NTRK2 Human, HEK refers to a recombinant human neurotrophic receptor tyrosine kinase 2 (NTRK2/TrkB) protein produced in human embryonic kidney (HEK293) cells. This protein is a high-affinity receptor for brain-derived neurotrophic factor (BDNF) and neurotrophin-4 (NT-4), playing critical roles in neuronal survival, synaptic plasticity, and nervous system development . Dysregulation of NTRK2 is associated with neurological disorders, cancers, and developmental conditions .
BDNF/TrkB Pathway: Activates downstream pathways (e.g., MAPK/ERK, PI3K/Akt) to regulate synaptic plasticity, neuronal differentiation, and survival .
Neurological Disorders: Loss-of-function mutations in NTRK2 are linked to epilepsy, autism, and neurodevelopmental delays . Hyperactivation due to oncogenic mutations drives tumorigenesis in leukemia and gliomas .
TrkB.T1: A truncated isoform lacking the kinase domain, implicated in glioma progression via altered cell migration and survival .
Full-Length TrkB: Critical for hippocampal long-term potentiation (LTP) and memory formation .
CRISPR/Cas9 Knockout: NTRK2–/– human neural progenitor cells showed reduced expression of neurogenic genes (e.g., NEUROD1, TUBB3) and upregulated glial markers (e.g., PDGFRA, DAB2), indicating TrkB’s role in suppressing gliogenesis .
Synaptic Plasticity: BDNF-NTRK2 signaling enhances synaptic strength in hippocampal neurons, validated using HEK293-derived recombinant protein .
Leukemia-Associated Mutations:
Entrectinib: Suppresses oncogenic TrkB signaling in leukemia models with A203T/R458G mutations .
Valproic Acid: Restores mTOR activity in NTRK2 T720I mutants, reversing synaptic deficits .
Recombinant human NTRK2 is a single polypeptide chain with glycosylation. It encompasses amino acids 32 to 430, resulting in a total of 632 amino acids. The molecular mass is 70.3 kDa. This protein is fused to a 233 amino acid hIgG-His-Tag at its C-terminus. Purification is achieved through proprietary chromatographic methods.
A colorless solution that has been sterilized by filtration.
The NTRK2 solution is provided at a concentration of 1 mg/ml. It is formulated in a solution of Phosphate-Buffered Saline (pH 7.4) containing 10% Glycerol.
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it should be frozen at -20°C. To ensure stability during long-term storage, the addition of a carrier protein (0.1% HSA or BSA) is recommended. Repeated freezing and thawing should be avoided.
The purity of this product, as determined by SDS-PAGE, is greater than 90%.
GP145-TrkB, trk-B, TRKB, NTRK2.
HEK293 Cells.
CPTSCKCSAS RIWCSDPSPG IVAFPRLEPN SVDPENITEI FIANQKRLEI INEDDVEAYV GLRNLTIVDS GLKFVAHKAF LKNSNLQHIN FTRNKLTSLS RKHFRHLDLS ELILVGNPFT CSCDIMWIKT LQEAKSSPDT QDLYCLNESS KNIPLANLQI PNCGLPSANL AAPNLTVEEG KSITLSCSVA GDPVPNMYWD VGNLVSKHMN ETSHTQGSLR ITNISSDDSG KQISCVAENL VGEDQDSVNL TVHFAPTITF LESPTSDHHW CIPFTVKGNP KPALQWFYNG AILNESKYIC TKIHVTNHTE YHGCLQLDNP THMNNGDYTL IAKNEYGKDE KQISAHFMGW PGIDDGANPN YPDVIYEDYG TAANDIGDTT NRSNEIPSTD VTDKTGREH LEPKSCDKTHT CPPCPAPELL GGPSVFLFPP KPKDTLMISR TPEVTCVVVD VSHEDPEVKF NWYVDGVEVH NAKTKPREEQ YNSTYRVVSV LTVLHQDWLN GKEYKCKVSN KALPAPIEKT ISKAKGQPRE PQVYTLPPSR DELTKNQVSL TCLVKGFYPS DIAVEWESNG QPENNYKTTP PVLDSDGSFF LYSKLTVDKS RWQQGNVFSC SVMHEALHNH YTQKSLSLSP GK.
NTRK2 (Neurotrophic Tyrosine Kinase Receptor Type 2) is a member of the trk family of tyrosine protein kinases that encode receptors for nerve growth factor-related proteins known as neurotrophins. The human NTRK2 gene has been mapped to chromosome 9, specifically near D9S1 on the proximal long arm of human chromosome 9q22 . The gene spans across approximately 355 kilobases and contains multiple exons that code for several isoforms of the TrkB receptor . Understanding this genomic organization is critical when designing genetic experiments involving NTRK2, particularly when considering potential genetic variants that might affect receptor function.
For controlled expression of NTRK2 in HEK293 cells, researchers commonly use doxycycline-inducible lentiviral expression systems. These systems typically employ vectors like pCW that express a puromycin resistance gene cassette under the control of the ubiquitous hPGK1 promoter . The NTRK2 constructs can include N-terminal HA-tags positioned between the signal peptide and the first amino acid of the mature TrkB receptor to facilitate detection . After transduction with lentiviral particles packaged in HEK293TN producer cells, stable cell lines are selected using puromycin (typically at 1-2 μg/ml). Expression of TrkB constructs is then induced by treating cells with doxycycline (1 mg/ml), providing temporal control over receptor expression .
Genetic variants within the NTRK2 locus have been associated with various phenotypes, including suicide attempts in depressed patients. A study examining 65 tagging single-nucleotide polymorphisms (SNPs) throughout the NTRK2 locus identified several risk alleles that showed association with suicide attempt among depressed patients . The genetic information content of NTRK2 can be clustered into 16 D′-based haplotype blocks, with certain SNPs showing significant interaction effects . When studying NTRK2 function in cellular models, researchers should consider how these genetic variants might influence receptor activity, signaling pathways, and cellular responses. This is particularly important when translating findings from cellular models to clinical applications.
NTRK2 fusion proteins that lack extracellular domains can become constitutively active through two main mechanisms. First, dimerization and transactivation may be forced by the fusion domain partner . Alternatively, abundance-dependent kinase domains may undergo autoactivation and become constitutively active, similar to their behavior in vitro . Research in HEK293 cells has demonstrated that high intracellular abundance is sufficient for neurotrophin-independent, constitutive activation of TrkB kinase domains. This finding helps explain how NTRK fusion proteins can function as oncogenes despite lacking protein domains traditionally considered essential for receptor activation. To investigate these mechanisms, researchers can employ site-directed mutagenesis of key residues (such as Y705 in the kinase domain) to block the cellular effects of these fusion proteins .
In HEK293 cells, both constitutively active TrkB kinase and the NTRK2-fusion oncogene SQSTM1-NTRK2 induce atypical cellular responses, including reduced actin filopodia dynamics, phosphorylation of focal adhesion kinase (FAK), and altered cell morphology . Importantly, these atypical cellular responses can be mimicked with just the intracellular kinase domain, which does not activate the traditional Trk-associated MAPK/ERK pathway . In glioblastoma-like U87MG cells, expression of TrkB or SQSTM1-NTRK2 reduces cell motility and causes dramatic transcriptome changes . These findings suggest that NTRK2 fusion proteins may drive oncogenesis through mechanisms distinct from those of ligand-activated native receptors. Researchers investigating these differences should employ comprehensive phenotypic assays, including cell morphology analysis, migration assays, and transcriptome profiling.
Expression of TrkB or SQSTM1-NTRK2 in glioblastoma-like U87MG cells causes drastic changes in the cellular transcriptome . To characterize these changes, researchers should employ RNA sequencing (RNA-seq) technology to provide a comprehensive view of differential gene expression. Comparison between cells expressing native TrkB, fusion proteins, and kinase-dead mutants can help identify genes specifically regulated by constitutive TrkB kinase activity. Additionally, pharmacological inhibition with clinically approved Trk inhibitors can confirm the specificity of observed transcriptome changes . Bioinformatic analysis should include pathway enrichment analysis to identify biological processes affected by NTRK2 fusion proteins, providing insights into their oncogenic mechanisms.
For studying NTRK2 effects on cell migration, researchers typically use a wound healing assay with silicone inserts. U87MG cells expressing TrkB constructs can be seeded at a density of 20,000 cells per well into a 2-well silicone insert positioned in a 35 mm μ-dish . After allowing cells to attach, doxycycline (1 mg/ml) is added to induce expression of the corresponding TrkB-related constructs. After 24 hours of induction, the silicone insert is removed, and cell migration into the gap is monitored using brightfield microscopy immediately after removal and at various timepoints thereafter . This approach allows quantitative assessment of migration rates and can reveal how different NTRK2 constructs affect cellular motility. When analyzing results, researchers should consider both the rate and pattern of migration, as these can provide insights into the underlying mechanisms.
Immunohistochemistry of NTRK-positive tumor samples has revealed that NTRK fusion proteins often localize to different cellular compartments than native Trk receptors . To study this differential localization, researchers should employ immunofluorescence microscopy with antibodies specific to the TrkB kinase domain or epitope tags incorporated into the fusion constructs. For optimal visualization, cells should be seeded on glass coverslips at a density of approximately 50,000 cells per 10 mm coverslip . After inducing expression with doxycycline, cells can be fixed and immunolabeled to determine subcellular localization. Co-staining with markers for specific cellular compartments (ER, Golgi, endosomes, etc.) can help identify the precise localization of fusion proteins and provide insights into their trafficking and signaling mechanisms.
When investigating NTRK2 kinase activity, several controls are critical for proper interpretation of results. First, kinase-deficient mutants generated by site-directed mutagenesis should be included to distinguish between kinase-dependent and -independent effects . Mutation of the Y705 residue in the kinase domain is particularly useful, as it blocks cellular effects and transcriptome changes induced by TrkB or SQSTM1-NTRK2 . Second, pharmacological inhibition with clinically approved Trk inhibitors provides an orthogonal approach to validate kinase-dependent effects. Third, comparison between ligand-induced activation of full-length receptors and constitutive activation of fusion proteins can reveal pathway-specific differences. Finally, time-course experiments are essential to distinguish between immediate and delayed responses, particularly when analyzing phosphorylation events and transcriptional changes.
Proximity labeling approaches like BioID or the newer UltraID technique offer powerful tools for identifying NTRK2 interacting proteins. UltraID provides superior labeling efficiency compared to earlier methods and can efficiently label proximal interactors in as little as 10 minutes . This rapid labeling capability allows researchers to detect interactors only at the time of pervanadate-induced activation of NTRK receptors, rather than over a preceding 24-hour period . When analyzing proximity labeling data, researchers should focus on proteins that are only seen in pervanadate-treated samples or whose bait-normalized spectral count value is significantly higher in treated samples. This approach has successfully identified key RTK-activated signaling proteins like PLCG1 and GRB2, as well as regulatory phosphatases like PTPN1 and PTPN11 .
Research on NTRK2 fusion proteins has significant implications for cancer therapy. Clinically approved Trk inhibitors have been shown to block the cellular effects and transcriptome changes induced by TrkB or SQSTM1-NTRK2 expression . This suggests that patients with tumors harboring NTRK2 fusions might benefit from targeted therapy with these inhibitors. Furthermore, understanding the atypical signaling mechanisms of NTRK2 fusion proteins may reveal novel therapeutic targets for cancers resistant to existing Trk inhibitors. Future research should focus on identifying biomarkers that predict response to Trk inhibitors and developing combination therapies that address resistance mechanisms. Additionally, the underglycosylated, atypical phospho-active Trk kinase signals observed in glioblastoma biopsies warrant further investigation as potential diagnostic markers .
The development of advanced proximity labeling technologies like UltraID represents a significant opportunity for deeper understanding of NTRK2 signaling dynamics. These techniques can capture transient interactions that occur specifically during receptor activation, providing insights into the temporal sequence of signaling events . Future applications might include comparing the interactomes of native NTRK2 receptors with fusion proteins to identify shared and distinct signaling partners. Similarly, comparing the interactomes of different NTRK family members (TrkA, TrkB, and TrkC) could reveal common mechanisms and receptor-specific pathways. Integration of proximity labeling data with phosphoproteomic analysis would provide a more comprehensive view of how NTRK2 activation reshapes cellular signaling networks, potentially identifying new therapeutic targets for NTRK2-driven diseases.
Neurotrophic Receptor Tyrosine Kinase 2 (NTRK2), also known as tropomyosin-related kinase B (TrkB), is a member of the neurotrophic tyrosine receptor kinase (NTRK) family. This kinase is a membrane-bound receptor that plays a crucial role in the development and function of the nervous system. The recombinant form of NTRK2, expressed in Human Embryonic Kidney (HEK) 293 cells, is widely used in research to study its biological functions and therapeutic potential.
NTRK2 is activated by binding to neurotrophins such as brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and neurotrophin-4/5 (NT-4/5) . Upon activation, NTRK2 undergoes autophosphorylation and initiates several downstream signaling pathways, including the MAPK pathway, leading to various cellular responses such as:
Abnormal expression of the NTRK2 gene is associated with several cancers, including colorectal, breast, papillary thyroid cancer, lung adenocarcinoma, and oral squamous cell carcinoma . Due to its role in cancer and nervous system development, NTRK2 is a target for therapeutic interventions and drug development.
The recombinant form of NTRK2 is expressed in HEK 293 cells, a widely used cell line derived from human embryonic kidney cells. This recombinant protein is typically tagged with a 6-His tag at the C-terminus and is purified to a high degree of purity (≥95% as determined by SDS-PAGE) . The recombinant NTRK2 protein is used in various biochemical and physiological studies to understand its function and to develop potential therapeutic agents.