Protein Structure: A receptor tyrosine kinase with extracellular ligand-binding domains and intracellular kinase domains .
Primary Ligand: Nerve Growth Factor (NGF), which binds to NTRK1 to activate downstream signaling pathways .
A study using rat C17.2 NSCs demonstrated that NTRK1 overexpression enhances differentiation into cholinergic neurons under NGF stimulation . Key results include:
| Parameter | NTRK1 Overexpression (NGF+) | Control (NGF+) | Fold Change |
|---|---|---|---|
| ChAT+ Cells | 45% | 15% | 3x |
| NeuN+ Neurons | Increased | Basal levels | N/A |
Mechanism: NTRK1 activation triggers SHC1/FRS2-mediated Ras-MAPK signaling, driving cell survival and differentiation .
NTRK1 knockdown in mice induces deficits in spatial memory and working/reference memory errors, as measured by:
Pre-treatment with O304 (AMPK activator) or rapamycin (autophagy inducer) restores mitophagy and neuronal viability in NTRK1-deficient mice, suggesting potential therapeutic targets .
| Intervention | Effect on Mitophagy | Outcome |
|---|---|---|
| O304 | ↑ LC3-II, ↑ PINK1 | Reverses neuronal apoptosis |
| Rapamycin | ↑ Autophagosome formation | Enhances ATP production |
NTRK1, a key player in the development and survival of nerve cells (neurons), is especially crucial for sensory neurons that transmit data related to touch, pain, and temperature. This protein, primarily found on the surface of cells like sensory neurons, acts as a kinase enzyme. It modifies the activity of various proteins by adding a phosphate group along with oxygen atoms at specific sites, a process known as phosphorylation. NTRK1 is activated upon binding with nerve growth factor beta, which triggers the protein to phosphorylate itself (autophosphorylation). This activated NTRK1 then phosphorylates other proteins, a crucial step for cell growth and survival.
Recombinant NTRK1 protein, produced in HEK cells from Rat, is a single, non-glycosylated polypeptide chain. This protein consists of 623 amino acids (specifically, amino acids 35 to 418) and has a molecular weight of 69 kDa. It is fused to a 239 amino acid long human IgG-His-tag at its C-terminus and is purified using proprietary chromatographic techniques.
The NTRK1 protein is supplied as a solution with a concentration of 1mg/ml. This solution is prepared in Phosphate-Buffered Saline (pH 7.4) and contains 10% Glycerol.
While NTRK1 remains stable for up to 1 week at a refrigerated temperature of 4°C, it is recommended to store it at a temperature below -18°C for optimal long-term storage. To further enhance its stability during long-term storage, consider adding a carrier protein like HSA or BSA at a concentration of 0.1%. It is important to avoid repeated freeze-thaw cycles to maintain the protein's integrity.
The biological activity of this protein is assessed based on its ability to inhibit NGF-induced proliferation in TF1 human erythroleukemic cells. This assay is carried out in the presence of 0.5 ng/ml of rat NGF. The ED50 for this inhibition is found to be within the range of ≤ 5 ng/ml.
Analysis using SDS-PAGE indicates that the purity of this protein is greater than 90%.
TRKA, TRK-A, NTRK-1.
SCRETCCPVG PSGLRCTRAG TLNTLRGLRG AGNLTELYVE NQRDLQRLEF EDLQGLGELR SLTIVKSGLR FVAPDAFHFT PRLSHLNLSS NALESLSWKT VQGLSLQDLT LSGNPLHCSC ALLWLQRWEQ EDLCGVYTQK LQGSGSGDQF LPLGHNNSCG VPSVKIQMPN DSVEVGDDVF LQCQVEGQAL QQADWILTEL EGTATMKKSG DLPSLGLTLV NVTSDLNKKN VTCWAENDVG RAEVSVQVSV SFPASVHLGK AVEQHHWCIP FSVDGQPAPS LRWFFNGSVL NETSFIFTQF LESALTNETM RHGCLRLNQP THVNNGNYTL LAANPYGQAA ASIMAAFMDN PFEFNPEDPI PVSFSPVDTN STSRDPVEKK DETP LEPKSC DKTHTCPPCP APELLGGPSV FLFPPKPKDT LMISRTPEVT CVVVDVSHED PEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLH QDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYT LPPSRDELTK NQVSLTCLVK GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHE ALHNHYTQKS LSLSPGKHHH HHH
NTRK1 (Neurotrophic Receptor Tyrosine Kinase 1), also known as TrkA, is a high-affinity receptor for nerve growth factor that plays essential roles in the development and function of the cholinergic nervous system in rats . The protein is expressed in specific and defined brain areas, including the striatum, basal forebrain, and paraventricular thalamic nucleus (PVT) . Beyond its neurological functions, NTRK1 also contributes to inflammatory processes and cell proliferation in other tissues.
In rat models, NTRK1 has been demonstrated to promote mesangial cell proliferation and inflammation in glomerulonephritis . Studies have shown that upregulation of NTRK1 enhances cell viability, proliferation, and expression of pro-inflammatory factors while activating the STAT3, p38, and ERK signaling pathways in mesangial cells . This makes NTRK1 an important target for research in both neurological and kidney disease models.
Unlike in some mouse models where NTRK1 knockout leads to neonatal lethality, rats exhibit a more complex phenotype when NTRK1 is manipulated, highlighting the importance of species-specific considerations in experimental design .
When detecting NTRK1 protein in rat tissues, researchers should carefully validate their antibodies for specificity. Based on comparative studies of commercial antibodies, not all options provide reliable results . Western blotting remains a foundational method for NTRK1 detection, but requires careful validation:
Antibody selection: Only certain commercial antibodies demonstrate true specificity. In one study, only one out of seven commercial antibodies showed adequate specificity in western blots, with specific bands absent in knockout samples .
Control samples: To ensure specificity, include positive controls (tissues known to express NTRK1, such as striatum or basal forebrain) and negative controls when possible.
Immunohistochemistry applications: For localization studies, antibodies that demonstrate specificity in western blotting should be further validated for immunohistochemistry. Distinct signals should be observable in regions with known NTRK1 expression, such as the striatum and basal forebrain .
Regional expression patterns: When validating detection methods, consider the characteristic expression pattern of NTRK1 in brain structures like the paraventricular thalamic nucleus (PVT), which shows differential expression with high levels in the anterior region and low levels in the posterior region .
For optimal reliability, researchers should perform preliminary validation studies with their specific antibodies and tissues of interest before proceeding with larger-scale experiments.
To establish a rat model for studying NTRK1 function, particularly in mesangial proliferative glomerulonephritis (MsPGN), follow these methodological approaches:
Standard MsPGN Rat Model:
Animal selection: Use male SD rats weighing 200-220g, housed under controlled conditions (22°C, 12-h/12-h light/dark cycle) .
Model induction: Administer a single injection of Thy1.1 monoclonal antibody (2.5 mg/kg) via the tail vein . This antibody is typically generated by OX-7 cells.
Timeline: For time-course studies, collect kidney tissues and isolate glomeruli at specific intervals (e.g., days 3, 7, and 14) after injection .
Controls: Include rats injected with equivalent volumes of normal saline as controls .
NTRK1 Knockdown Model:
Prepare lentiviral vectors: Use Ntrk1-RNAi lentivirus (1 × 10^9 TU/mL) for knockdown and negative control lentivirus for controls .
Administration protocol: Inject the lentivirus via the tail vein three times at 5-day intervals .
Disease induction: Following lentiviral treatment, inject Thy1.1 monoclonal antibody (2.5 mg/kg) via the tail vein .
Tissue collection: Euthanize rats on day 7 post-antibody injection to collect kidney tissues and isolate glomeruli .
These models allow for investigation of NTRK1's role in disease pathogenesis and potential therapeutic targeting. Researchers should monitor urinary protein levels and perform histological analyses (including periodic acid-Schiff staining and immunohistochemistry) to confirm successful model establishment .
NTRK1 regulates several important signaling pathways in rat tissues, particularly in mesangial cells:
STAT3 Pathway:
NTRK1 upregulation activates the STAT3 signaling pathway in mesangial cells, contributing to inflammatory responses and proliferation . This activation appears to be cell-type specific and context dependent.
MAPK Pathways:
p38 MAPK: NTRK1 promotes activation of the p38 signaling pathway, which can be experimentally verified through inhibition studies using SB202190 (p38 inhibitor) . When this pathway is blocked, the effects of NTRK1 on mesangial cell viability, proliferation, and inflammatory response are reversed .
ERK Pathway: NTRK1 upregulation also activates the ERK signaling pathway in rat mesangial cells. Treatment with PD98059 (ERK inhibitor) can reverse NTRK1-mediated effects on cell proliferation and inflammation .
These pathways function as mediators between NTRK1 activation and downstream cellular responses. When studying these pathways, researchers should employ appropriate inhibitors as experimental controls and validate pathway activation through phosphorylation-specific antibodies in western blotting experiments .
The interconnection between these pathways suggests complex regulatory mechanisms, and researchers should consider potential crosstalk when interpreting experimental results.
NTRK1 plays a significant role in MsPGN pathogenesis through multiple mechanisms:
Enhanced Mesangial Cell Proliferation:
NTRK1 expression is upregulated in the glomeruli of MsPGN rat models . This upregulation directly promotes mesangial cell proliferation, a hallmark of MsPGN pathology. Experimentally, knockdown of NTRK1 reduces mesangial cell proliferation in the glomeruli of MsPGN rats and decreases Ki67 expression in renal tubules .
Inflammatory Response Augmentation:
NTRK1 upregulation increases the expression of pro-inflammatory factors in glomeruli of MsPGN rats . When NTRK1 is knocked down, there is a corresponding reduction in these pro-inflammatory factors, suggesting a direct regulatory role in the inflammatory component of MsPGN .
Clinical Manifestations:
MsPGN rat models with elevated NTRK1 expression show increased urinary protein levels, indicating impaired renal function . NTRK1 knockdown reduces urine protein levels, providing evidence for its pathogenic role .
Signaling Pathway Activation:
NTRK1 activates multiple signaling cascades, including STAT3, p38, and ERK pathways, in MsPGN . These pathways collectively contribute to the proliferative and inflammatory phenotype observed in MsPGN. Inhibition of p38 and ERK pathways reverses the effects of NTRK1 on mesangial cell proliferation and inflammation, suggesting these are key mediators of NTRK1's pathogenic effects .
These findings position NTRK1 as a potential therapeutic target for MsPGN. By understanding its role in disease pathogenesis, researchers can develop targeted interventions to mitigate disease progression.
Studying NTRK1 knockdown in rat models presents several methodological challenges that researchers should consider:
Delivery Method Optimization:
Lentiviral vector administration requires careful optimization for efficient delivery. The reported protocol uses three injections at 5-day intervals via the tail vein , but tissue-specific targeting may require different approaches.
Alternative delivery methods such as adeno-associated virus (AAV) vectors or direct tissue injection may be needed for certain applications.
Knockdown Validation:
Researchers must thoroughly validate knockdown efficiency using multiple techniques:
qRT-PCR for mRNA level validation
Western blotting for protein level confirmation
Immunohistochemistry for spatial verification
Timing Considerations:
The timing of knockdown relative to disease induction is critical. In MsPGN models, lentiviral delivery preceded Thy1.1 antibody injection , but prevention versus treatment protocols may require different timelines.
Duration of knockdown effects must be established, as transient knockdown may yield different results than sustained knockdown.
Potential Compensatory Mechanisms:
NTRK family redundancy may lead to compensation by NTRK2 or NTRK3 when NTRK1 is knocked down.
Researchers should assess all three NTRK family members when studying knockdown effects.
Phenotypic Interpretation:
Complete knockdown versus partial knockdown may produce qualitatively different phenotypes.
Developmental versus acute knockdown effects may differ substantially due to NTRK1's role in development.
Control Design:
Proper controls must include:
Negative control lentivirus to account for viral effects
Sham operation controls to account for procedural effects
Non-target tissue controls to verify specificity
Addressing these challenges requires careful experimental design and validation at multiple levels to ensure reliable and interpretable results when studying NTRK1 knockdown effects.
Distinguishing between the three NTRK family members requires a multi-method approach:
mRNA Expression Analysis:
Quantitative real-time PCR (qRT-PCR) using gene-specific primers provides quantitative information about differential expression .
RNA sequencing data analysis reveals that NTRK1, NTRK2, and NTRK3 show distinct expression patterns across tissues. Based on human data (which may inform rat studies), these genes are typically downregulated in tumor tissues compared to normal tissues, with NTRK1, NTRK2, and NTRK3 showing significant downregulation in 20, 17, and 22 tumor types, respectively .
Protein Detection:
Western blotting with carefully validated antibodies specific to each NTRK family member is essential .
Immunohistochemistry allows for spatial resolution of expression patterns:
Genomic Profiling:
Analysis of genomic alterations reveals distinct patterns for each NTRK gene. In human studies that may inform rat research, alterations in NTRK1, NTRK2, and NTRK3 have been found in 645, 451, and 584 patients respectively, with only 21 individuals showing alterations in all three genes simultaneously .
Domain-Specific Mutation Analysis:
The three NTRK proteins have different domain structures with mutations clustering in specific regions:
NTRK1/2/3 mutations are primarily located in the Pkinase-Tyr and LRR-8 domains
The I-set domain in NTRK2/3 contains approximately 25% of mutations
The Ig-2 domain is specific to NTRK1 and contains about 5% of mutations
When designing experiments to distinguish between the three NTRK family members, researchers should employ multiple complementary approaches and include appropriate controls to confirm specificity of detection methods.
NTRK1 fusion genes have significant implications in cancer biology, though most data comes from human studies that can inform rat model development:
Fusion Gene Identification:
RNA sequencing is the gold standard for identifying NTRK1 fusion transcripts. Pipeline for RNA sequencing Data Analysis (PRADA) can identify fusion transcripts comprehensively with high confidence .
Confidence tiers (1-4) are used to rank fusion transcripts based on supporting evidence, with tier 1 representing strongest confidence .
Research Model Development:
To study NTRK1 fusions in rats, researchers can:
Generate transgenic rat models expressing specific human NTRK1 fusions
Use CRISPR-Cas9 to create endogenous fusions in rat genomes
Employ patient-derived xenograft approaches using immunocompromised rats
Clinical Relevance and Inhibitor Testing:
NTRK1 fusion genes are targetable by first-generation NTRK inhibitors like larotrectinib and entrectinib . Rat models can be used to:
Test efficacy of these inhibitors against specific NTRK1 fusions
Investigate mechanisms of resistance
Evaluate adverse effects of these inhibitors in vivo
Adverse Event Monitoring:
When testing NTRK inhibitors in rat models, researchers should monitor for specific adverse events identified in humans:
Most adverse events occur within the first month of treatment (60.5% for entrectinib; 46.9% for larotrectinib)
Late-onset adverse events can still occur after one year (10.3% for entrectinib; 8.9% for larotrectinib)
The Weibull distribution analysis suggests an "early failure type" pattern for both inhibitors, where adverse event incidence decreases over time
Translational Considerations:
When designing rat studies focused on NTRK1 fusions, consider overlapping adverse events identified for both entrectinib and larotrectinib, including renal impairment, taste disorder, disease progression, edema, and ascites .
Researchers often encounter contradictory data regarding NTRK1 function across different rat disease models. The following experimental approaches can help resolve these contradictions:
Comprehensive Model Comparison:
Direct side-by-side comparison of multiple disease models using standardized methods
Detailed characterization of baseline NTRK1 expression and activity in each model
Systematic documentation of strain, age, sex, and environmental variables that may influence results
Cell Type-Specific Analysis:
Contradictions may arise from cell type heterogeneity. Approaches to address this include:
Single-cell RNA sequencing to resolve cell-specific expression patterns
Cell type-specific conditional knockdown/knockout models
Co-localization studies with cell type-specific markers
Temporal Dynamics Assessment:
NTRK1 function may change over disease progression:
Distinguish between acute and chronic effects using inducible expression systems
Compare models at equivalent disease stages rather than absolute time points
Signaling Pathway Dissection:
Contradictions may result from differential pathway engagement:
Detailed phosphoproteomic analysis of downstream signaling in each model
Selective pathway inhibition using compounds like SB202190 (p38 inhibitor) and PD98059 (ERK inhibitor)
Genetic manipulation of specific pathway components to identify critical nodes
Statistical and Methodological Approaches:
Power analysis to ensure adequate sample sizes for detecting effects
Blinded assessment of outcomes to reduce bias
Use of multiple, complementary endpoint measurements
Meta-analysis of published literature with detailed examination of methodological differences
External Validation:
Collaborate with independent laboratories to reproduce key findings
Compare rat data with findings from other species including mice and human samples
Validate in vitro findings in vivo and vice versa
By systematically implementing these approaches, researchers can identify sources of contradiction and develop a more nuanced understanding of context-dependent NTRK1 functions across different rat disease models.
Optimal antibody-based detection of NTRK1 in rat tissue samples requires careful attention to several technical parameters:
Antibody Selection and Validation:
Commercial antibody evaluation: Only select antibodies that have been validated with proper controls. Evidence suggests only a small percentage of commercial antibodies show true specificity for NTRK1 .
Validation protocol: Test antibodies on tissues from NTRK1 knockout models or after NTRK1 knockdown to confirm specificity .
Cross-reactivity assessment: Verify that selected antibodies do not cross-react with NTRK2 or NTRK3.
Western Blotting Optimization:
Sample preparation: Fresh tissue extraction with protease inhibitors is critical for preserving NTRK1 integrity.
Protein loading: 30-50 μg of total protein per lane is typically optimal for detection.
Transfer conditions: Use PVDF membranes for better protein retention and signal strength.
Blocking: 5% non-fat milk in TBST for 1 hour at room temperature is generally effective.
Primary antibody incubation: Overnight at 4°C at optimized dilution (typically 1:500 to 1:1000).
Detection system: HRP-conjugated secondary antibodies with enhanced chemiluminescence provide good sensitivity.
Immunohistochemistry Protocol:
Fixation: 4% paraformaldehyde for 24-48 hours preserves antigenicity while maintaining tissue structure.
Antigen retrieval: Citrate buffer (pH 6.0) with heat-induced epitope retrieval improves detection.
Background reduction: Use 0.3% H₂O₂ to block endogenous peroxidase activity.
Antibody concentration: Titrate antibodies to determine optimal concentration (typically 1:100 to 1:500).
Signal amplification: Consider using tyramide signal amplification for low-abundance targets.
Controls: Include positive controls (striatum, basal forebrain) and negative controls .
Regional Considerations:
When performing immunohistochemistry, be aware of the expected regional expression patterns:
Strong signals should be present in striatum and basal forebrain .
The paraventricular thalamic nucleus (PVT) shows an anteroposterior gradient with higher expression in anterior regions .
Use these known expression patterns as internal controls for staining quality.
Following these optimized conditions will significantly improve the reliability and reproducibility of NTRK1 detection in rat tissue samples.
Accurate quantification of NTRK1 expression changes in rat models requires a multi-method approach:
mRNA Quantification Methods:
qRT-PCR analysis:
RNA-Seq analysis:
Normalize read counts appropriately (RPKM/FPKM/TPM)
Verify expression changes with differential expression analysis tools
Validate key findings with qRT-PCR
Protein Quantification Methods:
Western blotting:
ELISA assays:
Commercial NTRK1 ELISA kits can provide quantitative data
Generate standard curves for each experiment
Run samples in duplicate or triplicate
Immunohistochemistry Quantification:
Semi-quantitative scoring:
Establish clear scoring criteria (0-3+ or percentage positive cells)
Have multiple blinded observers score samples
Calculate inter-observer agreement statistics
Digital image analysis:
Capture images under standardized conditions
Use software to quantify staining intensity and distribution
Set consistent thresholds across all experimental groups
Report both intensity and percentage of positive area
Statistical Analysis:
Determine appropriate statistical tests based on data distribution
Calculate sample sizes using power analysis
Account for potential confounding variables
Consider using mixed-effects models for longitudinal studies
Validation Approaches:
Confirm expression changes using multiple techniques
Include appropriate positive and negative controls
Validate antibody specificity using knockdown/knockout samples
Correlate mRNA and protein expression changes when possible
By combining these methodological approaches and rigorous quantification techniques, researchers can accurately measure changes in NTRK1 expression in experimental rat models with high confidence and reproducibility.
Neurotrophic Receptor Tyrosine Kinase 1 (NTRK1), also known as Tropomyosin Receptor Kinase A (TrkA), is a protein encoded by the NTRK1 gene. This receptor is a member of the neurotrophic tyrosine kinase receptor (NTKR) family and plays a crucial role in the development and function of the nervous system . NTRK1 is primarily known for its high affinity binding to Nerve Growth Factor (NGF), which is essential for the survival, differentiation, and maintenance of neurons .
NTRK1 is a membrane-bound receptor that, upon binding with its ligand NGF, undergoes autophosphorylation. This phosphorylation activates several downstream signaling pathways, including the MAPK pathway, which is involved in cell differentiation and survival . The receptor is crucial for the development of sensory neurons and plays a significant role in specifying sensory neuron subtypes .
The NTRK1 receptor is essential for the development and survival of nerve cells, particularly sensory neurons that transmit information about sensations such as pain, temperature, and touch . Mutations in the NTRK1 gene can lead to various disorders, including congenital insensitivity to pain with anhidrosis (CIPA) and certain types of cancer, such as papillary thyroid carcinoma .
Recombinant NTRK1, particularly from rat models, is widely used in research to study its function and role in various biological processes. The recombinant form allows for detailed analysis of the receptor’s structure, function, and interactions with other proteins. It is also used to investigate the effects of mutations and to develop potential therapeutic interventions for related disorders.