indicated that active recombinant protein comprised approximately 2% of total protein in Sf9-derived membranes but only about 0.02% in HEK293-derived membranes
Quality considerations:
Bacterial expression systems often produce inclusion bodies requiring refolding
Mammalian systems provide more native-like post-translational modifications but lower yields
Sf9 cells offer a balance of reasonable eukaryotic modifications with higher expression levels
Optimization strategies:
High-density transfection protocols have been developed for Sf9 cells that significantly improve protein yields
Studies indicate that plasmid delivery efficiency can be high (>40-80% of DNA present within cells) within minutes of transfection
Temporary cell growth inhibition following high-density transfection may contribute to improved yield
These findings suggest that for many applications, the Sf9 expression system provides an optimal balance of protein yield, activity, and post-translational modifications for human AHCY production.
Structural biology studies with AHCY Human, Sf9 require careful planning to optimize sample preparation and experimental success:
Crystallography considerations:
The glycosylation heterogeneity may complicate crystallization
Consider enzymatic deglycosylation for homogeneous sample preparation
The C-terminal His-tag may require removal for optimal crystal packing
Inclusion of cofactor (NAD+) and/or substrate analogs can stabilize the protein
Cryo-EM preparations:
The relatively small size of AHCY (48.8 kDa) may present challenges for cryo-EM
Consider Fab complexes to increase particle size and provide fiducial markers
Successful Fab production in Sf9 cells has been demonstrated for structural studies
NMR studies:
Isotopic labeling in insect cells is more challenging than in bacterial systems
Consider domain-based approaches for larger proteins like AHCY
Selective labeling of specific residue types may be necessary
Protein engineering strategies:
Surface entropy reduction mutations may improve crystallizability
Domain truncations based on limited proteolysis can identify stable fragments
Consider engineering constructs with reduced intrinsic flexibility
In a recent study, researchers successfully expressed and purified Fab fragments from Sf9 cells for structural biology applications. The Fab heavy and light chain genes were synthesized and cloned into pFastBac Dual vector, with purification via metal ion affinity chromatography followed by size-exclusion chromatography . This approach could be adapted for generating antibody fragments against AHCY for structural studies.
Adenosylhomocysteinase (AHCY) is a crucial enzyme involved in the metabolism of S-adenosylhomocysteine (SAH). The recombinant form of this enzyme, produced in Sf9 Baculovirus cells, is widely used in research to study its function and role in various biological processes.
The human recombinant AHCY produced in Sf9 cells is a single, glycosylated polypeptide chain containing 441 amino acids, with a molecular mass of approximately 48.8 kDa . This enzyme is fused to a 6-amino acid His-Tag at the C-terminus, which facilitates its purification through chromatographic techniques .
AHCY catalyzes the reversible hydrolysis of S-adenosylhomocysteine (AdoHcy) into adenosine (Ado) and L-homocysteine (Hcy) . This reaction is crucial for maintaining the intracellular concentration of SAH, which is essential for transmethylation reactions . Transmethylation is a vital process in which methyl groups are transferred from one molecule to another, playing a significant role in DNA methylation, protein function, and lipid metabolism .
The regulation of SAH levels by AHCY is critical for various cellular processes. Methylation, controlled by AHCY, influences gene expression, protein function, and signal transduction . A deficiency in AHCY can lead to hypermethioninemia, a condition characterized by elevated levels of methionine in the blood .
The recombinant form of AHCY produced in Sf9 cells is used extensively in laboratory research. It allows scientists to study the enzyme’s structure, function, and interactions in a controlled environment. This research can lead to a better understanding of metabolic disorders and the development of potential therapeutic interventions .
AHCY (Human Recombinant, Sf9) is typically stored at 4°C for short-term use (2-4 weeks) and at -20°C for long-term storage. It is recommended to add a carrier protein, such as 0.1% HSA or BSA, to prevent degradation during storage . The enzyme should be handled carefully to avoid multiple freeze-thaw cycles, which can affect its stability and activity .