NANOG Human, His maintains ESC self-renewal by:
Synergizing with OCT4 and SOX2 to activate pluripotency genes (e.g., REX1) .
Forming prion-like condensates to bridge distant genomic loci, enabling chromatin reorganization .
Gene | Role | Reference |
---|---|---|
REX1 | Pluripotency maintenance | |
TAL1 | Inhibits hematopoiesis | |
POU3F1 | Anterior-posterior axis formation |
Reprogramming somatic cells: Overexpression of NANOG enhances induced pluripotent stem cell (iPSC) generation .
Ground-state pluripotency: Resets ESCs to a naive state independent of external signals .
Cancer Type | Role of NANOG | Clinical Implication |
---|---|---|
Glioblastoma | Maintains cancer stem cell (CSC) niche | Poor prognosis, drug resistance |
Colorectal | Promotes metastasis and recurrence | Diagnostic biomarker candidate |
Germ cell | Drives tumor initiation | Target for therapy |
High NANOG expression correlates with advanced-stage cancers and resistance to treatment .
Host systems: Bacterial (e.g., E. coli) or mammalian (e.g., HEK293) systems .
Purification: IMAC followed by size-exclusion chromatography (SEC) for high purity.
High yield: Simplified isolation from complex cellular lysates .
Versatility: Compatible with in vitro assays (e.g., electrophoretic mobility shift assays) .
High expression: Correlates with poor survival in glioblastoma, colorectal, and germ cell cancers .
NANOG p8 isoform: Elevated in CSCs; therapeutic target for reducing malignancy .
Epigenetic regulation: Promoter methylation silences NANOG during differentiation .
Heterogeneity: Subpopulations of ESCs/CSCs exhibit variable NANOG expression .
NANOG functions as a critical transcription factor in maintaining pluripotency and self-renewal in human embryonic stem cells. Methodologically, researchers should approach NANOG function through chromatin immunoprecipitation (ChIP) experiments combined with RNA-seq to identify direct target genes. NANOG's functional significance extends beyond simple maintenance of undifferentiated states, as it operates within complex transcriptional networks including OCT4 and SOX2 . For rigorous assessment of NANOG function, loss-of-function experiments using RNAi or CRISPR/Cas9 should be paired with gain-of-function approaches to establish causality in pluripotency maintenance.
NANOG contains an intact homeobox domain critical for DNA binding and transcriptional regulation . To study domain-specific functions, researchers should employ domain-deletion mutants and point mutations, particularly within the homeobox region. Methodologically, protein crystallography combined with molecular dynamics simulations provides insights into structural conformations that dictate NANOG's binding properties. When examining His-tagged NANOG constructs, researchers must verify that the tag does not interfere with the native protein structure through comparative functional assays of tagged versus untagged protein.
The distribution pattern of NANOG pseudogenes provides compelling evidence for evolutionary relationships among primates. Humans possess ten processed NANOG pseudogenes and one unprocessed pseudogene, while chimpanzees have nine unprocessed and one processed pseudogene . Researchers investigating evolutionary relationships should employ comparative genomics approaches, including:
Multiple sequence alignment of pseudogenes across species
Phylogenetic analysis to determine divergence time
Synteny analysis to examine genomic context conservation
The shared genomic locations of most NANOG pseudogenes between humans and chimpanzees strongly supports common ancestry, while human-specific NANOGP8 represents a post-divergence insertion event . Methodologically, researchers should utilize both maximum likelihood and Bayesian approaches for robust phylogenetic inference when studying these evolutionary patterns.
Distinguishing functional NANOG duplicates from pseudogenes requires multiple complementary approaches:
Transcriptional profiling to detect RNA expression
Examination of open reading frames for premature stop codons
Evolutionary conservation analysis through Ka/Ks ratios
Protein detection using specific antibodies
Functional rescue experiments in NANOG-deficient cells
NANOGP1 exemplifies a functional duplicate with an intact homeobox domain that has retained partial functional conservation with ancestral NANOG . In contrast, many NANOG pseudogenes contain disabling mutations. Methodologically, researchers should employ rigorously validated antibodies that can distinguish between NANOG variants, as demonstrated in studies that identified NANOGP8 expression in cancer cells .
Effective study of NANOG in cancer tissues requires a multi-modal approach:
RNA analysis: RT-PCR with primers that can discriminate between NANOG1 and NANOGP8
Protein detection: Modified immunofluorescence incorporating denaturation steps to reveal NANOG in protein complexes
In situ analysis: Polymer-based immunohistochemistry for tissue-level detection
Antibody validation: Preincubation with recombinant protein and shRNA knockdown controls
When analyzing cancer tissues, researchers should be aware that traditional immunostaining may underestimate NANOG presence due to protein-protein interactions masking epitopes. The modified denaturation protocol developed for NANOG detection revealed substantially more nuclear NANOG in cancer cells than conventional methods .
NANOG's role in tumor development extends beyond mere correlation, as demonstrated through functional knockdown studies. Research approaches should include:
RNAi-mediated knockdown followed by clonogenic assays
Assessment of proliferation, differentiation, and tumor formation capacity
Co-expression analysis with cancer stem cell markers like CD44
Epigenetic regulation studies using HDAC inhibitors like TSA
Studies have demonstrated that NANOG knockdown inhibits long-term clonal growth, reduces proliferation, and alters differentiation in cancer cells . Notably, NANOG expression appears enriched in CD44-positive putative cancer stem/progenitor cell populations . Methodologically, researchers should employ patient-derived xenografts rather than just cell lines to validate findings in more clinically relevant models.
Purification of His-tagged human NANOG requires specialized protocols due to its tendency to form aggregates and its sensitivity to proteolytic degradation:
Expression systems: Insect cell systems often yield better results than bacterial systems for full-length NANOG
Lysis conditions: Use of mild detergents with protease inhibitor cocktails
Purification strategy: Two-step approach with Ni-NTA affinity chromatography followed by size exclusion
Quality control: Assessment of protein integrity through western blotting and mass spectrometry
Researchers should validate that purified His-tagged NANOG retains DNA-binding activity through electrophoretic mobility shift assays (EMSA) with known NANOG response elements. For functional studies, researchers must confirm that the His-tag does not interfere with protein-protein interactions through comparative co-immunoprecipitation experiments.
Distinguishing between NANOG1 and NANOGP8 requires multi-level discrimination strategies:
At RNA level: PCR primers exploiting the 3'-UTR deletion in NANOGP8 (e.g., F2/R2 for NANOG1 vs. F2/R3 for both)
At protein level: Mass spectrometry to identify variant-specific peptides
At genomic level: CRISPR-based targeting of unique flanking sequences
Studies have demonstrated that cancer cells preferentially express NANOGP8 rather than NANOG1, with the latter being silenced via histone deacetylation rather than DNA methylation . Methodologically, researchers should include epigenetic modifier controls (TSA, 5'-aza-2'-deoxycytidine) when analyzing expression patterns in different cellular contexts.
Investigating NANOG's interactome requires sophisticated approaches:
Proximity labeling methods (BioID, APEX)
Cross-linking mass spectrometry to capture transient interactions
Co-immunoprecipitation followed by quantitative proteomics
Live-cell imaging of protein complex formation
Research has indicated that NANOG functions in higher-order protein-protein complexes, necessitating specialized detection methods . Advanced researchers should employ proximity-dependent biotinylation approaches to identify context-specific interactions in pluripotent versus cancer cells, as these may reveal mechanistic differences in NANOG function between these contexts.
Understanding the subfunctionalization of NANOGP1 requires sophisticated comparative approaches:
Single-cell RNA-seq of early embryonic development across great ape species
ChIP-seq for comparative target gene analysis
CRISPR-mediated knockout/knockin studies in species-specific pluripotent stem cells
Interspecies chimera formation to assess developmental competence
NANOGP1 shows high expression restricted to early epiblast cells and naïve-state pluripotent stem cells, suggesting developmental stage-specific functions . While NANOGP1 can induce naïve pluripotency, unlike NANOG, it is not required to maintain the undifferentiated status of human naïve pluripotent cells . This functional divergence represents a classic case of subfunctionalization following gene duplication, providing an excellent model for studying evolutionary developmental biology.
Antibody specificity represents a critical challenge in NANOG research due to its multiple pseudogenes and structural variants:
Validation approaches:
Testing on NANOG knockdown/knockout cells
Peptide competition assays
Comparison across multiple antibodies targeting different epitopes
Western blotting coupled with mass spectrometry validation
Advanced detection protocols:
Researchers should be aware that standard protocols may underdetect NANOG due to epitope masking in protein complexes, necessitating optimized procedures as demonstrated in cancer cell studies .
Addressing NANOG heterogeneity in tumors requires sophisticated analytical approaches:
Single-cell RNA-seq and spatial transcriptomics
Multiplex immunohistochemistry with cancer stem cell markers
Laser capture microdissection of NANOG-positive versus negative regions
Lineage tracing in patient-derived xenograft models
Studies have revealed that NANOG expression is heterogeneous at both inter- and intratumoral levels, with positive cells observed in both luminal and basal-like cells in prostate cancer . Methodologically, researchers should combine multiple detection methods and correlate with clinical outcomes to establish prognostic significance of heterogeneous NANOG expression patterns.
Nanog is a multidomain protein that contains a homeodomain responsible for DNA binding. This homeodomain allows Nanog to regulate the expression of genes involved in maintaining pluripotency and preventing differentiation. Nanog expression counteracts differentiation-promoting signals induced by extrinsic factors such as LIF (Leukemia Inhibitory Factor), Stat3, and BMP (Bone Morphogenetic Protein) .
Recombinant human Nanog with a His tag is produced using recombinant DNA technology. The His tag, a sequence of histidine residues, is added to the N-terminus of the Nanog protein to facilitate purification and detection. This recombinant form is typically expressed in Escherichia coli (E. coli) and purified using conventional chromatographic techniques .
Key Features:
Recombinant Nanog (Human, His Tag) is widely used in research to study the mechanisms of stem cell pluripotency and differentiation. It is also used in various assays to investigate the role of Nanog in cellular processes and to develop potential therapeutic applications targeting stem cell regulation.