NANOG is a homeodomain transcription factor essential for sustaining the self-renewal and undifferentiated state of ESCs. It works alongside Oct4 and Sox2 to form a regulatory network that suppresses differentiation signals . Aberrant NANOG expression is linked to germ cell tumors, breast cancer, and laryngeal carcinomas, where it promotes chemoresistance and metastasis .
The table below compares widely used NANOG monoclonal antibodies:
Pluripotency Maintenance: NANOG antibodies (e.g., H-2) identify interactions with Oct4 and Sox2 in ESC nuclei, critical for reprogramming somatic cells into induced pluripotent stem cells (iPSCs) .
Differentiation Markers: Downregulation of NANOG, detectable via WB or ICC, correlates with ESC differentiation .
Tumor Progression: Overexpression of NANOG in laryngeal precancerous lesions (detected by clone 67255-1-Ig) predicts malignant transformation, with 60% of dysplasias showing NANOG positivity .
Therapeutic Resistance: NANOG upregulates CD59 in immune-edited tumors, conferring resistance to trastuzumab-mediated complement-dependent cytotoxicity (CDC). Silencing NANOG restores CDC sensitivity .
Buffer Requirements: Clone eBioMLC-51 requires Foxp3 Staining Buffer for intracellular staining to prevent non-specific binding .
Cross-Reactivity: Clone H-2’s broad species reactivity makes it suitable for comparative studies across human, mouse, and rat models .
NANOG monoclonal antibodies are pivotal in:
The monoclonal NANOG antibody is produced from the hybridoma generated by fusing mouse myeloma cells with splenocytes from mice immunized with recombinant human NANOG protein. It is purified from mouse ascites using protein G, achieving a purity exceeding 95%. This unconjugated NANOG monoclonal antibody aligns with the mouse IgG1 isotype. It is suitable for various applications including ELISA, Western Blot, Immunocytochemistry, Immunofluorescence, and Flow Cytometry. The antibody can detect the NANOG protein in human, mouse, and rat samples.
NANOG, a transcription factor, plays a crucial role in the self-renewal of embryonic stem cells (ES), acting as a vital factor in maintaining pluripotency. Research has demonstrated the strict involvement of NANOG in the process of carcinogenesis, suggesting its potential as a prognostic marker for malignant tumors.
NANOG is a multidomain homeobox transcription factor that functions to maintain the undifferentiated state of pluripotent stem cells. It works by counteracting differentiation-promoting signals induced by extrinsic factors including LIF, Stat3, and BMP. As a key pluripotency regulator, NANOG expression is specifically localized to early embryos, the inner cell mass of the blastocyst, embryonic stem (ES) cells, and embryonic germ (EG) cells . Its expression is downregulated upon cellular differentiation, making it an essential marker for identifying pluripotent cell populations. NANOG functions alongside other pluripotency factors such as Oct4, SOX2, FoxD3, and Tcf3, creating a regulatory network that maintains stemness . The ability to reliably detect NANOG using specific monoclonal antibodies is therefore fundamental to understanding pluripotency mechanisms, monitoring differentiation protocols, and investigating developmental processes.
For human NANOG detection, antibodies like hNanog.2 clone recognize human-specific epitopes and have been validated for western blotting (≤1 μg/mL), immunohistochemistry (≤20 μg/mL), and immunocytochemistry (≤5 μg/mL) applications . These antibodies typically detect NANOG in human embryonic stem cells, NTERA cell lines, and various human cancer tissues.
For mouse NANOG detection, antibodies like eBioMLC51 clone have been specifically validated against mouse NANOG and detect a band at approximately 45 kDa in F9 embryonal carcinoma lysates but not in differentiated cell lines like NIH3T3 . The expression patterns detected by mouse-specific antibodies often overlap with but are not identical to Oct4 patterns, reflecting their distinct regulatory roles in maintaining pluripotency.
When designing experiments, researchers must carefully select the appropriate species-specific antibody to ensure optimal detection sensitivity and specificity, particularly in comparative studies of pluripotency mechanisms across species.
For optimal functionality of NANOG monoclonal antibodies, specific storage and handling procedures should be followed:
| Storage Duration | Recommended Temperature | Additional Considerations |
|---|---|---|
| Up to 1 month | 4°C | Maintain original formulation |
| Longer periods | -20°C | Avoid freeze-thaw cycles |
| Shelf life | 12 months at -20°C, 1 month at 4°C | Monitor expiration dates |
The standard formulation for many NANOG antibodies is 1mg/ml containing PBS (pH 7.4) with 0.1% Sodium Azide as a preservative . This formulation helps maintain antibody stability during storage. Most critical to antibody longevity is preventing repeated freeze-thaw cycles, which can cause protein denaturation and significantly reduce antibody functionality .
For routine experimental use, researchers should aliquot the stock antibody solution into single-use volumes upon receipt to minimize freeze-thaw events. When handling the antibody, use aseptic techniques and ensure all buffers and solutions are prepared with high-quality reagents. If diluting the antibody for specific applications, prepare fresh working solutions whenever possible rather than storing diluted antibody. Following these handling procedures will help maintain antibody specificity and sensitivity across multiple experiments.
Optimizing NANOG antibody concentrations requires method-specific titration based on established starting points:
For Western blotting optimization, researchers should prepare a dilution series (e.g., 1:250, 1:500, 1:1000, 1:2000) and test against positive control lysates from embryonic stem cells or NTERA cell lines . The optimal concentration provides clear detection of the expected 40-45 kDa band with minimal background. Using recommended immunoblot buffer conditions is critical for reproducible results .
For immunostaining applications, antibody titration should be performed with both positive controls (embryonic stem cells, testicular tissues) and negative controls (fully differentiated cells) . The optimal concentration yields strong nuclear signal in NANOG-expressing cells with minimal cytoplasmic or background staining. For tissue sections, testing both high and low pH antigen retrieval methods is recommended to determine optimal epitope accessibility .
Each new antibody lot should undergo validation titration to account for potential lot-to-lot variations in antibody concentration and affinity. This methodical approach ensures consistent, reproducible NANOG detection across experiments.
A comprehensive control strategy is essential for validating NANOG antibody specificity:
Positive Cellular Controls:
Negative Cellular Controls:
Tissue Controls:
Technical Controls:
Isotype-matched irrelevant antibody controls
Secondary antibody-only controls
Peptide competition assays to confirm epitope specificity
Multiple antibody clones targeting different NANOG epitopes
Genetic Controls:
NANOG knockdown/knockout cells (for reduced/absent signal)
NANOG overexpression systems (for enhanced signal)
Orthogonal Validation:
Correlation with NANOG mRNA expression
Co-staining with other pluripotency markers (Oct4, Sox2)
Functional assays of pluripotency correlating with NANOG expression
Implementation of this multi-layered control strategy ensures that observed NANOG staining patterns reflect true biological expression rather than technical artifacts or cross-reactivity with other proteins, providing a solid foundation for interpreting experimental results in pluripotency research.
The optimal protocol for NANOG ChIP experiments involves several critical steps:
Sample Preparation:
Culture embryonic stem cells under defined conditions maintaining pluripotency
Fix cells with 1% formaldehyde for 10-15 minutes at room temperature
Quench fixation with 125 mM glycine for 5 minutes
Harvest cells and wash thoroughly with ice-cold PBS
Resuspend in lysis buffer containing protease inhibitors
Sonicate chromatin to generate fragments of 200-500 bp
Immunoprecipitation:
Pre-clear chromatin with protein G beads
Incubate pre-cleared chromatin with NANOG antibody overnight at 4°C
Add protein G beads and incubate for 2-3 hours
Perform stringent washes to remove non-specific binding
Elute protein-DNA complexes and reverse crosslinks
Detection Methods:
For known targets: PCR amplification using primers spanning NANOG binding sites
For genome-wide analysis: Next-generation sequencing of immunoprecipitated DNA
Include input chromatin controls and IgG negative controls
Critical Optimization Parameters:
Sonication conditions require cell-type specific optimization
Antibody concentration may need adjustment based on NANOG expression levels
Washing stringency balances specificity against signal strength
As demonstrated in search result , this methodology successfully identifies NANOG-regulated genes. For human embryonic stem cells, researchers can use human-specific NANOG antibodies with appropriate primers for human NANOG target genes. This approach enables investigation of the transcriptional networks controlled by NANOG in maintaining pluripotency and regulating differentiation.
NANOG expression shows natural heterogeneity in pluripotent populations, making it an excellent marker for studying subpopulation dynamics. A comprehensive methodological approach includes:
Quantitative Single-Cell Analysis:
Immunofluorescence with precise calibration standards
Flow cytometry with careful gating strategies
Single-cell Western techniques for protein quantification
Combine with cell cycle markers to correlate NANOG expression with cell cycle phases
Multi-Dimensional Analysis:
Co-staining with other heterogeneously expressed factors (e.g., Rex1, Stella)
Correlation with surface markers for live cell isolation
Integration with functional assays of self-renewal and differentiation potential
Time-lapse imaging with NANOG reporter systems to track expression dynamics
Quantification Methods:
Mean fluorescence intensity measurements
Nuclear/cytoplasmic signal ratio analysis
Population distribution profiling using histogram analysis
Threshold-based classification into NANOG-high and NANOG-low subpopulations
Statistical Analysis:
Kernel density estimation for population distribution
Clustering algorithms to identify distinct subpopulations
Correlation analysis between NANOG and other pluripotency factors
Temporal analysis of population fluctuations
This methodological framework allows researchers to distinguish between genuinely heterogeneous NANOG expression patterns and technical artifacts. Since NANOG expression often overlaps with but is not identical to Oct4 expression patterns , comparative analysis of these factors provides insight into the complex regulatory networks governing pluripotency states. By quantifying NANOG heterogeneity under different culture conditions or perturbations, researchers can investigate mechanisms controlling pluripotency state transitions and lineage priming.
NANOG has emerged as a potential prognostic marker in multiple cancer types, including non-small cell lung cancer where its overexpression correlates with poor progression-free survival . A robust methodological framework for investigating NANOG in cancer includes:
This comprehensive approach enables reliable assessment of NANOG's prognostic significance across different cancer types. For non-small cell lung cancer specifically, where NANOG overexpression has been associated with poor clinical outcomes , standardized detection and quantification are essential for potential clinical application as a predictive biomarker that could guide treatment decisions.
Interpreting NANOG antibody results during cellular reprogramming requires careful consideration of temporal dynamics and heterogeneous expression patterns:
Temporal Expression Analysis:
NANOG expression typically appears after initial reprogramming factors (Oct4, Sox2, Klf4)
Early NANOG-positive cells may not represent fully reprogrammed iPSCs
Establishment of stable, homogeneous NANOG expression indicates completion of reprogramming
Time-course analysis with multiple time points is essential
Heterogeneity Interpretation:
Early reprogramming cultures show highly heterogeneous NANOG expression
Colonies with homogeneous NANOG expression correlate with successful reprogramming
NANOG-negative cells within positive colonies may indicate incomplete reprogramming
Quantitative analysis of expression level distributions provides insight into reprogramming efficiency
Comparative Marker Analysis:
Compare NANOG with early reprogramming markers (alkaline phosphatase, SSEA-1)
Correlate with late reprogramming markers (Tra-1-60, Tra-1-81)
Co-expression with endogenous Oct4 indicates advanced reprogramming
Analyze epigenetic modifiers that regulate NANOG expression
Functional Correlation:
Correlate NANOG expression patterns with functional pluripotency assays
Test differentiation potential of NANOG-positive subpopulations
Evaluate self-renewal capacity in relation to NANOG expression levels
Consider NANOG expression in the context of transcriptome-wide reprogramming
Technical Considerations:
Distinguish between endogenous and exogenous NANOG expression
Use antibodies that recognize relevant species-specific NANOG epitopes
Apply consistent staining and imaging parameters across time points
Include embryonic stem cell controls for calibrating expression levels
This methodological framework enables accurate interpretation of NANOG expression during the complex process of cellular reprogramming. NANOG serves not only as a marker of pluripotency but also as an active participant in the establishment of the pluripotent state, making its detection crucial for understanding reprogramming mechanisms and efficiency.
When encountering inconsistent NANOG antibody staining, implement this systematic troubleshooting approach:
Antibody Validation Issues:
Confirm antibody specificity using positive controls (BG01V human embryonic stem cells, Tera-2 cells)
Test multiple antibody lots to identify lot-specific inconsistencies
Verify the antibody recognizes the appropriate species (human vs. mouse NANOG)
Consider epitope accessibility in different sample preparation methods
Sample Preparation Variables:
Technical Parameter Optimization:
Titrate antibody concentration across a wider range
Adjust incubation time and temperature
Modify blocking conditions to reduce background
Test different detection systems (HRP vs. fluorescence)
Cell/Tissue-Specific Considerations:
Account for NANOG expression heterogeneity in pluripotent populations
Confirm cell/tissue viability and quality
Consider cell cycle effects on NANOG expression
Evaluate culture conditions that might affect pluripotency status
Quantification and Analysis Adjustment:
Standardize image acquisition parameters
Implement computational image analysis for objective quantification
Use appropriate statistical methods for heterogeneous populations
Establish clear positive/negative thresholds based on controls
Alternative Approaches:
Complement protein detection with mRNA analysis
Use reporter systems for live monitoring of NANOG expression
Apply single-cell techniques to address heterogeneity
Consider alternative antibody clones targeting different epitopes
By systematically investigating these variables, researchers can identify specific factors contributing to inconsistent staining and implement appropriate optimization strategies to achieve reproducible NANOG detection across experiments.
Integrating NANOG antibody detection with transcriptomic and epigenomic analyses requires a coordinated multi-omics approach:
Sequential Analysis Protocol:
Single-Cell Multi-Omics Integration:
Apply single-cell immunostaining for NANOG
Index sorting followed by single-cell RNA-seq
Computational integration of protein and transcriptome data
Trajectory analysis correlating NANOG expression with transcriptional states
ChIP-seq Optimization for NANOG:
Use optimized sonication parameters for 200-500bp fragments
Include appropriate controls (IgG, input chromatin)
Perform motif analysis on NANOG-bound regions
Integrate with histone modification ChIP-seq data
Data Integration Methods:
Correlation analysis between NANOG protein levels and mRNA expression
Identification of genes differentially expressed in NANOG-high vs. NANOG-low populations
Mapping NANOG binding sites to expression changes
Network analysis of NANOG-regulated gene modules
Validation Strategies:
Functional validation of key targets using CRISPR/Cas9
Reporter assays for NANOG-regulated enhancers
Perturbation studies followed by multi-omic profiling
Cross-species comparison of NANOG regulatory networks
This integrated approach provides comprehensive insights into how NANOG protein expression correlates with transcriptional and epigenetic states. By connecting NANOG binding events with gene expression changes and chromatin accessibility, researchers can elucidate the mechanistic basis of NANOG's role in maintaining pluripotency and regulating differentiation. The approach also enables identification of cell state-specific NANOG functions in heterogeneous populations.
Distinguishing between NANOG protein and its pseudogene products (particularly NANOGP8) is critical in cancer research, as pseudogene expression can confound interpretation of results. Implement these methodological approaches:
Antibody-Based Discrimination:
Select antibodies validated against specific NANOG epitopes that differ from pseudogene products
Test antibody specificity using cells expressing only canonical NANOG or pseudogene variants
Perform peptide competition assays with canonical and pseudogene-specific peptides
Use multiple antibodies targeting different epitopes to confirm results
Combined Protein-mRNA Analysis:
Design PCR primers distinguishing between NANOG and pseudogene transcripts
Perform parallel protein detection (Western blot/IHC) and transcript analysis
Correlate antibody staining patterns with transcript-specific expression
Apply in situ hybridization with probes specific to unique regions
Mass Spectrometry Validation:
Immunoprecipitate using NANOG antibodies
Perform mass spectrometry to identify peptide sequences
Analyze peptide sequences that differentiate canonical NANOG from pseudogenes
Quantify relative abundance of canonical vs. pseudogene-derived peptides
Genetic Manipulation Approaches:
Use CRISPR/Cas9 to specifically knock out canonical NANOG vs. pseudogenes
Create overexpression systems for canonical or pseudogene variants
Evaluate antibody reactivity in these genetic models
Assess functional differences between canonical and pseudogene products
Cancer Sample Analysis Strategy:
Include normal tissues as controls for canonical NANOG expression
Compare expression patterns in cancer vs. embryonic tissues
Correlate with other pluripotency markers that lack pseudogenes
Assess cellular localization (canonical NANOG is predominantly nuclear)
When investigating NANOG as a prognostic marker in cancer , these approaches ensure that observed associations with clinical outcomes reflect the protein of interest rather than pseudogene products. This distinction is particularly important given that NANOG and its pseudogenes may have different functional properties and clinical implications in cancer progression.
NANOG antibody applications are rapidly evolving alongside single-cell technologies, creating powerful new methodological approaches for pluripotency research:
Mass Cytometry (CyTOF) Integration:
Metal-conjugated NANOG antibodies enable simultaneous detection with dozens of other markers
Provides high-dimensional characterization of pluripotent states
Allows identification of rare intermediates during differentiation or reprogramming
Requires careful optimization of fixation, permeabilization, and antibody concentration
Single-Cell Protein-Transcriptome Analysis:
CITE-seq compatible NANOG antibodies for simultaneous protein and RNA detection
Correlation of NANOG protein levels with single-cell transcriptomes
Reveals post-transcriptional regulation mechanisms
Identifies cell states where protein and mRNA are discordant
Spatial Multi-Omics Applications:
In situ detection of NANOG in spatial context
Integration with spatial transcriptomics
Preservation of tissue architecture for developmental studies
Mapping NANOG expression domains in embryos or organoids
Live-Cell Single-Molecule Tracking:
Antibody fragments for tracking NANOG dynamics in living cells
Reveals transcription factor binding kinetics
Monitors NANOG mobility in different pluripotent states
Requires specialized antibody derivatives optimized for live-cell applications
Microfluidic Antibody-Based Sorting:
NANOG antibody-based selection of pluripotent populations
Integration with downstream single-cell analysis
Automated platforms for standardized processing
Enables functional testing of NANOG-expressing subpopulations
These emerging applications require continued optimization of NANOG antibodies for compatibility with new technological platforms. As these methods mature, they will provide unprecedented insights into the heterogeneity and dynamics of NANOG expression in pluripotent cells, cancer stem cells, and during development.
The translation of NANOG antibody applications from research to clinical diagnostics faces several challenges but holds significant promise:
Standardization Requirements:
Development of clinical-grade NANOG antibodies with rigorous validation
Establishment of standardized staining protocols for diagnostic pathology
Creation of reference standards for quantification
Implementation of automated staining and analysis platforms
Clinical Validation Challenges:
Technical Implementation Barriers:
Optimization for formalin-fixed paraffin-embedded clinical samples
Development of companion diagnostics for targeted therapies
Quality control measures for multi-center reproducibility
Training requirements for pathology interpretation
Emerging Clinical Applications:
NANOG detection in circulating tumor cells
Liquid biopsy approaches measuring soluble NANOG
Monitoring NANOG expression during treatment
Stratification of patients for clinical trials targeting cancer stem cells
Regulatory Considerations:
Validation requirements for FDA/EMA approval of diagnostic assays
Companion diagnostic development pathways
Laboratory developed test regulations
Integration into existing diagnostic workflows
The potential of NANOG as a prognostic marker in non-small cell lung cancer exemplifies its clinical relevance. Current research indicates that NANOG overexpression is significantly associated with poor progression-free survival, suggesting its utility for identifying patients who might benefit from alternative treatment strategies. As standardization and validation efforts progress, NANOG antibody-based diagnostics may become valuable tools for personalizing cancer treatment approaches.