Recombinant Mouse Beta-nerve growth factor (Ngf), partial (Active)

Shipped with Ice Packs
In Stock

Description

Molecular Characterization and Structure

Recombinant Mouse Beta-NGF (partial active) consists of amino acid residues 122–241 or 130–239 (species-dependent) and forms a non-covalent homodimer with three disulfide bonds essential for activity . Key structural parameters include:

ParameterValueSource
Molecular Weight12.4–13.5 kDa (monomer)
Sequence Homology90% with human, 95.8% with rat β-NGF
GlycosylationNone (produced in E. coli)
Quaternary StructureHomodimer with disulfide bonds

The mature form lacks the pro-domain (residues 1–121), which in proNGF induces apoptosis via p75NTR/sortilin receptors . Structural studies reveal stabilization of three loops in the mature region when the pro-domain is absent .

Biological Activity and Mechanism of Action

β-NGF signals through TrkA and LNGFR receptors, activating PI3K, Ras, and PLC pathways to regulate neuronal survival, differentiation, and synaptic plasticity . Its bioactivity is validated through:

  • TF-1 Cell Proliferation Assay: ED50 = 0.3–68.52 ng/ml, depending on expression system .

  • Neurite Outgrowth: Induces differentiation in PC12 cells and dorsal root ganglia (DRG) at 5 ng/ml .

  • B Lymphocyte Survival: Enhances B-cell proliferation and survival .

Assay TypeActivityED50Source
TF-1 ProliferationCell growth stimulation0.3–1.5 ng/ml
PC12 DifferentiationNeurite extension5 ng/ml
Mast Cell ActivationHistamine releaseLipid-bound form required

Production and Purification Methods

Produced in E. coli with >95% purity via affinity chromatography . Key production metrics:

ParameterSpecificationSource
Expression HostE. coli
Endotoxin Level<1.0 EU/µg
StorageLyophilized in Tris/NaCl buffer (pH 8)
Stability12 months at -80°C (lyophilized)

Pharmacological and Therapeutic Applications

Preclinical studies highlight its potential in:

  • Neurodegenerative Diseases:

    • Restores motor activity in MPTP-induced Parkinson’s models (1.78–2.86-fold improvement) .

    • Improves memory in scopolamine-induced amnesia models .

  • Nerve Regeneration:

    • Combined with bFGF in hydrogels enhances urethral muscle reinnervation .

  • Autoimmune Disorders: Modulates B-cell responses in inflammatory conditions .

Comparative Analysis with Other NGF Forms

FeatureRecombinant Mouse β-NGFproNGF
StructureMature form (no pro-domain)Includes pro-domain
Receptor BindingTrkA/LNGFRp75NTR/sortilin
Biological EffectNeuronal survivalApoptosis induction
GlycosylationNone2 N-linked, 2 O-linked glycans

Research Limitations and Future Directions

  • Delivery Challenges: Poor blood-brain barrier penetration limits CNS applications .

  • Stability Issues: Short plasma half-life (2–4 hours) necessitates nanoparticle encapsulation .

  • Therapeutic Potential: Ongoing trials for Alzheimer’s, spinal cord injury, and stress urinary incontinence .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered solution containing 20mM phosphate buffer (PB), 200mM sodium chloride (NaCl), adjusted to pH 8.0.
Form
Available in both liquid and lyophilized powder forms.
Lead Time
Typically, we can ship your order within 1-3 business days after receiving it. Delivery time may vary depending on the chosen purchasing method and location. Please consult your local distributor for specific delivery timeframes.
Note: All our proteins are shipped with standard blue ice packs. If dry ice shipping is required, please inform us in advance as additional fees will apply.
Shelf Life
The shelf life of our proteins is influenced by several factors including storage conditions, buffer composition, storage temperature, and the intrinsic stability of the protein itself.
Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. Lyophilized protein retains its stability for 12 months when stored at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. For multiple uses, aliquot to prevent repeated freeze-thaw cycles, which can degrade the protein's integrity.
Tag Info
Tag-Free
Synonyms
Ngf; Ngfb; Beta-nerve growth factor; Beta-NGF
Datasheet & Coa
Please contact us to get it.
Expression Region
130-239aa
Mol. Weight
12.4 kDa
Protein Length
Partial
Purity
Greater than 95% as determined by SDS-PAGE.
Research Area
Neuroscience
Source
E.coli
Species
Mus musculus (Mouse)
Target Names
Ngf
Uniprot No.

Target Background

Function
Nerve growth factor (NGF) plays a critical role in the development and maintenance of both the sympathetic and sensory nervous systems. It acts as an extracellular ligand for the NTRK1 and NGFR receptors, triggering cellular signaling cascades that regulate neuronal proliferation, differentiation, and survival. The immature NGF precursor (proNGF) functions as a ligand for the heterodimeric receptor formed by SORCS2 and NGFR, activating cellular signaling pathways 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). NGF also inhibits metalloproteinase-dependent proteolysis of platelet glycoprotein VI. It 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. A study investigated a long-distance signaling mechanism involving Porcine hemagglutinating encephalomyelitis virus-induced deficits in neurons. The researchers found that Ulk1 repression may result in limited NGF/TrkA retrograde signaling within activated Rab5 endosomes, potentially explaining the progressive failure of neurite outgrowth and survival. PMID: 29875237
  2. Mechanosensitive 'silent' nociceptors are characterized by the expression of the nicotinic acetylcholine receptor subunit alpha-3 (CHRNA3). The mechanically gated ion channel PIEZO2 is involved in NGF-induced mechanosensitivity in these neurons. PMID: 29241539
  3. Antibodies targeting NGF, TrkA, and p75 (also known as CD271) were utilized to explore the expression of these antigens in the non-decalcified young mouse femur. PMID: 29166838
  4. An inducible mouse model was developed to investigate the contribution of autocrine direct action of cleavage-resistant proNGF on systemic microvascular abnormalities in both retina and kidney, major targets for microvascular complications. PMID: 29253516
  5. Both in vivo mechanical loading and in vitro mechanical stretch were shown to induce a significant up-regulation of NGF in osteoblasts within 1 hour of loading. PMID: 28416686
  6. Nerve growth factor negatively regulates bone marrow granulopoiesis during small intestinal inflammation. PMID: 26683342
  7. A study suggests a common mechanism for selective follicular activation induced by a localized increase in NGF in interstitial cells, mediated via the mTOR signaling pathway. PMID: 28542147
  8. Nerve growth factor (NGF) signaling through neurotrophic tyrosine kinase receptor type 1 (TrkA) directs innervation of the developing mouse femur to promote vascularization and osteoprogenitor lineage progression. PMID: 27568565
  9. Mechanical stress-induced upregulation of NGF in colon SMC underlies the visceral hypersensitivity in bowel obstruction through increased gene expression and activity of tetrodotoxin-resistant Na channels in sensory neurons. PMID: 28079757
  10. Results indicate that perivascular nerves innervate neovessels as neovasculatures mature, and NGF accelerates the innervation of perivascular nerves in neovessels. PMID: 27493098
  11. These findings reveal a non-neuronal role for neurotrophins and identify a new regulatory pathway in insulin secretion that could be targeted to improve beta cell dysfunction. PMID: 27825441
  12. NGF signaling directly controls basal APP phosphorylation, subcellular localization, and BACE cleavage. PMID: 27076121
  13. NGF facilitates OVA with lowLPS-induced maturation of mouse BMDCs through LPS-up-regulated p75 NTR via activation of NF-kappaB pathways, providing another mechanism for the involvement of NGF in the Th2 response. PMID: 27437725
  14. NGF-OE mice exhibit age-dependent increases in Substance P and CGRP in the urothelium and hyperinnervation of the bladder. PMID: 27342083
  15. TNF-alpha upregulated Nerve Growth Factor [NGF] expression in synovial fibroblasts and macrophages, while IL-1beta upregulated NGF expression in synovial fibroblasts. These findings suggest that IL-1beta and TNF-alpha may regulate NGF signaling in Osteoarthritic joints, making them potential therapeutic targets for treating Osteoarthritis pain. PMID: 27635406
  16. Functional PAP(thorn) neurons are essential for the analgesic effect, which is mediated by NGF-trkA signaling. PMID: 27306411
  17. NGF negatively regulates growth cone retrograde actin flow on laminin. PMID: 26631553
  18. In E-Reeler retinas, NGF was significantly increased in retinal ganglion cells and glial cells. E-Reeler retinal bipolar cells and RGCs overexpress NGF and p75(NTR) as a protective endogenous response to Reelin deprivation. PMID: 26066836
  19. The NGF-induced up-regulation of TRPV1 via increased synthesis and release of endogenous CGRP leads to improved cardiac performance in I/R-injured diabetic heart. PMID: 25650182
  20. NGF effects on parasympathetic nerves may regulate airway smooth muscle. PMID: 25647301
  21. Ginger extract demonstrates a synaptogenic effect via NGF-induced ERK/CREB activation, resulting in memory enhancement. PMID: 25049196
  22. Proinflammatory cytokines in osteoarthritis (OA) joints and the increased mechanical loading of cartilage may mediate OA pain through the stimulation of NGF expression and release by chondrocytes. PMID: 24438745
  23. While highly conserved, NGF and proNGF of mouse and human origins exhibit distinct properties. Therefore, careful consideration is needed when conducting experiments involving cross-species systems. PMID: 25496838
  24. NGF demonstrates anti-oxidative and hepatoprotective effects, suggesting potential therapeutic applications in treating cholestatic liver diseases. PMID: 25397406
  25. Results suggest that the analgesic effect of CB1 activation may partly be attributed to the inhibition of NGF-induced sensitization of TRPV1. Additionally, the effect of CB1 activation is at least partly mediated by attenuation of NGF-induced increased PI3 signaling. PMID: 25088915
  26. NGF induces the removal of active caspase-3 in a lysosome-dependent manner. PMID: 24787014
  27. These results indicate that NGF exerts an antileishmanial effect by stimulating hydrogen peroxide production in macrophages. PMID: 24937592
  28. Iron accumulation induces NGF expression in hepatocytes, leading to LSEC defenestration via TrkA. PMID: 25460199
  29. Findings suggest that NGF overexpression in the ovary may be sufficient to cause both reproductive and metabolic alterations characteristic of polycystic ovary-like syndrome (PCOS), supporting the hypothesis that sympathetic hyperactivity may contribute to the development and/or progression of PCOS. PMID: 25211588
  30. Increased NGF concentrations abolish Sema3A-induced inhibitory effect on axon outgrowth, while they have no effect on Sema3A-induced collapse rate. PMID: 24338202
  31. Data suggests that proNGF may represent a novel pathway or possible mechanism underlying microglial toxicity in neuroinflammation, offering a potential target for therapeutic manipulation of neurodegenerative diseases. PMID: 24040063
  32. beta-NGF gene transfection promotes the differentiation of bone marrow stromal stem cells into neurons through regulation of AKT and MAPKs signaling pathways. PMID: 23934089
  33. Mechanical stimulation may attenuate NGFbeta signaling through Rac1. PMID: 23989259
  34. Independent of genotype, folate deficiency affects NGF levels in the frontal cortex, amygdala, and hippocampus. PMID: 23623989
  35. The calcineurin/NFAT pathway mediates the upregulation of PAI-1 by NGF. PMID: 23825664
  36. ProNGF/NGF imbalance triggers learning and memory deficits, neurodegeneration, and spontaneous epileptic-like discharges in transgenic mice. PMID: 23538417
  37. Elevated levels of NGF in target tissues stimulate sympathetic and sensory axonal sprouting. PMID: 23322532
  38. PIP5Kalpha acts as a negative regulator of nerve growth factor-induced neurite outgrowth by inhibiting PI3K/Akt signaling pathway in PC12 cells. PMID: 23538529
  39. Data suggest that dietary factors (e.g., olive pomace polyphenols) up-regulate NGF/TrkA (proto-oncogene trk) and BDNF/TrkB (brain derived neurotrophic factor/receptor) in hippocampus/olfactory bulb and down-regulate NGF/BDNF in frontal cortex/striatum. PMID: 23466052
  40. Research indicates that expression of NGF is down-regulated in wounded skin (epidermis) in diabetes. Among the growth factors investigated, only expression of NGF was down-regulated in healing skin wounds of diabetic mice compared to nondiabetic mice. PMID: 23426701
  41. ProNGF selectively promotes the growth of neurites from a subset of NGF-responsive neurons through a p75(NTR)-dependent mechanism during postnatal development when the axons of these neurons are ramifying within their targets in vivo. PMID: 23633509
  42. Overexpression of mouse NGF in urothelium & detrusor affected transcription of PAC1, VPAC1, VPAC2, PAPCAP, VIP & other peptides normally and in cyclophosphamide-induced cystitis. The changes were tissue- and disease-duration dependent. PMID: 22700375
  43. The HTM1 heterodimer of 2 NGF muteins binds p75 and TrkA on opposite sides of the heterodimer, but not 2 TrkA receptors, supporting the ligand passing of NGF from p75 to TrkA via a transient heteroreceptor complex of p75-NGF-TrkA. PMID: 22903500
  44. Our results indicate that BDNF-TrkB, but not NGF-TrkA signaling, is involved in brain repair after ICH. Early proper treadmill exercise may promote this repair process. PMID: 22999926
  45. Expression of NGF in the hippocampus, cortex, and adrenal gland of wild type animals tended to decrease following spaceflight. PMID: 22808101
  46. This study demonstrated that both TrkA and NGF support the survival of only a subset of basal forebrain cholinergic neurons during brain development. PMID: 23100411
  47. Letter: NGF-p75 and neuropsin/KLK8 pathways may cooperate in regulating epidermal homeostasis in inflamed skin. PMID: 22520925
  48. NGF derived from bronchial and alveolar epithelium plays a significant role in airway hyperresponsiveness after chronic exposure to mite antigen. PMID: 22168511
  49. Therapeutic potential of NGF for the prevention of cardiomyopathy in diabetic subjects. PMID: 22187379
  50. NGF exerts profibrotic activities in the airways by inducing type III collagen production in fibroblasts. PMID: 21816457

Show More

Hide All

Database Links

KEGG: mmu:18049

STRING: 10090.ENSMUSP00000102538

UniGene: Mm.1259

Protein Families
NGF-beta family
Subcellular Location
Secreted. Endosome lumen.
Tissue Specificity
Detected in submaxillary gland (at protein level). Highly expressed in male submaxillary gland. Levels are much lower in female submaxillary gland.

Q&A

What is recombinant mouse beta-NGF and how does it differ from full-length NGF?

Recombinant mouse beta-NGF (Ser122-Gly241) is a bioactive fragment of the full-length nerve growth factor protein, produced through bacterial expression systems. Unlike the full-length protein, this partial active form represents the mature, processed segment that retains full biological activity while offering advantages in consistency and solubility. The recombinant partial protein spans amino acids Ser122 to Gly241 of the native sequence, forming a homodimer with a molecular mass of approximately 13.5 kDa (14 kDa by apparent molecular mass in SDS-PAGE conditions). This partial construct captures the essential functional domain while removing regions that can introduce variability in experimental applications .

How does mouse beta-NGF compare structurally to NGF from other species?

Mouse beta-NGF shares significant structural homology with NGF from other mammalian species, exhibiting approximately 90% amino acid sequence identity with human beta-NGF and 95.8% with rat beta-NGF. These high conservation levels indicate the evolutionary importance of this signaling protein. The mature form of mouse beta-NGF is a homodimer composed of two 120 amino acid polypeptides that belongs to the cysteine-knot family of growth factors. These dimeric structures are stabilized by intramolecular disulfide bonds that create the characteristic "knot" configuration essential for receptor binding and biological activity. Despite the high homology, the species-specific differences can influence cross-reactivity in experimental systems, particularly in binding assays or when using antibodies targeted against specific epitopes .

What are the key structural features that enable beta-NGF's biological activity?

The biological activity of beta-NGF is attributable to several key structural features. As a member of the cysteine-knot family of growth factors, its three-dimensional configuration is maintained by conserved disulfide bridges that create a stable core structure. The homodimeric arrangement of the protein presents dual binding interfaces that enable it to interact simultaneously with its receptors (primarily β-NGFR/TrkA and p75NTR). The specific amino acid residues within the Ser122-Gly241 region form recognition motifs that enable selective binding to these receptors, triggering downstream signaling cascades. The recombinant form maintains these critical structural elements despite being expressed in E. coli systems, with proper refolding procedures ensuring the formation of native-like tertiary structure necessary for biological function .

How is the biological activity of recombinant mouse beta-NGF measured and quantified?

The biological activity of recombinant mouse beta-NGF is primarily quantified through cell proliferation assays using TF-1 human erythroleukemic cells. In this standardized bioassay, the effective dose that induces 50% of the maximal response (ED50) typically ranges from 0.3 to 1.5 ng/ml, providing a reliable metric for batch-to-batch comparison. The assay works by measuring the proliferative response of TF-1 cells, which express NGF receptors and demonstrate a dose-dependent growth response when exposed to bioactive NGF. Additional validation may include neurite outgrowth assays using PC12 cells or primary neuronal cultures, receptor binding studies, or phosphorylation analysis of downstream signaling molecules like ERK1/2 or AKT. For research applications requiring precise activity measurements, these functional assays are preferred over simple protein quantification methods like ELISA or Western blotting .

What are the optimal experimental conditions for using recombinant mouse beta-NGF in neural cell culture systems?

For neural cell culture applications, recombinant mouse beta-NGF should be reconstituted according to manufacturer guidelines, typically using a buffer containing 20mM Tris and 150mM NaCl at pH 8.0. The working concentration varies by application: for primary neuronal cultures, 50-100 ng/ml is typically effective; for neurite outgrowth in PC12 cells, 25-50 ng/ml is often sufficient; for survival assays with sympathetic neurons, 10-20 ng/ml may be adequate. The protein should be added to culture media containing serum substitutes rather than full serum (which may contain endogenous growth factors). To maintain activity during extended culture periods, replenishment every 2-3 days is recommended due to the relatively short half-life of the protein in culture conditions. For signaling studies, a minimum pre-incubation of 15-30 minutes is typically required before measuring downstream effects, though longer time points (6-24 hours) are necessary for observing morphological changes .

How does recombinant mouse beta-NGF contribute to B lymphocyte development and function?

Recombinant mouse beta-NGF plays multifaceted roles in B lymphocyte biology, functioning as both a growth and differentiation factor. It enhances B-cell survival by activating anti-apoptotic pathways, primarily through the PI3K/Akt signaling cascade that inhibits pro-apoptotic factors. In functional assays, concentrations of 10-50 ng/ml have been shown to significantly increase survival rates of isolated mouse B cells in serum-free conditions. Beyond survival, beta-NGF influences B-cell differentiation by modulating immunoglobulin production and enhancing the response to antigen stimulation. At the molecular level, NGF binding to TrkA receptors on B lymphocytes activates multiple signaling pathways, including MAPK and PLCγ, that regulate gene expression patterns essential for proper B-cell development. This neurotrophin-immune system crosstalk represents an important aspect of neuroimmune communication, with implications for understanding autoimmune conditions and inflammatory responses .

What are the critical considerations when designing dose-response experiments with recombinant mouse beta-NGF?

When designing dose-response experiments with recombinant mouse beta-NGF, researchers should implement a logarithmic concentration range spanning at least 0.01 ng/ml to 100 ng/ml to capture the complete response curve. This range encompasses the documented ED50 of 0.3-1.5 ng/ml for TF-1 cell proliferation while providing sufficient data points to calculate accurate EC50 values. Include appropriate positive controls (commercial NGF with verified activity) and negative controls (heat-inactivated NGF or irrelevant proteins). Time-course experiments should accompany dose-response studies, as different cellular responses occur at varying temporal scales: receptor phosphorylation (minutes), gene expression changes (hours), and morphological alterations (days). Vehicle controls must match the reconstitution buffer to account for any matrix effects. For maximal reproducibility, standardize serum conditions across experiments, as serum components may contain factors that potentiate or inhibit NGF signaling .

How should researchers approach experimental perturbations using NGF in signaling pathway studies?

When studying signaling pathways using NGF perturbations, researchers should employ a systematic experimental design that includes multiple doses of NGF (typically 0.1, 1, 10, and 100 ng/ml) to capture dose-dependent responses. Single and combinatorial approaches with other growth factors (such as EGF) can reveal pathway crosstalk and integration points. Gene knockdown or overexpression experiments should target both upstream receptors (TrkA, p75NTR) and downstream mediators (MAPK, PI3K) to delineate the signaling cascade comprehensively. For temporal analysis, implement a standardized time-course spanning immediate (5-15 minutes), early (30-60 minutes), and delayed (2-24 hours) responses to distinguish between primary signaling events and secondary adaptive responses. Quantitative readouts should include multiple analytical methods: phosphoprotein analysis via Western blotting or ELISA, transcriptional profiling through qPCR or RNA-seq, and functional assays appropriate to the cell type under investigation .

What methods can be used to verify the authenticity and purity of recombinant mouse beta-NGF preparations?

Verification of recombinant mouse beta-NGF authenticity and purity requires a multi-method analytical approach. SDS-PAGE analysis under both reducing and non-reducing conditions should demonstrate a predominant band at approximately 14 kDa (apparent molecular mass), with purity exceeding 95%. Mass spectrometry analysis can confirm the exact molecular weight (expected 13.5 kDa) and sequence coverage through peptide mapping. Endotoxin testing using the LAL method should verify levels below 1.0 EU per μg to prevent confounding inflammatory responses in sensitive experimental systems. Functional verification through bioassays, particularly the TF-1 cell proliferation assay with an expected ED50 of 0.3-1.5 ng/ml, provides critical confirmation of bioactivity. For immunological applications, Western blotting with anti-NGF antibodies that recognize species-specific epitopes can confirm identity, while size-exclusion chromatography can detect aggregation states that might affect activity. Researchers should maintain a reference standard from a validated lot against which new preparations can be compared .

What are the optimal storage conditions for maintaining the activity of recombinant mouse beta-NGF?

The optimal storage conditions for recombinant mouse beta-NGF vary depending on the form and intended use timeframe. Lyophilized protein demonstrates excellent stability, maintaining activity for up to 12 months when stored at temperatures between -20°C and -80°C, protected from light and moisture. Upon reconstitution, the protein solution exhibits significantly reduced stability, with recommended storage at 4-8°C for short-term use (2-7 days maximum). For intermediate-term storage of reconstituted protein, aliquoting into single-use volumes and freezing at temperatures below -20°C is recommended to minimize freeze-thaw cycles that can substantially degrade activity. When preparing aliquots, using low-protein-binding tubes and adding carrier proteins (such as 0.1% BSA) can prevent adsorption losses. The reconstitution buffer (typically 20mM Tris, 150mM NaCl, pH 8.0) should be maintained at these storage steps, as buffer exchanges may introduce destabilizing conditions that accelerate protein degradation .

How can researchers troubleshoot experiments when recombinant beta-NGF activity appears to be lower than expected?

When recombinant beta-NGF activity appears suboptimal, a systematic troubleshooting approach is essential. First, verify protein concentration using both Bradford/BCA assays and absorbance at 280nm to ensure accurate dosing. Check the storage history of the protein—excessive freeze-thaw cycles or extended periods at temperatures above -20°C can significantly reduce activity. Examine the reconstitution procedure—incomplete solubilization, incorrect buffer composition, or vigorous vortexing can all contribute to reduced activity. Analyze experimental conditions, particularly considering the presence of proteases, oxidizing agents, or extreme pH values that might degrade the protein. For cell-based assays, verify receptor expression levels in target cells, as receptor downregulation through continuous culture can reduce responsiveness. Implement positive controls using freshly prepared NGF from an alternative source or lot to differentiate between protein-specific and system-specific issues. Finally, consider potential interfering substances in the experimental system, such as serum components or concurrently administered factors that might antagonize NGF signaling .

What reconstitution protocols maximize the recovery and activity of lyophilized recombinant mouse beta-NGF?

To maximize recovery and activity during reconstitution of lyophilized recombinant mouse beta-NGF, researchers should follow a precise protocol. Begin by allowing the vial to equilibrate to room temperature before opening to prevent condensation that could affect protein solubility. Add the reconstitution buffer (typically sterile 20mM Tris, 150mM NaCl, pH 8.0) slowly along the sides of the vial rather than directly onto the protein cake. Gentle swirling or rotation for 15-30 minutes is preferable to vigorous vortexing or pipetting, which can cause denaturation through mechanical stress. If complete solubilization is not achieved after initial mixing, allow extended dissolution time at 4°C rather than increasing agitation intensity. For applications requiring high protein recovery, consider adding a carrier protein such as 0.1% pharmaceutical-grade BSA to the reconstitution buffer to prevent adsorption losses to container surfaces. After reconstitution, filter sterilization using low-protein-binding 0.22 μm filters can be performed, but protein concentration should be re-verified afterward to account for losses during filtration .

How can researchers design experiments to distinguish between TrkA and p75NTR-mediated effects of beta-NGF?

Distinguishing between TrkA and p75NTR-mediated effects of beta-NGF requires sophisticated experimental approaches targeting receptor-specific signaling. Pharmacological strategies employ selective inhibitors: K252a selectively blocks TrkA tyrosine kinase activity at 200-300 nM without affecting p75NTR signaling, while function-blocking antibodies against either receptor can provide complementary validation. Genetic approaches using siRNA knockdown, CRISPR/Cas9 gene editing, or cells derived from receptor-specific knockout animals offer more definitive differentiation. Mutant forms of NGF with altered binding affinities for each receptor represent another powerful approach—for example, NGF mutants R100E or R103E show reduced TrkA binding while maintaining p75NTR interaction. At the readout level, measuring distinct signaling pathways helps attribute responses to specific receptors: TrkA primarily activates ERK1/2, PI3K/Akt, and PLCγ pathways, while p75NTR uniquely activates JNK and NF-κB pathways. Combined approaches integrating multiple methods provide the most robust differentiation of receptor-specific contributions to observed biological responses .

What are the methodological considerations when using recombinant mouse beta-NGF in neuronal-glial co-culture systems?

When implementing recombinant mouse beta-NGF in neuronal-glial co-culture systems, several specialized methodological considerations become critical. The differential expression of NGF receptors across cell types necessitates careful experimental design—neurons typically express high levels of TrkA and p75NTR receptors, while certain glial populations (particularly microglia and Schwann cells) may express p75NTR but limited TrkA. Concentration optimization should be performed independently for co-culture systems compared to mono-cultures, as glial cells can secrete factors that potentiate NGF effects or express enzymes that degrade exogenous NGF. Serum-free or defined serum replacement media are strongly recommended to eliminate confounding effects from serum-derived growth factors. For long-term co-culture experiments, implement a consistent NGF replenishment schedule (typically every 48 hours) with concentration verification through ELISA to account for consumption or degradation. When analyzing cellular responses, use cell type-specific markers (MAP2 for neurons, GFAP for astrocytes, Iba1 for microglia) combined with phospho-specific antibodies to distinguish cell type-specific signaling events within the heterogeneous culture system .

How can parameter uncertainty in NGF signaling models be addressed through experimental design?

Addressing parameter uncertainty in NGF signaling models requires sophisticated experimental design strategies that systematically constrain model parameters. Instead of relying on single experimental conditions, researchers should implement a palette of complementary perturbations including dose gradients of NGF (typically 0.1, 1, 10, 100, and 500 ng/ml), combined with orthogonal manipulations such as receptor modulation through knockdown/overexpression approaches. Time-course measurements spanning multiple temporal scales (minutes to days) are essential for capturing both fast signaling events and slower transcriptional responses. Multi-parametric data collection combining proteomics, phosphoproteomics, and transcriptomics provides complementary constraints on model parameters. For particularly important parameters, directed experiments specifically designed to isolate and quantify individual rate constants may be necessary. Sensitivity analysis should guide experimental design by identifying parameters whose uncertainty most significantly impacts model predictions. Increasingly, Bayesian approaches to parameter estimation allow formal integration of prior knowledge with new experimental data, providing posterior probability distributions that quantify remaining uncertainty for each parameter after data integration .

How should researchers design control groups when using recombinant mouse beta-NGF in vivo?

When designing in vivo experiments with recombinant mouse beta-NGF, comprehensive control groups are essential for rigorous interpretation. Vehicle controls should receive identical volumes of the carrier solution used for NGF reconstitution, typically containing the same buffer components (20mM Tris, 150mM NaCl, pH 8.0) and any stabilizing proteins (such as BSA) but lacking NGF. Heat-inactivated NGF controls (prepared by heating at 95°C for 20 minutes) help distinguish between specific biological activity and non-specific protein effects. For studies examining NGF's neurotrophic functions, structurally related neurotrophins (BDNF, NT-3, or NT-4) at equimolar concentrations serve as important specificity controls. Dose-response designs incorporating multiple NGF concentrations (typically spanning 0.1-100 μg/kg) are preferable to single-dose approaches. Time-course controls capturing both acute (hours) and chronic (days-weeks) responses are necessary for understanding temporal dynamics. For genetic background effects, testing in multiple strains or both sexes helps establish the generalizability of observed responses. Finally, antagonist controls using TrkA inhibitors (such as GW441756) or function-blocking antibodies provide mechanistic validation of receptor-mediated effects .

What are the methodological approaches for studying NGF interactions with other neurotrophins in experimental systems?

Studying NGF interactions with other neurotrophins requires sophisticated methodological approaches that can detect complex signaling relationships. For receptor competition studies, pre-incubation protocols with varying concentrations of one neurotrophin followed by fixed concentrations of another can reveal competitive, additive, or synergistic interactions at the receptor level. Sequential application experiments (varying the order and timing of neurotrophin addition) help distinguish between receptor crosstalk and downstream signaling integration. At the molecular level, co-immunoprecipitation of receptors followed by Western blotting for phosphorylation sites can identify how one neurotrophin alters receptor activation by another. Transcriptional profiling through RNA-seq comparing responses to individual neurotrophins versus combinations identifies genes uniquely regulated by specific combinations. In neuronal systems, morphological analyses examining dendrite versus axon growth in response to localized neurotrophin gradients (using microfluidic chambers) reveal compartment-specific interactions. For in vivo studies, combinatorial genetic approaches with conditional deletion of multiple neurotrophin receptors, coupled with selective neurotrophin administration, provide the most definitive evidence of functional interactions .

What are the current limitations and challenges in research applications of recombinant mouse beta-NGF?

Current research with recombinant mouse beta-NGF faces several significant limitations and challenges that researchers must navigate. The relatively short half-life of the protein in experimental systems necessitates frequent replenishment, complicating long-term studies and increasing experimental variability. Species-specificity issues emerge when translating findings between model systems, as the 10% sequence divergence between mouse and human beta-NGF can affect receptor binding kinetics and downstream signaling efficiency. Technical challenges include batch-to-batch variability in specific activity despite consistent protein concentration measurements, requiring functional validation of each lot. The simplified bacterial expression system used for recombinant production lacks mammalian post-translational modifications that may subtly alter protein function compared to native NGF. From an experimental design perspective, the pleiotropic effects of NGF acting through multiple receptor systems (TrkA and p75NTR) and influencing diverse cellular processes complicates the interpretation of results, particularly in heterogeneous cell populations. Additionally, the field lacks standardized reporting of NGF activity units, with some manufacturers reporting mass concentration while others use biological activity units, creating confusion when comparing studies using products from different sources .

What emerging methodologies are enhancing precision in NGF signaling research?

Emerging methodologies are significantly advancing precision in NGF signaling research across multiple dimensions. Optogenetic approaches enabling light-controlled activation of TrkA receptors now permit unprecedented temporal and spatial precision in triggering NGF signaling, allowing researchers to isolate specific subcellular compartments for activation. CRISPR/Cas9-mediated genome editing facilitates precise modification of endogenous NGF receptors or downstream signaling components, creating cellular models with defined mutations that mirror human conditions. Single-cell transcriptomics and proteomics reveal previously obscured cell-to-cell variability in NGF responses within seemingly homogeneous populations, providing insights into cellular decision-making processes. Computational modeling integrating multi-omics datasets now enables prediction of network-level responses to NGF perturbations, facilitating experimental design optimization. Advanced imaging techniques combining super-resolution microscopy with biosensors for second messengers allow real-time visualization of NGF signaling dynamics at the molecular scale. Microfluidic platforms creating precisely controlled NGF gradients enable detailed studies of growth cone guidance and axonal pathfinding. These methodological advances collectively enhance our ability to dissect the complex and dynamic nature of NGF signaling with unprecedented precision and temporal resolution .

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.