NTRK2 Human produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain containing 407 amino acids (32-430a.a) and having a molecular mass of 45.2kDa.
NTRK2 is fused to an 8 amino acid His tag at C-terminus and purified by proprietary chromatographic techniques.
Neurotrophic Receptor Tyrosine Kinase 2, also known as NTRK2, is a tyrosine-protein kinase receptor involved in the development and maturation of the central and peripheral nervous systems. It regulates neuronal survival, migration, proliferation, differentiation, and synapse formation and plasticity. NTRK2 plays a role in learning and memory by regulating both short-term synaptic function and long-term potentiation. Known substrates for TRK family receptors include SHC1, PI-3 kinase, and PLC-gamma-1.
Human NTRK2, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain containing 407 amino acids (32-430a.a). It has a molecular mass of 45.2kDa. NTRK2 is fused to an 8 amino acid His tag at the C-terminus and purified using proprietary chromatographic techniques.
The NTRK2 solution is supplied at a concentration of 0.5mg/ml and contains 10% glycerol in phosphate-buffered saline (pH 7.4).
For short-term storage (2-4 weeks), the product can be stored at 4°C. For long-term storage, it is recommended to store the product frozen at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity is determined to be greater than 95% by SDS-PAGE analysis.
GP145-TrkB, trk-B, TRKB, NTRK2
Sf9, Baculovirus 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 VTDKTGREHL EHHHHHH
NTRK2 (Neurotrophic Tyrosine Kinase Receptor Type 2) is a gene that encodes the tropomyosin receptor kinase B (TrkB), a member of the transmembrane tyrosine kinase family . The primary function of TrkB is critical for the development and maintenance of the nervous system . This receptor has high affinity for several neurotrophins, particularly brain-derived neurotrophic factor (BDNF), as well as NT3 and NT4/5 . TrkB signaling plays vital roles in neuronal differentiation, growth, and survival throughout human development and adulthood . Dysregulation of TrkB is associated with a spectrum of conditions including neurodegenerative diseases, psychiatric disorders, and certain types of cancer .
NTRK2 polymorphisms demonstrate significant associations with cognitive and emotional processes. Research using the Lazarus-type stress-coping inventory, ego aptitude scale (EAS), and social adaptation self-evaluation scale (SASS) has revealed that specific NTRK2 single-nucleotide polymorphisms (SNPs) significantly influence stress-coping strategies . For example, the study of five NTRK2 SNPs (rs11140800, rs1187286, rs1867283, rs1147198, and rs10868235) showed significant associations with cognitive strategies, problem-solving, confrontive-coping, seeking social support, distancing, and positive reappraisal . These polymorphisms also demonstrate significant impact on all seven ego-related factors measured by the EAS, including critical, nurturing, mature, rational, natural, intuitive, and adaptive attitudes . Methodologically, researchers investigating these associations should employ standardized psychological assessment tools alongside genetic testing to establish reliable correlations.
Multiple experimental models exist for studying NTRK2 function in human neural context:
Human neural progenitor cell lines: The ReNcell VM line has been identified as expressing high levels of TrkB with robust activity, making it suitable for NTRK2 studies .
CRISPR/Cas9-modified cell lines: Researchers have successfully generated NTRK2-deficient (NTRK2−/−) ReNcell VM using CRISPR/Cas9 gene editing technology . This provides a reproducible and readily available cell culture system for studying TrkB's role during human neural differentiation.
Transcriptomic analysis: Global transcriptomic analysis of NTRK2−/− cells has revealed major changes in expression of specific genes responsible for neurogenesis, neuronal development, and glial differentiation .
When selecting an experimental model, researchers should consider their specific research questions and the limitations of each system. The CRISPR/Cas9-modified ReNcell VM line offers significant advantages for mechanistic studies, particularly for analyzing TrkB isoforms and validating antibodies and pharmacological agents targeting the TrkB pathway .
NTRK2 signaling appears to play a dual role in neural cell fate determination. Transcriptomic analysis of NTRK2-deficient human neural progenitor cells has revealed that key neurogenic transcription factors are massively downregulated in NTRK2−/− cells . Concurrently, early glial progenitor markers are significantly enriched in NTRK2−/− cells compared to NTRK2+/+ cells . This indicates a previously undescribed inhibitory role of TrkB on glial differentiation in addition to its well-established pro-neurogenesis function .
To investigate this balance experimentally, researchers should:
Utilize cell-type specific markers to track differentiation trajectories
Employ time-course experiments to determine critical developmental windows
Incorporate pathway analysis to understand downstream mediators of this balance
Consider single-cell approaches to capture heterogeneity in response to NTRK2 signaling
The understanding of NTRK2's role in cell fate determination has significant implications for neural development studies and potential therapeutic approaches for conditions with aberrant neuron/glia ratios.
The molecular mechanisms by which NTRK2 contributes to stress resilience involve complex genomic and non-genomic changes that facilitate neural adaptations to stress. Research indicates that NTRK2 signaling considerably affects the actions of antidepressants and may be integral to the brain's plasticity under stress loads . BDNF/NTRK2-stimulated intracellular signaling has been shown to be crucial for antidepressant effects, with stimulation in the hippocampus, prefrontal cortex, and anterior cingulate cortex being vital for these responses .
Methodologically, researchers investigating these mechanisms should:
Employ animal models with conditional NTRK2 knockout in specific brain regions
Utilize human neural cells with NTRK2 variants to study signaling differences
Incorporate stress paradigms that model human psychological stress
Measure both immediate signaling changes and long-term adaptations
Consider epigenetic modifications as mediators of lasting effects
The TRKB receptor (encoded by NTRK2) has a regulatory role in neural differentiation and maintains specific neuron populations in areas such as the human prefrontal cortex, which is critical for stress processing and emotional regulation .
Differentiating between NTRK2 isoforms requires a multi-faceted approach:
Isoform-specific genetic manipulation: CRISPR/Cas9 technology can be used to selectively modify specific exons or regulatory regions that control the expression of different TrkB isoforms . The generated NTRK2−/− ReNcell VM model represents a valuable starting point for re-introduction of specific isoforms to determine their individual contributions .
Transcriptomic profiling: RNA-seq analyses comparing cells expressing different TrkB isoforms can reveal distinct transcriptional signatures and downstream pathways activated by each isoform .
Proteomic approaches: Mass spectrometry and co-immunoprecipitation studies can identify isoform-specific protein-protein interactions that may explain functional differences.
Functional assays: Researchers should employ multiple functional readouts including neuronal differentiation, survival, neurite outgrowth, and electrophysiological properties to comprehensively characterize isoform-specific effects.
Domain-specific antibodies and inhibitors: Validation of isoform-specific detection and manipulation tools is essential for accurate experimentation.
The human neural cell line with NTRK2 knockout provides an excellent system for these investigations as it allows controlled re-expression of specific isoforms in a human neural context .
Research has identified significant associations between specific NTRK2 polymorphisms and various stress-coping strategies in humans. A study examining five NTRK2 SNPs (rs11140800, rs1187286, rs1867283, rs1147198, and rs10868235) found the following specific associations:
NTRK2 SNP | Associated Stress-Coping Strategies | P-value |
---|---|---|
rs11140800 | Cognitive strategies | <0.05 |
rs11140800 | Confrontative coping | <0.01 |
rs11140800 | Seeking social support | <0.05 |
rs1187286 | Cognitive strategies | <0.05 |
rs1187286 | Planned problem-solving | <0.01 |
rs1187286 | Distancing | <0.05 |
rs1867283 | Positive reappraisal | <0.05 |
rs10868235 | Cognitive strategies | <0.05 |
rs10868235 | Confrontative coping | <0.01 |
These findings suggest that variations in the NTRK2 gene significantly influence how individuals respond to and manage stress . To study these associations, researchers should employ standardized psychological assessment tools like the Lazarus-type stress-coping inventory alongside genotyping methodologies . Additionally, investigating these associations in different populations and under various stress conditions would provide a more comprehensive understanding of the gene-environment interactions that influence stress coping.
NTRK2 knockout in human neural progenitor cells (ReNcell VM) leads to substantial transcriptomic alterations that affect neural development pathways . Global transcriptomic analysis has revealed major changes in the expression of specific genes responsible for:
Neurogenesis: Key neurogenic transcription factors are massively downregulated in NTRK2−/− cells .
Neuronal development: Genes involved in neuronal maturation, axon guidance, and synapse formation show altered expression patterns .
Glial differentiation: Early glial progenitor markers are significantly enriched in NTRK2−/− cells compared to NTRK2+/+ cells, suggesting an inhibitory role of TrkB on glial differentiation .
To investigate these transcriptomic changes, researchers should:
Perform RNA-sequencing at different time points during neural differentiation
Validate key findings with qPCR and protein-level analyses
Use pathway analysis tools to identify affected signaling networks
Correlate transcriptomic changes with morphological and functional phenotypes
Consider chromatin immunoprecipitation sequencing (ChIP-seq) to identify direct transcriptional targets
These findings highlight the critical role of NTRK2 in regulating cell fate decisions during human neural development, with important implications for understanding neurodevelopmental disorders associated with NTRK2 dysfunction .
Generating reliable NTRK2-deficient human neural cell lines requires careful methodological considerations:
CRISPR/Cas9 targeting strategy: Researchers should design guide RNAs targeting conserved exons present in all functional NTRK2 isoforms to ensure complete knockout . Multiple guide RNAs should be tested to identify those with highest efficiency and specificity.
Cell line selection: The ReNcell VM immortalized human neural progenitor stem cell line has been identified as expressing high levels of TrkB with robust activity, making it ideal for NTRK2 knockout studies .
Validation approaches:
Genomic validation: Confirm mutations using sequencing to verify frameshift or nonsense mutations
Transcript validation: Use RT-PCR and RNA-seq to confirm absence of wild-type transcripts
Protein validation: Western blotting with antibodies targeting different domains of TrkB
Functional validation: Confirm lack of response to BDNF stimulation through phosphorylation assays
Control considerations: Generate and maintain isogenic control lines that have undergone the same CRISPR/Cas9 process but retain NTRK2 function to account for potential off-target effects .
Phenotypic characterization: Comprehensive assessment of proliferation, differentiation capacity, and electrophysiological properties to understand the full impact of NTRK2 deficiency.
This approach has successfully generated NTRK2−/− ReNcell VM lines that provide a reproducible and readily available cell culture system for studying TrkB's role during human neural differentiation .
To effectively study relationships between NTRK2 genetics and psychological traits, researchers should implement the following methodological approaches:
Subject selection and assessment:
Recruit ethnically homogeneous samples to minimize population stratification effects
Screen participants using standardized psychiatric interviews like the Mini-International Neuropsychiatric Interview (MINI) and SCID-II questionnaire
Apply standardized psychological measures such as the Lazarus-type Stress-Coping Inventory (SCI), Ego Aptitude Scale (EAS), and Social Adaptation Self-Evaluation Scale (SASS)
Genetic analysis:
Statistical approaches:
Apply appropriate corrections for multiple testing
Consider gene-environment interactions in analyses
Use multivariate approaches to account for correlations between psychological traits
A study employing these methods successfully identified significant associations between NTRK2 polymorphisms and various psychological traits related to stress coping and social adaptation, demonstrating the viability of this methodological approach .
Translating NTRK2 research findings from animal models to human applications presents several challenges:
Species-specific differences in NTRK2 expression and function:
Human NTRK2 has distinct expression patterns and potentially different isoform ratios compared to animal models
Regulatory elements controlling NTRK2 expression may differ significantly between species
Methodological limitations:
Experimental system considerations:
Solutions and advances:
Development of human neural cell lines with NTRK2 modifications using CRISPR/Cas9 provides more translatable models
Integration of findings across levels (genetic associations, cellular models, and clinical observations) improves translational validity
Use of induced pluripotent stem cells (iPSCs) from patients with NTRK2 variants allows for personalized modeling
The generation of NTRK2−/− ReNcell VM using CRISPR/Cas9 gene editing technology represents an important advance in addressing these challenges, providing a reproducible human neural cell model to study NTRK2 function .
Human lineage mutations (HLMs) in genes like NTRK2 may have played significant roles in human evolution and speciation. Around 4% of the Homo sapiens genome differs from our closest relative, the chimpanzee (Pan troglodytes), including approximately 35 million single nucleotide variations and 90 Mb regions with structural variations . These genetic differences cover almost all evolutionary events that distinguish humans from other primates .
To investigate the evolutionary significance of HLMs in NTRK2:
Comparative genomic approaches: Researchers should compare NTRK2 sequences across primates, focusing on mutations unique to humans that may have contributed to cognitive adaptations .
Functional analysis of human-specific variants: Using CRISPR/Cas9 to introduce human-specific NTRK2 variants into non-human primate cells or replace human NTRK2 sequences with ancestral versions can reveal functional differences .
RNA-protein binding analysis: Examining how human-specific NTRK2 variants affect RNA-protein interactions may reveal mechanisms by which these mutations influence gene regulation and neural development .
Enrichment analysis methodology: Researchers should compare observed versus expected numbers of influential HLMs in NTRK2, calculating fold enrichment to determine evolutionary significance .
Understanding how HLMs in NTRK2 contributed to human neurological evolution could provide insights into unique aspects of human cognition and behavior, particularly relating to stress resilience and social adaptation capacities that differentiate humans from other primates .
The development of NTRK2-targeted therapeutics for neuropsychiatric disorders has advanced significantly based on our understanding of TrkB's role in neural function and stress resilience. NTRK2 demonstrates antidepressant-like effects, with the TRKB receptor being stimulated in the hippocampus, prefrontal cortex, and anterior cingulate cortex during antidepressant treatment . This receptor activation is vital for producing antidepressant effects, and BDNF/NTRK2-stimulated intracellular signaling considerably affects the actions of these medications .
Current methodological approaches for developing NTRK2-targeted therapeutics include:
High-throughput screening: Identification of small molecules that specifically modulate TrkB signaling without affecting related receptors.
Structure-based drug design: Using crystallographic data of the TrkB receptor to design molecules that can modulate its activity with high specificity.
NTRK2 variant analysis: Examining how specific NTRK2 polymorphisms affect treatment response to develop personalized therapeutic approaches .
Target validation using knockout models: The newly developed NTRK2−/− human neural cell lines provide valuable tools for validating potential therapeutic targets in the TrkB pathway and testing candidate compounds .
Downstream pathway targeting: Identification of critical nodes in TrkB signaling pathways that could be targeted to achieve therapeutic effects while minimizing side effects.
The generation of human NTRK2-deficient neural cell lines offers significant advantages for screening and validating pharmacological agents targeting the TrkB pathway in a human cellular context , potentially accelerating the development of more effective therapeutics for neuropsychiatric disorders.
Several promising research directions for NTRK2 in human neuroscience warrant exploration:
Single-cell analysis of NTRK2 function: Investigating cell-type specific roles of TrkB using single-cell transcriptomics and proteomics could reveal nuanced functions in different neural populations.
NTRK2 in neural circuit development: Leveraging optogenetics and chemogenetics in combination with NTRK2 manipulation to understand how TrkB signaling shapes functional neural circuits.
Epigenetic regulation of NTRK2: Exploring how environmental factors influence NTRK2 expression and function through epigenetic mechanisms could provide insights into gene-environment interactions in neuropsychiatric disorders.
NTRK2 in human brain organoids: Studying NTRK2 function in three-dimensional brain organoids could bridge the gap between cellular models and in vivo human brain development.
Therapeutic targeting of specific TrkB domains: Developing approaches that selectively modulate particular functions of TrkB while preserving others could minimize off-target effects in therapeutic applications.
NTRK2 in neuroinflammation: Investigating the role of TrkB in mediating interactions between neural and immune cells could reveal new mechanisms relevant to neurodegenerative and neuropsychiatric conditions.
Neurotrophic Receptor Tyrosine Kinase 2 (NTRK2), also known as TrkB, is a critical protein in the development and maintenance of the mammalian nervous system. It belongs to the neurotrophic factor family of related polypeptides and serves as a receptor for brain-derived neurotrophic factor (BDNF) and neurotrophin-4 (NTF4). NTRK2 plays a pivotal role in neuron survival, proliferation, migration, differentiation, and synapse formation and plasticity .
NTRK2 is a membrane-bound receptor tyrosine kinase. Upon binding with its ligands, such as BDNF, it undergoes homodimerization, autophosphorylation, and activation. This activation triggers downstream signaling pathways, including the MAPK pathway, which leads to various cellular responses essential for nervous system development and function .
NTRK2 is predominantly expressed in the brain, with enhanced expression in regions such as the cerebral cortex, cerebellum, basal ganglia, hypothalamus, midbrain, amygdala, choroid plexus, hippocampal formation, and spinal cord. It is also found in the thyroid gland and other tissues. The protein is localized mainly to the plasma membrane and intracellular compartments .
Mutations and abnormal expression of the NTRK2 gene are associated with several neurological and psychiatric disorders, including obesity, mood disorders, and developmental and epileptic encephalopathy. Additionally, NTRK2 has been implicated in various cancers, such as colorectal, breast, papillary thyroid cancer, lung adenocarcinoma, and oral squamous cell carcinoma .
Recombinant NTRK2 proteins are widely used in research to study their role in neurobiology and disease. These proteins are also being explored as potential therapeutic targets for treating neurological disorders and cancers. The development of NTRK2 inhibitors and other modulators holds promise for future therapeutic interventions .