LENG9 antibodies are polyclonal or monoclonal reagents that bind specifically to the LENG9 protein, a 501-amino-acid protein encoded by the LENG9 gene located on chromosome 19q13.42 . These antibodies facilitate the detection and functional analysis of LENG9 in diverse experimental contexts, including cancer biology and reproductive health studies.
Applications: Validated for Western blot (WB), immunohistochemistry (IHC), immunofluorescence (IF), and ELISA .
| Application | Tissues/Cell Lines Detected | Dilution Range |
|---|---|---|
| WB | HeLa, Y79 cells | 1:500–1:2000 |
| IHC | Kidney, brain, heart, liver, lung, placenta | 1:20–1:200 |
| IF/ICC | HeLa, MCF-7 cells | 1:200–1:800 |
LENG9 is upregulated in HPV-integrated cervical tumors, where it promotes oncogenic progression:
Functional Role: Overexpression of LENG9 in cervical cancer cells (CaSki, SiHa) enhances proliferation, migration, and invasion. Knockdown inhibits these processes .
Mechanism: LENG9 interacts with CDC5L (cell cycle regulator) and B4GALT2 (apoptosis modulator), suggesting roles in tumor survival and growth .
LENG9 is linked to endometrial receptivity and fertility:
Expression: Upregulated in receptive endometrium tissues during menstrual cycles, correlating with miR-30d activity .
Clinical Relevance: Dysregulation may contribute to endometriosis and infertility .
Targeting LENG9 could inhibit tumor progression, though no clinical trials are yet reported .
HPV Integration: LENG9 amplification via HPV-human hybrid structures drives cervical cancer aggressiveness .
Metal-Ion Binding: GO analysis identifies LENG9’s molecular function in metal-ion interactions, critical for protein interactions .
Evolutionary Conservation: LENG9 is conserved in mammals and bony fish but absent in birds or invertebrates .
LENG9 (leukocyte receptor cluster member 9) is a protein of significant research interest in human immunology and cell biology. In humans, the canonical protein consists of 501 amino acid residues with a molecular mass of 53.2 kDa. It belongs to the leukocyte receptor cluster (LRC) family and is widely expressed across multiple tissues, with notable expression in the bronchus, vagina, and appendix tissues . The protein's extensive tissue distribution suggests potential functional importance in various biological processes, making it a valuable target for antibody development to study its expression patterns, interactions, and possible roles in normal and pathological conditions.
LENG9 antibodies are employed across multiple immunodetection applications in research settings. The most widely used applications include:
Immunohistochemistry (IHC): For detecting LENG9 expression in tissue sections
Western Blot (WB): For protein identification and semi-quantitative analysis
Immunofluorescence (IF): For subcellular localization studies
Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative detection
The selection of application depends on the specific research question, with many antibodies being validated for multiple techniques. Most commercial LENG9 antibodies are specifically tested for human reactivity, though orthologs have been identified in mouse, rat, bovine, frog, zebrafish, and chimpanzee models .
When validating a new LENG9 antibody for research use, a comprehensive approach following established validation procedures is essential. A recommended methodological workflow includes:
Cell line selection: Use proteomic databases like PaxDB to identify cell lines with relatively high LENG9 expression that are amenable to genetic modification .
CRISPR/Cas9 knockout: Generate a LENG9 knockout in the selected cell line to create a definitive negative control .
Initial screening: Test the antibody by immunoblot comparing parental and knockout cell lines to verify specificity .
Expression analysis: Perform quantitative immunoblots on a panel of cell lines to identify those with the highest LENG9 expression levels for subsequent validation experiments .
Advanced validation: Use the validated cell lines to screen for antibody specificity in:
This methodical approach ensures antibody specificity and reduces the risk of misinterpreting results due to non-specific binding.
Determining the optimal working dilution for LENG9 antibodies requires a systematic titration approach specific to each application. While manufacturer recommendations provide starting points (e.g., 1:50-1:200 for immunohistochemistry and 0.25-2 μg/mL for immunofluorescence) , researchers should:
Perform dilution series: Test a range of dilutions around the manufacturer's recommendation (e.g., 1:25, 1:50, 1:100, 1:200, 1:400 for IHC).
Include controls: Always run positive controls (tissues known to express LENG9) and negative controls (LENG9 knockout tissues/cells and secondary-only controls).
Evaluate signal-to-noise ratio: The optimal dilution provides the strongest specific signal with minimal background.
Consider blocking optimization: If background remains problematic, test different blocking reagents (BSA, normal serum, commercial blockers) while maintaining the same antibody dilution.
Document conditions: Record all experimental parameters, including sample preparation methods, incubation times/temperatures, and detection systems for reproducibility.
This methodical approach ensures reliable and specific LENG9 detection while conserving valuable antibody resources.
Several critical factors can significantly impact LENG9 antibody performance in immunohistochemistry:
Fixation method and duration: Overfixation can mask epitopes while underfixation can compromise tissue morphology. For LENG9 detection, standard 10% neutral buffered formalin fixation for 24-48 hours is generally appropriate, though optimization may be required.
Antigen retrieval technique: Heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) should be tested to determine which best exposes LENG9 epitopes without causing tissue degradation.
Tissue section thickness: Optimal thickness is typically 4-5 μm; thicker sections may increase background while thinner sections may have insufficient antigen.
Blocking protocol: Thorough blocking of endogenous peroxidase activity and non-specific binding sites is essential for clean results.
Incubation conditions: Primary antibody incubation at 4°C overnight often produces better results than shorter incubations at room temperature.
Detection system sensitivity: For low-abundance targets like LENG9, amplification systems such as polymer-based detection may yield better results than standard ABC methods.
Counterstaining intensity: Overly intense hematoxylin counterstaining can obscure positive DAB staining, particularly for nuclear or low-expression targets.
Systematic optimization of these factors is necessary for reproducible LENG9 detection in tissue samples.
When encountering non-specific binding with LENG9 antibodies, a structured troubleshooting approach should be implemented:
Verify antibody specificity: Compare results with LENG9 knockout controls to confirm the observed pattern is truly non-specific binding rather than unexpected LENG9 expression .
Optimize blocking: Increase blocking duration (60-90 minutes) and test alternative blocking agents (5% BSA, 5-10% normal serum matched to secondary antibody species, commercial blockers).
Adjust antibody concentration: Dilute the primary antibody further if background is high while maintaining adequate incubation time.
Modify washing steps: Increase the number and duration of washes (e.g., 5 washes × 5 minutes each) with gentle agitation in PBS-T (0.1% Tween-20).
Pre-adsorb antibody: For polyclonal antibodies, pre-adsorption against tissue powder from the same species can reduce cross-reactivity.
Test alternative buffers: Try different diluents for the primary antibody (PBS with 1% BSA, TBS with 0.1% Tween-20).
Consider sample preparation: Some fixatives can increase background; test fresh samples or alternative fixation methods if possible.
Evaluate secondary antibody: Test different lots or sources of secondary antibody, and ensure it's appropriately matched to the primary.
Systematic documentation of these optimization steps helps establish reliable protocols for future experiments.
Investigating LENG9 protein interactions requires sophisticated molecular techniques:
Co-immunoprecipitation (Co-IP):
Use validated LENG9 antibodies to pull down LENG9 and associated proteins
Analyze precipitated complexes by mass spectrometry to identify interaction partners
Confirm interactions using reverse Co-IP with antibodies against identified partners
Proximity Ligation Assay (PLA):
Enables detection of protein-protein interactions in situ with high sensitivity
Requires antibodies against LENG9 and suspected interaction partners from different species
Produces fluorescent signals only when proteins are within 40 nm of each other
FRET/BRET Analysis:
For studying dynamic interactions in living cells
Requires fusion of fluorescent/bioluminescent proteins to LENG9 and potential partners
Measures energy transfer between proteins in close proximity
Crosslinking Mass Spectrometry:
Chemical crosslinking stabilizes transient interactions
Crosslinked complexes containing LENG9 are purified and analyzed by mass spectrometry
Provides information about interaction interfaces
Yeast Two-Hybrid Screening:
For systematic screening of potential LENG9 interaction partners
Can be followed by targeted validation using above methods in mammalian cells
Each method has strengths and limitations, and combining multiple approaches provides the most robust characterization of LENG9 interactomes.
Integrating LENG9 antibody-based protein data with genomic and transcriptomic findings requires a multi-omics approach:
Correlation analysis:
Quantify LENG9 protein levels using validated antibodies across multiple samples
Measure LENG9 mRNA expression in the same samples
Calculate correlation coefficients to identify potential post-transcriptional regulation
Cell type-specific expression:
Use LENG9 antibodies for immunohistochemistry or flow cytometry to identify cell populations expressing the protein
Compare with single-cell RNA-seq data to identify discrepancies suggesting post-transcriptional regulation
Create cell type-specific expression maps integrating both data types
Response to genetic perturbations:
Analyze LENG9 protein expression changes following CRISPR-based modification of suspected regulatory elements
Compare with transcriptional responses to identify translation-specific effects
Pathway integration:
Combine LENG9 antibody-based interaction data with pathway information from transcriptomics
Use network analysis tools to identify functional modules containing LENG9
Validate key nodes through targeted protein and transcript measurements
Disease-associated variants:
Assess how genomic variants in LENG9 affect protein expression, localization, or interactions
Use LENG9 antibodies to characterize protein-level consequences of transcriptionally-detected alterations
This integrated approach provides a more complete understanding of LENG9 biology than any single data type alone.
Multiplex immunofluorescence involving LENG9 antibodies requires careful planning and optimization:
Antibody compatibility assessment:
Test for cross-reactivity between all antibodies in the multiplex panel
Ensure all primary antibodies are raised in different species or are of different isotypes
Verify that detection systems (secondary antibodies or direct conjugates) don't cross-react
Spectral overlap management:
Select fluorophores with minimal spectral overlap
Consider brightness differences between targets (assign brighter fluorophores to less abundant targets like LENG9)
Include single-color controls for spectral unmixing during analysis
Sequential staining protocol development:
Determine optimal order of antibody application (generally from weakest to strongest signal)
Test whether heat-mediated antibody stripping between rounds affects LENG9 epitope integrity
Validate complete stripping by re-probing with secondary antibodies alone
Signal amplification considerations:
For low-abundance LENG9 detection, evaluate tyramide signal amplification (TSA) or other amplification methods
Ensure amplification doesn't increase background or cause signal bleeding into other channels
Autofluorescence management:
Implement appropriate autofluorescence quenching protocols
Consider spectral unmixing algorithms during analysis to separate true signal from autofluorescence
Validation controls:
Compare multiplex results with single-plex staining to ensure consistent LENG9 detection
Include biological controls (LENG9 knockout tissues) to confirm specificity in the multiplex context
These considerations enable reliable detection of LENG9 alongside other markers of interest in complex tissue samples.
Accurate quantification of LENG9 expression across tissues and cell types requires a multi-modal approach:
Western blot quantification:
Use validated LENG9 antibodies with recombinant LENG9 protein standards for absolute quantification
Normalize to total protein (measured by stain-free technology or housekeeping proteins)
Create standard curves spanning the expected range of LENG9 expression
Include the following quantification controls in each experiment:
| Control Type | Purpose | Implementation |
|---|---|---|
| Positive control | Verify detection system | Cell line with known high LENG9 expression |
| Negative control | Confirm specificity | LENG9 knockout cell line |
| Loading control | Ensure equal protein loading | Total protein measurement or stable reference protein |
| Standard curve | Enable absolute quantification | Purified recombinant LENG9 in serial dilutions |
Immunohistochemistry quantification:
Use digital image analysis with validated algorithms for LENG9 detection
Implement H-score or other semi-quantitative scoring systems
Calibrate using cell lines with known LENG9 expression levels embedded in control blocks
Flow cytometry:
Use directly conjugated LENG9 antibodies or validated secondary detection systems
Include quantification beads to convert fluorescence intensity to molecules of equivalent soluble fluorochrome (MESF)
Use median fluorescence intensity (MFI) for population comparisons
Mass cytometry (CyTOF):
Label LENG9 antibodies with rare earth metals for highly multiplexed analysis
Enables simultaneous phenotyping of cells while quantifying LENG9 expression
Particularly valuable for heterogeneous tissues with multiple cell populations
Targeted mass spectrometry:
Develop selected reaction monitoring (SRM) or parallel reaction monitoring (PRM) assays
Use stable isotope-labeled peptide standards for absolute quantification
Provides orthogonal validation of antibody-based methods
Combining multiple quantification approaches provides the most robust assessment of LENG9 expression across different biological contexts.
Studying LENG9 in non-human models requires careful consideration of evolutionary conservation and antibody cross-reactivity:
Cross-reactivity assessment:
Sequence homology analysis:
Align LENG9 protein sequences across species to identify conserved and divergent regions
Select antibodies targeting conserved epitopes for cross-species applications
Consider custom antibody development for highly divergent regions
Model-specific considerations:
| Model Organism | Key Considerations | Recommended Approaches |
|---|---|---|
| Mouse/Rat | High homology to human LENG9 | Many human LENG9 antibodies may cross-react; validate with knockout controls |
| Zebrafish | Moderate sequence divergence | Test multiple antibodies; consider raising antibodies against zebrafish-specific sequences |
| Xenopus | Greater evolutionary distance | Focus on highly conserved domains; validate with morpholino knockdown |
| Chimpanzee | Very high homology | Most human antibodies should work; ethical considerations limit availability |
Alternative approaches when antibodies fail:
CRISPR/Cas9 knock-in of epitope tags (FLAG, HA, etc.) to endogenous LENG9
Creation of fluorescent protein fusions for live imaging
Use of proximity labeling approaches (BioID, APEX) to study LENG9 interactomes
Functional conservation assessment:
Compare LENG9 localization patterns across species using validated antibodies
Assess whether LENG9 interactions are conserved through co-immunoprecipitation
Determine if expression patterns during development are maintained across species
These approaches enable robust comparative studies of LENG9 biology across evolutionary diverse model systems.
Epitope mapping for LENG9 antibodies requires systematic analysis to precisely identify antibody binding sites:
Peptide array analysis:
Synthesize overlapping peptides (typically 15-20 amino acids with 5-10 amino acid overlaps) covering the entire LENG9 sequence
Immobilize peptides on membranes or glass slides
Probe with LENG9 antibodies and detect binding patterns
Identify peptides showing positive signals to narrow down epitope regions
Recombinant fragment analysis:
Generate a series of truncated LENG9 constructs
Express and purify these fragments
Test antibody binding by Western blot or ELISA
Progressively narrow down the epitope region
Alanine scanning mutagenesis:
Once a general epitope region is identified, create point mutations changing each amino acid to alanine
Test antibody binding to each mutant
Identify critical residues required for antibody recognition
Hydrogen/deuterium exchange mass spectrometry:
Compare hydrogen/deuterium exchange rates of LENG9 alone versus LENG9-antibody complex
Regions protected from exchange in the complex represent potential epitopes
Provides structural insights about the antibody-antigen interaction
X-ray crystallography or cryo-EM:
For highest resolution epitope mapping
Determine 3D structure of antibody-LENG9 complex
Identifies precise atomic interactions at the binding interface
Computational approaches:
Use existing structural data or predicted models of LENG9
Apply epitope prediction algorithms to identify surface-exposed regions
Guide experimental approaches based on predictions
Epitope information helps explain cross-reactivity patterns, predict potential interference with protein function, and enables strategic selection of antibody pairs for sandwich assays.
Rigorous evaluation criteria ensure selection of optimal LENG9 antibodies for specific research applications:
Validation documentation assessment:
Application-specific considerations:
| Application | Critical Evaluation Criteria | Preferred Antibody Characteristics |
|---|---|---|
| Western Blot | Band specificity, signal-to-noise ratio | Recognizes denatured epitopes, high affinity |
| IHC/IF | Background level, subcellular localization pattern | Compatible with fixation, recognizes native epitopes |
| IP/Co-IP | Precipitation efficiency, non-specific binding | High affinity, minimal cross-reactivity |
| Flow Cytometry | Cell surface vs. intracellular accessibility | Recognizes native epitopes, direct conjugates preferred |
| ELISA | Sensitivity, dynamic range | Pairs well with detection antibody, high affinity |
Antibody format evaluation:
Monoclonal vs. polyclonal: Monoclonals provide higher reproducibility; polyclonals may offer better sensitivity
Host species: Consider compatibility with other antibodies in multiplex applications
Clonality: For monoclonals, compare different clones recognizing distinct epitopes
Conjugation: Evaluate need for direct conjugates vs. secondary detection
Reproducibility considerations:
Assess lot-to-lot consistency data if available
Check recombinant vs. immunized animal-derived antibodies (recombinant generally offers higher reproducibility)
Evaluate production scale and long-term availability for longitudinal studies
Supporting literature evaluation:
Review publications using the antibody for similar applications
Assess quality of methods and controls in published studies
Contact authors for additional information if needed
These systematic evaluation criteria help researchers select antibodies most likely to yield reliable, reproducible results for their specific LENG9 studies.
Systematic comparison of LENG9 antibodies requires standardized evaluation protocols:
Side-by-side testing framework:
Application-specific performance metrics:
For Western blot:
Signal-to-noise ratio at equivalent concentrations
Specificity (absence of non-specific bands)
Sensitivity (detection limit using diluted samples)
Reproducibility (consistency across replicate blots)
For IHC/IF:
Background levels in negative control tissues
Signal intensity in positive control tissues
Subcellular localization precision
Staining pattern consistency across tissue replicates
For immunoprecipitation:
Capture efficiency (percentage of input LENG9 recovered)
Purity of precipitated material (non-specific co-precipitants)
Compatibility with downstream applications
Quantitative scoring system:
Develop a weighted scoring rubric for each performance attribute
Assign numerical scores to each antibody across criteria
Calculate composite scores for objective comparison
| Performance Attribute | Weight | Scoring Criteria | Example |
|---|---|---|---|
| Specificity | 40% | 1-5 scale based on background/non-specific binding | Ab1: 4, Ab2: 2, Ab3: 5 |
| Sensitivity | 30% | 1-5 scale based on detection limits | Ab1: 3, Ab2: 5, Ab3: 2 |
| Reproducibility | 20% | 1-5 scale based on replicate consistency | Ab1: 5, Ab2: 3, Ab3: 4 |
| Cost-effectiveness | 10% | 1-5 scale based on price per application | Ab1: 2, Ab2: 4, Ab3: 3 |
| Weighted Total | 100% | Sum of weighted scores | Ab1: 3.7, Ab2: 3.3, Ab3: 3.8 |
Blind evaluation:
Code antibodies to prevent bias during evaluation
Have multiple researchers independently assess performance
Compare scores and resolve discrepancies through consensus
This structured comparison approach enables objective selection of the best LENG9 antibody for specific research applications.
Comprehensive documentation ensures experimental reproducibility and facilitates troubleshooting:
Antibody metadata documentation:
Manufacturer and catalog number
Lot number (critical for reproducibility assessment)
Clone name/number for monoclonals
Host species and antibody isotype
Immunogen details (peptide sequence, fusion protein design)
Production method (hybridoma, recombinant, polyclonal)
Validation documentation:
Specificity controls used (knockout, knockdown, peptide blocking)
Cross-reactivity testing results
Application-specific validation data
References to published validation studies
Experimental protocol documentation:
Complete step-by-step procedures with exact buffer compositions
Antibody concentration and dilution calculation method
Incubation times and temperatures
Detection systems with complete details
Image acquisition parameters (exposure times, gain settings)
Quantification methods and software used
Structured reporting format:
Laboratory notebooks with consistent antibody information sections
Electronic records with standardized fields for antibody details
Protocol management systems linking antibody information to methods
Sample tracking connected to antibody lot information
Reporting standards for publication:
Follow journal-specific antibody reporting requirements
Include all validation data in supplements if not in main text
Provide RRID (Research Resource Identifier) for each antibody
Deposit detailed protocols in repositories like protocols.io
This comprehensive documentation approach ensures experimental reproducibility, facilitates troubleshooting, and enhances the scientific value of research using LENG9 antibodies.
LENG9 antibodies can provide crucial insights into tissue-specific expression patterns through several advanced approaches:
Multi-tissue profiling:
Apply validated LENG9 antibodies to tissue microarrays containing diverse normal and diseased tissues
Quantify expression levels and subcellular localization patterns across tissues
LENG9 shows notable expression in bronchus, vagina, and appendix tissues, suggesting specific biological roles in these contexts
Single-cell resolution analysis:
Combine LENG9 antibodies with single-cell technologies:
Mass cytometry (CyTOF) with metal-conjugated LENG9 antibodies
Imaging mass cytometry for spatial context
CODEX multiplexed imaging for simultaneous detection of LENG9 and cell type markers
Correlate LENG9 protein expression with single-cell transcriptomics data
Developmental trajectory mapping:
Apply LENG9 antibodies to tissue samples across developmental timepoints
Trace expression changes during tissue differentiation and maturation
Identify potential roles in tissue-specific developmental processes
Pathological condition analysis:
Compare LENG9 expression between normal and diseased tissues
Evaluate expression changes in inflammatory conditions, cancer progression, or tissue remodeling
Assess potential as a diagnostic or prognostic biomarker
Signaling context integration:
Combine LENG9 detection with phospho-specific antibodies against signaling molecules
Map LENG9 expression to specific signaling states in different tissues
Identify tissue-specific regulatory networks involving LENG9
These approaches could reveal previously unknown functions of LENG9 in specific tissue contexts and potentially identify new therapeutic targets or diagnostic markers.
Emerging trends in antibody validation represent critical advances that should be applied to LENG9 research:
Genetic knockout validation as the gold standard:
Orthogonal validation methods:
Combine antibody-based detection with mass spectrometry
Compare protein levels detected by antibodies with targeted MS-based peptide quantification
Correlate with independent measures like LENG9 mRNA levels while accounting for post-transcriptional regulation
Automated high-throughput validation pipelines:
Standardized testing across hundreds of conditions and fixation methods
Machine learning algorithms to assess staining patterns and predict specificity
Cloud-based repositories of validation data accessible to researchers
Independent validation initiatives:
Multi-laboratory blind testing of antibody performance
Preregistered validation studies to prevent publication bias
Community-driven validation databases with standardized metrics
Application-specific validation:
Recognition that antibody performance varies across applications
Validation data must match the intended research application
Development of application-specific validation standards and metrics
Recombinant antibody technologies:
Shift toward sequence-defined recombinant antibodies for LENG9
Elimination of batch-to-batch variation inherent to hybridoma or serum-derived antibodies
Genetic engineering to optimize performance for specific applications
Implementing these emerging validation approaches for LENG9 antibodies will significantly enhance research reproducibility and reliability.
Computational methods are increasingly essential for extracting maximum value from LENG9 antibody-based experiments:
Image analysis algorithms:
Automated segmentation of cells/tissues in LENG9 immunofluorescence images
Quantification of expression levels across heterogeneous samples
Subcellular localization pattern recognition and classification
Correlation of LENG9 expression with morphological features
Multi-omics data integration:
Computational frameworks linking LENG9 protein data with:
Transcriptomics (RNA-seq, single-cell RNA-seq)
Genomics (variant effects, regulatory elements)
Proteomics (interaction networks, post-translational modifications)
Metabolomics (pathway activities)
Bayesian networks to infer causal relationships
Machine learning for pattern recognition:
Supervised learning to identify LENG9 expression patterns associated with biological states
Unsupervised learning to discover novel LENG9-related tissue or cell classifications
Deep learning approaches for feature extraction from complex tissue images
Predictive modeling:
Structural modeling of LENG9 to predict antibody epitopes
Simulation of antibody-antigen interactions for optimized selection
In silico prediction of cross-reactivity risks
Knowledge graph approaches:
Integration of LENG9 antibody data with published literature
Automated hypothesis generation through network analysis
Identification of knowledge gaps and prioritization of experiments
Reproducibility enhancement tools:
Automated protocol generation based on antibody characteristics
Statistical power calculators specific to antibody-based experiments
Digital notebook systems capturing all experimental parameters
These computational approaches transform antibody-based data from descriptive observations into mechanistic insights and predictive models.