Neurotrophin-3 (NT-3), a member of the nerve growth factor (NGF) family, is critical for neuronal survival, synaptogenesis, and muscle innervation in mice. It binds high-affinity receptors TrkC and TrkB, influencing both central and peripheral nervous system development . In recombinant forms, mouse NT-3 is widely used in research to study neuroprotection, neuromuscular junction formation, and regenerative therapies .
Neuronal Survival: Promotes survival of sensory and motor neurons during development .
Synaptic Plasticity: Enhances hippocampal plasticity and learning/memory in adult mice .
Muscle Function: Supports neuromuscular junction maintenance and prevents age-related sarcopenia .
NT-3 knockout (KO) mice exhibit severe phenotypes, highlighting its essential role in neurodevelopment and muscle function:
NT-3 KO Mice: Show ~40% reduction in hippocampal CA1 synapses and impaired learning .
TrkC KO Mice: Mimic NT-3 KO phenotypes, confirming receptor dependency .
Adeno-associated virus (AAV)-mediated NT-3 gene therapy demonstrates therapeutic potential in neuromuscular and age-related disorders:
Recombinant Mouse Neurotrophin-3, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 119 amino acids. With a molecular weight of 13.6 kDa, this purified protein is obtained through proprietary chromatographic techniques.
Biological activity, assessed by the dose-dependent proliferation of BaF3 cells transfected with the TrkB receptor, typically falls within the range of 1-10 ng/ml. This corresponds to a specific activity of 100,000-1,000,000 units/mg.
YAEHKSHRGE YSVCDSESLW VTDKSSAIDI RGHQVTVLGE IKTGNSPVKQ YFYETRCKEA RPVKNGCRGI DDKHWNSQCK TSQTYVRALT SENNKLVGWR WIRIDTSCVC ALSRKIGRT.
NT-3 (Neurotrophin-3) is a growth factor with significant roles in nervous system development, myelination, neuroprotection, and regeneration. Additionally, NT-3 possesses immunomodulatory and anti-inflammatory properties that make it valuable for studying neurological disorders. In mouse models, NT-3 has demonstrated therapeutic potential for conditions characterized by demyelination and axonal damage, such as multiple sclerosis and peripheral neuropathies . The ability to manipulate NT-3 expression through gene therapy or genetic knockout approaches allows researchers to investigate its functional impact on disease progression and neuronal integrity.
Several key mouse models are utilized in NT-3 research, each serving distinct research purposes:
Experimental Autoimmune Encephalomyelitis (EAE) mice: These serve as a chronic relapsing mouse model of multiple sclerosis, allowing researchers to study NT-3's effects on central nervous system demyelination and inflammation .
Sh3tc2-/- mice: This model replicates Charcot-Marie-Tooth Type 4C (CMT4C), a demyelinating peripheral neuropathy, facilitating investigation of NT-3's effects on peripheral nerve myelination and function .
Conditional NT-3 knockout mice: Generated by crossing mice expressing CRE recombinase under the Synapsin I promoter with mice containing floxed NT-3 genes, these models allow for neuron-specific deletion of NT-3 to study its role in synaptic transmission and plasticity .
These models enable researchers to investigate NT-3's functions across different neurological contexts and disease states.
The method of NT-3 delivery varies based on research objectives and the target nervous system:
For central nervous system (CNS) research focused on conditions like multiple sclerosis, investigators commonly use intramuscular injections of self-complementary AAV1 vectors (scAAV1.tMCK.NT-3) into the gastrocnemius muscle. This approach results in sustained systemic production of NT-3, with measurable levels appearing in the serum within 7 weeks post-delivery . This peripheral administration strategy effectively influences CNS pathology while avoiding the challenges of direct CNS delivery.
In peripheral nervous system studies, such as those involving Charcot-Marie-Tooth models, researchers similarly employ intramuscular injections of AAV1.tMCK.NT-3 vectors. This technique has demonstrated efficacy in improving both histopathological outcomes and functional measures like nerve conduction velocity . The peripheral delivery method allows for targeting both motor and sensory components of peripheral nerves.
The timing of delivery is also crucial - for example, in EAE models, administration typically occurs 3 weeks post-disease induction, while in Sh3tc2-/- mice, treatment often begins at 4 weeks of age .
Designing robust functional outcome measures for NT-3 gene therapy studies requires a comprehensive, multi-modal approach that captures both behavioral and electrophysiological changes:
Behavioral Testing Protocol:
Rotarod Assessment: Implement accelerating rotarod protocols (starting at 5 rpm with constant acceleration of 0.5 rpm/s) to evaluate motor function and balance. Testing should be conducted at baseline (pre-treatment), mid-point (3 months post-treatment), and end-point (6 months post-treatment). Include an acclimation run at least 24 hours before experimental runs, and record the best performance from three consecutive trials .
Grip Strength Evaluation: Utilize a digital grip strength meter to assess bilateral simultaneous hindlimb strength. Each mouse should perform three consecutive trials, with the average force measurement included in the analysis .
Electrophysiological Measurements:
Nerve Conduction Studies: Conduct under 2% isoflurane anesthesia while maintaining body temperature on a heating pad. Use disposable needle electrodes and established EMG systems to measure nerve conduction velocity, particularly in the sciatic nerve .
Experimental Design Considerations:
Include appropriate control groups (wild-type and untreated disease model mice)
Ensure adequate sample sizes (n=15-16 per group minimum based on successful studies)
Implement blind testing protocols to eliminate observer bias
Account for sex differences in analysis, as female mice often demonstrate different baseline performance and treatment responses than males
This comprehensive assessment approach provides complementary data points that collectively reflect the multifaceted impact of NT-3 gene therapy on neuromuscular function.
The design of adeno-associated virus (AAV) vectors for NT-3 expression requires careful consideration of several critical parameters to optimize transgene expression, tissue tropism, and therapeutic efficacy:
Vector Serotype Selection:
AAV1 has demonstrated particular efficacy for NT-3 delivery in neuromuscular applications. This serotype shows robust transduction of muscle tissue while allowing sufficient systemic distribution of the expressed neurotrophin .
Promoter Selection:
The tissue-specific muscle creatine kinase (tMCK) promoter is frequently employed to drive NT-3 expression in neuromuscular studies. This promoter ensures strong, sustained expression specifically in muscle tissue, creating an effective biological pump system for NT-3 production .
Vector Configuration:
Self-complementary AAV (scAAV) configurations are preferred over single-stranded designs, as demonstrated in recent studies utilizing scAAV1.tMCK.NT-3. This configuration bypasses the rate-limiting step of second-strand synthesis, accelerating the onset of transgene expression .
Dosage Optimization:
Effective protocols typically utilize 1 × 10^11 viral genome (vg) doses for intramuscular delivery into the gastrocnemius muscle. This dosage has demonstrated sufficient NT-3 production without apparent vector-related toxicity .
Administration Route:
Intramuscular injection into the gastrocnemius muscle has proven effective, creating a biological pump that produces measurable NT-3 levels detectable in serum through ELISA testing .
Transgene Optimization:
The human NT-3 coding sequence is commonly employed, which maintains cross-species efficacy while allowing distinction between endogenous and vector-derived neurotrophin for research purposes .
These parameters collectively determine the success of AAV-mediated NT-3 gene therapy in experimental mouse models.
Assessing the differential impact of NT-3 on neuronal versus glial pathology requires sophisticated histopathological and molecular analyses:
Histopathological Approaches:
Peripheral Nerve Analysis: Conduct qualitative and quantitative studies of peripheral nerves (sciatic and tibial) using both light and electron microscopy. Specific measurements should include:
Central Nervous System Analysis: For CNS tissues (brain and spinal cord), implement:
Neuromuscular Junction (NMJ) Analysis:
Evaluate NMJ innervation status using fluorescent staining techniques to distinguish between:
Fully innervated NMJs
Partially innervated NMJs
Denervated NMJs
This provides insight into the functional integration of neural improvements .
Molecular and Cellular Characterization:
Inflammatory Profiling: Measure expression of pro-inflammatory cytokines in brain and spinal cord tissues to assess neuroinflammatory impacts
Immune Cell Analysis: Quantify regulatory T cells in spleens and lymph nodes using flow cytometry to determine systemic immunomodulatory effects
Cell-Specific Markers: Employ immunostaining for neuronal, oligodendrocyte, and Schwann cell-specific markers to identify cell type-specific responses to NT-3 therapy
By implementing this comprehensive analytical approach, researchers can delineate the distinct effects of NT-3 on neurons versus supporting glial cells in both central and peripheral nervous systems.
Confirming successful NT-3 gene deletion in conditional knockout mouse models requires a multi-layered validation approach:
Genomic PCR Verification:
The most direct method utilizes PCR primers designed to flank the loxP sites to detect the deletion event. As illustrated in the research data, primers P1 (5′-TTACCTGCTCATGAAGAAGCCTTGTTGAGC-3′) and P3 (5′-GCATGGTTTCTGGCAGTCATAGATGCTTCC-3′) can be positioned to generate amplification products of distinctly different sizes depending on deletion status. Before Cre-mediated recombination, these primers would be approximately 5kb apart and would not generate a visible PCR product under standard cycling conditions. After successful deletion, the primers would be positioned only 200bp apart, yielding a detectable 200bp band when analyzed via agarose gel electrophoresis .
Recommended PCR Cycling Conditions:
Initial denaturation: 94°C for 30 seconds
Annealing: 65°C for 30 seconds
Extension: 72°C for 30 seconds
Validation of Cre Expression Timing:
To confirm appropriate spatiotemporal activity of the Cre recombinase, researchers should implement reporter systems such as crossing Cre-expressing lines with lacZ reporter mice. This approach can verify:
The onset of Cre expression (initial weak expression at E11.5 with robust expression by E12.5 for Synapsin I promoter-driven Cre)
The cell-type specificity of the deletion (strongest expression in differentiated neurons for neuronal-specific deletions)
Functional Validation:
Beyond genomic confirmation, functional assays should be employed to verify the physiological consequences of NT-3 deletion, such as:
Protein-level verification through ELISA or Western blot analysis to confirm reduced NT-3 expression
Electrophysiological studies to assess functional impacts on target systems
This multi-modal validation strategy ensures both genetic and functional confirmation of successful conditional NT-3 gene deletion.
Accurate measurement of NT-3 levels in mouse serum following gene therapy requires careful optimization of collection, processing, and analytical procedures:
Blood Collection Protocol:
Collect blood samples from anesthetized mice at defined timepoints (typically at study endpoints, approximately 6-7 months post-gene delivery)
Ensure consistent collection methodology (cardiac puncture or submandibular bleeding) across all experimental groups
Process samples immediately to prevent protein degradation
Serum Processing:
Allow blood to clot at room temperature for 30 minutes
Centrifuge at 2,000g for 15 minutes at 4°C
Carefully collect the serum supernatant
Either analyze immediately or store at -80°C (avoiding freeze-thaw cycles)
NT-3 Quantification by ELISA:
The enzyme-linked immunosorbent assay (ELISA) represents the gold standard for NT-3 quantification in serum samples:
Use commercially available ELISA kits specifically validated for mouse NT-3 detection
Include appropriate standards ranging from 0-1000 pg/mL
Run all samples in duplicate or triplicate to ensure reproducibility
Include internal quality controls in each assay run
Calculate NT-3 concentrations using standard curve interpolation
Data Interpretation Considerations:
Establish baseline NT-3 levels in wild-type and untreated disease model mice
Expected NT-3 levels following successful AAV1.tMCK.NT-3 gene therapy should show measurable elevation above baseline
Correlate serum NT-3 levels with functional and histopathological outcomes to establish dose-response relationships
This methodological approach ensures reliable quantification of circulating NT-3 following gene therapy, providing crucial pharmacodynamic data to interpret therapeutic efficacy.
Advanced histopathological techniques are essential for quantifying remyelination and axonal protection in NT-3 treated mouse models:
Tissue Preparation Protocol:
Perfusion and Fixation:
Transcardial perfusion with 4% paraformaldehyde
Post-fixation of dissected tissues (peripheral nerves, spinal cord, brain) for 24-48 hours
For electron microscopy: Fixation in 2.5% glutaraldehyde followed by 1% osmium tetroxide
Processing and Sectioning:
For light microscopy: Paraffin embedding with 5-10μm sections
For electron microscopy: Resin embedding with ultrathin (70-90nm) sections
For immunofluorescence: Cryoprotection and 10-30μm frozen sections
Quantitative Assessment Techniques:
Peripheral Nerve Analysis:
G-ratio Measurement: Calculate the ratio of axon diameter to total fiber diameter (including myelin) using digital imaging software. Normal G-ratios range from 0.6-0.7; deviations indicate abnormal myelination .
Axon Size Distribution: Generate histograms categorizing axons by diameter (typically in 1μm bins from 1-15μm). NT-3 treatment typically increases the proportion of axons in the 3-6μm diameter range, indicating improved axonal maturation .
Schwann Cell Density: Quantify using S100β immunostaining or through morphological identification in semithin sections, measured as cells per mm² .
Central Nervous System Analysis:
Myelin Quantification: Employ Luxol Fast Blue staining or myelin basic protein (MBP) immunohistochemistry with digital image analysis to calculate percentage of myelinated area .
Axonal Integrity Assessment: Quantify using SMI-31 or SMI-32 immunostaining to detect phosphorylated and non-phosphorylated neurofilaments respectively .
Inflammatory Infiltrate Quantification: Use H&E staining or immunohistochemistry for CD3+ T cells, CD11b+ macrophages/microglia to assess inflammatory burden .
Neuromuscular Junction Analysis:
These comprehensive histopathological approaches provide quantitative assessment of NT-3's effects on myelination and axonal integrity in multiple neural compartments.
Analysis of sex-specific differences in response to NT-3 treatment requires methodical experimental design and statistical approaches:
Experimental Design Requirements:
Balanced Sex Distribution:
Consistent Age Matching:
Estrous Cycle Consideration:
Document estrous cycle stage during functional testing in female mice
Consider pooling data across cycle stages or stratifying by cycle phase depending on research questions
Statistical Analysis Framework:
Two-Way ANOVA Approach:
Use treatment status (NT-3 treated vs. untreated) as the first factor
Use sex (male vs. female) as the second factor
Test for main effects of both treatment and sex
Critically evaluate treatment × sex interaction effects
Post-Hoc Testing:
Implement Tukey's or Bonferroni correction for multiple comparisons
Conduct specific contrasts between:
Treated males vs. untreated males
Treated females vs. untreated females
Treated males vs. treated females
Untreated males vs. untreated females
Interpretation Guidelines:
Based on published findings, researchers should be aware that:
Female mice generally demonstrate better baseline performance on functional tests like rotarod across all experimental groups
This sex difference is statistically significant in untreated cohorts
NT-3 gene therapy responses may show sex-specific patterns, though published data suggests no significant sex-specific response to NT-3 therapy in some models
Data Presentation:
Present sex-specific data in supplementary figures with separate analysis while maintaining combined data in main figures when no significant interaction effects are observed .
This analytical framework ensures rigorous evaluation of potential sex-specific responses to NT-3 treatment, addressing an important dimension of experimental variability.
Analyzing longitudinal functional outcomes following NT-3 gene therapy requires sophisticated statistical approaches that account for repeated measures and between-subject factors:
Recommended Statistical Models:
Mixed-Effects Linear Models:
Include fixed effects for treatment group, time point, and their interaction
Incorporate random effects for individual subjects to account for within-subject correlation
Specify appropriate covariance structure (typically unstructured or autoregressive)
This approach accommodates missing data points more effectively than traditional repeated-measures ANOVA
Repeated Measures ANOVA:
Appropriate when complete data is available for all subjects at all time points
Include between-subjects factor (treatment group) and within-subjects factor (time)
Test for main effects and interaction effects
Use Mauchly's test to verify sphericity assumption and apply Greenhouse-Geisser correction when violated
Key Time Point Comparisons:
For NT-3 gene therapy studies, critical comparisons include:
Baseline (pre-treatment, typically 1 month of age)
Mid-point assessment (3 months post-treatment)
Sample Analysis Table for Rotarod Performance:
Time Point | NT-3 Treated (n=15) | Untreated Controls (n=16) | p-value |
---|---|---|---|
Baseline | 47.33 ± 2.09 s | 51.63 ± 2.25 s | 0.1750 |
Mid-point | 53.20 ± 1.74 s | 45.38 ± 2.31 s* | 0.0112 |
End-point | 48.67 ± 2.26 s | 41.69 ± 2.45 s* | 0.0381 |
*Significantly different from baseline (p < 0.05)
Post-Hoc Analysis:
For significant interaction effects, conduct pairwise comparisons with appropriate adjustment for multiple comparisons (Bonferroni or Tukey)
Calculate percent change from baseline for each group at each time point
Compare treatment effect sizes at different time points to assess temporal dynamics of therapeutic response
This statistical framework provides robust analysis of longitudinal functional outcomes while accounting for the complex data structure inherent in NT-3 gene therapy studies.
Reconciling discrepancies between electrophysiological and behavioral outcomes in NT-3 mouse studies requires systematic analysis of potential mechanistic explanations:
Common Discrepancy Patterns:
Improved Electrophysiology Without Behavioral Gains:
May indicate subclinical neural improvements insufficient to manifest as functional changes
Could reflect compensatory mechanisms masking behavioral deficits despite improved conduction
Behavioral Improvements Without Electrophysiological Changes:
May suggest central adaptation or compensation at supraspinal levels
Could indicate improvements in non-assessed neural parameters
Reconciliation Approaches:
Comprehensive Neural Pathway Analysis:
Temporal Resolution Enhancement:
Implement more frequent assessment time points to detect transient changes
Consider that electrophysiological improvements may precede behavioral changes
Analyze recovery slopes rather than single time point comparisons
Multi-Parameter Correlation Analysis:
Calculate correlation coefficients between electrophysiological measures and behavioral outcomes
Generate scatter plots with regression lines to visualize relationships
Identify potential threshold effects where electrophysiological improvements must reach certain levels before behavioral changes emerge
Methodological Sensitivity Assessment:
Evaluate whether behavioral tests are sufficiently sensitive to detect subtle improvements
Consider ceiling or floor effects in behavioral assays
Implement more challenging behavioral paradigms to detect subtle functional changes
Interpretation Framework:
When discrepancies occur, researchers should consider that:
NT-3 effects on synaptic transmission and plasticity may be distinct from effects on axonal conduction
The neuron-specific knockout studies demonstrated that neuronal NT-3 is not required for synaptic transmission or certain forms of hippocampal plasticity
Peripheral NT-3 delivery may produce different effects than endogenous neuronal NT-3 production
Both central and peripheral effects of NT-3 may contribute to behavioral outcomes through distinct mechanisms
This systematic approach to reconciling discrepancies provides deeper insight into NT-3's multifaceted mechanisms of action across different neural systems.
Based on current research findings, several promising translational pathways are emerging from NT-3 mouse model research:
Multiple Sclerosis Applications:
NT-3 gene therapy has demonstrated significant potential for treating multiple sclerosis through multiple mechanisms:
Improved remyelination and axonal protection in the central nervous system
Reduced expression of pro-inflammatory cytokines in brain and spinal cord
Increased regulatory T cell populations in peripheral immune tissues
These combined neuroprotective and immunomodulatory effects resulted in improved clinical scores and functional performance in EAE mouse models
The translational implication is particularly relevant for progressive MS forms where current therapies show limited efficacy. AAV-delivered NT-3 could potentially address both the inflammatory and neurodegenerative components of chronic progressive MS .
Peripheral Neuropathy Applications:
For hereditary peripheral neuropathies like Charcot-Marie-Tooth disease, NT-3 therapy has shown promise through:
Improved myelination of peripheral nerves
Enhanced regenerative activity in distal nerve segments
Increased Schwann cell density and differentiation
Preservation of neuromuscular junction innervation
Significant improvements in nerve conduction velocity and functional outcomes
These findings suggest potential applications for various demyelinating peripheral neuropathies, including CMT subtypes, diabetic neuropathy, and chemotherapy-induced peripheral neuropathy.
Delivery System Innovation:
The intramuscular delivery approach using AAV1.tMCK vectors represents a significant translational advantage, as it:
Avoids the need for direct neural tissue injection
Creates a sustainable biological pump for continuous NT-3 production
Has demonstrated long-term efficacy (6+ months) in mouse models
Utilizes AAV vectors with established safety profiles in human clinical applications
This delivery system could potentially be adapted for human use with minimal modification, leveraging existing clinical experience with AAV vectors.
As research progresses, these translational applications hold considerable promise for addressing currently untreatable or poorly managed neurological conditions.
Several methodological improvements could significantly enhance the reliability and translational value of NT-3 mouse model research:
Standardized Outcome Measure Protocols:
Establish consensus protocols for behavioral testing, including standardized:
Rotarod acceleration parameters
Grip strength measurement techniques
Timing and frequency of assessments
Environmental conditions during testing
Develop consistent electrophysiological recording protocols with:
Standardized electrode placement
Uniform stimulation parameters
Consistent temperature control measures
Reproducible analysis algorithms
Enhanced Dosing and Delivery Optimization:
Implement dose-response studies with multiple vector concentrations
Evaluate alternative muscle groups for intramuscular delivery
Compare serotype efficacy (beyond AAV1) for NT-3 delivery
Investigate the impact of delivery timing relative to disease onset
Improved Disease Model Characterization:
Establish more precise natural history studies of untreated disease models
Incorporate genetically diverse background strains to assess treatment robustness
Develop more chronic progressive models that better mimic human disease timelines
Implement aging studies to determine efficacy in older animals
Advanced Analytical Approaches:
Utilize unbiased stereological techniques for histopathological quantification
Implement machine learning algorithms for automated analysis of complex histological datasets
Develop computational models to predict treatment outcomes based on baseline characteristics
Incorporate single-cell RNA sequencing to identify cell type-specific responses to NT-3 therapy
Enhanced Reproducibility Measures:
Pre-register experimental protocols and analysis plans
Implement larger sample sizes based on appropriate power calculations
Utilize multicenter collaborative approaches to validate key findings
Report comprehensive negative and neutral findings alongside positive results
Translational Readiness Improvements:
Incorporate humanized mouse models expressing human NT-3 receptors
Develop human-equivalent dosing models
Establish clear biomarkers correlating with functional improvements
Validate findings across multiple disease models to demonstrate mechanism generalizability
These methodological enhancements would collectively strengthen the reliability and translational potential of NT-3 mouse model research.
Despite significant progress, several critical limitations persist in understanding the molecular mechanisms of NT-3 action in mouse models:
Receptor Specificity and Downstream Signaling:
Current research has not fully elucidated the relative contributions of different NT-3 receptors (TrkC, p75NTR, and TrkB with lower affinity) to therapeutic outcomes. This limitation creates uncertainties regarding:
Which receptor subtype mediates specific aspects of NT-3's therapeutic effects
How receptor expression patterns influence tissue-specific responses
Whether different disease states alter receptor availability or signaling efficiency
The detailed intracellular signaling cascades that translate receptor activation into therapeutic outcomes
Cell Type-Specific Responses:
Limited understanding exists regarding how different neural cell populations respond to NT-3 therapy:
The differential response of oligodendrocytes versus neurons to NT-3 in demyelinating conditions
Whether NT-3's effects on Schwann cells are direct or mediated through axonal signals
How NT-3 influences microglial and astroglial activation states
The mechanisms by which NT-3 affects T cell populations and regulatory immune cells
Systemic Versus Local Effects:
The relative contribution of systemic versus localized NT-3 effects remains poorly understood:
How circulating NT-3 crosses the blood-brain barrier, particularly during inflammation
Whether peripheral immune modulation or direct CNS effects predominate in MS models
The transport mechanisms that deliver muscle-produced NT-3 to peripheral nerves
The concentration gradients necessary for therapeutic efficacy in different tissues
Temporal Dynamics:
Limited data exists on the temporal aspects of NT-3 signaling:
The optimal therapeutic window for intervention in different disease models
Whether continuous versus pulsatile NT-3 delivery produces different outcomes
How long-term NT-3 exposure affects receptor expression and sensitivity
The sustainability of therapeutic effects after treatment cessation
Genetic and Environmental Modifiers:
The impact of genetic background and environmental factors on NT-3 efficacy remains understudied:
How genetic variants in NT-3 signaling pathways affect therapeutic responses
Whether environmental factors like exercise or diet modify NT-3 effectiveness
How aging influences NT-3 receptor expression and downstream signaling efficiency
Addressing these limitations would provide crucial insights into NT-3's mechanisms of action and help optimize therapeutic strategies for clinical translation.
Neurotrophin-3 (NT-3) is a member of the neurotrophin family, which includes nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin-4 (NT-4). These proteins play crucial roles in the development, maintenance, and function of the nervous system by promoting the survival, differentiation, and growth of neurons.
NT-3 is a protein that is encoded by the NTF3 gene. The protein is initially synthesized as a precursor molecule, which is then cleaved to produce the mature, biologically active form. The mature NT-3 protein consists of 119 amino acids and has a molecular weight of approximately 13.6 kDa .
NT-3 exerts its effects by binding to specific receptors on the surface of target cells. The primary receptor for NT-3 is TrkC (tropomyosin receptor kinase C), although it can also interact with TrkA and TrkB receptors to a lesser extent. Upon binding to its receptor, NT-3 activates intracellular signaling pathways that promote neuronal survival, differentiation, and growth .
NT-3 is known to selectively promote the differentiation and survival of specific neuronal subpopulations in both the central and peripheral nervous systems. It has been shown to mediate neurite outgrowth and induce phosphorylation of ERK1/2 MAPK in TrkC-transfected cells . These activities are essential for the proper development and function of the nervous system.
Recombinant NT-3 is produced using various expression systems, including E. coli. The recombinant protein is typically purified to high levels of purity (>95%) and is free from endotoxins, making it suitable for research applications . The recombinant mouse NT-3 protein is used in various experimental settings to study its effects on neuronal cells and to investigate potential therapeutic applications for neurodegenerative diseases and nerve injuries.
Recombinant NT-3 has been widely used in research to study its role in neuronal development and function. It has been employed in cell culture experiments to promote neurite outgrowth and to investigate the signaling pathways activated by NT-3. Additionally, recombinant NT-3 has potential therapeutic applications in the treatment of neurodegenerative diseases, such as Alzheimer’s disease, and in promoting nerve regeneration following injury .