Recombinant Human Beta-nerve growth factor (NGF), partial (Active)

Shipped with Ice Packs
In Stock

Description

Biological Activity

β-NGF binds tropomyosin receptor kinase A (TrkA) and p75 neurotrophin receptor (p75NTR), activating pathways like PI3K/Akt and MAPK/Erk. Functional assays demonstrate its potency:

Activity Across Sources

SourceED₅₀ (Cell Proliferation)Specific ActivityKey Assay
R&D Systems 0.2–2 ng/mLNot specifiedTF-1 erythroleukemic cells
Proteintech 0.5–3.0 ng/mL≥500,000 IU/mgNeurite outgrowth (PC12)
BPS Bioscience 0.5 ng/mLNot specifiedTF-1 cells
Irvine Scientific ≤5 ng/mLNot specifiedTF-1 cells
  • Mechanistic Insights:

    • Induces TrkA phosphorylation in PC12 cells, confirming receptor activation .

    • Promotes survival of retinal ganglion cells (RGCs) in vivo at 180–540 µg/mL doses .

    • Enhances histamine release in mast cells and modulates immune responses .

Key Uses

  • Neuronal Studies: Differentiation of PC12 cells and dorsal root ganglia (DRG) .

  • Ophthalmology: Topical application reduces RGC apoptosis in glaucoma models .

  • Immunology: Regulates B-cell differentiation and neutrophil survival .

  • ELISA Standards: Carrier-free formulations minimize interference .

Therapeutic Potential

  • Prevents secondary neurodegeneration in optic nerve injury models .

  • Attenuates astrocyte activation and inflammation in diabetic corneas .

Research Findings and Clinical Relevance

  • Neuroprotection: Topical β-NGF (540 µg/mL) preserved 70% of RGCs in partial optic nerve transection models, outperforming lower doses .

  • Immune Modulation: Enhances chemotaxis of polymorphonuclear leukocytes and suppresses B-cell apoptosis .

  • Cross-Species Reactivity: Active in murine, rat, and human systems .

Challenges and Considerations

  • Structural Sensitivity: Proper disulfide bond formation is critical; misfolding in bacterial systems (E. coli) necessitates refolding protocols .

  • Dose-Dependent Effects: Higher concentrations (≥540 µg/mL) may paradoxically reduce efficacy in some models .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered 20 mM Phosphate Buffered Saline (PBS), 250 mM Sodium Chloride (NaCl), pH 7.0.
Form
Lyophilized powder
Lead Time
Typically, we can ship the products within 5-10 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery times.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend centrifuging the vial briefly before opening to ensure the contents are at the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. To ensure long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting the solution for storage at -20°C/-80°C. Our default final glycerol concentration is 50% and can be used as a reference.
Shelf Life
The shelf life is influenced by various factors, including storage conditions, buffer components, storage temperature, and the inherent stability of the protein itself. Generally, the shelf life of liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
Beta nerve growth factor; Beta NGF; Beta-nerve growth factor; Beta-NGF; HSAN5; MGC161426; MGC161428; Nerve growth factor (beta polypeptide); Nerve growth factor; Nerve growth factor beta; Nerve growth factor beta polypeptide; Nerve growth factor beta subunit; NGF; NGF_HUMAN; NGFB; NID67
Datasheet & Coa
Please contact us to get it.
Expression Region
122-239aa
Mol. Weight
13.5 kDa
Protein Length
Partial
Purity
Greater than 95% as determined by SDS-PAGE.
Research Area
Neuroscience
Source
Mammalian cell
Species
Homo sapiens (Human)
Target Names
NGF
Uniprot No.

Target Background

Function
Nerve growth factor (NGF) plays a crucial role in the development and maintenance of the sympathetic and sensory nervous systems. It acts as an extracellular ligand for the NTRK1 and NGFR receptors, initiating cellular signaling cascades that regulate neuronal proliferation, differentiation, and survival (Probable). The immature NGF precursor (proNGF) functions as a ligand for the heterodimeric receptor formed by SORCS2 and NGFR, activating cellular signaling cascades that lead to inactivation of RAC1 and/or RAC2, reorganization of the actin cytoskeleton, and neuronal growth cone collapse. Unlike mature NGF, proNGF promotes neuronal apoptosis (in vitro). It also inhibits metalloproteinase-dependent proteolysis of platelet glycoprotein VI. NGF binds lysophosphatidylinositol and lysophosphatidylserine between the two chains of the homodimer. The lipid-bound form promotes histamine release from mast cells, unlike the lipid-free form.
Gene References Into Functions
  1. This study reports the effects of secretome-enriched conditioned medium (CM) of neuronally differentiated PC12 cells on the neuronal differentiation of human mesenchymal stem cells via an NGF-like mechanism. PMID: 29527653
  2. Single nucleotide polymorphisms in the NGF gene (rs6330) and NGFR gene (rs2072446 and rs734194) are associated with ischemic stroke. The NGF rs6330*T and NGFR rs2072446*T minor alleles might be nominated as a risk factor for developing ischemic stroke, while the NGFR rs734194*G minor allele could be a protective factor against this disease, at least in the Armenian population. PMID: 29499660
  3. This study examines neurotrophic factors and hippocampal activity in post-traumatic stress disorder (PTSD). PMID: 29799860
  4. SNRPA may contribute to gastric cancer (GC) progression via NGF and could serve as a prognostic biomarker for GC. PMID: 30039889
  5. Results demonstrate that NGF signaling is strongly linked to pathological and regenerative processes in human teeth. PMID: 28465581
  6. This study suggests an interaction between NGF, GDNF, and MMP-9 during the transition to malignancy in prostate cancer (PC). This interaction may play a role in regulating PC cell differentiation, tumor invasion, progression, and the aggressiveness of PC. PMID: 28237042
  7. The anti-tumor activity of oleuropein against hepatocellular carcinoma could be attributed to influencing the pro-NGF/NGF balance by affecting MMP-7 activity without altering the gene expression of NGF. PMID: 29476769
  8. Co-expression patterns of NGF and heme oxygenase-1 might be used as prognostic indicators for gastric carcinoma patients. PMID: 28679437
  9. This review briefly addresses the peripheral and central sensitization mechanisms of airway neurons and focuses on NGF signaling and its role in cough hypersensitivity. PMID: 28494216
  10. First-trimester plasma nerve growth factor levels are lower in patients who subsequently develop preeclampsia. PMID: 27513943
  11. This study provides evidence of variation in plasmatic beta-NGF expression during the progression of dementia. PMID: 27802234
  12. NGF is functionally linked to beta-catenin, promoting the migration of human ovarian cancer cells via the WNT/beta-catenin pathway. PMID: 27835587
  13. Varicella zoster virus DNA replication is regulated, in part, by an NGF pathway that is PI3-kinase-independent. PMID: 27683235
  14. Many studies indicate that the presence of NGF alone cannot induce cell carcinogenesis, both in normal neuronal and non-neuronal cells/tissues. However, the possibility that the co-expression of NGF and pro-carcinogenic molecules might lead to different consequences cannot be excluded. PMID: 27439311
  15. This study reports the effect and underlying mechanisms of NGF/BDNF on the production of NPW in PC12 cells and the hypothalamus. PMID: 28249734
  16. These data support a role for islet NGF in fine-tuning insulin secretion. PMID: 27424144
  17. The results of this study indicate that dysmenorrhea pain severity is partly genetically determined by chromosome 1p13.2, near the nerve growth factor locus. PMID: 27454463
  18. Findings suggest that IL-1beta and TNF-alpha regulate Nerve Growth Factor expression and production in synovial macrophages and fibroblasts in osteoarthritic joints. PMID: 28677145
  19. These results advance our understanding of the conformational properties of proNGF and NGF, providing a rationale for the diverse biological effects of NGF and proNGF at the molecular level. PMID: 28798232
  20. The results of this study suggest that proNGF protein levels may enhance the diagnostic accuracy of currently used CSF biomarker panels. PMID: 26825093
  21. Neuroimmune-endocrine events may lead to overactivity of the sympathetic nervous system, triggering a cascade of pathological conditions in the ovary in polycystic ovary syndrome (PCOS). Data suggest that women with PCOS exhibit a reduction in CRH and NGF; this reduction in CRH/NGF could be influenced by the sympathetic nervous system and may reflect a deficit in neuronal stress-adaptation in PCOS patients (CRH = corticotropin releasing hormone). PMID: 27908212
  22. The rare nerve growth factor-beta (NGFB) mutation R221W causes a selective loss of thinly myelinated fibers, particularly unmyelinated C-fibers. Carriers of this mutation exhibit altered pain sensation. PMID: 27146986
  23. NGF expression was positively correlated with disease severity and visceral hypersensitivity in irritable bowel syndrome patients. PMID: 27862119
  24. This study investigates a stage-related modulation of beta-NGF and its receptors in the inflammatory process of osteoarthritis (OA). PMID: 28253191
  25. BDNF and NGF serum levels are reduced in the early and moderate glaucoma stages, suggesting that both factors could be further investigated as potential circulating biomarkers for early glaucoma detection. PMID: 28068360
  26. Overexpression of ARMS blocked NGF-mediated secretion without affecting basal secretion, while a decrease in ARMS resulted in potentiation. Similar effects were observed with synembryn-B, a protein that interacts directly with ARMS. Downstream of ARMS and synembryn-B are Galphaq and Trio proteins, which modulate the activity of Rac1 in response to NGF. PMID: 26966186
  27. Data show that the standardized nerve growth factor (NGF) concentration was negatively correlated with continuous pain, neuropathic pain, and total score, while the standardized S100 proteins S100A8/A9 concentration was positively correlated with present pain intensity and continuous pain. PMID: 27936243
  28. Results indicate that NGF inhibited CRT translocation induced by mitoxantrone. NGF's effect on CRT translocation could have implications for immunotherapy, potentially lessening the effectiveness of this treatment approach. PMID: 28260038
  29. NGF stimulates the generation of neurons, but not neuronal progenitors, from embryonic stem cells and influences the proportion of specific types of neurons in cultures of differentiating embryonic stem cells. PMID: 28364186
  30. NGF attenuates these responses, both in vivo and in vitro. Therefore, NGF therapy may represent a novel approach for managing diabetic keratopathy. PMID: 27978558
  31. The NGF signaling pathway presents a potential target for developing molecularly targeted therapies. PMID: 27792755
  32. These data suggest a positive feedback loop through which NGF-mediated upregulation of p75(NTR) can contribute to the chemo-resistance of Triple-negative breast cancer cells. PMID: 27577679
  33. Based on current developments, this review provides not only a comprehensive overview of the biological effects of NGF-TrkA-p75NTR on cancer cells and their microenvironment, but also explains why NGF and its receptors are poised to evoke significant interest as promising therapeutic anti-cancer targets in the coming decade. PMID: 27264679
  34. NGF/CD133 might be an effective and potent therapeutic target for pancreatic cancer metastasis, particularly in perineural invasion. PMID: 27654574
  35. The analysis of covariance (ANCOVA) indicated that the mean serum GDNF and NTF3 levels of ADHD patients were significantly higher than those of controls. However, serum BDNF and NGF levels did not show any significant differences between groups. PMID: 27561780
  36. All patients had serum neurotrophin (NT-3, BDNF, NGF) concentrations determined. PMID: 27367919
  37. Serum NGF does not differentiate between recurrent acute pancreatitis and chronic pancreatitis. PMID: 27020638
  38. NGF FLIPs TrkA onto the death TRAIL in neuroblastoma cells. PMID: 26962689
  39. Major depression patients had similar serum NGF levels to controls. PMID: 27008247
  40. Data demonstrate the involvement and modulation of nerve growth factor and its receptors in chronic obstructive pulmonary disease and its staging. PMID: 26408608
  41. This study showed that IL-17, in addition to stimulating an inflammatory response, negatively regulates the action of NGF and NGF R in the polar forms of leprosy. PMID: 26616164
  42. Locally increased estrogen levels and inflammation may cause increased NGF production in the uterus of patients with adenomyosis. PMID: 25519715
  43. This study is the first to comprehensively assess the enhancement of neural differentiation of bone marrow mesenchymal stem cells following transfection with bFGF and NGF. PMID: 26572749
  44. Data suggest MMP7 (matrix metalloproteinase 7) in follicular fluid cleaves proNGF (pro-nerve growth factor) in the ovarian follicle; both MMP7 and proNGF appear to be products of granulosa cells; processing of proNGF to NGF appears to regulate apoptosis. PMID: 26457789
  45. NGF promotes renal fibrosis via TGF-beta1-signaling activation, suggesting that high NGF serum levels in kidney diseases could contribute to worsening renal fibrosis. PMID: 26066770
  46. This study suggests that the circadian rhythm in the esophagus may be important for the mediation of and/or the response to erosive damage in GERD patients. PMID: 26337663
  47. Intratumoral nerve growth factor expression is not associated with perineural invasion in patients with resected extrahepatic cholangiocarcinoma. PMID: 26547754
  48. NGF plays a role in modulating trkANGFR/p75NTR in alphaSMA-expressing conjunctival fibroblasts from human ocular cicatricial pemphigoid. PMID: 26569118
  49. NGF-induced tyrosine kinase independent TrkA signaling through CD44 is sufficient to maintain tumor aggressiveness in breast cancer. PMID: 25840418
  50. Urinary NGF, but not BDNF, levels decreased significantly after hyaluronic acid therapy. PMID: 24614892

Show More

Hide All

Database Links

HGNC: 7808

OMIM: 162030

KEGG: hsa:4803

STRING: 9606.ENSP00000358525

UniGene: Hs.2561

Involvement In Disease
Neuropathy, hereditary sensory and autonomic, 5 (HSAN5)
Protein Families
NGF-beta family
Subcellular Location
Secreted. Endosome lumen.

Q&A

How is the biological activity of rhβ-NGF validated in experimental settings?

Multiple complementary assays are employed to validate rhβ-NGF biological activity:

  • PC12 Cell Differentiation: The gold standard assay measures neurite outgrowth in rat pheochromocytoma cells. Functional rhβ-NGF induces neuronal differentiation at concentrations of 5-50 ng/mL, with optimal effects typically observed at 25-50 ng/mL .

  • TrkA Receptor Phosphorylation: Confirmation of activation of the high-affinity receptor tyrosine kinase (TrkA) through phosphorylation assays. This can be confirmed through Western blotting or ELISA-based approaches .

  • TF-1 Cell Proliferation: The human erythroleukemic cell line TF-1 responds mitogenically to rhβ-NGF with typical ED50 values of 0.2-5 ng/mL .

  • Trk Receptor Inhibition: Selective tyrosine kinase inhibitors (e.g., K-252a at 100 nM) can block rhβ-NGF effects on PC12 cells, confirming specificity of the observed differentiation .

  • In vivo testing: Assessment of neurotrophic activity on superior cervical ganglia (SCG) in newborn mice, measuring ganglia hypertrophy and neuronal effects .

These validation methods should be applied sequentially, beginning with in vitro assays before progressing to more complex in vivo assessments .

What expression systems are most effective for producing bioactive rhβ-NGF?

Several expression systems have been employed for rhβ-NGF production, each with distinct advantages and limitations:

Expression SystemAdvantagesLimitationsTypical Yield
Mammalian cells (CHO, HeLa)Proper post-translational modifications, highest biological activityHigher cost, slower growth≈21.5 ± 2.9 mg/liter
Escherichia coliHigh yield, lower costRequires refolding from inclusion bodies, variable activityVariable
Saccharomyces cerevisiaeModerate yield, eukaryotic processingMay have non-human glycosylation patternsNot specified
Insect cellsGood protein foldingComplex glycosylation differencesNot specified

For research requiring the highest biological activity comparable to native NGF, mammalian expression systems utilizing CHO or HeLa cells are recommended, particularly when coupled with high-density culture methods like miniPERM bioreactors . The selection of the expression system should align with the specific research objectives, considering the trade-off between yield and biological activity .

How should researchers design optimal experiments to evaluate rhβ-NGF efficacy?

Designing rigorous experiments with rhβ-NGF requires careful consideration of several methodological factors:

  • Power Analysis: Conduct a priori power analysis to determine appropriate sample sizes for detecting expected effect sizes. This is particularly important for costly in vivo experiments with rhβ-NGF .

  • Dose-Response Relationships: Establish complete dose-response curves (typically ranging from 0.1-100 ng/mL) rather than testing single concentrations. This approach reveals optimal dosing and potential biphasic effects .

  • Time-Course Studies: Monitor temporal responses, as NGF effects can vary significantly across time points. Neurite outgrowth assays typically require 48-72 hours for optimal visualization, while receptor phosphorylation occurs within minutes to hours .

  • Appropriate Controls: Include:

    • Vehicle controls (matched to protein carrier solution)

    • Positive controls (commercial NGF with established activity)

    • Specificity controls (TrkA inhibitors like K-252a)

    • Heat-inactivated rhβ-NGF (negative control)

  • Machine Learning Approaches: For complex experimental designs, consider Bayesian Optimal Experimental Design (BOED) with machine learning to optimize experimental parameters and maximize information gain with minimal experimental runs .

  • ANCOVA Considerations: When baseline measurements are identical for all experimental units (e.g., zero expression of a gene prior to treatment), focus on measuring predictive covariates that might influence response variability rather than relying solely on difference-in-differences approaches .

The experimental design should include thorough validation of rhβ-NGF activity before proceeding to more complex biological questions .

What are the critical species-specific differences in NGF structure and function researchers should consider?

Species differences in NGF amino acid sequence significantly affect receptor binding and biological activity:

  • Receptor Binding Site Variations: The amino acid sequence of rabbit NGF differs from other species primarily at receptor binding sites. In spontaneous-ovulating species, alanine and proline residues that interact with the high-affinity receptor are replaced by serine .

  • Low-Affinity Receptor Interactions: In rabbits, asparagine and methionine are substituted by lysine at the low-affinity receptor binding site, potentially altering signaling dynamics .

  • Cross-Species Activity: Though human, mouse, and rat β-NGF proteins are generally cross-reactive, the efficacy may vary. The biological activity of rhβ-NGF in human peripheral neuropathies has not consistently matched the effects of murine NGF (mNGF) in some studies .

  • Functional Implications: These structural differences can impact:

    • TrkA receptor binding affinity

    • p75 receptor interactions

    • Downstream signaling pathway activation

    • Tissue-specific responses

When designing cross-species experiments, researchers should validate the activity of the specific NGF variant in their target system, as receptor binding differences may lead to unexpected variations in biological outcomes .

What methodological approaches can overcome the challenges of proNGF contamination in rhβ-NGF preparations?

The presence of unprocessed proNGF forms can significantly confound experimental results as proNGF preferentially binds to p75 receptors, opposing the neurotrophic activity of mature NGF . Researchers can implement several strategies to address this issue:

  • Host Cell Selection: Employ cell lines with efficient proNGF processing capabilities. HeLa TetOff clones have demonstrated complete processing of proNGF, with culture medium devoid of unprocessed forms .

  • Purification Strategy:

    • Implement affinity chromatography steps specifically designed to separate mature NGF from proNGF

    • Consider size-exclusion chromatography as a final polishing step to eliminate high molecular weight forms

    • Validate purity using both reducing and non-reducing SDS-PAGE

  • Quality Control Assays:

    • Western blot analysis with antibodies specific to mature NGF and proNGF domains

    • Mass spectrometry to confirm molecular weight and sequence integrity

    • Functional assays that distinguish mature NGF from proNGF activities

  • Serum Considerations: Optimize serum concentration in the culture medium (e.g., using 5% rather than higher concentrations) to reduce serum contaminants while maintaining adequate protein production. This approach not only reduces purification challenges but also lowers production costs .

  • Scale-up Strategy: Implement miniPERM bioreactors for high-density culture of mammalian cells, which can yield approximately 100-fold higher recombinant protein concentrations than conventional flask cultures while simplifying initial purification steps .

Thorough characterization of rhβ-NGF preparations is essential, as the ratio of mature NGF to proNGF can significantly influence experimental outcomes, particularly in studies of neurotrophic and neuroprotective effects .

How can researchers optimize rhβ-NGF reconstitution and storage protocols to maintain maximum biological activity?

Proper handling of rhβ-NGF is critical for maintaining biological activity throughout experimental procedures:

  • Reconstitution Protocol:

    • Centrifuge the vial before opening to collect all material

    • Reconstitute lyophilized rhβ-NGF at 0.1-0.1 mg/mL in sterile buffer

    • For carrier-free preparations, use sterile PBS

    • For preparations with carrier protein, use PBS containing at least 0.1% human or bovine serum albumin

    • Gently pipette solution down the sides of the vial rather than directly onto the protein

    • Allow several minutes for complete reconstitution

    • Do not vortex the solution as this can denature the protein

  • Storage Recommendations:

    • For prolonged storage, prepare working aliquots in 0.1% BSA solution

    • Store at -80°C to maintain stability

    • Avoid repeated freeze-thaw cycles which significantly reduce activity

    • Use manual defrost freezers rather than auto-defrost types

  • Quality Control:

    • Verify protein concentration after reconstitution

    • Perform activity assays (e.g., TF-1 cell proliferation) with each new preparation

    • Monitor purity through reducing and non-reducing SDS-PAGE

    • Test for endotoxin contamination using Kinetic LAL assays (acceptable limit ≤ 0.1 EU/μg)

  • Carrier Protein Considerations:

    • BSA enhances protein stability and increases shelf-life

    • For applications where carrier proteins may interfere (e.g., certain immunoassays), use carrier-free formulations

    • When using carrier-free preparations, prepare fresh working solutions more frequently

Researchers should validate each new lot of reconstituted rhβ-NGF in their specific experimental system before conducting full-scale studies to ensure consistent biological activity .

What are the key considerations for designing experiments to evaluate rhβ-NGF effects on neuronal differentiation and survival?

Designing robust experiments to assess rhβ-NGF effects on neuronal systems requires attention to several critical parameters:

  • Cell Line Selection:

    • PC12 cells: Standard model for neuronal differentiation, but responses may vary between subclones

    • Primary neurons: More physiologically relevant but higher variability

    • Dorsal root ganglia (DRG): Useful for studying sensory neuron responses

    • TF-1 cells: For proliferation assays rather than differentiation

  • Culture Conditions:

    • Substrate coating affects neuronal attachment and differentiation (collagen, poly-L-lysine, laminin)

    • Serum levels influence baseline differentiation and may contain NGF-like factors

    • Cell density affects paracrine signaling and resource competition

    • Culture duration should be optimized for each endpoint (48-72h typical for differentiation assays)

  • Experimental Design Considerations:

    • Include concentration gradients (typically 0.1-100 ng/mL) to establish dose-response relationships

    • Implement time-course analyses to capture both early signaling events and later morphological changes

    • Use parallel cultures for different analytical techniques rather than sequential sampling

    • Consider machine learning approaches like Bayesian Optimal Experimental Design to maximize information gain

  • Quantification Methods:

    • Neurite outgrowth: Length, number, branching patterns (automated image analysis recommended)

    • Cell viability: MTT/XTT assays, live/dead staining, or flow cytometry

    • Molecular markers: GAP-43, βIII-tubulin, synaptic proteins

    • Receptor activation: TrkA phosphorylation status by Western blot or ELISA

  • Controls and Validation:

    • Positive controls: Commercial NGF preparations with established activity

    • Negative controls: Heat-inactivated rhβ-NGF, vehicle only

    • Specificity controls: TrkA inhibition with K-252a (100 nM)

    • Biological replication: Minimum 3 independent experiments with technical replicates

  • Statistical Analysis:

    • A priori power analysis to determine sample size

    • Account for nested data structure (cells within wells within plates)

    • Consider non-linear dose-response modeling rather than simple parametric tests

    • Implement robust statistical methods resistant to outliers and non-normal distributions

By systematically addressing these factors, researchers can develop experimental protocols that maximize the reproducibility and translational relevance of rhβ-NGF studies in neuronal systems .

How do researchers effectively assess the translational potential of rhβ-NGF from in vitro to in vivo models?

Bridging the gap between in vitro findings and in vivo applications represents a significant challenge in rhβ-NGF research. A systematic approach includes:

  • Sequential Testing Strategy:

    • Begin with in vitro characterization (PC12 differentiation, TrkA activation)

    • Progress to ex vivo systems (tissue explants like superior cervical ganglia)

    • Advance to in vivo models with clear, measurable endpoints

    • Validate findings across multiple models before translational applications

  • In Vivo Model Selection Criteria:

    • Relevance to human pathophysiology

    • Conservation of NGF signaling pathways

    • Feasibility of intervention and assessment

    • Ethical considerations and regulatory compliance

  • Pharmacological Parameters:

    • Bioavailability and tissue distribution studies

    • Dosing regimens based on in vitro EC50/ED50 values with allometric scaling

    • Half-life determination and clearance mechanisms

    • Route of administration optimization (systemic vs. local delivery)

  • Translational Endpoints:

    • Morphological: Histological analysis of ganglia hypertrophy, neuronal density, and innervation patterns

    • Biochemical: TrkA protein expression levels, substance P mRNA upregulation

    • Functional: Behavioral assessments, electrophysiological measurements

    • Safety: Monitor for hyperalgesia/allodynia, mast cell degranulation, immune responses

  • Experimental Design Considerations:

    • Implement blinded assessment protocols

    • Include appropriate vehicle controls with carrier proteins

    • Use positive controls (e.g., purified mouse NGF) for comparison

    • Power analysis to determine minimum animal numbers needed

    • Consider sex as a biological variable

  • Translational Gap Analysis:

    • Document species-specific differences in NGF sequence and receptor binding

    • Compare dose-response relationships between in vitro and in vivo settings

    • Assess differences between recombinant and native NGF effects

    • Identify potential biomarkers that correlate across systems

A particularly important consideration is that while some studies report injection-site pain with NGF administration, research has demonstrated that properly produced rhβ-NGF can exert neurotrophic effects comparable to murine NGF without apparent hyperalgesia, suggesting optimization of preparation methods can minimize adverse effects .

Implementing this systematic approach not only enhances scientific rigor but also increases the probability of successfully translating promising in vitro findings to clinically relevant applications .

How can researchers address batch-to-batch variability in rhβ-NGF preparations?

Batch-to-batch variability presents a significant challenge in rhβ-NGF research. Implementing the following methodological approaches can help minimize this variability:

  • Standardized Production Protocol:

    • Maintain consistent cell passage numbers for production cultures

    • Control serum concentration and source across production runs

    • Standardize induction conditions and harvest timing

    • Implement rigorous documentation of all production parameters

  • Comprehensive Quality Control:

    • Purity Assessment: ≥95% by reducing and non-reducing SDS-PAGE

    • Endotoxin Testing: ≤0.1 EU/μg by Kinetic LAL

    • Biological Activity: Standardized ED50 determination in TF-1 cell proliferation assay (≤5 ng/mL)

    • Molecular Identity: Mass spectrometry fingerprinting against reference standards

  • Reference Standard System:

    • Establish an internal reference standard from a well-characterized batch

    • Perform side-by-side testing of new batches against this standard

    • Calculate relative potency ratios rather than relying on absolute values

    • Consider implementing the WHO International Standard for NGF when available

  • Stability Monitoring:

    • Implement real-time and accelerated stability testing programs

    • Monitor activity retention at defined time points

    • Establish expiration dating based on stability data

    • Document storage condition effects on activity retention

  • Advanced Analytical Characterization:

    • Circular dichroism to assess secondary structure

    • Size-exclusion chromatography to detect aggregation

    • Receptor binding assays to evaluate functional epitope integrity

    • Western blot with epitope-specific antibodies to confirm structural integrity

  • Validation Across Multiple Bioassays:

    • PC12 differentiation (qualitative and quantitative assessment)

    • TrkA phosphorylation (receptor activation)

    • TF-1 proliferation (standardized potency)

    • Implement at least two orthogonal bioassays for each batch

By implementing these methodological approaches, researchers can significantly reduce batch-to-batch variability and establish more reproducible experimental systems for investigating rhβ-NGF biology and therapeutic applications .

What advanced analytical methods can researchers employ to characterize rhβ-NGF structural integrity and functionality?

Comprehensive characterization of rhβ-NGF requires a multi-modal analytical approach that addresses both structural integrity and functional activity:

  • Primary Structure Analysis:

    • Mass Spectrometry: MALDI-TOF or ESI-MS for molecular weight confirmation

    • Peptide Mapping: Enzymatic digestion followed by LC-MS/MS to verify amino acid sequence

    • N-terminal Sequencing: Edman degradation to confirm correct processing of signal peptide

    • Disulfide Bond Mapping: Non-reducing peptide analysis to verify correct disulfide pairing

  • Higher-Order Structure Characterization:

    • Circular Dichroism (CD): Assessment of secondary structure elements

    • Fluorescence Spectroscopy: Intrinsic tryptophan fluorescence for tertiary structure evaluation

    • Differential Scanning Calorimetry (DSC): Thermal stability assessment

    • Size-Exclusion Chromatography (SEC): Detection of aggregates and dimeric state

  • Functional Characterization:

    • Surface Plasmon Resonance (SPR): Kinetic binding parameters to TrkA and p75 receptors

    • Cell-Based Reporter Assays: MAPK/ERK pathway activation

    • Phospho-specific Western Blotting: TrkA phosphorylation at specific tyrosine residues

    • Cell Differentiation Quantification: Automated image analysis of neurite outgrowth

  • Comparative Analysis:

    • Parallel Bioassays with Reference Standard: Calculate relative potency

    • Cross-Species Activity Assessment: Comparative testing in human, mouse, and rat cell systems

    • Native vs. Recombinant Comparison: Side-by-side testing with purified natural NGF

    • Structure-Activity Relationship Studies: Correlation of structural parameters with bioactivity

  • Advanced Biophysical Methods:

    • Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS): Protein dynamics and solvent accessibility

    • Small-Angle X-ray Scattering (SAXS): Solution structure analysis

    • Ion Mobility Mass Spectrometry: Conformational integrity assessment

    • Atomic Force Microscopy (AFM): Visualization of protein aggregates and morphology

  • Stability-Indicating Methods:

    • Forced Degradation Studies: Exposure to extreme pH, oxidation, temperature

    • Accelerated Stability Testing: Activity retention under stressed conditions

    • Aggregation Analysis: SEC-MALS for molecular weight distribution

    • Receptor Binding After Stress: Functional epitope integrity assessment

Implementation of these analytical methods provides researchers with a comprehensive understanding of rhβ-NGF structural characteristics and their relationship to biological activity, enabling more informed experimental design and interpretation of results .

How might machine learning approaches enhance experimental design for rhβ-NGF research?

The application of machine learning (ML) to rhβ-NGF research offers promising avenues for optimizing experimental design and data analysis:

  • Bayesian Optimal Experimental Design (BOED):

    • Formalize scientific questions into probabilistic models

    • Simulate experimental outcomes across possible design parameters

    • Identify designs that maximize information gain with minimal experimental runs

    • Update experimental parameters based on incoming data in an adaptive design framework

  • Parameter Optimization for Cell-Based Assays:

    • Identify optimal combinations of rhβ-NGF concentration, exposure time, and cell density

    • Reduce experimental iterations through predictive modeling

    • Minimize reagent usage while maximizing statistical power

    • Account for complex interactions between experimental variables

  • Image Analysis for Morphological Outcomes:

    • Automated quantification of neurite outgrowth parameters

    • Deep learning segmentation of neuronal structures in complex cultures

    • Classification of cell differentiation states based on morphological features

    • Unbiased detection of subtle phenotypic changes

  • Multi-Parameter Optimization:

    • Balance competing objectives (e.g., maximizing differentiation while minimizing toxicity)

    • Incorporate cost and time constraints into experimental planning

    • Identify minimal sets of experiments needed to answer specific research questions

    • Optimize across multiple readouts simultaneously

  • Translational Prediction Models:

    • Develop algorithms to predict in vivo outcomes based on in vitro data

    • Identify key parameters that correlate across experimental systems

    • Reduce animal usage through improved predictive modeling

    • Incorporate historical data to improve prediction accuracy

  • Implementation Considerations:

    • Start with simulation studies to validate ML approaches before applying to wet-lab experiments

    • Build collaborative teams with ML expertise and domain knowledge

    • Maintain transparency in ML model design and validation

    • Make experimental code and data openly available to increase reproducibility

As demonstrated in recent work, ML-based BOED methods can help design experiments that more efficiently determine which models best account for biological behavior and more efficiently characterize parameters within preferred models, potentially accelerating rhβ-NGF research while reducing resource requirements .

What novel therapeutic applications of rhβ-NGF are emerging from recent research?

Recent research has expanded the potential therapeutic applications of rhβ-NGF beyond traditional neurodegenerative conditions:

  • Reproductive Medicine:

    • Ovulation Induction: In induced-ovulating species, β-NGF has been identified as an important factor in ovulation, with research showing that recombinant rabbit β-NGF (rrβ-NGF) added to seminal doses administered intravaginally can induce ovulation with a delayed LH peak, leading to plasma progesterone increase, gestation, and delivery .

    • Sperm Function Enhancement: Addition of rrβ-NGF (1 μg/mL) to rabbit ejaculate improved sperm motility parameters without affecting viability, suggesting potential applications in assisted reproductive technologies .

  • Immune System Modulation:

    • B-Lymphocyte Development: NGF is involved in the growth, differentiation, and survival of B lymphocytes, potentially offering therapeutic avenues for immune disorders .

    • Mast Cell Regulation: NGF increases mast cell numbers and induces histamine release, which may have applications in inflammatory and allergic conditions .

    • Neutrophil Survival: NGF suppresses apoptosis of murine peritoneal neutrophils, suggesting potential applications in neutropenia or wound healing .

  • Ocular Surface Disorders:

    • Corneal Neuropathic Pain: Targeting corneal nerve regeneration in dry eye disease and post-surgical neuropathic pain.

    • Neurotrophic Keratopathy: Promoting corneal epithelial healing and sensory innervation restoration .

  • Peripheral Nerve Injury:

    • Enhanced Regeneration Protocols: Optimized delivery systems for sustained release of rhβ-NGF at injury sites.

    • Combination Therapies: Synergistic effects with other neurotrophic factors or biomaterials .

  • Central Nervous System Applications:

    • Basal Forebrain Cholinergic Neurons: Trophic support for neurons affected in Alzheimer's disease and other dementias.

    • Neuroprotective Strategies: Prevention of neuronal loss in neurodegenerative conditions .

  • Pain Modulation:

    • Improved Formulations: Development of rhβ-NGF preparations that maintain neurotrophic effects while minimizing hyperalgesia/allodynia.

    • Targeted Delivery: Selective activation of regenerative pathways while avoiding pain sensitization mechanisms .

The diversity of these emerging applications highlights the pleiotropic nature of NGF as a cytokine with roles extending well beyond the nervous system, offering promising research directions for conditions ranging from infertility to immune disorders and beyond .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.