The synthesis of FTL recombinant monoclonal antibodies involves:
Cloning: FTL-specific antibody genes are inserted into expression vectors.
Transfection: Host cells (e.g., CHO or HEK293) are transfected with the vector.
Expression: Antibodies are secreted into cell culture supernatant.
Purification: Affinity chromatography isolates the antibody for use .
Lot-to-Lot Consistency: Defined genetic sequences eliminate variability .
Scalability: In vitro methods enable large-scale production .
Engineering Flexibility: Isotype-switching or species-switching for multiplexing experiments .
FTL recombinant monoclonal antibodies are utilized across diverse platforms:
Rapid Test Kits: Serve as positive controls for filarial antigen detection (e.g., Brugia Rapid™ and BLF Rapid®) .
Gold Nanoparticle Conjugation: Confirm antigenicity of test lines in stored rapid tests .
Application | Example | Source |
---|---|---|
QC Reagent | Replaces patient serum in filarial diagnostic kit validation | |
Immunoaffinity Purification | Captures recombinant BmSXP protein from mixed pools |
Western Blot (WB): Detects endogenous FTL at 1:500–1:1000 dilution .
Immunohistochemistry (IHC): Identifies FTL expression in tumor tissues (e.g., gastric, breast, prostate cancers) .
Flow Cytometry (FC): Analyzes FTL surface expression at 1:50–1:200 dilution .
Recombinant monoclonal antibodies (rmAbs) against BmSXP and BmR1 filarial proteins demonstrated:
Diagnostic Utility: rmAbs (e.g., Ab5B, Ab4-Fc) validated rapid test sensitivity and specificity .
Conjugation Efficiency: Gold-labeled rmAbs confirmed antigen integrity post-storage .
A novel workflow isolated antigen-specific antibody-secreting cells (ASCs) from convalescent COVID-19 patients, enabling rapid recombinant antibody production within 10 days. Key outcomes:
High-Throughput Screening: Generated 36 recombinant mAbs from five donors .
Functional Selection: Screened for neutralizing activity prior to cloning .
The FTL recombinant monoclonal antibody is synthesized through a meticulous process involving in vitro cloning. FTL antibody genes are seamlessly integrated into expression vectors, which are subsequently introduced into host cells. This creates an environment that facilitates the expression of the recombinant antibody within a cell culture medium. Following expression, the antibody is meticulously purified from the supernatant of the transfected host cells using affinity chromatography. This antibody exhibits a specific binding affinity for the human FTL protein, as demonstrated in ELISA, IHC, and FC applications.
Ferritin light chain (FTL) is an essential component of the ferritin protein complex, playing a pivotal role in the storage, regulation, and release of iron within cells. This function is crucial for maintaining iron homeostasis, safeguarding against oxidative stress, and supporting various cellular metabolic processes.
Ferritin light chain (FTL) serves as a vital component in the storage and regulation of iron within cells, ensuring its presence in a soluble, non-toxic, and readily available form. This function is critical for maintaining iron homeostasis. Iron is absorbed in the ferrous form and subsequently deposited as ferric hydroxides after oxidation. FTL also plays a role in delivering iron to cells. In the developing kidney, FTL facilitates iron uptake within capsule cells.
FTL (Ferritin Light Chain) is a crucial subunit of the ferritin protein complex that stores iron in a soluble, non-toxic, readily available form. It plays a fundamental role in iron homeostasis within cells. The FTL protein is involved in several key biological processes, including:
Iron uptake in the ferrous form and deposition as ferric hydroxides after oxidation
Delivery of iron to cells throughout the body
Mediation of iron uptake in capsule cells of the developing kidney
Delivery to lysosomes via the cargo receptor NCOA4 for autophagic degradation and release of iron
These functions make FTL an important research target for studies involving iron metabolism, oxidative stress responses, and related pathological conditions.
FTL recombinant monoclonal antibodies have been validated for multiple research applications, with specific validation depending on the manufacturer and antibody clone. Common validated applications include:
IHC (Immunohistochemistry) on formalin-fixed paraffin-embedded (FFPE) tissues
ELISA (Enzyme-Linked Immunosorbent Assay) for protein quantification
When selecting an antibody for your specific research needs, it's important to verify that the antibody has been validated for your intended application and target species.
The key differences between mouse monoclonal and recombinant monoclonal FTL antibodies involve their production methods, consistency, and application advantages:
Mouse Monoclonal FTL Antibodies:
Produced using traditional hybridoma technology
Generated by immunizing mice with the target antigen (FTL protein or fragment)
Antibody-producing B cells from the mouse are fused with myeloma cells to create hybridomas
Each hybridoma cell line produces a single antibody specificity
Examples include mouse monoclonal antibody FTL/1386 (ab218400)
Recombinant Monoclonal FTL Antibodies:
Produced using in vitro cloning and recombinant DNA technology
The antibody genes are incorporated into expression vectors and introduced into host cells
Expression occurs in a cell culture environment
Purified from the supernatant of transfected host cells through affinity chromatography
Can be produced in different host species (e.g., rabbit IgG)
Recombinant antibodies typically offer greater batch-to-batch consistency, reduced animal use, and can be engineered for enhanced performance in specific applications.
Optimizing immunohistochemistry protocols with FTL recombinant antibodies requires careful consideration of several factors:
Antigen Retrieval:
For FFPE tissues, heat-induced epitope retrieval is typically necessary. Based on validated protocols:
Boil tissue sections in 10mM Citrate Buffer, pH 6.0, for 10-20 minutes
Allow sections to cool at room temperature for 20 minutes before proceeding
Antibody Dilution:
Start with the manufacturer's recommended dilution range (typically 1:50-1:200 for IHC)
Perform a dilution series to determine optimal antibody concentration
For ab218400, successful staining has been reported at 0.2-0.5 μg/ml
Incubation Conditions:
Optimal incubation time appears to be 30 minutes to overnight at 4°C
Room temperature incubation (30 minutes) has been validated for some antibodies
Detection Systems:
Select an appropriate detection system based on your species and isotype (e.g., HRP-polymer for rabbit IgG)
Include appropriate blocking steps to minimize background
Controls:
Always include positive tissue controls (human pancreas and testis tissues have shown positive staining)
Include negative controls (primary antibody omission and isotype controls)
Consider using FTL knockout cell lines as specificity controls when available
When using FTL antibodies for Western blot applications, researchers should consider these methodological aspects:
Sample Preparation:
Use appropriate lysis buffers that preserve protein structure
Include protease inhibitors to prevent degradation
Determine optimal protein loading (20 μg per lane has been validated)
Running Conditions:
Use reducing conditions for optimal results with FTL antibodies
Ensure appropriate percentage gel selection (FTL has a predicted band size of 20 kDa)
Antibody Concentration:
Higher concentrations may increase background signal
Expected Results:
The predicted band size for FTL is 20 kDa
Some antibodies may also detect additional bands at 31 kDa and 35 kDa
Validate specificity using knockout cell lines when possible
Detection Systems:
Both chemiluminescence and fluorescent detection systems have been validated
For fluorescent detection, secondary antibodies such as IRDye® 800CW and IRDye® 680RD have been successfully used
Proper controls are essential for validating antibody specificity in FTL-related research:
Positive Controls:
Well-characterized cell lines with known FTL expression (HeLa, A549, HepG2)
Human tissue samples known to express FTL (liver, pancreas, testis)
Recombinant FTL protein for western blot positive controls
Negative Controls:
FTL knockout cell lines (several are commercially available):
Primary antibody omission controls
Isotype controls matching the primary antibody's species and isotype
Loading Controls:
Peptide Competition:
Consider performing peptide competition assays using the immunizing peptide to confirm binding specificity
The most stringent specificity validation comes from comparing wild-type to knockout samples, as demonstrated in the published validation data showing antibody reactivity in wild-type HeLa cells but signal loss in FTL knockout HeLa cells .
FTL recombinant antibodies can be powerful tools for investigating iron metabolism disorders through several advanced research approaches:
Tissue Expression Analysis:
IHC analysis of patient tissues can reveal altered FTL expression patterns in hereditary hyperferritinemia-cataract syndrome, neuroferritinopathy, and other iron storage disorders
Comparative analysis between healthy and diseased tissues can identify cell-specific changes in FTL expression
Protein-Protein Interaction Studies:
Co-immunoprecipitation using FTL antibodies can identify altered interactions between FTL and:
Subcellular Localization Studies:
Immunofluorescence with FTL antibodies can reveal changes in ferritin localization in disease states
Co-localization with lysosomal markers can assess ferritinophagy efficiency
Quantitative Analysis:
Western blotting and ELISA with FTL antibodies allow quantitative assessment of FTL expression levels
Flow cytometry can be used to assess cellular FTL content on a single-cell level
Genetic Modification Models:
FTL knockout cell lines can serve as models for studying the functional consequences of FTL deficiency
Complementation studies with wild-type or mutant FTL can assess functional impacts of specific mutations
These approaches enable comprehensive investigation of molecular mechanisms underlying iron metabolism disorders and potential therapeutic interventions.
Studying FTL in autophagy and ferritinophagy requires specific methodological considerations:
Induction of Ferritinophagy:
Iron chelation with deferoxamine (DFO) or other chelators to induce ferritinophagy
Starvation conditions to trigger general autophagy
Use of specific autophagy inducers like rapamycin
Dual Immunostaining Approach:
Co-staining for FTL and NCOA4 (the selective cargo receptor for ferritinophagy)
Co-staining for FTL and autophagy markers (LC3, p62)
Co-staining for FTL and lysosomal markers (LAMP1, LAMP2)
Live-Cell Imaging Considerations:
Transfection with fluorescently tagged FTL constructs
Use of lysosomal dyes in combination with immunofluorescence
Time-lapse imaging to capture dynamic ferritinophagy processes
Quantitative Assessments:
Western blot analysis of FTL degradation kinetics under ferritinophagy-inducing conditions
Flow cytometry to quantify cellular FTL levels during ferritinophagy
ELISA-based quantification of FTL in cellular compartments
Inhibitor Studies:
Use of autophagy inhibitors (bafilomycin A1, chloroquine) to block lysosomal degradation
Assessment of FTL accumulation when lysosomal function is inhibited
Comparison between general autophagy inhibition and specific ferritinophagy inhibition
Knockout/Knockdown Verification:
NCOA4 knockout/knockdown to confirm specificity of ferritinophagy process
Autophagy-related gene knockouts to determine pathway dependencies
These methodological approaches allow for comprehensive characterization of FTL's role in ferritinophagy and the broader autophagy pathway.
Multiplexed imaging with FTL recombinant antibodies enables simultaneous visualization of multiple targets, providing spatial context for understanding FTL's role in cellular processes:
Sequential Multiplexing Approach:
Use FTL antibodies in combination with antibodies to other proteins of interest
Ensure antibodies are from different host species or isotypes to prevent cross-reactivity
Consider using specifically validated antibody panels for iron metabolism studies
Spectral Unmixing Strategies:
Use fluorophores with minimal spectral overlap
Employ spectral imaging systems capable of separating closely overlapping fluorophores
Validated fluorophore combinations include IRDye® 800CW and IRDye® 680RD
Multi-epitope Ligand Cartography (MELC):
Use FTL antibodies in iterative immunofluorescence imaging cycles
Photobleach fluorophores between cycles to build high-parameter tissue maps
Combine with image analysis tools for quantitative spatial analysis
Mass Cytometry Imaging:
Conjugate FTL antibodies to rare earth metals for mass cytometry imaging (IMC)
Allows for highly multiplexed imaging (30+ markers) with minimal signal overlap
Particularly useful for complex tissue microenvironments
Proximity Ligation Assays:
Combine FTL antibodies with antibodies against potential interaction partners
Detect protein-protein interactions within intact cells or tissues
Particularly useful for studying FTL interactions with FTH or NCOA4
Validation Controls:
Include single-stain controls for each antibody
Include fluorescence minus one (FMO) controls for accurate gating and analysis
These multiplexed approaches significantly enhance our understanding of FTL's spatial relationships with other cellular components in both normal and pathological conditions.
Understanding potential sources of error is crucial for accurate interpretation of FTL antibody-based experiments:
Common Causes of False Positives:
Common Causes of False Negatives:
Researchers should systematically evaluate these factors when troubleshooting unexpected results, giving particular attention to proper controls and validated protocols.
Quantitative analysis of FTL expression requires platform-specific approaches and careful normalization:
Western Blot Quantification:
Use digital imaging systems for densitometric analysis
Normalize FTL band intensity to loading controls (GAPDH, alpha-tubulin)
Construct standard curves using recombinant FTL protein for absolute quantification
Use FTL knockout samples to determine background signal threshold
Immunohistochemistry Quantification:
Employ digital pathology software for automated scoring
Quantify parameters such as:
Percentage of positive cells
Staining intensity (0, 1+, 2+, 3+)
H-score calculation (% of 1+ cells × 1) + (% of 2+ cells × 2) + (% of 3+ cells × 3)
Compare to validated positive controls (human pancreas, testis)
Flow Cytometry Analysis:
Gate populations based on forward/side scatter properties
Establish negative gates using FTL knockout cells or isotype controls
Quantify median fluorescence intensity (MFI) for population analysis
Consider fluorescence minus one (FMO) controls for precise gating
ELISA Data Analysis:
Generate standard curves using purified recombinant FTL
Ensure samples fall within the linear range of the standard curve
Account for matrix effects through sample dilution series
Calculate intra- and inter-assay coefficients of variation for quality control
Cross-Platform Data Integration:
Normalize data to common reference samples across platforms
Use relative quantification when comparing between methods
Consider statistical approaches like z-score normalization for multi-platform comparisons
Validate findings using orthogonal methods
These quantitative approaches enhance experimental rigor and reproducibility in FTL expression studies.
When facing contradictory results across different detection methods for FTL, a systematic approach to resolution is necessary:
Methodological Analysis:
Evaluate each method's sensitivity limits and dynamic range
Consider epitope accessibility differences between techniques
Assess whether the antibodies used recognize different epitopes
Review whether post-translational modifications affect epitope recognition
Validation Hierarchy:
Consider results from multiple methods with this hierarchy of confidence:
Methods using multiple antibodies recognizing different epitopes
Methods with comprehensive positive and negative controls
Methods without adequate controls (lowest confidence)
Sample-Specific Considerations:
Different subcellular localization may affect detection efficiency
Sample preparation differences may alter epitope exposure
Protein complexes may mask epitopes in certain assays
Isoform-specific expression may cause apparent contradictions
Resolution Strategies:
Documentation Practices:
Record detailed methodological parameters for each experiment
Document lot numbers of antibodies used
Maintain comprehensive records of all controls
Consider pre-registering experimental protocols to minimize bias
FTL recombinant antibodies offer valuable tools for investigating the role of iron dysregulation in neurodegenerative conditions:
Neuroferritinopathy Studies:
IHC analysis of brain tissues to detect abnormal FTL accumulation
Co-staining with markers of neurodegeneration to establish spatial relationships
Quantitative analysis of FTL expression in different brain regions
Detection of mutant FTL forms associated with specific disorders
Iron-Related Neurodegeneration:
Assessment of FTL expression in Parkinson's, Alzheimer's, and ALS models
Correlation of FTL levels with markers of oxidative stress
Investigation of FTL's role in neuronal iron homeostasis
Monitoring changes in FTL expression during disease progression
Methodological Considerations:
Optimize tissue fixation to preserve brain tissue architecture
Use antigen retrieval optimized for neural tissues (citrate buffer, pH 6.0)
Consider autofluorescence quenching for aged brain tissues
Include age-matched controls for comparative analysis
Cellular Models:
Use of patient-derived iPSCs differentiated into neurons
Transfection with wild-type or mutant FTL to study functional consequences
Live-cell imaging to track FTL dynamics in neuronal cells
These approaches enable investigation of iron dysregulation as both a contributor to and consequence of neurodegenerative processes, potentially revealing new therapeutic targets.
Developing effective multiplex assays with FTL antibodies requires careful attention to several technical aspects:
Antibody Compatibility:
Select antibodies from different host species or isotypes
Verify lack of cross-reactivity between antibodies
Test for epitope competition when using multiple antibodies against the same protein
Validate each antibody individually before combining into multiplex format
Signal Optimization:
Titrate each antibody to determine optimal working concentration
Balance signal intensities across all targets
Consider sequential detection for targets with vastly different abundance
Optimize detection reagents for each antibody in the panel
Panel Design for Iron Biology:
Effective multiplex panels might include:
Technical Validation:
Perform single-stain controls for each antibody
Include fluorescence minus one (FMO) controls
Data Analysis Considerations:
Account for spectral overlap through compensation or unmixing
Develop standardized gating strategies for flow cytometry
Establish quantitative metrics for image-based multiplexing
Apply appropriate statistical methods for multiparameter data
These considerations help ensure robust and reliable results when incorporating FTL antibodies into multiplex experimental platforms.
Several cutting-edge technologies are poised to expand the research applications of FTL recombinant antibodies:
Single-Cell Proteomics:
Integration of FTL antibodies into single-cell mass cytometry (CyTOF)
Analysis of FTL expression heterogeneity at single-cell resolution
Correlation with other iron metabolism proteins at single-cell level
Development of FTL-targeted antibody-oligonucleotide conjugates for CITE-seq
Advanced Imaging Techniques:
Super-resolution microscopy for nanoscale localization of FTL
Expansion microscopy for enhanced visualization of FTL distribution
Correlative light and electron microscopy to relate FTL localization to ultrastructure
Light sheet microscopy for 3D visualization of FTL in whole tissues or organoids
Engineered Antibody Variants:
Site-specific conjugation methods for better preservation of antibody function
Nanobody or single-chain variable fragment (scFv) derivatives for improved tissue penetration
Bispecific antibodies targeting FTL and interaction partners simultaneously
Intrabodies for live-cell tracking of FTL dynamics
Artificial Intelligence Applications:
Machine learning algorithms for automated analysis of FTL staining patterns
Deep learning approaches for multiplexed image analysis
Predictive modeling of FTL expression based on multiparameter data
Computer vision for high-throughput screening applications
These emerging technologies will provide unprecedented insights into FTL biology and iron homeostasis, potentially leading to novel therapeutic strategies for iron-related disorders.
Improving reproducibility in FTL antibody-based research requires systematic standardization efforts:
Antibody Validation Standards:
Adoption of minimum validation requirements (knockout controls, multiple applications)
Standardized reporting of validation data in publications
Independent verification of antibody specificity by third parties
Creation of publicly accessible validation datasets
Protocol Standardization:
Development of consensus protocols for common applications
Standardized fixation and antigen retrieval methods for tissues
Uniform sample preparation procedures for cell lines
Standardized reporting formats for experimental conditions
Reference Materials:
Creation of reference cell lines with defined FTL expression levels
Development of standard recombinant FTL protein preparations
Generation of standard tissue microarrays for IHC validation
Establishment of digital reference images for staining patterns
Data Reporting Requirements:
Minimum information standards for FTL antibody experiments
Mandatory reporting of antibody catalog numbers and lot numbers
Comprehensive documentation of controls used
Full disclosure of image acquisition and processing parameters
Community Resources:
Central database of validated FTL antibodies and applications
Repository of optimized protocols for different experimental contexts
Collaborative quality assessment programs
Cross-laboratory validation initiatives
These standardization efforts would significantly enhance data reproducibility and comparability across different laboratories, accelerating progress in FTL-related research fields.