Recombinant NTF4 is available in two formulations:
Purpose: Enhances stability and shelf-life for cell culture or ELISA standards.
Reconstitution: 50 μg/mL in PBS containing ≥0.1% serum albumin .
Purpose: Ideal for applications where BSA might interfere (e.g., structural studies).
NTF4 activates TrkB receptors, triggering downstream pathways such as ERK1/2 MAPK and PI3K/Akt, which regulate neuronal survival and differentiation . Key functional data:
Neurogenesis: NTF4 enhances neuronal differentiation in embryonic neural stem cells by suppressing Notch and IL-6 signaling .
Neuroprotection: Reduces apoptosis in dopamine neurons and improves motor neuron innervation .
Glaucoma: Mutations in NTF4 (e.g., p.Gly157Ala) correlate with impaired TrkB activation and disease progression .
Pulmonary Fibrosis: Dysregulated TrkB/NTF4 signaling contributes to fibroblast proliferation .
Mechanistic Insights:
Disease Associations:
Human Neurotrophin-4 (NT-4) is a member of the NGF family of neuronal and epithelial growth factors. The protein is synthesized as a 210 amino acid (aa) precursor comprising:
The structural hallmark of NT-4, like all neurotrophins, is the characteristic arrangement of six conserved cysteine residues that form three disulfide bonds, known as the cysteine knot. This structure has also been found in other growth factors such as PDGF . Mature human NT-4 shares 48-52% amino acid sequence identity with human β-NGF, BDNF, and NT-3 .
Recombinant NT-4 retains the key functional properties of endogenous NT-4 when properly produced and folded. Most commercially available recombinant human NT-4 is produced in insect cell lines like Spodoptera frugiperda (Sf21) using baculovirus expression systems . The functionality of recombinant NT-4 can be verified through:
TrkB receptor activation assays - Proper recombinant NT-4 should induce receptor dimerization and activation of TrkB
ERK1/2 MAPK phosphorylation - As demonstrated in TrkB-transfected HEK-293 cells, where NT-4 induces concentration-dependent ERK1/2 activation that can be blocked by K252a (TrkB inhibitor)
Neuronal survival assays - Effective concentration for promoting neuronal survival typically ranges from 0.3-3 ng/mL
When using recombinant NT-4, the biological activity should be validated through these functional assays rather than assumed based solely on protein concentration.
NT-4 has several unique characteristics that distinguish it from other neurotrophins:
Receptor specificity: NT-4 binds primarily to TrkB (shared with BDNF) and to p75NTR (which binds all neurotrophins)
Expression pattern: NT-4 is expressed at highest levels in prostate, with lower levels in thymus, placenta, and skeletal muscle . It is also expressed in neurons of the superior cervical, stellate and celiac ganglion
Sequence homology: While maintaining the conserved neurotrophin structure, NT-4 shares only 48-52% amino acid identity with other human neurotrophins
Evolutionary conservation: Human NT-4 shares 91% and 95% amino acid sequence identity with mouse and rat NT-4/5, respectively, indicating strong evolutionary conservation
Unique functions: Despite activating the same TrkB receptor as BDNF, NT-4 has distinct physiological roles due to differences in expression patterns and possibly signaling dynamics
For optimal stability and activity of recombinant NT-4, follow these guidelines:
Storage:
Use a manual defrost freezer and avoid repeated freeze-thaw cycles
Store lyophilized protein at -20°C to -70°C for up to 12 months from receipt date
After reconstitution, store at 2-8°C for up to 1 month under sterile conditions
For longer storage after reconstitution, aliquot and store at -20°C to -70°C for up to 6 months
Reconstitution:
For standard preparations (with carrier protein): Reconstitute at 50 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin
For carrier-free preparations: Reconstitute at 50 μg/mL in sterile PBS
For working solutions below 50 μg/mL, BSA (0.1 mg/ml) should be added to prevent protein adsorption to tubes and loss of activity
Always centrifuge all protein preparations before use (10,000 × g for 5 minutes)
Note that repeated freezing/thawing cycles may result in loss of activity
Several complementary methods can be used to detect NT-4 with different advantages:
Immunoassays (ELISA):
Sandwich immunoassays using specific antibodies like biotinylated anti-human NT-4 (e.g., BAF268) allow quantitative detection
This approach offers high sensitivity and specificity for protein-level expression
In situ hybridization:
Using DIG-labeled RNA probes prepared from human NT-4 cDNA fragments
Example protocol: Use primers targeting nucleotide positions 374-839 from the first ATG of NT-4
Visualization with alkaline phosphatase-conjugated anti-DIG (1:2,500) and NBT/BCIP color development (5.5 hr, 30°C)
This method allows spatial localization of NT-4 mRNA in tissue sections
Western blot:
For detecting NT-4 protein expression in cell lysates
Use phosphorylation-specific antibodies when studying signaling pathways activated by NT-4 (e.g., anti-phospho-ERK1/2 for downstream signaling)
RT-PCR:
For NT-4 mRNA detection
Primer design targeting conserved regions is critical for specificity
Example: Primers like NTF4-SF (5′-CTGCAGCTGGCGGCAGTCC-3′) and NTF4-SR (5′-ATTACCCTCAAGTTGCTCCA-3′) can generate a 466 bp fragment
To verify the biological activity of recombinant NT-4:
TrkB receptor phosphorylation assay:
Neuronal survival assay:
Neurite outgrowth measurement:
Calcium current measurements:
Recent research has revealed that NT-4 plays a complex dual role in breast cancer:
Pro-metastatic effects:
NT-4 promotes epithelial-mesenchymal transition (EMT), cell motility, and invasiveness of breast cancer cells in vitro and in vivo
Mechanistically, NT-4 activates PRKDC/AKT and ANXA1/NF-κB pathways to stabilize SNAIL protein
This leads to decreased E-cadherin levels, promoting metastatic potential
Anti-proliferative effects:
NT-4 inhibits cell proliferation while promoting cellular apoptosis in vitro
It inhibits xenograft tumorigenicity in vivo
NT-4 increases ANXA1 phosphorylation and sumoylation and its interaction with importin β
This leads to nuclear import and retention of ANXA1, which activates the caspase-3 apoptosis cascade
Clinical significance:
This dual role suggests that NT-4 may contribute to early metastasis of breast cancer and could serve as a prognostic marker.
Studies have identified multiple heterozygous mutations in the NTF4 gene associated with primary open-angle glaucoma (POAG):
Mutation profile:
Seven different heterozygous mutations in the NTF4 gene (C7Y, E84K, A88V, R90H, R206W, R206Q, and R209G) have been identified in POAG patients
These mutations account for approximately 1.7% of POAG cases
The mutations were found in both late juvenile and adult-onset POAG, with age at diagnosis varying from 36 to 80 years
Most patients with NTF4 mutations had elevated intraocular pressure (IOP) ranging from 25 to 40 mmHg, though some had normal pressure readings
Functional impact:
Structural analysis and molecular modeling showed that these mutations affect the NT-4 protein structure and its interaction with TrkB
The mutations were found to impair NT-4 signaling, which may affect retinal ganglion cell survival
This supports the hypothesis that factors compromising neuronal survival, rather than just elevated intraocular pressure, contribute to POAG pathogenesis
Evidence strength:
These findings provide strong genetic evidence linking NTF4 variants to POAG and highlight the importance of neurotrophic factors in glaucoma pathogenesis.
NT-4 plays distinct but overlapping roles with other neurotrophins in neuronal development and survival:
Developmental roles:
Studies comparing TrkB−/− and Bdnf−/−/Ntf4−/− mice show that NT-4 contributes to geniculate ganglion development
At E11.5, neuron numbers were reduced by 31% in Bdnf−/−/Ntf4−/− mice compared to wild-type
By E12.5, neuron counts were reduced by 48% in Bdnf−/−/Ntf4−/− mice and 81% in TrkB−/− mice
By E13.5, neuron numbers were reduced by 80% in Bdnf−/−/Ntf4−/− mice and 87% in TrkB−/− mice
These findings suggest that NT-4 and BDNF together account for most, but not all, TrkB-dependent neuronal development
Functional specificity:
Despite activating the same TrkB receptor as BDNF, NT-4 has unique functions
NT-4 promotes dendritic outgrowth and calcium currents in cultured mesencephalic dopamine neurons
It promotes growth and remodeling of adult motor neuron innervation
NT-4 serves as an anterograde survival factor for postsynaptic cells and protects against apoptotic neuronal death
Signaling mechanisms:
NT-4 binds to TrkB with high affinity, inducing receptor dimerization and activation
It also binds to p75NTR, which can modulate TrkB signaling
NT-4-induced TrkB signaling can augment NMDA receptor activity, potentially increasing neuronal sensitivity to excitotoxic cell death
These mechanisms contribute to both pro-survival and pro-apoptotic effects depending on the cellular context
Different experimental models offer advantages for specific aspects of NT-4 research:
In vitro neuronal models:
Primary neuron cultures (especially mesencephalic dopamine neurons) for studying NT-4's effects on neuronal survival, neurite outgrowth, and electrophysiological properties
TrkB-transfected cell lines (e.g., HEK-293) for studying receptor activation and signaling pathways
Organotypic brain slice cultures for studying NT-4's effects in a more intact neural circuit environment
Cancer cell models:
Breast cancer cell lines for studying NT-4's dual role in proliferation/apoptosis and metastasis
Xenograft models in immunodeficient mice to evaluate NT-4's effects on tumor growth and metastasis in vivo
Patient-derived organoids to assess NT-4's effects in a more clinically relevant context
Genetic models:
TrkB−/− and Bdnf−/−/Ntf4−/− knockout mice for studying developmental roles
Conditional knockout or knockin models to study tissue-specific or temporal NT-4 functions
CRISPR/Cas9-engineered cell lines with specific NT-4 pathway mutations
Glaucoma models:
Transgenic mice expressing human NTF4 mutations identified in POAG patients
Ex vivo retinal explant cultures for studying NT-4's effects on retinal ganglion cell survival
Induced pluripotent stem cell (iPSC)-derived retinal organoids from POAG patients with NTF4 mutations
The choice of model should align with the specific research question and consider the expression of relevant receptors and signaling molecules.
Distinguishing between NT-4 and BDNF functions requires careful experimental design:
Receptor-based approaches:
Utilize specific blocking antibodies that selectively inhibit NT-4 or BDNF binding to TrkB
Design peptide competitors that mimic specific binding epitopes unique to each neurotrophin
Compare signaling kinetics and duration by time-course analysis after stimulation with equivalent concentrations of NT-4 versus BDNF
Genetic approaches:
Use NT-4-specific knockout models compared to BDNF-specific knockouts
Employ RNA interference targeting either NT-4 or BDNF specifically
Analyze phenotypes in Bdnf−/−/Ntf4−/− double knockout versus single knockouts to identify unique versus overlapping functions
Functional assays:
Compare concentration-response relationships for both neurotrophins across multiple endpoints
Examine context-dependent effects in different cell types that may respond differently to each neurotrophin
Analyze differential effects on specific TrkB phosphorylation sites or downstream signaling pathways
Expression analysis:
Compare temporal and spatial expression patterns of NT-4 and BDNF in your experimental system
Use in situ hybridization to identify cells expressing one or both factors
Examine co-expression with TrkB to identify potential autocrine/paracrine signaling mechanisms
When investigating NT-4's role in disease:
Control for genetic background effects:
When using transgenic or knockout models, ensure appropriate genetic background controls
Consider using multiple founder lines or different genetic backgrounds to confirm phenotypes
For human studies, account for population stratification in genetic association analyses
Consider developmental timing:
NT-4 has important developmental roles, so distinguishing between developmental versus acute effects is critical
Use inducible systems (e.g., tamoxifen-inducible Cre-loxP) to control the timing of NT-4/TrkB manipulation
Include detailed temporal analyses to determine when NT-4 signaling becomes disrupted in disease models
Account for compensatory mechanisms:
Loss of NT-4 may trigger compensatory upregulation of other neurotrophins
Assess levels of related neurotrophins (especially BDNF) and their receptors
Consider using acute versus chronic manipulations to distinguish primary from compensatory effects
Address cell type specificity:
NT-4 affects multiple cell types beyond neurons, including immune cells and epithelial cells
Use cell type-specific promoters for targeted manipulation of NT-4 signaling
Employ methods like single-cell RNA-seq to identify cell populations differentially affected by NT-4
Validate in human samples:
Confirm findings from model systems in relevant human tissue samples
Use patient-derived cells or organoids when possible
Consider NT-4 polymorphisms and mutations identified in human diseases as experimental variables
Common technical challenges with recombinant NT-4 include:
Loss of activity during storage/handling:
Avoid repeated freeze-thaw cycles by storing as single-use aliquots
Add carrier protein (0.1 mg/ml BSA) for diluted solutions to prevent adsorption to tubes
Always centrifuge before use (10,000 × g for 5 min) to remove aggregates
Validate activity of each lot before use in critical experiments
Batch-to-batch variability:
Include positive controls with known activity in each experiment
Consider testing multiple lots or sources when initiating new studies
Establish internal standards and functional validation protocols
Quantify activity rather than relying solely on protein concentration
Receptor cross-reactivity:
NT-4 activates both TrkB and p75NTR receptors
Include appropriate receptor blocking controls to determine specific receptor contributions
Consider using receptor-selective mutants or inhibitors when studying specific signaling pathways
Verify receptor expression in your experimental system before interpreting NT-4 effects
Species differences:
Human NT-4 shares 91% and 95% amino acid identity with mouse and rat NT-4, respectively
While highly conserved, these differences may affect cross-species activities
When possible, use species-matched NT-4 for your experimental system
Be cautious when interpreting evolutionary studies or cross-species experiments
Optimization strategies for NT-4 treatments:
Concentration optimization:
Perform concentration-response experiments covering a wide range (typically 0.1-100 ng/mL)
The effective concentration for neuronal effects is typically 0.3-3 ng/mL, but this varies by assay
Include both low (sub-threshold) and high (potentially saturating) concentrations
Consider receptor density in your system - higher expression may require different concentrations
Treatment duration:
For signaling studies (e.g., TrkB phosphorylation), use short timepoints (5-60 minutes)
For neurite outgrowth, longer treatments (24-72 hours) are typically needed
For survival effects, both acute (24-48 hours) and chronic (5-14 days) treatments may be informative
Consider pulse treatments versus continuous exposure to model physiological conditions
Experimental design considerations:
Include time-matched controls for each concentration tested
For long-term treatments, consider media replenishment schedules and protein stability
When comparing NT-4 with other neurotrophins, match molar concentrations rather than weight/volume
For complex endpoints (e.g., gene expression), perform detailed time-course experiments
Validation approach:
Establish clear quantitative readouts for activity (e.g., % survival, neurite length)
Use multiple complementary assays to confirm effects (e.g., morphological plus biochemical)
Include positive controls (e.g., BDNF) and negative controls in each experiment
Document lot-specific activity for reproducibility across experiments
Emerging research directions for NT-4 include:
Neuroprotective strategies:
NT-4's ability to promote neuronal survival makes it a candidate for treating neurodegenerative disorders
Studies suggest potential applications in protecting retinal ganglion cells in glaucoma, especially given the association of NTF4 mutations with POAG
The distinct properties of NT-4 versus BDNF may provide advantages in specific neuroprotective contexts
Neural tissue engineering:
NT-4 can be used to enhance the neurotrophic capacity of stem cells for neural tissue engineering
Combined with other growth factors, NT-4 contributes to axonal growth in engineered neural tissues
Integration of NT-4 into biomaterials may provide sustained delivery for nerve regeneration
Cancer therapeutics:
The dual role of NT-4 in cancer (promoting metastasis while inhibiting proliferation) presents complex but potentially valuable therapeutic opportunities
Targeting NT-4-mediated signaling pathways might be effective in preventing early metastasis
The prognostic value of NT-4 expression suggests potential applications in personalized medicine approaches
Immune modulation:
NT-4 is secreted by activated T cells and granulocytes at sites of inflammation
Its role in tissue regeneration in inflammatory contexts suggests potential applications in immune-mediated diseases
Further research into NT-4's immune functions may reveal novel therapeutic applications
NT-4 can form heterodimers with other neurotrophins, which has important functional implications:
Heterodimer formation:
NT-4 can form heterodimers with BDNF or NT-3, in addition to forming homodimers
The mature protein is secreted as a homodimer but can also associate with other neurotrophins under physiological conditions
Functional consequences:
Heterodimers may exhibit unique receptor binding properties compared to homodimers
They could potentially activate different combinations of Trk receptors simultaneously
This might allow for more complex signaling outcomes than possible with homodimers alone
Research considerations:
When studying NT-4 in systems expressing multiple neurotrophins, consider possible heterodimer effects
Experimental designs should account for endogenous neurotrophin expression
Purified heterodimers versus homodimers can be used to dissect specific signaling outcomes
Technical approaches:
Co-expression systems can be used to generate neurotrophin heterodimers
Biochemical techniques such as co-immunoprecipitation can detect heterodimer formation
Binding studies with purified heterodimers can assess receptor interactions and activation potential
This area represents an emerging frontier in neurotrophin research with potential implications for both basic science and therapeutic applications.