AmCyan is a human codon-optimized variant of the wild-type Anemonia majano cyan fluorescent protein (AmCyan1), engineered for enhanced brightness and stability in mammalian systems . Key characteristics include:
Parameter | Value | Source |
---|---|---|
Excitation Maximum | 458 nm | |
Emission Maximum | 489 nm | |
Molecular Weight | ~25 kDa (AmCyan1 variant) | |
Destabilization Domain | N-terminal DD tag (for controlled degradation) |
The DD tag allows conditional stabilization of AmCyan fusion proteins via Shield1 ligand binding, enabling precise temporal control of protein expression .
The AmCyan monoclonal antibody is produced in mice using recombinant AmCyan protein as the immunogen . Key features include:
Attribute | Specification | Source |
---|---|---|
Isotype | IgG (unconjugated) | |
Host Species | Mouse | |
Purification Method | Affinity chromatography (Protein G/immunogen) | |
Clonality | Monoclonal (e.g., clone 5F9) |
This antibody exhibits high specificity for AmCyan-tagged proteins, enabling detection in Western blot (WB) and immunoprecipitation (IP) .
The antibody is primarily used for:
Western Blotting: Detects AmCyan-tagged proteins at dilutions of 1:500–1:5000 .
Immunoprecipitation: Effective at dilutions of 1:100–1:200 .
Validation of Expression: Confirms successful tagging and stability of fusion proteins .
WB: Load lysates containing AmCyan-tagged proteins, transfer to PVDF membrane, and incubate with primary antibody (1:5000) followed by HRP-conjugated secondary antibody .
IP: Use antibody-conjugated beads to pull down AmCyan-tagged complexes for downstream analysis .
The antibody shows cross-reactivity with AmCyan and AmCyan-tagged proteins but no nonspecific binding to untagged targets .
In silkworm-based systems, AmCyan-tagged antibodies exhibited distinct N-glycosylation profiles compared to CHO-derived counterparts, influencing Fc-mediated effector functions like antibody-dependent cellular cytotoxicity (ADCC) .
AmCyan1 is a human codon-optimized variant of the wild-type Anemonia majano cyan fluorescent protein (AmCyan) that exhibits enhanced emission characteristics with excitation and emission maxima of 458nm and 489nm, respectively. Unlike other commonly used fluorescent proteins such as GFP (Green Fluorescent Protein), AmCyan offers distinct spectral properties that make it valuable for multicolor imaging applications. The protein's structure has been engineered for improved brightness and stability in mammalian expression systems, allowing for efficient detection in various experimental contexts .
The optimization process for AmCyan involved modifications to enhance folding efficiency, reduce aggregation tendency, and improve quantum yield. These improvements make AmCyan particularly suitable for applications requiring distinct fluorescent signatures that can be easily differentiated from other commonly used fluorophores such as GFP, YFP, and RFP in multiplexed experimental designs.
AmCyan monoclonal antibodies are typically mouse-derived immunoglobulins (IgG) that specifically recognize and bind to AmCyan protein or AmCyan-tagged fusion proteins. These antibodies are generated using recombinant AmCyan as the immunogen, resulting in highly specific recognition properties. The clone 5F9, a commonly used AmCyan monoclonal antibody, demonstrates high affinity and specificity for the target protein .
The antibodies function through specific epitope recognition on the AmCyan protein structure, allowing researchers to detect and isolate AmCyan-tagged proteins from complex biological samples. They maintain their binding specificity across various experimental conditions, making them reliable tools for multiple applications including Western blotting (with dilution ranges of 1:300-5000) and immunoprecipitation procedures (with typical dilution ranges of 1:100-200) .
DD-AmCyan1 represents an advanced application of the AmCyan fluorescent protein, where the protein is tagged on its N-terminus with the ProteoTuner destabilization domain (DD). This system provides a powerful means for studying protein dynamics and regulation through controlled protein stability. The presence of this destabilization domain causes rapid, proteasomal degradation of the fluorescent fusion protein under normal conditions .
The key advantage of this system is the ability to regulate protein levels through the addition of Shield1, a membrane-permeant ligand. When Shield1 is added to the medium, it binds to the destabilization domain and protects the fusion protein from degradation, allowing researchers to rapidly induce protein expression and monitor subsequent effects. This provides temporal control over protein expression without altering transcriptional activity, enabling more precise studies of protein function and cellular responses. The kinetics of Shield1-mediated protection typically show detectable protein levels within 15-30 minutes after treatment, with maximum accumulation occurring within 4-12 hours, depending on the specific experimental system .
AmCyan monoclonal antibodies serve multiple key functions in molecular and cellular research:
Protein Detection and Quantification: These antibodies enable precise detection of AmCyan-tagged proteins in Western blotting applications, with recommended dilutions ranging from 1:300 to 1:5000 depending on expression levels and detection methods .
Protein Isolation and Characterization: Through immunoprecipitation (IP) procedures, researchers can isolate AmCyan-tagged protein complexes from cell lysates, facilitating studies of protein-protein interactions and complex formation. Typical IP applications use dilution ranges of 1:100 to 1:200 .
Flow Cytometry Applications: AmCyan's spectral properties make it valuable in multicolor flow cytometry panels, particularly when combined with markers like CD8 for immune cell phenotyping and functional studies .
Protein Localization Studies: Using immunofluorescence techniques, AmCyan monoclonal antibodies can help visualize the subcellular localization of tagged proteins, particularly in cases where direct fluorescence from the AmCyan tag might be insufficient.
Protein Dynamics Research: When used with systems like DD-AmCyan1, these antibodies enable studies of protein turnover, stability, and temporal regulation within cellular contexts .
Designing effective Western blotting protocols for AmCyan monoclonal antibodies requires careful consideration of several experimental parameters:
Sample Preparation:
Lyse cells in a buffer containing 0.01M TBS (pH 7.4) with protease inhibitors to prevent protein degradation
Denature proteins at 95°C for 5 minutes in sample buffer containing SDS and reducing agents
Load 20-50 μg of total protein per lane for standard detection of AmCyan-tagged proteins
Electrophoresis and Transfer:
Use 10-12% polyacrylamide gels for optimal resolution of AmCyan-tagged proteins (approximately 27 kDa plus the mass of the fused protein of interest)
Transfer proteins to PVDF or nitrocellulose membranes at 100V for 1 hour or 30V overnight for larger fusion proteins
Antibody Incubation:
Block membranes with 5% non-fat dry milk or BSA in TBST for 1 hour at room temperature
Dilute primary AmCyan monoclonal antibody to 1:2000-1:5000 in blocking buffer
Incubate overnight at 4°C with gentle agitation
Wash membranes 3-5 times with TBST
Incubate with appropriate HRP-conjugated secondary antibody (anti-mouse IgG) at 1:5000-1:10000 dilution for 1 hour at room temperature
Detection and Analysis:
Develop using enhanced chemiluminescence detection reagents
Expose to X-ray film or digital imaging system
Expected band size should correlate with the molecular weight of your target protein plus 27 kDa (AmCyan)
Incorporating AmCyan into multicolor flow cytometry panels requires careful consideration of spectral overlap and compensation strategies:
Panel Design Considerations:
Spectral Properties: AmCyan's excitation maximum at 458nm and emission maximum at 489nm place it between FITC and PE in the spectral range, requiring appropriate laser excitation (typically 405nm or 445nm) .
Compensation Matrix: Due to potential spillover into FITC and PE channels, comprehensive compensation controls are essential. Single-stained controls for each fluorochrome in your panel should be prepared using the same cells or compensation beads.
Compatible Fluorochromes: Optimal multicolor panels might include combinations of AmCyan with fluorophores such as Pacific Blue, APC, PerCP-Cy5.5, and PE-Cy7 to minimize spectral overlap.
Example 6-Color Panel for T Cell Analysis:
Fluorochrome | Marker | Excitation Laser | Emission Filter |
---|---|---|---|
AmCyan | CD8 | 405nm | 500/50nm |
Pacific Blue | CD3 | 405nm | 450/50nm |
FITC | CD45RA | 488nm | 530/30nm |
PE | CCR7 | 561nm | 585/42nm |
PerCP-Cy5.5 | CD4 | 488nm | 695/40nm |
APC | CD25 | 640nm | 670/30nm |
Protocol Optimization:
Store CD8 AmCyan antibody at 2-8°C protected from light to maintain stability
Titrate antibody concentrations to determine optimal staining index
Include appropriate isotype controls to establish background fluorescence levels
Implement consistent gating strategies based on fluorescence-minus-one (FMO) controls
Preserving the functional integrity of AmCyan monoclonal antibodies requires careful attention to storage and handling protocols:
Storage Recommendations:
Store antibody vials at 2-8°C in the dark to prevent photobleaching
Do not freeze conjugated forms as this may compromise functionality
For long-term storage beyond one year, aliquot the antibody to minimize freeze-thaw cycles
The antibody remains stable until the expiration date when stored as directed
Handling Protocols:
Equilibrate antibodies to room temperature before opening vials
Centrifuge briefly to collect solution at the bottom of the vial
Avoid repeated freeze-thaw cycles that can cause protein denaturation and loss of binding capacity
When diluting, use buffers containing stabilizing proteins such as 1% BSA
For maximum stability, store working dilutions at 4°C for up to one week
Buffer Compatibility:
The storage buffer composition of 0.01M TBS (pH 7.4) with 1% BSA, 0.02% Proclin300, and 50% Glycerol provides optimal stability through:
Physiological pH maintenance
Prevention of non-specific binding
Antimicrobial protection
The choice of fixation and permeabilization methods significantly impacts the performance of AmCyan monoclonal antibodies in various applications:
Fixation Approaches:
Fixation Method | Impact on AmCyan Detection | Recommended Applications |
---|---|---|
4% Paraformaldehyde | Preserves fluorescence and epitope accessibility | Immunofluorescence, flow cytometry |
Methanol/Acetone | May diminish fluorescent signal but can enhance epitope exposure | Western blotting sample preparation |
Glutaraldehyde | Strong protein crosslinking that may mask epitopes | Not recommended for AmCyan detection |
Permeabilization Strategies:
For intracellular staining, 0.1-0.5% Triton X-100 provides adequate permeabilization while preserving antibody-epitope interactions
Saponin (0.1%) offers more gentle permeabilization for detecting membrane-associated AmCyan-tagged proteins
When performing flow cytometry, commercial permeabilization kits specifically designed for preserving fluorescent protein signals should be considered
Protocol Optimization:
For best results, optimize fixation time (typically 10-20 minutes at room temperature)
Test different permeabilization durations to balance adequate access with epitope preservation
Include controls using native AmCyan fluorescence to assess signal retention after fixation
Rigorous validation of AmCyan monoclonal antibody specificity is essential for generating reliable experimental data:
Essential Validation Steps:
Positive and Negative Controls: Include both AmCyan-expressing samples and non-expressing controls in all experiments. The absence of signal in negative controls confirms specificity.
Blocking Peptide Experiments: Pre-incubation of the antibody with recombinant AmCyan protein should abolish or significantly reduce detection signal in Western blotting or immunofluorescence.
Molecular Weight Verification: In Western blotting applications, bands should appear at the expected molecular weight of AmCyan (27 kDa) plus any fused protein of interest.
Cross-Reactivity Assessment: Test against other fluorescent proteins (especially those with similar spectral properties) to confirm lack of cross-reactivity.
Knockdown/Knockout Validation: In systems where AmCyan expression can be manipulated, demonstrate corresponding changes in antibody detection signal.
Documentation Protocol:
Record lot-specific validation data
Maintain detailed protocols that produce consistent results
Document optimization parameters including antibody dilutions and incubation conditions
Consider secondary validation methods that don't rely solely on antibody-based detection
Western blotting with AmCyan monoclonal antibodies may present several technical challenges that can be systematically addressed:
Possible Causes: Insufficient blocking, excessive antibody concentration, inadequate washing
Solutions:
Possible Causes: Low expression levels, protein degradation, insufficient transfer
Solutions:
Possible Causes: Protein degradation, post-translational modifications, non-specific binding
Solutions:
Freshly prepare samples and maintain cold chain
Use denaturing conditions that disrupt protein complexes
Include phosphatase inhibitors if phosphorylation affects migration
For fusion proteins, verify expected molecular weight calculations
Possible Causes: Antibody degradation, variable expression levels, protocol inconsistencies
Solutions:
Accurate quantification and interpretation of AmCyan flow cytometry data requires systematic approaches to analysis:
Gating Strategy Development:
Initial FSC/SSC Gating: Begin with forward/side scatter to identify the population of interest and exclude debris.
Singlet Selection: Apply FSC-H vs. FSC-A gating to eliminate doublets that could skew fluorescence readings.
Viability Discrimination: Include a viability dye in a non-overlapping channel to exclude dead cells that may exhibit autofluorescence.
FMO Controls: Use fluorescence-minus-one controls to establish proper boundary positions for AmCyan-positive populations.
AmCyan-Specific Gating: Set positive gates based on negative controls and the expected distribution pattern.
Quantification Approaches:
Percentage Positivity: Report the percentage of cells within the defined positive gate.
Mean/Median Fluorescence Intensity (MFI): Calculate the average signal intensity of positive populations to assess expression levels.
Staining Index: Calculate using the formula: (MFI positive - MFI negative) / (2 × SD of negative population).
Data Normalization Strategies:
Use consistent instrument settings between experiments or implement calibration beads.
Apply compensation matrices based on single-stained controls to correct for spectral overlap.
Consider standardized measures like Molecules of Equivalent Soluble Fluorochrome (MESF) for cross-experiment comparisons.
Statistical Analysis Guidelines:
For comparing populations, apply appropriate statistical tests (t-test, ANOVA) to MFI values rather than percentages when assessing expression levels.
For multivariate data, consider dimensionality reduction techniques (tSNE, UMAP) to identify patterns across multiple parameters.
Report both biological and technical replicates to establish reproducibility .
When faced with discrepancies between results obtained using AmCyan monoclonal antibodies across different experimental platforms, researchers should implement systematic troubleshooting approaches:
Systematic Analysis Framework:
Cross-Platform Validation:
Confirm protein expression using complementary methods (e.g., direct fluorescence observation, Western blotting, and flow cytometry)
Document specific experimental conditions for each platform that may influence antibody performance
Epitope Accessibility Assessment:
Different experimental conditions may affect epitope exposure
Test alternative fixation/permeabilization methods across platforms
Consider native versus denatured protein conformations in different applications
Reagent Consistency Verification:
Use the same antibody lot across all experimental platforms
Prepare fresh working dilutions for each experiment
Document storage conditions and freeze-thaw cycles
Sample Preparation Harmonization:
Standardize lysis buffers and protein extraction protocols
Align fixation protocols when possible
Control for post-translational modifications that might affect epitope recognition
Resolution Strategy for Common Contradictions:
Contradiction Type | Possible Cause | Resolution Approach |
---|---|---|
Positive WB, Negative IF | Epitope masking in fixed samples | Test alternative fixation methods; use epitope retrieval techniques |
Positive Flow, Negative WB | Conformation-dependent epitope | Use native gel conditions for Western blotting |
Variable signal intensity | Differential expression levels | Quantify and normalize to housekeeping genes/proteins |
Detection in unexpected locations | Proteolytic cleavage or alternative splicing | Use C- and N-terminal targeted antibodies to map protein fragments |
Documentation Requirements:
Maintain detailed records of experimental conditions for each platform
Document antibody dilutions, incubation times, and detection methods
Record instrument settings and analysis parameters
Implement standardized reporting forms to facilitate cross-experiment comparisons
The integration of AmCyan monoclonal antibodies with super-resolution microscopy offers powerful approaches for visualizing protein localization and dynamics beyond the diffraction limit:
Optimal Super-Resolution Techniques for AmCyan Detection:
Structured Illumination Microscopy (SIM):
Achieves ~100 nm resolution through illumination pattern manipulation
Compatible with standard AmCyan fluorescence detection
Requires careful management of photobleaching through anti-fade mounting media
Optimal for multicolor imaging combining AmCyan with spectrally distinct fluorophores
Stimulated Emission Depletion (STED) Microscopy:
Achieves ~30-80 nm resolution through selective deactivation of fluorophores
AmCyan's photophysical properties make it moderately suitable for STED applications
Requires optimization of depletion laser parameters to prevent photobleaching
Consider using anti-AmCyan primary antibodies with STED-optimized secondary antibodies for improved performance
Single Molecule Localization Microscopy (PALM/STORM):
Achieves ~20-30 nm resolution through sequential activation and localization of single molecules
Direct PALM applications with photoswitchable variants of AmCyan show promise
Alternatively, use AmCyan monoclonal antibodies with photoswitchable secondary antibodies for STORM approaches
Protocol Optimization Considerations:
For fixed-cell applications, optimize fixation to preserve both AmCyan fluorescence and epitope accessibility
Implement drift correction strategies for long-acquisition super-resolution imaging
Consider oxygen scavenging systems to reduce photobleaching during extended imaging sessions
For multicolor super-resolution, carefully select complementary fluorophores with minimal spectral overlap
Technical Parameters for Optimal Imaging:
Technique | Recommended Buffer | Mounting Medium | Key Optimization Parameters |
---|---|---|---|
SIM | PBS pH 7.4 | Anti-fade with glycerol base | Laser power, grating rotation steps |
STED | PBS with 10% glycerol | TDE-based medium | Depletion laser power, time gating |
PALM/STORM | Oxygen-scavenging buffer with MEA | No mounting (imaging buffer) | Activation laser intensity, frame rate |
Despite their utility, AmCyan monoclonal antibodies face several limitations in multiplexed experimental systems that researchers should consider:
Spectral Overlap Challenges:
The emission spectrum of AmCyan (peak at 489nm) shows partial overlap with FITC and GFP signals, requiring careful compensation in flow cytometry and confocal microscopy
In multicolor imaging, this spectral proximity can limit the number of distinctly resolvable channels
Alternative fluorophores with more discrete spectral properties may be preferable for highly multiplexed systems
Technical Limitations:
Sensitivity variations between detection platforms can lead to inconsistent performance across experimental systems
The requirement for clone 5F9 specificity may limit flexibility in antibody host species selection for co-staining experiments
Potential cross-reactivity with structurally similar cyan fluorescent protein variants requires thorough validation
System-Specific Constraints:
Experimental System | Limitation | Mitigation Strategy |
---|---|---|
Flow Cytometry | Compensation complexity with FITC/GFP | Use alternative fluorophores in PE or APC channels |
Multiplex Immunohistochemistry | Limited antibody host diversity | Sequential staining with intervening stripping steps |
Live Cell Imaging | Potential interference with endogenous fluorescence | Background subtraction and autofluorescence controls |
Mass Cytometry | Incompatibility with metal-based detection | Consider alternative tagging strategies |
Emerging Alternative Approaches:
Spectral flow cytometry with unmixing algorithms may better resolve AmCyan from spectrally similar fluorophores
Quantum dot-conjugated secondary antibodies offer narrower emission spectra for improved multiplexing
Proximity ligation assays can overcome some multiplexing limitations while providing enhanced sensitivity
Cyclic immunofluorescence methods allow for sequential staining and imaging of numerous markers on the same sample
CRISPR-based genome editing technologies offer powerful approaches to enhance research involving AmCyan-tagged proteins, enabling precise genomic integration and controlled expression:
Endogenous Tagging Strategies:
CRISPR/Cas9-mediated homology-directed repair (HDR) enables precise insertion of AmCyan tags at endogenous gene loci
This approach maintains native promoter control and expression levels, avoiding artifacts associated with overexpression
Design considerations include:
Optimal cut site selection near the desired integration location
Construction of repair templates with ~800bp homology arms flanking the AmCyan sequence
Strategic placement of the tag (N-terminal, C-terminal, or internal) to minimize functional disruption
Inducible Expression Systems:
CRISPR interference (CRISPRi) and CRISPR activation (CRISPRa) systems can be combined with AmCyan reporters to create tunable expression models
dCas9-based systems fused to transcriptional regulators allow temporal control of AmCyan-tagged protein expression
These approaches complement the DD-AmCyan1 system by enabling regulation at the transcriptional rather than post-translational level
Multiplexed Functional Genomics:
CRISPR screens using AmCyan as a selectable marker or readout enable high-throughput functional genomics
Single-cell analysis of CRISPR-edited populations with AmCyan reporters allows correlation between genetic perturbations and phenotypic outcomes
Implementation of multi-color fluorescent protein systems (including AmCyan) facilitates tracking of multiple genetic modifications simultaneously
Technical Implementation Framework:
Design Phase:
Select appropriate guide RNAs using validated design tools
Engineer repair templates with AmCyan sequence optimized for the target organism
Include flexible linker sequences to minimize interference with protein function
Validation Requirements:
Confirm precise integration using PCR and sequencing
Verify protein functionality through appropriate functional assays
Compare expression levels to endogenous untagged protein
Test antibody recognition of the integrated tag using immunoprecipitation and Western blotting
Advanced Applications:
Time-lapse microscopy of AmCyan-tagged proteins in CRISPR-edited cells for dynamic studies
Combinatorial CRISPR editing with multiple fluorescent tags for interaction studies
Integration with optogenetic systems for spatiotemporal control of protein function