The NTRK2 Recombinant Monoclonal Antibody is a laboratory-generated antibody that specifically binds to TrkB, a transmembrane tyrosine kinase receptor activated by brain-derived neurotrophic factor (BDNF) and neurotrophin-4 (NT-4). TrkB regulates neuronal development and survival through signaling pathways like MAPK/ERK, PI3K-AKT, and PLCγ . Recombinant monoclonal antibodies are produced using in vitro cloning techniques to ensure high specificity and batch-to-batch consistency .
These antibodies are synthesized using recombinant DNA technology, enabling precise engineering of antigen-binding sites. Key features include:
Western Blotting: Detects TrkB isoforms (TrkB-FL: ~145 kDa; TrkB-T1: ~90 kDa) in human neural cells and brain tissues .
ELISA/Cytometric Bead Array: Quantifies TrkB levels with sensitivity ranges of 0.313–250 ng/mL .
Immunofluorescence: Localizes TrkB in neurons and glial cells (e.g., SH-SY5Y cells) .
Validates TrkB’s role in neurogenesis vs. gliogenesis using CRISPR/Cas9-edited NTRK2–/– cells .
Blocks caspase-3 activation in hypoxic-ischemic injury models (e.g., 29D7 antibody) .
Identifies TrkB isoforms and their kinase activity in signaling pathways .
Genetic knockout of NTRK2 in ReNcell VM cells revealed:
Downregulation: Neurogenic transcription factors (NHLH1, NEUROD1) .
Mechanism: TrkB suppresses gliogenesis while promoting neurogenesis .
Agonist antibodies (e.g., 29D7) enhance ERK1/2 and AKT phosphorylation, improving neuronal survival post-injury .
TrkB-targeted therapies are explored for neurodegenerative diseases and NTRK fusion-positive cancers .
Parameter | Proteintech MP01108-3 | Creative Biolabs 29D7 |
---|---|---|
Detection Range (ELISA) | 3.91–250 ng/mL | Not specified |
Sensitivity (Cytometric) | 0.313–40 ng/mL | N/A |
Key Applications | Cytometric bead array, Western blot | Neuronal survival assays |
Reactivity | Human | Human, Mouse |
TrkB (Tropomyosin receptor kinase B), encoded by the NTRK2 gene, is a receptor tyrosine kinase crucial for the development and maturation of the central and peripheral nervous systems. Its functions encompass regulating neuronal survival, proliferation, migration, differentiation, synapse formation, and synaptic plasticity. TrkB serves as a receptor for brain-derived neurotrophic factor (BDNF) and neurotrophin-4 (NTF4). While less efficiently, it can also bind neurotrophin-3 (NTF3), influencing neuronal survival. Ligand binding induces TrkB homodimerization, autophosphorylation, and activation, leading to the recruitment and/or phosphorylation of downstream effectors, including SHC1, FRS2, SH2B1, SH2B2, and PLCG1. These effectors trigger distinct, overlapping signaling cascades. Through SHC1, FRS2, SH2B1, and SH2B2, TrkB activates the GRB2-Ras-MAPK pathway, regulating neuronal differentiation (including neurite outgrowth). It also controls the Ras-PI3 kinase-AKT1 pathway, primarily governing growth and survival. PLCG1 activation and downstream protein kinase C pathways regulate synaptic plasticity, contributing to learning and memory processes by influencing both short-term synaptic function and long-term potentiation. PLCG1-mediated NF-κB activation upregulates genes involved in cell survival, suppressing anoikis (apoptosis due to loss of cell-matrix interactions). TrkB may additionally participate in neurotrophin-dependent calcium signaling in glial cells and mediate neuron-glia communication.
The NTRK2 gene and its protein product TrkB have been implicated in various physiological and pathological processes, as detailed in the following research studies:
NTRK2 is one of the members of the neurotrophin receptor family, which includes TrkA and TrkC. All Trk proteins have a conserved structure with a signal peptide, two cysteine-rich domains, leucine-rich motifs, and two Ig-like domains in the extracellular portion . NTRK2 is activated by neurotrophins including BDNF (Brain Derived Neurotrophic Factor), Neurotrophin-3 (NT-3), and Neurotrophin-4 (NT-4) . Upon activation, NTRK2 mediates numerous signaling events that regulate neuron physiology, cell proliferation and survival, axon and dendrite growth and patterning, and synaptic plasticity . NTRK2 dysregulation has been associated with neurological disorders, psychiatric diseases, and certain cancers, making it an important target for neurological research .
Human NTRK2 has 7 isoforms with predicted molecular weights ranging from 35 to 93 kDa . The two primary isoforms are the full-length receptor tyrosine kinase (TrkB.FL) containing the complete signaling domain, and the kinase-deficient truncated isoform (TrkB.T1) . Research has shown that contrary to expectations, TrkB.T1 is the predominant isoform expressed in gliomas . TrkB.T1 contains a unique 11-amino acid C-terminus that distinguishes it from other isoforms . Understanding these isoform differences is crucial for experimental design and interpretation, as they have distinct signaling capabilities and potentially different functions in normal and pathological conditions.
NTRK2 is found primarily in the nervous system, with high expression in neurons, astrocytes, and oligodendrocyte precursors . It is also expressed in the pancreas, skeletal muscles, and kidney . The truncated form of TrkB (lacking the tyrosine kinase domain) is found in heart, ovary, and spleen . This distribution pattern is important to consider when selecting positive control tissues and interpreting antibody staining results across different experimental systems.
Validation should include multiple complementary approaches: 1) Western blot analysis to confirm detection of expected molecular weight proteins (35-93 kDa depending on isoform) ; 2) Testing in tissues or cell lines with known NTRK2 expression patterns; 3) Using NTRK2-deficient models as negative controls, such as CRISPR/Cas9-generated NTRK2-/- cell lines ; 4) Comparing antibodies targeting different epitopes; 5) Performing peptide competition assays; and 6) Verifying isoform specificity, particularly when distinguishing between TrkB.FL and TrkB.T1 . Additionally, researchers should consider creating recombinant antibodies with different Fc regions (e.g., rabbit) to minimize mouse-on-mouse artifacts in immunohistochemistry applications .
Epitope selection is critical because NTRK2 has multiple isoforms with different domains and undergoes extensive post-translational modifications . Researchers must consider: 1) Whether the epitope is in a region common to all isoforms or specific to certain isoforms (e.g., the kinase domain in TrkB.FL or unique C-terminus of TrkB.T1) ; 2) The 11 potential N-glycosylation sites that may affect antibody binding ; 3) The accessibility of the epitope in different applications (native vs. denatured protein); 4) Species homology if working with model organisms; and 5) Potential cross-reactivity with other Trk family members due to conserved domains.
To differentiate between NTRK2 isoforms, researchers should: 1) Select antibodies targeting isoform-specific regions, such as the kinase domain for TrkB.FL or the unique 11-amino acid C-terminus for TrkB.T1 ; 2) Use Western blotting with appropriate controls to distinguish isoforms by molecular weight; 3) Complement antibody-based methods with RT-qPCR using isoform-specific primers ; 4) Consider the predominance of TrkB.T1 in certain tissues such as gliomas ; and 5) Use recombinant isoform proteins as positive controls. This multi-method approach increases confidence in isoform-specific detection.
Researchers should anticipate potential cross-reactivity with: 1) Other Trk family members (TrkA/NTRK1 and TrkC/NTRK3) due to homology in conserved domains; 2) Related receptor tyrosine kinases; 3) Non-specific binding to tissues with high expression of similar proteins; and 4) Mouse-on-mouse artifacts when using mouse monoclonal antibodies on mouse tissues . To address these issues, researchers can use knockout controls, peptide competition assays, and recombinant antibodies with different Fc regions to minimize host-specific artifacts as mentioned in the search results .
For optimal NTRK2 immunohistochemistry, researchers should: 1) Select fixation protocols appropriate for neural tissues (typically 4% paraformaldehyde); 2) Optimize antigen retrieval methods specific to the epitope and tissue preparation; 3) Use appropriate blocking solutions to reduce background; 4) Include proper controls, particularly NTRK2 knockout tissues/cells and tissues with known expression patterns; 5) When working with mouse tissues, consider recombinant antibodies with rabbit Fc regions to avoid mouse-on-mouse artifacts ; and 6) Validate staining patterns with multiple antibodies targeting different epitopes to confirm specificity.
For optimal Western blotting of NTRK2: 1) Consider sample preparation methods that preserve protein integrity, such as using protease inhibitors; 2) Select appropriate lysis buffers based on the cellular localization of the target; 3) Use gradient gels (e.g., 4-12% Bis-Tris) to resolve different isoforms effectively ; 4) Include positive controls with known NTRK2 expression and negative controls from knockout cells ; 5) Transfer to PVDF membranes at appropriate voltage and time (e.g., 90 min at 80V) ; 6) Block with 5% BSA in TBST ; and 7) Validate antibody specificity against recombinant NTRK2 isoforms to confirm band identity.
For flow cytometry applications with NTRK2 antibodies, researchers should: 1) Optimize fixation and permeabilization protocols depending on whether targeting extracellular or intracellular domains; 2) Use appropriate fluorophore-conjugated secondary antibodies or directly conjugated primary antibodies; 3) Include isotype controls to assess non-specific binding; 4) Validate antibody performance using positive control cell populations; 5) Use NTRK2 knockout cells as negative controls; 6) Consider cell sorting approaches to isolate NTRK2-positive populations for further analysis; and 7) If working with transfected cells, use mCherry or GFP positive cells as selection markers as shown in CRISPR/Cas9 NTRK2 knockout experiments .
To study NTRK2 activation states, researchers should: 1) Use phospho-specific antibodies targeting key phosphorylation sites of NTRK2; 2) Design time-course experiments following BDNF, NT-3, or NT-4 stimulation ; 3) Include inhibitor controls for specificity of activation; 4) Consider the differences in signaling between full-length TrkB.FL (with kinase activity) and truncated TrkB.T1 (kinase-deficient) ; 5) Examine downstream signaling pathways like PI3K that are implicated in TrkB.T1 signaling in glioma ; 6) Use both Western blotting and immunocytochemistry to assess activation in both population and single-cell levels; and 7) Compare activation patterns in wild-type versus NTRK2 knockout models to confirm signaling specificity .
CRISPR/Cas9 technology provides robust validation tools for NTRK2 antibodies by: 1) Generating true negative controls through complete NTRK2 gene knockout ; 2) Creating isoform-specific knockouts to validate isoform-selective antibodies; 3) Enabling epitope tagging of endogenous NTRK2 for antibody-independent detection; 4) Allowing precise quantification of antibody specificity by comparing signal in wild-type versus knockout cells; 5) Generating cell lines with defined NTRK2 mutations to test antibody sensitivity to specific variants; and 6) Creating model systems to study the functional consequences of NTRK2 loss, as demonstrated in studies showing that NTRK2 knockout decreases neurogenesis and favors glial progenitors .
To study NTRK2 in neural differentiation, researchers should: 1) Use CRISPR/Cas9-mediated NTRK2 knockout in neural progenitor cells to assess differentiation effects ; 2) Employ qPCR to measure expression of neurogenic transcription factors and glial progenitor markers ; 3) Perform comprehensive transcriptomic analysis to identify global gene expression changes associated with NTRK2 loss ; 4) Combine NTRK2 antibodies with markers of neuronal and glial lineages for co-localization studies; 5) Use time-course experiments to track NTRK2 expression during differentiation; 6) Consider the different roles of TrkB.FL versus TrkB.T1 in differentiation outcomes; and 7) Validate key findings across multiple neural progenitor cell models.
When facing discrepancies between protein and mRNA expression data, researchers should: 1) Consider post-transcriptional regulation mechanisms affecting NTRK2; 2) Verify that antibodies and PCR primers target the same isoforms ; 3) Examine potential methodological differences in sensitivity between techniques; 4) Use absolute quantification methods to directly compare transcript and protein levels; 5) Analyze protein stability and turnover rates; 6) Consider that whole gene NTRK2 expression might mask isoform-specific changes, as seen in glioma research where similar whole gene expression showed distinct isoform patterns ; and 7) Validate findings using multiple antibodies targeting different epitopes to confirm protein expression patterns.
For studying NTRK2 splice variants in cancer: 1) Use isoform-specific antibodies and PCR primers to distinguish between TrkB.FL and TrkB.T1 ; 2) Consider that TrkB.T1 is the predominant isoform in gliomas, contrary to previous assumptions about TrkB.FL ; 3) Employ both in vitro cell models and in vivo xenograft approaches to assess oncogenic effects; 4) Perform comprehensive transcriptomic and pathway analysis to identify isoform-specific signaling mechanisms ; 5) Assess the effects of specific isoforms on downstream pathways such as PI3K, Akt, and STAT3 signaling ; 6) Include proper controls of normal neural cells/tissues for comparison; and 7) Consider the paradoxical finding that high TrkB.FL expression correlates with better prognosis in gliomas .
To reduce high background in NTRK2 immunostaining: 1) Optimize antibody concentration through careful titration; 2) Increase blocking time and concentration (5% BSA as used in protocols) ; 3) Add appropriate detergents to reduce non-specific hydrophobic interactions; 4) Extend washing steps and increase washing buffer volumes; 5) For mouse tissues, use specialized blocking techniques or consider recombinant antibodies with rabbit Fc regions as mentioned in the search results ; 6) For multiple labeling experiments, test for potential cross-reactivity between secondary antibodies; 7) Use proper controls to identify the source of background (secondary-only, isotype controls); and 8) Consider using more specific detection systems like tyramide signal amplification for weak signals while maintaining low background.
For inconsistent Western blot results: 1) Verify sample preparation consistency, including lysis buffer composition and protease inhibitor use; 2) Consider NTRK2's 11 potential N-glycosylation sites which may affect migration and detection ; 3) Optimize protein loading amounts (typically 20-50 μg total protein); 4) Test different membrane types (PVDF vs. nitrocellulose); 5) Adjust transfer conditions for high molecular weight isoforms; 6) Use fresh samples as NTRK2 may be sensitive to freeze-thaw cycles; 7) Test multiple antibody concentrations and incubation conditions; and 8) Include positive controls such as brain tissue lysates or recombinant NTRK2 protein on each blot to confirm detection consistency.
When antibodies fail to detect expected NTRK2 expression: 1) Verify NTRK2 transcript expression using qPCR with appropriate primers ; 2) Consider alternative antibodies targeting different epitopes; 3) Test more sensitive detection methods such as amplified detection systems; 4) Optimize antigen retrieval for fixed tissues; 5) Examine whether post-translational modifications might mask the epitope; 6) Consider that expression levels might vary between isoforms, with TrkB.T1 predominating in some tissues ; 7) Use enrichment methods such as immunoprecipitation before detection; and 8) Validate antibody reactivity with recombinant NTRK2 protein controls.
When facing discrepancies between different NTRK2 antibodies: 1) Compare the epitopes targeted by each antibody and consider isoform specificity; 2) Evaluate the validation data for each antibody, including knockout controls ; 3) Consider that antibodies targeting different domains may reveal different aspects of NTRK2 biology; 4) Test under multiple experimental conditions (native vs. denatured proteins); 5) Verify results with orthogonal techniques (RT-qPCR, RNA-seq) ; 6) Consider the expression patterns of different isoforms across tissues; 7) Use NTRK2 knockout models as definitive negative controls ; and 8) Consult literature for reported discrepancies with specific antibody clones.
Single-cell approaches can revolutionize NTRK2 isoform research by: 1) Revealing cell type-specific expression patterns of TrkB.FL versus TrkB.T1 that may be masked in bulk tissue analysis ; 2) Identifying rare cell populations with unique NTRK2 isoform signatures; 3) Correlating NTRK2 isoform expression with cell state and differentiation status ; 4) Mapping the heterogeneity of NTRK2 expression in complex tissues such as brain tumors ; 5) Combining protein and transcript detection at single-cell resolution to resolve post-transcriptional regulation; 6) Linking NTRK2 isoform expression to functional outcomes in individual cells; and 7) Providing insight into how NTRK2 isoform balance affects cell fate decisions during neural development .
Emerging applications for NTRK2 antibodies in therapeutic development include: 1) Companion diagnostics to identify patients with NTRK2-dependent tumors; 2) Therapeutic antibodies targeting specific NTRK2 isoforms, particularly TrkB.T1 which predominates in gliomas ; 3) Antibody-drug conjugates for targeted delivery to NTRK2-expressing cells; 4) In vivo imaging agents for monitoring NTRK2 expression in tumors; 5) Tools for evaluating the efficacy of TrkB modulators, including recent psychedelic compounds that act as positive allosteric modulators of TrkB ; 6) Monitoring treatment responses in NTRK2-targeted therapies; and 7) Development of isoform-specific inhibitors based on detailed understanding of NTRK2 splice variant functions in disease .
Advancing methodologies to improve NTRK2 detection specificity include: 1) Developing recombinant antibodies with enhanced specificity for particular NTRK2 isoforms; 2) Creating nanobodies or aptamers with reduced size for improved tissue penetration; 3) Implementing multiplexed detection systems to simultaneously visualize multiple NTRK2 isoforms; 4) Developing proximity ligation assays to detect specific NTRK2 protein-protein interactions; 5) Utilizing CRISPR-based tagging of endogenous NTRK2 for antibody-independent visualization ; 6) Implementing machine learning algorithms for automated analysis of NTRK2 expression patterns in imaging data; and 7) Developing more sensitive methods to detect low abundance NTRK2 isoforms in challenging sample types.
For translational research with NTRK2 antibodies, researchers should consider: 1) Validating antibodies in human tissues that match the disease being studied; 2) Accounting for differences in NTRK2 isoform expression between model systems and human pathology ; 3) Understanding the prognostic implications of specific isoforms, such as high TrkB.FL correlating with better outcomes in gliomas ; 4) Standardizing detection protocols across research sites for reproducibility; 5) Correlating NTRK2 detection with clinical outcomes in patient samples; 6) Developing quantitative scoring systems for NTRK2 expression in clinical samples; 7) Addressing confounding factors such as tissue processing variations in multicenter studies; and 8) Considering how NTRK2 isoform expression might influence response to targeted therapies.