The FAM53A Antibody, Biotin Conjugated is a specialized immunoreagent designed for detecting the FAM53A protein (Family With Sequence Similarity 53 Member A) in research applications. This antibody is chemically linked to biotin, a vitamin that enables high-affinity binding to streptavidin or avidin, facilitating signal amplification in assays such as ELISA, Western blotting, and immunohistochemistry (IHC) .
Target: FAM53A, a protein implicated in cellular processes like proliferation, migration, and tumor suppression .
Conjugation: Biotin, enabling compatibility with streptavidin-enzyme or fluorophore conjugates .
Host: Rabbit-derived polyclonal antibody, ensuring broad epitope recognition .
Biotin conjugation enhances detection sensitivity by leveraging streptavidin’s high binding affinity () . Common uses include:
ELISA: Paired with streptavidin-HRP for signal amplification .
Western Blot: Indirect detection via biotin-streptavidin systems .
Immunohistochemistry (IHC): Localizes FAM53A in tissues (e.g., nuclear/cytoplasmic staining in human skeletal muscle) .
FAM53A exhibits dual roles in breast cancer, depending on p53 status:
p53-Positive Cells: Acts as a tumor suppressor by inhibiting migration, invasion, and proliferation via MEK/ERK pathway modulation .
p53-Negative Cells: Potential therapeutic target due to elevated expression promoting oncogenic traits .
Correlation with p53: FAM53A levels inversely correlate with p53 status in clinical samples .
Pathway Modulation: Downregulates Snail, cyclin D1, and MMP9; upregulates E-cadherin and p21 .
Major suppliers include:
Abbexa Ltd: Offers biotin-conjugated FAM53A antibodies validated for ELISA .
CUSABIO TECHNOLOGY LLC: Provides custom conjugates with >95% purity .
Biotin Interference: Endogenous biotin in samples may cause false signals; blocking kits are recommended .
Storage: Repeated freeze-thaw cycles degrade antibody efficacy; aliquot for long-term stability .
Further studies are needed to:
FAM53A, also known as Dorsal neural-tube nuclear protein, is a 398 amino acid nuclear protein that plays a crucial role in neural development by specifying dorsal cell fates within the neural tube. It is particularly expressed in dorsal regions of the midbrain, hindbrain, diencephalon, and spinal neural tube during embryonic development. It is also expressed at lower levels in the branchial arches, telencephalon, heart, and somites of developing embryos . The protein's involvement in dorsomedial patterning of the third ventricle suggests it may be a critical factor in brain development research . Understanding FAM53A function has implications for developmental neurobiology and potentially for neurological disorders, as the gene is located on chromosome 4, which also contains the Huntingtin gene associated with Huntington's disease .
FAM53A antibodies enable researchers to investigate several key developmental processes, including:
Neural tube dorsal-ventral patterning mechanisms
Cell fate specification in the developing central nervous system
Molecular signaling hierarchies in neural differentiation
Embryonic brain region formation, particularly in the midbrain and hindbrain
Potential interactions between neural development and somitogenesis
Studies tracking FAM53A expression patterns through immunohistochemistry can reveal temporal and spatial dynamics of neural development, potentially uncovering new insights into how the vertebrate nervous system establishes its complex architecture during embryogenesis .
When designing immunohistochemistry experiments with biotin-conjugated FAM53A antibodies, consider these methodological approaches:
For paraffin-embedded tissues (IHC-P):
Use optimal fixation methods that preserve antigen structure (4% paraformaldehyde is often suitable)
Include antigen retrieval steps (citrate buffer pH 6.0 or EDTA buffer pH 9.0)
Apply recommended dilution ratios (1:200-400) to optimize signal-to-noise ratio
Incorporate appropriate blocking steps to minimize non-specific binding
Use streptavidin-conjugated detection systems compatible with biotin conjugation
Include both positive controls (tissues known to express FAM53A) and negative controls (primary antibody omission)
For frozen sections (IHC-F):
Use freshly prepared cryosections (10-14 μm thickness)
Consider shorter fixation times compared to paraffin sections
Optimize incubation times and temperatures empirically for your specific tissue
For all IHC applications, developmental stage-specific positive controls are crucial since FAM53A expression varies temporally during embryogenesis.
For rigorous Western blotting experiments using biotin-conjugated FAM53A antibodies, implement these critical controls:
Positive control tissue/cell lysates: Use neural tissue known to express FAM53A (e.g., embryonic brain samples) to validate antibody performance
Negative control lysates: Include samples from tissues with minimal FAM53A expression
Loading controls: Probe for housekeeping proteins (β-actin, GAPDH) to normalize expression levels
Molecular weight validation: Confirm detection at the expected molecular weight (~45 kDa for FAM53A)
Dilution optimization: Test multiple antibody dilutions (1:300-5000 range) to determine optimal signal-to-background ratio
Blocking optimization: Evaluate different blocking agents to reduce background from the biotin-streptavidin detection system
Signal inhibition control: Pre-incubation with immunizing peptide should abolish specific signal
Secondary reagent control: Include lanes with only streptavidin-conjugated detection reagent to identify endogenous biotin-containing proteins
Comprehensive controls ensure reliable interpretation of FAM53A expression data and enable troubleshooting of technical issues.
Optimizing dual immunofluorescence protocols for co-localization studies involving biotin-conjugated FAM53A antibodies requires careful consideration of several technical parameters:
Sequential immunostaining approach:
First detect the non-biotinylated antibody using standard indirect immunofluorescence
Block any free binding sites with excess unconjugated secondary antibody
Apply biotin-conjugated FAM53A antibody
Detect with fluorophore-conjugated streptavidin (e.g., Alexa Fluor 488 or 594)
Alternative detection strategies:
Consider using tyramide signal amplification (TSA) to enhance detection sensitivity of low-abundance FAM53A protein
Employ streptavidin-conjugated quantum dots for photostable signal and multiplexing capability
Spectral compatibility assessment:
Select fluorophores with minimal spectral overlap
Include single-stained controls for each fluorophore to establish appropriate imaging parameters
Consider linear unmixing algorithms during image acquisition if spectral overlap cannot be avoided
Z-stack acquisition:
Collect optical sections through the entire nucleus to properly evaluate nuclear localization of FAM53A
Use deconvolution algorithms to enhance resolution of subcellular co-localization
Quantitative co-localization analysis:
Apply Manders' or Pearson's correlation coefficients to quantify the degree of co-localization
Use appropriate software (ImageJ with co-localization plugins, Imaris, or similar tools) for objective analysis
This approach enables precise spatial relationship analysis between FAM53A and other developmental markers while accounting for the biotin conjugation of the primary antibody.
While FAM53A is primarily studied for its role in neural development, investigating its potential interactions with chromatin may provide insights into its regulatory mechanisms. When adapting biotin-conjugated FAM53A antibodies for ChIP applications, consider:
Crosslinking optimization:
Test different formaldehyde concentrations (0.5-2%) and incubation times
Consider dual crosslinking with disuccinimidyl glutarate (DSG) followed by formaldehyde for optimal capture of protein-DNA interactions
Sonication parameters:
Optimize sonication conditions to generate DNA fragments of 200-500 bp
Confirm fragmentation efficiency by agarose gel electrophoresis
Pre-clearing strategy:
Implement rigorous pre-clearing with protein G beads to minimize non-specific binding
Include additional blocking steps with BSA and salmon sperm DNA
Immunoprecipitation approach:
Use streptavidin-coated magnetic beads rather than agarose beads for higher efficiency
Include IgG controls matched to the host species (rabbit) of the FAM53A antibody
Consider a two-step IP approach: first capture with anti-FAM53A, then capture with streptavidin
Washing stringency:
Implement progressively stringent washing buffers to minimize background
Monitor washing efficiency through measurement of unbound biotinylated antibody
Elution considerations:
Use biotin competition elution rather than standard SDS elution to specifically release biotin-conjugated complexes
Consider on-bead digestion protocols to minimize background
The biotin conjugation provides both advantages (high-affinity streptavidin capture) and challenges (endogenous biotin competition) that must be carefully addressed in experimental design.
FAM53A shows variable conservation across species, which has important implications for cross-species applications of antibodies. Based on available data:
Confirmed reactivity:
Predicted reactivity:
Conservation considerations:
The epitope region spanning amino acids 255-340/398 appears to be relatively conserved among vertebrates
The immunogen range may determine cross-reactivity success with different species
Experimental validation requirements:
When working with species beyond human and dog, researchers should:
Perform careful validation studies including Western blotting to confirm reactivity
Consider epitope mapping to identify regions of highest conservation
Include appropriate positive and negative controls from the target species
Alternative approaches for non-reactive species:
Custom antibody generation using species-specific peptides
Use of gene expression analysis rather than protein detection
Consideration of alternative FAM53 family members (FAM53B, FAM53C) that may serve similar functions
Researchers should carefully evaluate sequence homology in the epitope region before applying these antibodies to evolutionary studies or when working with non-validated species.
Rigorous validation of FAM53A antibody specificity is essential for generating reliable data, particularly when studying different neural tissues. Implement these complementary validation strategies:
Genetic validation approaches:
Use CRISPR/Cas9 knockout controls where FAM53A has been deleted
Compare antibody staining in wild-type versus knockdown tissues (siRNA, shRNA)
Confirm loss of signal in knockout/knockdown samples by multiple detection methods
Biochemical validation:
Perform peptide competition assays using the immunizing peptide
Conduct Western blotting to confirm detection of a single band at the expected molecular weight
Validate using recombinant FAM53A protein as a positive control
Cross-methodological validation:
Compare protein detection with mRNA expression patterns using in situ hybridization
Correlate antibody staining with transgenic reporter lines (if available)
Use multiple antibodies targeting different epitopes of FAM53A
Tissue-specific considerations:
Evaluate background staining in tissues known to lack FAM53A expression
Test fixation-dependent epitope masking by comparing multiple fixation protocols
Assess developmental stage-specific expression patterns to confirm alignment with published transcriptomic data
Signal quantification:
Implement quantitative image analysis to compare signal-to-background ratios across tissues
Establish threshold criteria for positive versus negative staining
This multi-faceted validation approach ensures confidence in experimental results and facilitates troubleshooting if inconsistencies arise during research.
High background is a common challenge when working with biotin-conjugated antibodies due to endogenous biotin in tissues. Here's a systematic approach to minimize background:
Pre-analytical considerations:
Block endogenous biotin/avidin binding sites using commercial biotin/avidin blocking kits
Implement additional blocking steps with 5% BSA or 5-10% normal serum from the same species as secondary reagents
Consider using non-fat dry milk as an alternative blocking agent for Western blots
Washing optimization:
Increase washing duration and frequency between steps
Add low concentrations of detergent (0.05-0.1% Tween-20) to washing buffers
Consider using high-salt washing steps to reduce non-specific ionic interactions
Antibody dilution refinement:
Tissue-specific adaptations:
For tissues with high endogenous biotin (like brain, kidney, liver):
Consider alternative detection methods or non-biotinylated primary antibodies
Pre-absorb tissues with streptavidin followed by biotin before applying the biotinylated antibody
Quench endogenous peroxidase activity thoroughly if using HRP-conjugated streptavidin
Alternative detection strategies:
Consider tyramide signal amplification to allow higher primary antibody dilutions
Use fluorescent streptavidin conjugates with spectral properties distinct from autofluorescent components
Sample preparation modifications:
Adjust fixation protocols to better preserve antigenicity while reducing background
Incorporate additional permeabilization steps for optimal nuclear antigen access
Implementing these strategies systematically while changing only one variable at a time will help identify the source of background issues.
Detecting low-abundance FAM53A in developing neural tissues requires specialized approaches to enhance sensitivity without introducing artifacts:
Signal amplification methods:
Implement tyramide signal amplification (TSA) which can increase sensitivity 10-100 fold
Use polymer-based detection systems that carry multiple enzyme molecules per antibody binding event
Consider sequential application of biotin-streptavidin layers for multi-layered amplification
Sample preparation optimization:
Test multiple fixatives to identify optimal epitope preservation conditions
Compare heat-induced versus enzymatic antigen retrieval methods
Consider shorter fixation times to minimize epitope masking
Instrumentation considerations:
Use high-sensitivity detection systems (PMT-based confocal microscopy, sCMOS cameras)
Implement spectral unmixing to distinguish specific signal from tissue autofluorescence
Consider super-resolution microscopy for improved signal discrimination
Protocol adaptations:
Extend primary antibody incubation time (overnight at 4°C or up to 48-72 hours)
Adjust incubation temperature profiles (e.g., room temperature versus 4°C)
Use of orbital shakers to improve antibody penetration
Concentration methods:
For Western blotting, implement immunoprecipitation before gel electrophoresis
Use gradient gels to better resolve proteins in the expected molecular weight range
Consider loading higher protein amounts when working with tissues having low FAM53A expression
Comparative analysis approaches:
Implement ratiometric imaging comparing FAM53A to housekeeping proteins
Perform digital image analysis with appropriate thresholding algorithms
Use software that can detect signal above calculated background levels
These approaches can be combined and optimized depending on the specific experimental context and biological question being addressed.
Distinguishing genuine FAM53A staining from artifacts requires systematic analysis and appropriate controls:
Pattern analysis criteria:
True FAM53A expression should show nuclear localization consistent with its function
Expression patterns should correlate with known developmental domains (dorsal neural tube, midbrain, hindbrain)
Staining should show biological coherence (e.g., gradients rather than random cell positivity)
Signal intensity should correlate with known expression levels in different tissues
Control-based validation:
Compare staining patterns with in situ hybridization data for FAM53A mRNA
Analyze knockout/knockdown tissues to confirm signal specificity
Evaluate multiple antibodies targeting different FAM53A epitopes
Technical artifact identification:
Edge artifacts: Stronger staining at tissue margins suggests diffusion limitations
Necrotic tissue: Non-specific binding to damaged tissue regions
Nuclear trapping: Some secondary antibodies can be non-specifically retained in nuclei
Biotin-rich regions: May show false positivity with inadequate blocking
Quantitative approaches:
Implement computational image analysis to objectively measure signal characteristics
Calculate signal-to-noise ratios across different experimental conditions
Use threshold-based segmentation to distinguish positive cells from background
Contextual interpretation:
Compare FAM53A patterns with related developmental markers
Evaluate consistency across multiple developmental timepoints
Assess biological plausibility based on known developmental processes
When analyzing quantitative FAM53A expression data across developmental stages, consider these statistical approaches:
Normalization strategies:
Normalize FAM53A expression to appropriate housekeeping proteins/genes
Consider geometric mean of multiple reference genes for more stable normalization
Implement tissue-specific normalization factors when comparing across different neural regions
Time-series analysis methods:
Apply repeated measures ANOVA for longitudinal developmental studies
Consider mixed-effects models when analyzing multiple embryos across timepoints
Implement time-series clustering to identify patterns in expression trajectories
Spatial analysis approaches:
Use spatial statistics (Moran's I, Getis-Ord) to quantify expression clustering
Apply gradient analysis methods to quantify dorsal-ventral or anterior-posterior patterns
Consider dimensionality reduction techniques for complex spatial datasets
Comparative statistical methods:
Utilize non-parametric tests (Mann-Whitney, Kruskal-Wallis) when normality cannot be assumed
Apply multiple comparison corrections (Bonferroni, Benjamini-Hochberg) for statistical rigor
Consider ANCOVA when controlling for covariates like tissue size or cell density
Visualization approaches:
Generate heatmaps to visualize expression across developmental stages and tissues
Use principal component analysis (PCA) plots to identify major sources of variation
Implement box-and-whisker plots to display distribution characteristics at each timepoint
Power analysis considerations:
Conduct a priori power analyses to determine appropriate sample sizes
Report effect sizes alongside p-values for better interpretation of biological significance
Consider Bayesian approaches for studies with limited sample availability
The choice of statistical methods should be guided by experimental design, data distribution characteristics, and the specific developmental hypotheses being tested.
Single-cell technologies offer transformative approaches to understanding FAM53A function with unprecedented resolution:
Single-cell transcriptomics applications:
scRNA-seq to identify cell populations expressing FAM53A within heterogeneous neural tissues
Trajectory analysis to map FAM53A expression changes during differentiation pathways
Co-expression network analysis to identify genes functionally related to FAM53A
Spatial transcriptomics integration:
Combining antibody detection with in situ sequencing to correlate protein and mRNA localization
Spatial transcriptomics platforms (Visium, Slide-seq) to map FAM53A expression in tissue context
Integration of spatial data with developmental atlases to place FAM53A in anatomical context
Single-cell proteomics approaches:
Mass cytometry (CyTOF) with FAM53A antibodies for high-dimensional protein analysis
Single-cell Western blotting to quantify FAM53A protein levels in individual cells
Proximity ligation assays to detect FAM53A protein interactions at single-cell resolution
Functional genomics at single-cell level:
CRISPR screens with single-cell readouts to identify FAM53A-dependent pathways
Single-cell ATAC-seq to correlate FAM53A expression with chromatin accessibility states
Perturb-seq approaches to measure transcriptional consequences of FAM53A modulation
Computational integration strategies:
Multi-omics data integration algorithms to connect FAM53A genomics, transcriptomics, and proteomics
Trajectory inference methods to map FAM53A dynamics during development
Machine learning approaches to predict FAM53A function from single-cell profiles
These emerging technologies provide complementary approaches to traditional antibody-based methods and may reveal previously unrecognized aspects of FAM53A biology in neural development.
FAM53A research has several promising applications for understanding neurodevelopmental disorders:
Structural brain abnormalities:
FAM53A's role in dorsal neural tube development suggests potential involvement in cortical malformations
The protein's expression in the developing midbrain and hindbrain indicates possible relevance to cerebellum-related disorders
Disruptions in dorsal-ventral patterning could contribute to neural tube defects
Genetic disorder associations:
Pathway integration:
FAM53A likely participates in developmental signaling pathways relevant to neurodevelopmental disorders
Integration with known pathways (Wnt, Sonic Hedgehog, BMP) could provide mechanistic insights
FAM53A binding partners may include proteins already implicated in neurodevelopmental conditions
Therapeutic target exploration:
Understanding FAM53A regulation could identify intervention points for developmental disorders
Modulation of FAM53A-dependent pathways might offer approaches for neural repair or regeneration
Small molecule screens targeting FAM53A interactions could yield therapeutic candidates
Biomarker potential:
FAM53A expression patterns might serve as diagnostic markers for specific developmental disorders
Changes in FAM53A levels could indicate pathway disruptions in patient-derived neural organoids
Longitudinal assessment might help predict developmental trajectories
These applications highlight the importance of developing specific and reliable FAM53A antibodies for both research and potential clinical applications in neurodevelopmental disorders.