NANOG-TAT Human

NANOG-TAT Human Recombinant
Shipped with Ice Packs
In Stock

Description

NANOG Human Recombinant produced in E.Coli is a single, non-glycosylated, Polypeptide chain containing 304 amino acids and c-terminal 13 amino acid TAT peptide having a molecular mass of 36.1kDa.
The NANOG is purified by proprietary chromatographic techniques.

Product Specs

Introduction
NANOG is a homeobox transcription factor composed of multiple domains. Its primary function is to preserve the undifferentiated state of pluripotent stem cells. NANOG expression works against the differentiation-inducing signals produced by external factors like LIF, Stat3, and BMP. Cell differentiation can proceed once NANOG expression is reduced. NANOG expression is regulated by proteins such as the transcription factors Oct4, SOX2, FoxD3, Tcf3, and the tumor suppressor p53.
Description
Recombinant Human NANOG, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 304 amino acids. It includes a C-terminal 13 amino acid TAT peptide and has a molecular weight of 36.1 kDa. The purification of NANOG is carried out using proprietary chromatographic methods.
Physical Appearance
Sterile Filtered solution.
Formulation
The NANOG protein solution is prepared with PBS and 50mM Arginine.
Stability
For optimal storage, the product should be kept at 4°C if the entire vial will be used within 2-4 weeks. For longer storage periods, freezing at -20°C is recommended. The addition of a carrier protein (0.1% HSA or BSA) is suggested for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
The purity is determined to be greater than 95.0% based on SDS-PAGE analysis.
Synonyms
NANOG, Homeobox protein NANOG, Homeobox transcription factor Nanog, hNanog.
Source
Escherichia Coli.
Amino Acid Sequence

VDPACPQSL PCFEASDCKE SSPMPVICGP EENYPSLQMS SAEMPHTETV SPLPSSMDLL IQDSPDSSTS PKGKQPTSAE NSVAKKEDKV PVKKQKTRTV FSSTQLCVLN DRFQRQKYLS LQQMQELSNI LNLSYKQVKT WFQNQRMKSK RWQKNNWPKN SNGVTQKASA PTYPSLYSSY HQGCLVNPTG NLPMWSNQTW NNSTWSNQTQ NIQSWSNHSW NTQTWCTQSW NNQAWNSPFY NCGEESLQSC MQFQPNSPAS DLEAALEAAG EGLNVIQQTT RYFSTPQTMD LFLNYSMNMQ PEDVGGYGRK KRRQRRR

Q&A

What is NANOG-TAT and how does it differ from native NANOG?

NANOG-TAT is a recombinant fusion protein comprising human NANOG protein linked to a 13-residue TAT peptide at the C-terminus . The native NANOG protein functions as a regulatory transcription factor associated with pluripotency maintenance in embryonic stem cells, while its expression is suppressed in differentiated adult tissues . The critical difference lies in the addition of the TAT peptide, which enables the protein to directly penetrate cell membranes through a process called protein transduction . This property allows NANOG-TAT to enter primary and transformed cells without viral vectors or transfection reagents, representing an alternative methodology for introducing transcription factors into cells . The human recombinant NANOG-TAT is synthesized as a 304 amino acid polypeptide with the additional TAT peptide, resulting in a 36.2 kDa protein that retains the biological activity of native NANOG while gaining cell-penetrating capabilities .

What are the critical domains of NANOG and their functional significance?

NANOG protein contains several functionally distinct domains that contribute to its role in pluripotency maintenance. The homeodomain (HD) region serves as the primary DNA-binding domain, directly interacting with target gene promoters such as the OCT4 promoter . Crystallographic studies have revealed specific amino acid residues within the homeodomain that are crucial for DNA recognition and binding affinity . The C-terminal domain of NANOG has been identified as particularly important for protein-protein interactions, including its association with Rad51, which influences DNA damage repair mechanisms in stem cells . The N-terminal domain contributes to protein stability and potentially to transactivation functions. Mutation studies have demonstrated that specific residues, such as L122 in the homeodomain, can significantly enhance DNA binding affinity, protein stability, and biological function when substituted (e.g., L122A mutation) . The functional significance of these domains extends beyond pluripotency maintenance to include effects on cell proliferation, senescence suppression, and DNA damage response modulation, making domain-specific analyses essential for understanding NANOG's multifaceted roles .

What cellular processes does NANOG-TAT influence in different cell types?

NANOG-TAT exerts diverse effects across various cell types, with its influence extending beyond pluripotency regulation. In NIH 3T3 fibroblast cells, NANOG-TAT administration induces three-dimensional growth and formation of cell foci, indicating anchorage-independent growth similar to cellular transformation . This effect is concentration-dependent and reversible upon withdrawal of NANOG-TAT . In primary human adult dermal fibroblasts (MP-hADFs), NANOG-TAT treatment results in increased proliferation rates, morphological changes toward more spindle-like shapes, and reduced cytoplasm-to-nucleus ratios . Notably, NANOG-TAT suppresses senescence-associated β-galactosidase activity in these cells, effectively preventing cellular senescence . In embryonic stem cells, NANOG interacts with DNA damage repair machinery, specifically binding to Rad51 and inhibiting Rad51-promoted DNA damage repair, which maintains the high basal level of γH2AX in ES cells . This interaction is specific to NANOG, as other core stemness factors like Oct4 and Sox2 fail to activate γH2AX or interact with Rad51 . The capacity of NANOG-TAT to modulate these diverse cellular processes makes it a valuable tool for investigating cellular reprogramming, senescence, and DNA damage response mechanisms across different experimental models.

How should NANOG-TAT concentration and exposure time be optimized for experimental designs?

Optimizing NANOG-TAT concentration and exposure time is crucial for achieving desired biological effects while minimizing potential toxicity or off-target effects. Research indicates that NANOG-TAT effects are strictly dependent on both concentration and duration of exposure . For inducing anchorage-independent growth in NIH 3T3 cells, maximal numbers of foci were observed at a concentration of 50 nM NANOG-TAT applied for 5 days . When working with primary human fibroblasts, a concentration of 100 nM NANOG-TAT has been effectively used to enhance proliferation and suppress senescence . To determine optimal conditions for a specific cell type or experimental goal, researchers should implement a dose-response matrix experiment, testing concentrations ranging from 25-200 nM and exposure times from 24 hours to 7 days. Cell viability assays (MTT or ATP-based) should be performed in parallel to identify any concentration-dependent cytotoxicity. For long-term experiments, researchers should consider whether continuous exposure or intermittent treatment would better serve their experimental goals, as evidence suggests that NANOG-TAT effects can be reversible upon withdrawal . Importantly, researchers should validate NANOG-TAT activity in their specific experimental system using immunoblotting to confirm intracellular protein presence and downstream target gene expression analysis to verify functional activity.

What methods are effective for tracking NANOG-TAT cellular uptake and activity?

Tracking NANOG-TAT cellular uptake and activity requires a multi-modal approach to confirm both physical presence and functional activity of the protein. For direct visualization of uptake, researchers can use fluorescently labeled NANOG-TAT (conjugated with fluorophores like FITC or Alexa dyes) combined with confocal microscopy to track the protein's cellular localization over time. Immunocytochemistry using antibodies against either NANOG or the TAT peptide provides an alternative visualization method without protein modification. To quantify uptake kinetics, flow cytometry analysis of cells treated with fluorescently labeled NANOG-TAT at various time points can determine the percentage of positive cells and the intensity of uptake. For functional validation, researchers should assess downstream effects through multiple approaches: qRT-PCR analysis of NANOG target genes (such as OCT4), chromatin immunoprecipitation (ChIP) to confirm binding to known genomic targets, and phenotypic assays appropriate to the cell type (such as proliferation measurements or senescence markers) . Western blot analysis of cell lysates can confirm the presence of intact NANOG-TAT protein and should include time-course experiments to determine protein stability after uptake. Additionally, functional readouts such as suppression of senescence-associated β-galactosidase activity in fibroblasts provide clear evidence of biological activity .

How can NANOG-TAT be used for cellular reprogramming experiments?

NANOG-TAT offers a non-genetic approach to cellular reprogramming that bypasses concerns associated with viral vector integration or prolonged ectopic gene expression. For reprogramming experiments, researchers should implement a sequential protocol that begins with optimization of cell density (typically 1-5 × 10^4 cells/cm²) in appropriate culture vessels. The standard reprogramming cocktail containing NANOG-TAT (100 nM) should be supplemented with other key reprogramming factors – either as TAT-fusion proteins (OCT4-TAT, SOX2-TAT, LIN28-TAT) or through transient transfection approaches . The duration of NANOG-TAT treatment should be determined empirically, but typically ranges from 7-21 days with media changes and fresh protein addition every 48 hours. Culture conditions should be progressively shifted from somatic cell maintenance to stem cell supportive conditions (including appropriate substrate coating, growth factors, and small molecules like vitamin C or valproic acid to enhance reprogramming efficiency). Reprogramming progress should be monitored through morphological changes, pluripotency marker expression (SSEA-4, TRA-1-60), and functional assays such as alkaline phosphatase staining . For enhancing reprogramming efficiency, consider using the NANOG L122A mutant variant, which has demonstrated superior DNA binding affinity and promotes reprogramming into ground-state pluripotency from the primed state . The protein transduction approach allows precise temporal control, enabling researchers to withdraw NANOG-TAT at specific stages to investigate its role in initiation versus maintenance of reprogramming.

How does NANOG-TAT suppress cellular senescence at the molecular level?

NANOG-TAT suppresses cellular senescence through multiple interconnected molecular mechanisms centered on cell cycle regulation. Primary research evidence demonstrates that NANOG transduction into primary human fibroblasts results in complete suppression of senescence-associated β-galactosidase (SA-β-gal) activity, a key biomarker of cellular senescence . At the molecular level, NANOG exerts its anti-senescence effects primarily through downregulation of the cyclin-dependent kinase inhibitor p27KIP1 (also known as CDKN1B) . In synchronized mouse embryonic fibroblasts (MEFs), NANOG-TAT treatment consistently decreased p27KIP1 expression at both mRNA and protein levels, with significant reduction observed as early as 5 hours post-treatment and maintained through 21 hours . This downregulation of p27KIP1 enables cell cycle progression by relieving inhibition of cyclin-CDK complexes that drive the G1/S transition. Additionally, NANOG may influence other senescence regulators, although analysis of p53, p16INK4a, and p21 expression showed no significant changes in response to NANOG-TAT treatment in MEFs . The senescence-suppressing effect translates to remarkable proliferative capacity, with NANOG-TAT-treated human primary fibroblasts achieving approximately 8×10^11 cells after 10 passages from an initial 250,000 cells, compared to only 1.5×10^9 cells in control conditions – representing a >500-fold enhancement in expansion potential .

What is the role of NANOG in DNA damage response regulation?

NANOG plays a previously unrecognized role in DNA damage response regulation through direct interaction with DNA repair machinery. Research has identified NANOG as a novel inhibitor of Rad51, a key protein involved in homologous recombination-mediated DNA repair . Through both in vivo co-immunoprecipitation assays and in vitro pulldown experiments, the C-terminal domain of NANOG was shown to directly interact with Rad51, while the N-terminal and DNA-binding domains did not demonstrate this interaction . This interaction has significant functional consequences, as NANOG overexpression dramatically retards γH2AX removal after DNA damage induction and increases the percentage of DNA in comet tails, indicating impaired DNA repair . The specificity of this effect was confirmed as overexpression of other core stemness factors (Oct4 and Sox2) failed to activate γH2AX or interact with Rad51 . Mechanistically, NANOG impedes the binding of Rad51 to single-stranded DNAs and reduces homologous recombination efficiency . The inhibitory effect on Rad51 is not restricted to embryonic stem cells, as vector-mediated overexpression or direct protein delivery of either full-length NANOG or its C-terminal domain into somatic cancer cells demonstrated strong inhibition of Rad51 activity . This Rad51 inhibition mechanism may contribute to the distinctive genomic instability and DNA damage response characteristics observed in pluripotent stem cells.

How does the L122A mutation enhance NANOG function in pluripotency maintenance?

The L122A mutation in NANOG represents a structure-guided enhancement that significantly improves multiple aspects of NANOG function. Crystallographic analysis of human NANOG homeodomain bound to OCT4 promoter DNA revealed that leucine at position 122 is involved in DNA recognition and is likely functionally important . When this residue was substituted with alanine (L122A), the mutation enhanced DNA binding affinity and increased protein stability . The molecular basis for this enhancement appears to relate to optimized protein-DNA interaction geometry, as the mutation occurs within the DNA-recognition interface of the homeodomain . Functionally, when overexpressed in epiblast stem cells or human induced pluripotent stem cells, the L122A mutant demonstrated enhanced capacity for reprogramming into ground-state pluripotency compared to wild-type NANOG . This mutation represents an example of rational protein engineering based on structural insights, where a single amino acid substitution yields improved functionality across multiple parameters. For researchers investigating pluripotency mechanisms or developing enhanced reprogramming protocols, the L122A variant offers a potentially superior tool compared to wild-type NANOG-TAT, particularly for applications requiring robust and efficient induction of ground-state pluripotency . These findings highlight the value of structure-function studies in optimizing transcription factors for stem cell research applications.

How can researchers leverage NANOG-TAT for expansion of primary human cells?

NANOG-TAT provides a powerful tool for expanding primary human cells while maintaining their characteristics and avoiding senescence. To implement a NANOG-TAT-based expansion protocol, researchers should first establish baseline growth curves and senescence markers for their specific primary cell type under standard conditions . The optimal NANOG-TAT concentration (typically 100 nM for fibroblasts) should be determined through pilot experiments assessing both proliferation enhancement and cell viability . Culture medium should be supplemented with NANOG-TAT every 48-72 hours during medium changes to maintain consistent intracellular protein levels. Cell density should be carefully managed, as NANOG-TAT-treated cells exhibit higher proliferation rates and altered contact inhibition properties; typically, cells should be passaged when reaching 70-80% confluence to prevent overgrowth . For long-term expansion experiments, researchers should implement a comprehensive quality control regimen including periodic assessment of senescence markers (SA-β-gal staining), karyotype analysis to monitor genomic stability, and functional assays specific to the cell type to ensure maintained phenotype . This approach has demonstrated remarkable results with human primary adult dermal fibroblasts, achieving >500-fold greater cell numbers after 10 passages compared to control conditions . The expansion of primary human cells through transient transduction of NANOG protein has broad applications for in vitro expansion of cells with limited proliferative capacity, potentially including primary cells for regenerative medicine applications.

What considerations are important when using NANOG-TAT in cancer cell research?

When employing NANOG-TAT in cancer cell research, several important considerations must be addressed to ensure valid and interpretable results. First, baseline endogenous NANOG expression should be determined in the target cancer cell line, as many cancer cells already express NANOG at variable levels, which may influence their response to exogenous NANOG-TAT . Dose-response experiments are essential, as cancer cells may have altered sensitivity to NANOG-TAT compared to normal cells, potentially requiring adjustment of standard concentrations (50-100 nM) used in other cell types . The reversibility of NANOG-TAT effects should be carefully assessed, as demonstrated in NIH 3T3 cells where foci formation induced by NANOG-TAT was reversed after protein withdrawal . This reversibility has important implications for experimental design, particularly for assessing NANOG's role in cancer cell phenotypes. Researchers should monitor changes in DNA damage response, as NANOG directly interacts with Rad51 to inhibit homologous recombination-mediated DNA repair, which could affect cancer cell sensitivity to DNA-damaging chemotherapeutics or radiation . Comprehensive analysis should include assessment of cell cycle distribution, apoptosis markers, migration/invasion capabilities, and cancer stem cell markers to fully characterize NANOG-TAT effects. The C-terminal domain of NANOG, which mediates Rad51 interaction, may be particularly relevant for cancer studies focused on DNA damage repair mechanisms and could be used independently of the full-length protein in some experimental designs .

What methods can be used to assess NANOG-TAT effects on cell cycle regulation?

Comprehensive assessment of NANOG-TAT effects on cell cycle regulation requires integration of multiple experimental approaches. Flow cytometry-based cell cycle analysis using propidium iodide or DAPI staining should be performed at multiple time points following NANOG-TAT treatment to determine changes in the proportion of cells in G0/G1, S, and G2/M phases . For more precise analysis, researchers can implement cell synchronization protocols (serum starvation for G0, aphidicolin for S phase) prior to NANOG-TAT treatment to track progression through specific cell cycle phases . Quantitative real-time PCR and Western blot analyses should target key cell cycle regulators, particularly cyclin-dependent kinase inhibitors like p27KIP1, which has been specifically implicated in NANOG-mediated cell cycle effects . The following table summarizes key molecular targets for assessment:

Cell Cycle RegulatorFunctionNANOG-TAT Effect
p27KIP1 (CDKN1B)CDK inhibitor, G1/S checkpointDownregulated
Cyclins (D1, E, A)Cell cycle progressionPotential upregulation
CDK2, CDK4, CDK6Cell cycle progressionActivity likely increased
p53Cell cycle arrest, apoptosisNo significant change
p16INK4aCDK inhibitor, senescenceNo significant change
p21 (CDKN1A)CDK inhibitor, p53 targetNo significant change
EdU or BrdU incorporation assays provide direct measurement of DNA synthesis rates and S-phase entry, while time-lapse microscopy can determine actual cell division rates and mitotic duration. For comprehensive mechanistic insight, researchers should perform chromatin immunoprecipitation (ChIP) assays to identify whether NANOG directly regulates cell cycle gene promoters or enhancers. Additionally, phospho-specific antibodies can be used to monitor the phosphorylation status of retinoblastoma protein (Rb), a key indicator of G1/S transition regulation .

What future research directions could advance our understanding of NANOG-TAT applications?

Several promising research directions could significantly advance our understanding of NANOG-TAT applications and mechanisms. Structure-guided protein engineering approaches, exemplified by the L122A mutation, represent a fertile area for developing enhanced NANOG variants with improved functionality for specific applications . High-resolution temporal studies tracking NANOG-TAT effects from immediate (minutes to hours) to long-term (days to weeks) time frames could reveal previously unrecognized dynamic aspects of its action. Investigation of cell type-specific responses to NANOG-TAT across a broader spectrum of primary and transformed cells would help establish predictive frameworks for its application in diverse cellular contexts. Systematic analysis of NANOG-TAT interaction with the cellular proteome through techniques like BioID or proximity labeling could identify novel protein partners beyond the known interactions with pluripotency factors and Rad51 . Development of conditional or inducible NANOG-TAT variants would enable more precise temporal control in experimental applications, potentially through incorporation of photocleavable linkers or chemically-responsive elements. Comparison studies between wild-type NANOG-TAT and domain-specific variants could help dissect the contribution of different protein regions to specific cellular outcomes, particularly the C-terminal domain's role in Rad51 inhibition . Finally, exploration of NANOG-TAT effects on cellular metabolism, epigenetic landscapes, and three-dimensional genome organization could reveal broader regulatory roles beyond the currently established functions in pluripotency, proliferation, and DNA damage response .

Product Science Overview

NANOG: The Transcription Factor

NANOG is a homeobox transcription factor that plays a crucial role in the self-renewal of embryonic stem cells and the maintenance of pluripotency . It works in concert with other key transcription factors such as OCT4 and SOX2 to prevent differentiation and maintain the stem cell state . NANOG is essential during the early stages of embryogenesis and is involved in the formation of the epiblast, a structure that gives rise to the embryo proper .

TAT Peptide: A Delivery Vehicle

The TAT peptide, derived from the HIV-1 virus, is known for its ability to transduce across cell membranes. This property makes it an effective delivery vehicle for various molecules, including proteins, into cells . When fused with NANOG, the TAT peptide facilitates the efficient delivery of the NANOG protein into cells, enhancing its biological activity and potential therapeutic applications .

Recombinant Production

Recombinant human NANOG-TAT is typically produced in Escherichia coli (E. coli) expression systems. The protein is synthesized as a single chain, non-glycosylated polypeptide consisting of 318 amino acids . The molecular mass of NANOG-TAT is approximately 36.2 kDa, as determined by SDS-PAGE analysis . The production process involves the fusion of the NANOG cDNA sequence with the TAT peptide sequence, followed by purification using chromatographic techniques .

Biological Activity and Applications

NANOG-TAT retains the biological activity of the native NANOG protein, making it a valuable tool for research in stem cell biology and regenerative medicine . It has been shown to enhance cell migration and downregulate the expression of certain cell cycle inhibitors, such as p27, in cancer cells . This highlights its potential role as a therapeutic target for various malignancies, including cervical cancer .

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 2024 Thebiotek. All Rights Reserved.