ACE2 (18-740) Human is a 723-amino-acid polypeptide (excluding tags) produced in mammalian expression systems (e.g., HEK293 or baculovirus) . It retains the extracellular catalytic domain but excludes the transmembrane (residues 741–761) and cytoplasmic (762–805) regions . Key features include:
Neutralization Assays: Competes with membrane-bound ACE2 for spike binding, reducing viral infectivity (EC₅₀ ≤100 ng/mL) .
Structural Biology: Used in cryo-EM and crystallography to map ACE2-spike interactions .
Kinetic Parameters: Hydrolyzes Mca-YVADAPK(Dnp)-OH with specific activity >250 pmol/min/μg .
Therapeutic Potential: Recombinant ACE2 (APN01) has entered clinical trials for COVID-19 and pulmonary hypertension .
Storage: Stable at -80°C in PBS (pH 7.4) with 10–20% glycerol .
Solubility: Maintains activity in aqueous buffers (e.g., 40 mM Tris-HCl, 110 mM NaCl) .
COVID-19 Therapy: Intranasal ACE2 (18-740) reduced brain SARS-CoV-2 titers and mortality in murine models .
RAAS Modulation: Soluble ACE2 lowers Ang II levels, attenuating lung injury and hypertension .
Feature | ACE2 (18-740) | Truncated ACE2 (dACE2) |
---|---|---|
Viral Binding | Yes | No |
Enzymatic Activity | Retained | Lost |
Tissue Distribution | Broad (lungs, heart, gut) | Lung epithelium, bile duct |
RAAS Modulation: ACE2 (18-740) counterbalances ACE by degrading Ang II, offering protection against hypertension and fibrosis .
Allosteric Regulation: Dynamic structural changes enable spike protein binding and catalytic activity .
Genetic Variants: Seven ACE2 polymorphisms (e.g., Lys26Arg, Asn638Ser) alter SARS-CoV-2 affinity but are rare in populations .
Angiotensin-converting enzyme 2 (ACE2) is an enzyme found on the surface of cells in various organs, including the intestines, arteries, lungs, heart, and kidneys. ACE2 serves as an entry point for SARS coronaviruses, including SARS-CoV-2, the virus responsible for COVID-19. The coronavirus spike (S) glycoprotein, a class I viral fusion antigen found on the outer envelope of the virus, plays a crucial role in viral infection. It does this by binding to host cell receptors and facilitating the fusion of viral and cellular membranes. The S1 subunit of the spike protein contains two main domains: the N-terminal domain and the C-terminal domain. One or both of these domains can act as a receptor-binding domain. SARS-CoV and MERS-CoV both utilize their C-domain to bind to their respective receptors. ACE2 is a type I transmembrane protein with an extracellular N-terminal domain containing the catalytic site and an intracellular C-terminal tail. Structurally, ACE2 consists of a signal peptide, a transmembrane domain, and a single metalloproteinase active site containing a HEXXH zinc-binding domain. Functionally, ACE2 acts as a mono-carboxypeptidase, an enzyme that cleaves peptide bonds at the carboxyl terminus of an amino acid. ACE2 degrades Angiotensin I to produce the nonapeptide Angiotensin 1-9 and Angiotensin II to create the heptapeptide Angiotensin 1-7.
This recombinant ACE2 protein is produced in HEK293 cells and corresponds to the human ACE2 protein sequence from amino acids 18 to 740. It is fused to an IgG-His tag at the N-terminus, adding 242 amino acids and resulting in a total molecular weight of 110.8 kDa. This ACE2 protein specifically binds to the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein.
This product is supplied as a 0.5 mg/ml solution of ACE2 Human protein in phosphate-buffered saline (PBS) with a pH of 7.4 and 10% glycerol.
ACE-2 Human Recombinant Protein is shipped with ice packs to maintain a cool temperature during transport. Upon receipt, it should be stored at -20°C (-4°F).
The purity of this protein is greater than 90% as determined by SDS-PAGE analysis.
The biological activity of this protein is determined by its ability to bind to the SARS-CoV-2 Spike RBD in a functional ELISA assay. The ED50 value, representing the concentration at which 50% binding is observed, is less than or equal to 100 ng/ml. The specific activity, defined as the amount of enzyme required to hydrolyze 1.0 picomole of the substrate Mca-YVADAPK(Dnp)-OH per minute at a pH of 7.5 and a temperature of 25°C, is greater than 250 picomoles/min/microgram.
HEK293 Cells
DGS QSTIEEQ AKTFLDKFNH EAEDLFYQSS LASWNYNTNI TEENVQNMNN AGDKWSAFLK EQSTLAQMYP LQEIQNLTVK LQLQALQQNG SSVLSEDKSK RLNTILNTMS TIYSTGKVCN PDNPQECLLL EPGLNEIMAN SLDYNERLWA WESWRSEVGK QLRPLYEEYV VLKNEMARAN HYEDYGDYWR GDYEVNGVDG YDYSRGQLIE DVEHTFEEIK PLYEHLHAYV RAKLMNAYPS YISPIGCLPA HLLGDMWGRF WTNLYSLTVP FGQKPNIDVT DAMVDQAWDA QRIFKEAEKF FVSVGLPNMT QGFWENSMLT DPGNVQKAVC HPTAWDLGKG DFRILMCTKV TMDDFLTAHH EMGHIQYDMA YAAQPFLLRN GANEGFHEAV GEIMSLSAAT PKHLKSIGLL SPDFQEDNET EINFLLKQAL TIVGTLPFTY MLEKWRWMVF KGEIPKDQWM KKWWEMKREI VGVVEPVPHD ETYCDPASLF HVSNDYSFIR YYTRTLYQFQ FQEALCQAAK HEGPLHKCDI SNSTEAGQKL FNMLRLGKSE PWTLALENVV GAKNMNVRPL LNYFEPLFTW LKDQNKNSFV GWSTDWSPYA DQSIKVRISL KSALGDKAYE WNDNEMYLFR SSVAYAMRQY FLKVKNQMIL FGEEDVRVAN LKPRISFNFF VTAPKNVSDI IPRTEVEKAI RMSRSRINDA FRLNDNSLEF LGIQPTLGPP NQPPVSLEPK SCDKTHTCPP CPAPELLGGP SVFLFPPKPK DTLMISRTPE VTCVVVDVSHEDPEVKFNWY VDGVEVHNAK TKPREEQYNS TYRVVSVLTV LHQDWLNGKE YKCKVSNKAL PAPIEKTISK AKGQPREPQVYTLPPSRDEL TKNQVSLTCL VKGFYPSDIA VEWESNGQPE NNYKTTPPVL DSDGSFFLYS KLTVDKSRWQ QGNVFSCSVMHEALHNHYTQ KSLSLSPGKH HHHHH
ACE2 (18-740) refers to the extracellular domain (amino acids 18-740) of human Angiotensin-converting enzyme 2, which plays a central role in vascular, renal, and myocardial physiology. Unlike its homolog ACE, ACE2 converts angiotensin I to Ang1-9 and angiotensin II to Ang1-7. This domain is critically important because it contains the catalytic site responsible for these conversions and serves as the primary binding site for SARS coronaviruses. In knockout studies, ace2-/ace2- mice demonstrate severely reduced cardiac contractility, highlighting ACE2's importance in heart function regulation . The recombinant ACE2 (18-740) protein has become an essential tool for researchers studying cardiovascular physiology, COVID-19 pathogenesis, and therapeutic development against SARS-CoV-2.
Mammalian expression systems are strongly preferred for producing high-quality ACE2 (18-740) Human recombinant protein. Commercial sources typically use HEK 293 cells for expression, as seen in multiple product descriptions . Mammalian systems are essential because ACE2 requires proper glycosylation and other post-translational modifications for correct folding and function. When evaluating expression quality, researchers should expect >95% purity by SDS-PAGE analysis and endotoxin levels below 1 EU/μg . Alternative expression systems like bacterial or insect cells may yield higher quantities but typically produce proteins with altered glycosylation patterns that might not fully recapitulate the native human ACE2 functions, particularly for binding studies with viral proteins.
For optimal preservation of ACE2 (18-740) Human activity, the recombinant protein should typically be stored at -80°C for long-term storage, with working aliquots kept at -20°C to minimize freeze-thaw cycles. When handling the protein for experiments, it should be thawed gently on ice and maintained at 4°C during experimental preparations. The buffer composition is crucial for stability – typically PBS or HEPES-based buffers with controlled pH (7.2-7.4) are recommended, potentially with low concentrations of stabilizing agents. For certain applications like Surface Plasmon Resonance assays, the protein may require desalting using appropriate molecular weight cutoff columns (e.g., Zeba Spin 7K MWCO columns) . Researchers should validate protein activity after any modification to storage or handling protocols by assessing enzymatic function or binding capabilities.
Engineering ACE2 (18-740) for enhanced viral binding requires strategic amino acid substitutions at the ACE2/SARS-CoV-2 interface. Research has identified three key mutations—T27L, H34V, and N90E (collectively termed the "LVE" variant)—that dramatically improve binding affinity across multiple SARS-CoV-2 variants . The T27L and H34V substitutions optimize interactions with SARS-CoV-2 RBD amino acids 473 and 456, respectively, while N90E eliminates an N-linked glycosylation site that creates steric hindrance at the binding interface . Molecular modeling using software like Protean 3D with DFIRE scoring algorithms can predict optimal mutations based on stabilizing interactions. The effectiveness of these engineered variants is remarkable, with binding affinities improving from the nanomolar range (for wild-type ACE2) to the picomolar or even femtomolar range for engineered variants, representing up to 7,000-fold enhancement in binding strength for certain SARS-CoV-2 variants .
Surface Plasmon Resonance (SPR) provides the most accurate and comprehensive analysis of binding affinities between ACE2 (18-740) and SARS-CoV-2 spike proteins. The methodology requires careful preparation: proteins should be desalted on appropriate columns (e.g., Zeba Spin 7K MWCO), and binding assays conducted in HBS-N buffer containing EDTA and Tween 20, with flow rates of approximately 30 μL/min . Typical experimental parameters include 120 seconds for association and 180 seconds for dissociation, with reference-subtracted SPR binding curves analyzed using a 1:1 binding model to derive kinetic parameters and dissociation constants (KD) . The table below shows comparative binding affinities for different ACE2 constructs against various SARS-CoV-2 variants:
ACE-2 Construct | SARS-CoV-2 Variant | KD Value |
---|---|---|
Wild-type ACE-2 Fc (Genscript) | Wuhan variant, S1 subunit | 3.0 nM |
LiVE/STR chimera | Alpha variant B.1.1.7, S1 subunit | 378 pM |
LiVE-Longer YTE chimera | Omicron BA.2 spike protein trimer | 78 fM |
LiVE-Longer YTE chimera | Omicron BQ.1.1 spike protein trimer | 1.81 pM |
Wild-type ACE-2 Fc (ACRO) | Omicron XBB.1 spike protein trimer | 22.4 nM |
LiVE-Longer YTE chimera | Omicron XBB.1 spike protein trimer | 215 pM |
This systematic approach enables quantitative comparison of binding affinities across different viral variants and ACE2 constructs .
ACE2 (18-740) can be incorporated into therapeutic development through several advanced strategies. One promising approach involves creating chimeric ACE2/Fc fusion proteins with enhanced binding properties. These chimeras combine an engineered ACE2 domain (containing mutations like T27L, H34V, and N90E) with modified antibody domains . Two key variants include an "Fc-silent" version (containing L234S/L235T/G236R mutations, termed "STR") that minimizes antibody-dependent enhancement (ADE) risk, and a "live longer" Fc-silent version with YTE mutations (M252Y/S254T/T256E) that increase FcRn receptor binding for extended biological half-life . These chimeric constructs have demonstrated ultrahigh binding affinities (in the picomolar to femtomolar range) to a wide variety of SARS-CoV-2 variants, including emerging Omicron sublineages . The therapeutic potential of these constructs should be validated through surrogate viral neutralization tests and live virus neutralization assays with physiologically relevant models like airway organoids, which better recapitulate human respiratory tissues than simple cell culture systems .
When designing viral binding studies with ACE2 (18-740), several essential controls must be incorporated for result validation. First, researchers should include both positive and negative binding controls: a validated wild-type ACE2-Fc construct (such as those from GenScript or ACRO) serves as a positive control with established binding properties (KD ~3-16 nM for the Wuhan variant) , while an irrelevant protein of similar size and structure provides a negative control to assess non-specific binding. Second, concentration-dependent binding assays should include a range spanning at least three orders of magnitude (e.g., 0.0064-100 μg/ml as described in viral neutralization studies) . Third, multiple viral variants should be tested in parallel to assess binding specificity, particularly when evaluating engineered ACE2 variants designed for "variant-agnostic" binding. Finally, when conducting SPR or similar binding assays, researchers should perform reciprocal experimental setups (immobilizing either ACE2 or viral protein) to ensure binding parameters are consistent regardless of which partner is immobilized.
A comprehensive neutralization assay design for testing ACE2 (18-740) against SARS-CoV-2 should employ a multi-tiered approach. Initially, surrogate Viral Neutralization Tests (sVNT) provide efficient screening capability. The GenScript C-PASS system described in the literature uses an ELISA-based approach with HRP-conjugated recombinant SARS-CoV-2 RBD fragment and human ACE2 receptor protein . For definitive analysis, researchers should progress to live virus neutralization assays in appropriate biosafety level 3 facilities. A methodologically sound approach involves challenging human airway organoids (AOs) with virus (MOI = 0.1) in the presence of ACE2 proteins at concentrations ranging from 0.0064 to 100 μg/ml (n=3 replicates per concentration) . After 24 hours, media should be replaced (including fresh ACE2 proteins), followed by supernatant harvesting 24 hours later for viral load quantification . This design enables generation of complete dose-response curves and determination of IC50 values for neutralization activity, allowing direct comparison between different ACE2 constructs or against therapeutic antibodies.
Multiple factors can impact experimental reproducibility when working with ACE2 (18-740), requiring careful experimental design and quality control. Batch-to-batch variation in protein production is a primary concern—researchers should verify >95% purity by SDS-PAGE and HPLC for each batch and perform comparative binding assays to confirm consistent functionality. Storage conditions significantly affect stability, with freeze-thaw cycles potentially degrading activity; researchers should use fresh aliquots whenever possible and validate protein activity before critical experiments. Buffer composition can alter protein conformation and activity, necessitating consistent preparation; for certain applications like SPR, desalting procedures using specific columns (e.g., Zeba Spin 7K MWCO) are recommended . Post-translational modifications, particularly glycosylation patterns, affect ACE2 functionality; the N90 glycosylation site specifically impacts SARS-CoV-2 binding, highlighting the importance of consistent expression systems . Finally, experimental conditions (temperature, incubation times, reagent concentrations) must be meticulously standardized and documented to enable meaningful comparisons across experiments and between laboratories.
The molecular basis for differential binding of ACE2 (18-740) to various SARS-CoV-2 variants lies in specific amino acid interactions at the binding interface. Molecular modeling studies have revealed that certain amino acid positions in both ACE2 and the viral receptor binding domain (RBD) are critical determinants of binding affinity . In wild-type ACE2, residues T27 and H34 interact with SARS-CoV-2 RBD amino acids at positions 455, 453, 473, and 456 . Mutations in these regions, either in the viral RBD (as occurs in variants like Alpha, Delta, and Omicron) or in engineered ACE2 constructs (such as the T27L and H34V substitutions), can significantly alter binding energetics. The T27L and H34V substitutions in engineered ACE2 variants optimize interactions with conserved SARS-CoV-2 RBD residues, creating a "variant-agnostic" binding profile . Additionally, the N90E substitution eliminates glycosylation-related steric hindrance at the binding interface . The effectiveness of these strategic substitutions is demonstrated by the dramatic improvement in binding affinities, from nanomolar KD values for wild-type ACE2 to picomolar or femtomolar values for engineered variants, representing up to several thousand-fold enhancement in binding strength .
ACE2 (18-740) can serve as a powerful tool for predicting the binding properties of emerging SARS-CoV-2 variants through a systematic analytical approach. Researchers can express and purify the spike protein RBD or S1 subunit from new variants, then perform quantitative binding studies using Surface Plasmon Resonance (SPR) . By comparing binding kinetics and affinities (KD values) to those of established variants, researchers can rapidly classify new variants according to their ACE2 binding properties. This approach is particularly valuable when combined with engineered "variant-agnostic" ACE2 constructs like the LVE variant, which maintains high affinity across multiple viral lineages . The methodology should follow established protocols: proteins desalted on appropriate columns, binding assays conducted in HBS-N buffer with EDTA and Tween 20, flow rates of 30 μL/min, and standard association/dissociation times . This approach allows for rapid assessment of new variants even before extensive clinical data becomes available, potentially identifying variants with enhanced transmission capabilities due to altered ACE2 binding properties.
ACE2 (18-740)-based therapeutics offer several significant advantages over monoclonal antibodies for COVID-19 treatment or prevention. First, engineered ACE2 variants demonstrate "variant-agnostic" binding properties, maintaining high affinity across multiple SARS-CoV-2 variants including those that escape monoclonal antibody neutralization . This is because ACE2-based therapeutics target the conserved, functionally critical receptor-binding interface that viruses cannot easily mutate without compromising infectivity. Second, chimeric ACE2/Fc fusion proteins can be designed with "Fc-silent" domains (containing L234S/L235T/G236R mutations) that minimize the risk of antibody-dependent enhancement (ADE) of infection . Third, these constructs can be further engineered with YTE mutations (M252Y/S254T/T256E) in the Fc domain to increase FcRn receptor binding, extending biological half-life 3-4 fold, particularly beneficial for nasal or aerosol administration . Fourth, ACE2-based therapeutics potentially offer dual benefits by not only neutralizing the virus but also replacing ACE2 enzymatic activity that may be disrupted during infection. Finally, the ultrahigh binding affinities achieved with engineered ACE2 variants (reaching femtomolar KD values for some variants) suggest potential for higher potency at lower doses compared to many monoclonal antibodies .
When designing experiments to compare multiple engineered ACE2 (18-740) variants, researchers should implement a comprehensive, multi-parameter assessment approach. First, binding affinity characterization should employ Surface Plasmon Resonance (SPR) as described in the literature, with consistent experimental conditions across all variants and multiple viral targets representing diverse SARS-CoV-2 lineages . The resulting kinetic parameters and KD values provide quantitative metrics for direct comparison. Second, researchers should assess neutralization potency using both surrogate assays (sVNT) and live virus neutralization with physiologically relevant models like airway organoids, testing each variant across a standardized concentration range (e.g., 0.0064-100 μg/ml) to generate complete dose-response curves and determine IC50 values . Third, stability testing should evaluate thermal stability, resistance to proteolytic degradation, and shelf-life under various storage conditions. Fourth, for variants intended as therapeutics, pharmacokinetic studies should assess half-life in circulation and in target tissues, particularly for constructs with modifications designed to extend biological persistence . Finally, manufacturability assessment should evaluate expression yield, purification efficiency, and consistency across batches. This multi-parameter approach enables comprehensive comparison between variants to identify optimal candidates for specific research or therapeutic applications.
Optimizing ACE2 (18-740) expression requires systematic refinement of multiple parameters to achieve maximum yield without compromising activity. For expression system selection, HEK 293 cells have proven most effective for producing properly folded and glycosylated ACE2 . Vector design should incorporate strong promoters (such as CMV), optimized Kozak sequences, and efficient secretion signals, along with suitable purification tags (typically His-tag) . For transient transfection approaches, researchers should optimize transfection reagent type and DNA:reagent ratios through small-scale pilot experiments before scaling up. Cell culture conditions significantly impact yield—researchers should evaluate different media formulations (including those supplemented with protein expression enhancers), feeding strategies, and harvest timing. Temperature optimization can enhance proper folding; many researchers find that lowering culture temperature to 30-32°C during the expression phase improves the yield of correctly folded protein. For purification, a multi-step approach typically yields best results: IMAC using Ni-NTA for initial capture of His-tagged protein, followed by size exclusion chromatography to separate monomeric ACE2 from aggregates, and possibly ion exchange chromatography for final polishing. Throughout optimization, researchers should assess not only total protein yield but also specific activity (enzymatic function or viral binding capacity) to ensure that increased production doesn't come at the cost of reduced functionality.
Verifying the structural integrity of purified ACE2 (18-740) requires a multi-technique analytical approach addressing different aspects of protein quality. SDS-PAGE and Western blotting provide basic assessments of size, purity (which should exceed 95%) , and immunoreactivity. Size exclusion chromatography coupled with multi-angle light scattering (SEC-MALS) can determine the oligomeric state and detect aggregation. Circular dichroism (CD) spectroscopy offers information about secondary structure content, while differential scanning fluorimetry (DSF) or differential scanning calorimetry (DSC) assess thermal stability and can identify stabilizing buffer conditions. Mass spectrometry provides precise molecular weight determination and can confirm post-translational modifications, particularly glycosylation patterns which are critical for ACE2 function. Native mass spectrometry can further verify the intact structure and complex formation. Functional assays are equally important: enzymatic activity assays measuring conversion of angiotensin I to Ang1-9 or angiotensin II to Ang1-7 confirm catalytic function, while binding assays (SPR or ELISA-based) with SARS-CoV-2 RBD verify receptor functionality. For comprehensive validation, researchers should compare their purified ACE2 against commercial standards with established quality metrics. These combined approaches provide a thorough assessment of ACE2 structural integrity, essential for reliable experimental outcomes.
The most promising future research directions for ACE2 (18-740) Human span both basic science and translational applications. In therapeutics development, the creation of multivalent ACE2-based molecules could further enhance binding avidity and neutralization potency beyond the already impressive femtomolar affinities achieved with engineered variants . Next-generation ACE2 chimeras combining the viral-binding domain with novel functional domains—such as immunomodulators or tissue-targeting moieties—could expand therapeutic applications beyond direct viral neutralization. For drug delivery, ACE2-based targeting strategies could enable selective delivery of therapeutic payloads to tissues expressing SARS-CoV-2 spike protein. In diagnostics, engineered high-affinity ACE2 variants could serve as superior capture agents for ultrasensitive viral detection assays. For basic virology research, ACE2 (18-740) provides a powerful tool for studying viral evolution and predicting the emergence of variants with altered transmissibility. In structural biology, continued refinement of our understanding of the ACE2-viral interface could reveal new opportunities for therapeutic intervention. Finally, broader application of the protein engineering strategies that yielded high-affinity ACE2 variants could inform the development of engineered receptors for other emerging pathogens, creating a platform technology for rapid response to future pandemic threats.
The engineering of high-affinity ACE2 (18-740) variants has yielded several key lessons applicable to both COVID-19 research and broader protein engineering efforts. First, the remarkable success of the "LVE" (T27L, H34V, N90E) variant demonstrates that strategic mutations targeted to the protein-protein interface can dramatically enhance binding affinity, achieving improvements of several thousand-fold (from nanomolar to femtomolar KD values) . Second, the N90E mutation specifically highlights how glycosylation can create steric hindrance at binding interfaces; eliminating glycosylation sites can significantly improve protein-protein interactions when the glycan impedes binding . Third, the "variant-agnostic" binding profile of engineered ACE2 variants emphasizes the value of targeting conserved viral regions that cannot easily mutate without compromising viral fitness . Fourth, the creation of chimeric ACE2/Fc fusion proteins with modified Fc domains (STR or YTE variants) demonstrates how combining optimized binding domains with engineered functional domains can create multifunctional therapeutic candidates with enhanced properties like extended half-life . Finally, the systematic integration of computational modeling (using tools like Protean 3D with DFIRE scoring) with experimental validation (SPR and viral neutralization assays) provides a powerful template for rational protein engineering across diverse applications . These lessons extend beyond COVID-19 research to inform protein engineering strategies for other receptor-ligand systems and therapeutic targets.
ACE2 (18-740) research contributes significantly to preparedness for future coronavirus outbreaks through multiple pathways. First, the development of "variant-agnostic" high-affinity ACE2 variants provides a versatile platform for rapidly responding to novel coronaviruses that utilize ACE2 as an entry receptor . Unlike antibodies that might lose efficacy against new viral strains, engineered ACE2-based molecules target the conserved receptor-binding interface that all ACE2-utilizing coronaviruses must maintain. Second, the methodologies established for characterizing ACE2-viral interactions—including SPR binding assays, surrogate neutralization tests, and airway organoid infection models—create a standardized toolkit for rapidly assessing new viral threats . Third, the chimeric ACE2/Fc fusion protein designs, particularly those with Fc-silent domains to prevent antibody-dependent enhancement, offer templates for developing prophylactic agents that could be deployed early in an outbreak . Fourth, the molecular understanding gained from ACE2-SARS-CoV-2 structural studies informs predictions about potential mutations that might emerge in future outbreaks. Finally, the "LiVE-Longer YTE" chimera specifically designed for nasal administration represents an approach to blocking viral entry at the primary site of infection, potentially providing a model for rapid-response prophylactics against future respiratory pathogens . Collectively, these advances create both technological platforms and conceptual frameworks that can significantly accelerate response times to future coronavirus outbreaks.
ACE-2 Construct | SARS-CoV-2 Variant | KD Value | Relative Affinity |
---|---|---|---|
Wild-type ACE-2 Fc | Omicron BQ.1.1 spike | 12.6 nM | 1× |
LiVE-Longer YTE chimera | Omicron BQ.1.1 spike | 1.81 pM | ~7,000× |
Wild-type ACE-2 Fc | Omicron BA4.6 spike | 27.1 nM | 1× |
LiVE-Longer YTE chimera | Omicron BA4.6 spike | 845 pM | ~32× |
Second, researchers should compare both kinetic parameters (kon and koff rates)—affinity improvements driven by faster association rates may have different functional implications than those driven by slower dissociation rates. Third, the relationship between affinity and neutralization potency is not always linear; beyond certain affinity thresholds, other factors like steric accessibility or concentration at sites of viral entry may become limiting. Fourth, variant-specific differences can provide insights into viral evolution—consistent binding by engineered ACE2 variants across multiple viral variants demonstrates successful targeting of conserved viral features . Finally, researchers should consider physiological context—femtomolar affinities observed in purified protein systems may translate differently in complex biological environments with competing interactions and diffusion limitations.
Appropriate statistical analysis of ACE2 binding and neutralization data requires careful consideration of experimental design and data characteristics. For binding kinetics determined by SPR, non-linear regression using a 1:1 binding model is typically applied to derive association rate (kon), dissociation rate (koff), and equilibrium dissociation constant (KD) parameters . Goodness-of-fit should be evaluated using residual plots and chi-square values. When comparing multiple ACE2 variants or viral strains, researchers should perform at least three independent experiments and report mean values with standard deviations or standard errors. For neutralization assays, dose-response curves should be analyzed using four-parameter logistic regression to determine IC50 values (concentration providing 50% inhibition) . Statistical significance of differences between ACE2 variants can be assessed using parametric tests (ANOVA with appropriate post-hoc comparisons) for normally distributed data or non-parametric alternatives (Kruskal-Wallis) when normality assumptions are violated. For correlation analyses between binding affinity and neutralization potency, Spearman's rank correlation is often more appropriate than Pearson's correlation due to the non-linear relationship between these parameters. Power analysis should guide sample size determination, particularly for resource-intensive experiments like live virus neutralization assays with airway organoids . Finally, researchers should clearly report both statistical significance (p-values) and effect sizes to communicate both the reliability and magnitude of observed differences.
Computational modeling provides powerful insights into ACE2-viral interactions that complement experimental approaches. Molecular dynamics simulations can reveal dynamic aspects of the binding interface not captured in static crystal structures, identifying transient interactions and conformational changes that influence binding energetics. Molecular docking with knowledge-based scoring functions (like DFIRE described in the research) enables rapid screening of potential ACE2 mutations and prediction of their effects on binding affinity . More sophisticated approaches like free energy perturbation calculations can provide quantitative estimates of binding energy changes caused by specific mutations. Sequence-based evolutionary analysis can identify conserved regions in viral spike proteins that are constrained due to functional requirements, representing ideal targets for engineered ACE2 variants. Network analysis of amino acid interactions at the binding interface can identify key residues that contribute disproportionately to binding stability, as demonstrated by the identification of T27L, H34V, and N90E substitutions in engineered ACE2 variants . Glycan modeling can predict steric effects of glycosylation on binding interactions, explaining observations like the improved binding seen with the N90E mutation that eliminates a glycosylation site . By integrating these computational approaches with experimental validation, researchers can develop more accurate predictive models and accelerate the rational design of improved ACE2 variants for both research and therapeutic applications.
Angiotensin Converting Enzyme 2 (ACE2) is a crucial enzyme in the renin-angiotensin system (RAS), which plays a significant role in regulating cardiovascular and renal functions. The recombinant form of human ACE2, specifically the segment spanning amino acids 18-740, has garnered attention for its potential therapeutic applications and its role in various physiological processes.
ACE2 is a monocarboxypeptidase that metabolizes several peptides, including the degradation of angiotensin II (Ang II) into angiotensin 1-7 (Ang 1-7). Ang II is known for its vasoconstrictive and proliferative effects, while Ang 1-7 exerts vasodilatory and antiproliferative actions through the Mas receptor . This balance between Ang II and Ang 1-7 is critical for maintaining cardiovascular homeostasis.
The recombinant human ACE2 (18-740 a.a.) is often produced in HEK293 cells and includes a His-tag for purification purposes . This segment of ACE2 retains its enzymatic activity and binding affinity, making it suitable for research and therapeutic applications.
Cardiovascular Diseases: ACE2 has shown promise in treating heart failure and hypertension. By converting Ang II to Ang 1-7, ACE2 helps mitigate the adverse effects of Ang II, such as myocardial hypertrophy, fibrosis, and diastolic dysfunction . Recombinant ACE2 has been explored as a potential therapy for heart failure, with studies indicating its ability to suppress pressure overload-induced heart failure .
Acute Respiratory Distress Syndrome (ARDS): ACE2 has been implicated in the pathogenesis of ARDS. A pilot clinical trial investigated the use of recombinant human ACE2 (GSK2586881) in patients with ARDS. The study found that ACE2 administration was well-tolerated and modulated RAS peptides, although it did not significantly change acute physiology or clinical outcomes .
COVID-19: ACE2 is known to be the functional receptor for the SARS-CoV-2 virus, which causes COVID-19. The virus binds to ACE2 on the surface of host cells, facilitating its entry and subsequent infection . This has led to research into the potential use of recombinant ACE2 to block viral entry and reduce the severity of COVID-19.
The discovery of ACE2 in 2000 marked a significant advancement in understanding the RAS . Since then, extensive research has been conducted to explore its physiological roles and therapeutic potential. Recombinant human ACE2 (18-740 a.a.) has become a valuable tool in this research, enabling scientists to study its effects in various disease models and develop novel therapeutic strategies.