NANOG Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Overview of NANOG Protein

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 .

Key NANOG Monoclonal Antibody Clones

The table below compares widely used NANOG monoclonal antibodies:

CloneHostIsotypeTarget EpitopeApplicationsKey Features
eBioMLC-51MouseIgG1Mouse NanogWB, ICCDetects ~45 kDa band in F9 cells; requires Foxp3 Staining Buffer for ICC .
H-2MouseIgG2aC-terminus (AA 151-305)WB, IP, IF, ELISACross-reactive with mouse, rat, and human NANOG .
hNanog.2MouseIgG1Human NanogWB, IHC, ICCValidated in NTERA cell lysates and FFPE tissues .
P5A10ATMouseIgG2aHuman NANOG (AA 1-154)WB, IHCPurified via protein-G; used in cancer studies .
67255-1-IgMouseIgG2aFull-length human NanogWB, IF, FCDetects both native (~39 kDa) and modified (~48 kDa) isoforms .

Stem Cell Biology

  • 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 .

Cancer Research

  • 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 .

Technical Considerations

  • 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 .

Clinical Implications

NANOG monoclonal antibodies are pivotal in:

  • Diagnostics: Identifying NANOG as a biomarker in germ cell tumors and squamous cell carcinomas .

  • Therapeutic Development: Targeting NANOG to overcome resistance in antibody-based cancer therapies .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

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.

Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. For specific delivery time, please consult your local distributors.
Synonyms
Embryonic stem cell specific homeobox protein (Nanog) antibody; ENK antibody; FLJ12581 antibody; hNanog antibody; Homeobox protein NANOG antibody; Homeobox transcription factor Nanog antibody; homeobox transcription factor Nanog-delta 48 antibody; NANOG antibody; Nanog homeobox antibody; NANOG_HUMAN antibody
Uniprot No.

Target Background

Function
NANOG is a transcription regulator critically involved in the proliferation and self-renewal of inner cell mass and embryonic stem (ES) cells. It plays a crucial role in imposing pluripotency on ES cells, preventing their differentiation towards extraembryonic endoderm and trophectoderm lineages. NANOG blocks bone morphogenetic protein-induced mesoderm differentiation of ES cells by physically interacting with SMAD1 and interfering with the recruitment of coactivators to the active SMAD transcriptional complexes. It functions as both a transcriptional activator and repressor, exhibiting optimal binding to the DNA consensus sequences 5'-TAAT[GT][GT]-3' or 5'-[CG][GA][CG]C[GC]ATTAN[GC]-3'. NANOG binds to the POU5F1/OCT4 promoter and can autorepress its own expression in differentiating ES cells. This autorepression occurs through binding to its promoter following interaction with ZNF281/ZFP281, leading to the recruitment of the NuRD complex and subsequent repression of expression. When overexpressed, NANOG promotes cells to enter the S phase and enhance proliferation.
Gene References Into Functions
  1. The critical roles of NANOG and its pseudogene NANOGP8 in cancer progression highlight the significance of their association with exosomes, potentially allowing exosomal NANOG to serve as a powerful diagnostic biomarker. Variations in NANOG/NANOGP8 gene sequences in exosomal DNA include an insertion into the 3' UTR. PMID: 29787607
  2. Elevated expression of the pluripotency factor NANOG. PMID: 29845283
  3. The interaction of Nanog with the AR signaling axis might contribute to the regulation of Ovarian cancer stem cells. Additionally, androgen may promote stemness characteristics in ovarian cancer cells by activating the Nanog promoter. PMID: 29716628
  4. Research indicates a correlation between Nanog expression, HBsAg, differentiation, and TNM stage in hepatocellular carcinoma (HCC). Nanog may serve as an unfavorable prognostic biomarker for HCC. PMID: 29198990
  5. NANOG could be a potential biomarker for the early diagnosis of urothelial carcinoma of the bladder. PMID: 29279584
  6. These findings demonstrate that analysis of IHC expression patterns of MK and NANOG in pretreatment biopsy specimens can provide a more definitive prognosis prediction for each oral squamous cell carcinoma (OSCC) patient, aiding clinicians in developing more precise individualized treatment modalities. PMID: 29113102
  7. Chicken egg-white extracts promote OCT4 and NANOG expression and telomeres growth in 293T cells. PMID: 28838341
  8. Rectal tumor tissue OCT4 (p<0.001), SOX2 (p=0.003), and NANOG (p<0.001) expressions were found to be higher than those in adjacent tissue. PMID: 29214774
  9. Data show that lung adenocarcinoma SPC-A1 cells differentiated by a two-stage induction (TSI) method lost stem cell characteristics, including absent expression of OCT4 and Nanog. PMID: 27588392
  10. Our data suggest that 3D culture increases the expression of Nanog through the relaxation of actin cytoskeleton, which mediates reduced Suv39h1 and H3K9me3 levels. PMID: 28276635
  11. Results show that NANOG could reverse the effects of stem cell senescence and restore the myogenic differentiation potential of senescent MSCs. PMID: 28125933
  12. MACC1-induced tumor progression in colorectal cancer acts, at least in part, via the newly discovered MACC1/Nanog/Oct4 axis. PMID: 26758557
  13. RNAi-mediated silencing of NANOG in SKOV-3 reversed the expression of mesenchymal cell markers and restored expression of E-cadherin. Susceptibility to cisplatin increased in SKOV-3 cells upon down-regulation of NANOG, and reversible results were obtained in Moody cells post-overexpression of NANOG. PMID: 27884977
  14. NANOG enabled reactivation of the ROCK and Transforming Growth Factor (TGF)-beta pathways—both of which were impaired in senescent cells—leading to ACTIN polymerization, MRTF-A translocation into the nucleus, and serum response factor (SRF)-dependent myogenic gene expression. PMID: 27350449
  15. High NANOG expression is associated with brain neoplasms. PMID: 28933914
  16. Super-enhancers at the Nanog locus differentially regulate neighboring pluripotency-associated genes, particularly DPPA3. PMID: 27681417
  17. Our data reveal that SATB2 in alveolar bone mesenchymal stem cells (AB-BMSCs) associates with their age-related properties, and prevents AB-BMSCs senescence via maintaining Nanog expression. PMID: 27632702
  18. High NANOG expression is associated with Multidrug Resistance in breast and cervical cancer. PMID: 28716899
  19. miR-612 has a suppressive role on hepatocellular carcinoma cell stemness via Sp1/Nanog signaling pathway. PMID: 27685621
  20. These data reveal an overexpression of NANOG and other markers of pluripotency and stemness in meningiomas. PMID: 28345785
  21. The NANOG deficiency affected multiple genes, particularly, supressed drug-resistance via down-regulated ABCG2 in Eca109 cells, and caused G1 arrest by down-regulated cyclin D1 (CCND1) expression. PMID: 28601640
  22. Endogenous Plastic Somatic (ePS) cells in a latent state, i.e. lacking SOX2, OCT3/4 and NANOG (SON) expression, in non-diseased breast specimens through immunohistochemical analysis of previously identified ePS-specific biomarkers (CD73(+), EpCAM(+) and CD90(-)). PMID: 27705752
  23. Data suggest that C-terminal truncated hepatitis B virus X protein (HBx-DeltaC1) enhances liver cancer stem cell (CSC) properties through Stat3/Nanog cascade, providing insight for the therapeutic intervention for hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC). PMID: 28186991
  24. Nanog directly repressed transcription of the miR-200c and miR-200b genes in colon cancer cells, inducing epithelial-mesenchymal transformation. PMID: 28163188
  25. The LGR5-expressing fraction of CD54+ cells represents gastric cancer CSCs, in which LGR5 is closely associated with stemness and EMT core genes. PMID: 28033430
  26. Data show that Nanog homebox (NANOG) but not sex-determining region Y-box2 (SOX2) and octamer-binding protein 4 (OCT4) expression was overexpressed in the endometrium of women with intrauterine adhesion (IUA). PMID: 28253866
  27. The NANOG transcription was significantly upregulated by ETV4 overexpression. PMID: 28412366
  28. Collectively, these findings demonstrate a novel role of YBX1 in maintaining the stemness of CSCs and metastasis, unveiling YBX1 as a promising therapeutic target for NSCLC treatments. PMID: 28400280
  29. The early response of pluripotency genes OCT4 and NANOG to the differentiation-inducing stimuli is mediated by dynamic changes in chromatin marks, while DNA methylation is acquired in the later stages of neurogenesis. PMID: 28601081
  30. USP21 specifically regulates the Lys48-linked polyubiquitination and stability of NANOG. PMID: 27956178
  31. To our knowledge, this is the first report on lineage reprogramming of TILs using protein stem cell transcription factors delivered directly to the nucleus. PMID: 27084449
  32. Nanog expression is a prognostic biomarker for triple-negative breast cancer. PMID: 27300169
  33. Stat3 was correlated with NANOG-mediated EMT. PMID: 26801672
  34. miR-760 was proved to be functionally associated with NANOG via regulating its expression. PMID: 27133070
  35. SIRT-1 and NANOG are highly correlated biological markers for diagnosis and prognosis follow-up in patients with adenocarcinoma. PMID: 27540973
  36. Renal cell carcinoma patients with low Nanog and Oct4 expressions in tumor tissues had significantly higher survival rates (p < 0.05). High Nanog and Oct4 expressions may be potential therapeutic targets. PMID: 26631537
  37. ANOG was regulated by the extracellular IGF signaling pathway via STAT3 phosphorylation in colorectal cancer (CRC). This coincides with the observation that IGF receptor IGF-1R is often increasingly expressed in malignant metastatic colon cancer. Taken together, our data define the crucial functions of the IGF/STAT3/NANOG/Slug signaling axis in the progression of CRC. PMID: 26840943
  38. Nanog is a positive regulator of cervical cancer dedifferentiation. PMID: 26936116
  39. Data show that long intergenic non-protein coding RNA ROR may act as a competitive endogenous RNAs (ceRNAs), effectively becoming a sink for microRBA miR-145, thereby activating the derepression of core transcription factors Nanog. PMID: 26636540
  40. ALKBH5 overexpression decreased NANOG mRNA methylation, increased NANOG levels, and increased the percentage of BCSCs, phenocopying the effect of hypoxia. PMID: 27001847
  41. Expression levels of OCT4, SOX2, and NANOG were evaluated by immunohistochemistry with tissue microarray slides of 436 OSCC, 362 corresponding tumor-adjacent normal (CTAN) tissues, and 71 normal uvula epithelium tissues. PMID: 26211876
  42. Nanog possesses an important function in BCSC. PMID: 26339994
  43. Our study provides new insights into the function of DPPA5 and NANOG regulation in human pluripotent stem cells. PMID: 26661329
  44. Available data demonstrate that NANOG is strictly involved in the process of carcinogenesis and is a potential prognostic marker for malignant tumors. PMID: 26618281
  45. The disruption of Nanog expression results in less proliferation, invasiveness, migration, more chemosensitivity, and reversal of EMT in HepG2 cells, indicating that Nanog plays crucial roles in influencing the malignant phenotype of HepG2 cells. PMID: 26676719
  46. The promoter activity of Nanog and Oct4 was upregulated, and beta-catenin was observed to bind to these promoters during H. pylori infection, while a Wnt/beta-catenin inhibitor suppressed promoter activity and binding. PMID: 26940070
  47. Ectopic expression of the Oct-4 gene in hAFMSCs with high self-renewal ability could upregulate Nanog and Sox-2 gene expression. PMID: 25385323
  48. Nanog is an oncogene with multiple roles in promoting tumorigenesis and metastasis. PMID: 26073077
  49. The positive correlation among Oct-4, Nanog, and beta-catenin suggests their coordinated role in maintaining proliferation in oral carcinoma cells. PMID: 24700702
  50. Oct3/4 and Nanog represent probable CSC markers in HNSCC, which contribute to the development of DNM in part by enhancing cell motility and invasiveness. PMID: 26483189

Show More

Hide All

Database Links

HGNC: 20857

OMIM: 607937

KEGG: hsa:79923

STRING: 9606.ENSP00000229307

UniGene: Hs.635882

Protein Families
Nanog homeobox family
Subcellular Location
Nucleus.
Tissue Specificity
Expressed in testicular carcinoma and derived germ cell tumors (at protein level). Expressed in fetal gonads, ovary and testis. Also expressed in ovary teratocarcinoma cell line and testicular embryonic carcinoma. Not expressed in many somatic organs and

Q&A

What is NANOG and why is it a critical target for monoclonal antibodies in stem cell research?

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.

How do mouse anti-human and mouse anti-mouse NANOG monoclonal antibodies differ in applications and specificity?

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.

What are the optimal storage and handling procedures for maintaining NANOG monoclonal antibody functionality?

For optimal functionality of NANOG monoclonal antibodies, specific storage and handling procedures should be followed:

Storage DurationRecommended TemperatureAdditional Considerations
Up to 1 month4°CMaintain original formulation
Longer periods-20°CAvoid freeze-thaw cycles
Shelf life12 months at -20°C, 1 month at 4°CMonitor 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.

How should researchers optimize NANOG antibody concentrations for different detection methods?

Optimizing NANOG antibody concentrations requires method-specific titration based on established starting points:

Detection MethodRecommended Starting ConcentrationOptimization ApproachExpected Results
Western Blot1:500-1:1,000 or 1 μg/mL Serial dilution series~40-45 kDa band
Immunohistochemistry1:50 or 1-15 μg/mL Titration across tissue typesNuclear staining in pluripotent cells
Immunocytochemistry5-15 μg/mL Cell type-specific optimizationNuclear localization
ChIP5 μg per 5×10^6 cells Input chromatin titrationEnrichment of NANOG targets

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.

What controls are essential for validating NANOG antibody specificity in pluripotency research?

A comprehensive control strategy is essential for validating NANOG antibody specificity:

  • Positive Cellular Controls:

    • BG01V human embryonic stem cells (validated for both human and mouse NANOG antibodies)

    • Tera-2 human embryonic lung carcinoma cell line (strong NANOG expression)

    • F9 embryonal carcinoma cells (mouse NANOG expression)

  • Negative Cellular Controls:

    • NIH3T3 fibroblasts (confirmed NANOG-negative)

    • Fully differentiated somatic cells

    • Differentiated derivatives of ESCs (showing downregulation)

  • Tissue Controls:

    • Testicular cancer sections (positive control)

    • Normal testis (positive in specific cell populations)

    • 13 d.p.c. mouse embryos (developmental control)

    • Adult somatic tissues (negative 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.

What is the optimal protocol for using NANOG antibodies in chromatin immunoprecipitation (ChIP) experiments?

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:

    • Use 5 μg NANOG antibody per 5×10^6 cells

    • 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.

How can NANOG antibodies be employed to investigate heterogeneity in pluripotent stem cell populations?

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.

What approaches should be used to investigate NANOG expression in cancer tissues for prognostic applications?

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.

How should researchers interpret NANOG antibody results when investigating cellular reprogramming?

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.

What are the most effective troubleshooting strategies for inconsistent NANOG antibody staining?

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:

    • Optimize fixation conditions (duration, fixative concentration)

    • Test multiple antigen retrieval methods (high vs. low pH)

    • Ensure consistent cell culture conditions for stem cells

    • Minimize time between tissue collection and fixation

  • 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.

How can researchers integrate NANOG antibody-based detection with transcriptomic and epigenomic analyses?

Integrating NANOG antibody detection with transcriptomic and epigenomic analyses requires a coordinated multi-omics approach:

  • Sequential Analysis Protocol:

    • Divide cell populations based on NANOG antibody staining (flow sorting)

    • Process NANOG-high and NANOG-low populations for RNA-seq and epigenomic profiling

    • Perform ChIP-seq using NANOG antibodies (5 μg per 5×10^6 cells)

    • Integrate with ATAC-seq to identify accessible chromatin regions

  • 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.

What methodological approaches can distinguish between NANOG protein and its pseudogene products in cancer research?

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.

How are NANOG antibody applications evolving with advances in single-cell technologies?

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.

What are the major challenges and future research directions for NANOG antibody applications in clinical diagnostics?

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:

    • Large-scale studies correlating NANOG expression with patient outcomes

    • Determination of clinically relevant cutoff values for "overexpression"

    • Integration with established diagnostic markers

    • Prospective studies evaluating predictive value for treatment response

  • 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.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.