b NGF Human

Beta Nerve Growth Factor Human Recombinant
Shipped with Ice Packs
In Stock

Description

Mechanism of Action

β-NGF Human binds two primary receptors to mediate downstream signaling:

ReceptorAffinitySignaling Pathways ActivatedBiological Outcomes
TrkA (High-affinity)~20 pMPI3K/Akt, Ras/MAPK, PLC-γNeuronal survival, differentiation
p75NTR (Low-affinity)~10 nMNF-κB, JNKApoptosis or survival (context-dependent)

Proteolytic cleavage of proNGF (precursor) by serine proteases yields mature β-NGF, which exhibits cross-reactivity across human, mouse, and rat models .

Neurotrophic Effects

  • Neuronal Survival: Essential for sympathetic and sensory neuron maintenance; apoptosis occurs upon NGF withdrawal .

  • Central Nervous System: Supports cholinergic neurons in the basal forebrain, with implications for Alzheimer’s disease .

Immune Modulation

  • B-Cell Proliferation: Induces dose-dependent DNA synthesis in human B lymphocytes (EC₅₀ = 0.6 ng/mL) .

  • Mast Cell Activation: Enhances histamine release and lipid mediator production, amplifying inflammatory pain .

Reproductive Roles

  • Ovulation Induction: Seminal β-NGF triggers ovulation in induced ovulators (e.g., llamas) .

  • Gestational Regulation: Serum β-NGF levels rise significantly during pregnancy, correlating with glucose metabolism and fetal development .

Therapeutic Trials

ConditionPhaseDoseOutcomeSide Effects
Diabetic PolyneuropathyIII0.1 μg/kg SCNo significant improvement vs. placeboInjection-site hyperalgesia
HIV-Associated NeuropathyII0.3 μg/kg SCReduced pain scoresTransient myalgia
Alzheimer’s DiseasePreclinicalN/AReduced β-amyloid toxicity in rodent models N/A

Analytical Advances

  • IA-LC–MS/MS Assays: Quantify total NGF (tNGF) and proNGF in serum, revealing a 78× increase in β-NGF during primate gestation .

  • Human Pregnancy: Median tNGF levels rise from 1.53 ng/mL (non-pregnant) to 2.71 ng/mL (third trimester) .

Key Studies

  1. B-Cell Differentiation: β-NGF enhances IgM secretion 5-fold in human B cells, with minimal effect on IgG or IgE .

  2. ProNGF vs. Mature NGF: ProNGF exhibits distinct bioactivity, including increased placental association in miscarriages .

  3. Cross-Species Activity: Human β-NGF retains ~90% homology with rodent variants, enabling translational research .

Challenges

  • Side Effects: Dose-dependent pain at injection sites limits clinical utility .

  • Assay Sensitivity: Traditional ELISAs underperform compared to mass spectrometry (IA-LC–MS/MS) .

Product Specs

Introduction
Nerve Growth Factor-beta (NGF-β) plays a crucial role in stimulating nerve growth. The NGF complex regulates the growth and differentiation of sympathetic and specific sensory neurons. Genetic mutations affecting the NGF-β gene have been linked to hereditary sensory and autonomic neuropathy type 5 (HSAN5). Furthermore, abnormal expression of this gene is associated with allergic rhinitis.
Description
Recombinant Human Nerve Growth Factor-beta, produced in E. coli, is a non-glycosylated homodimer formed by two identical polypeptide chains. Each chain contains 121 amino acids and has a molecular weight of 13.6 kDa, resulting in a total molecular weight of 27.2 kDa for the dimer. The two chains are linked by a disulfide bond. The purification process of NGF-b involves proprietary chromatographic techniques.
Physical Appearance
White, lyophilized (freeze-dried) powder, sterile-filtered.
Formulation
The lyophilized beta-NGF protein was prepared by freeze-drying a 0.2 μm filtered solution containing 0.1% Trifluoroacetic Acid (TFA).
Solubility
For reconstitution, it is recommended to dissolve the lyophilized NGF-b in sterile 18 MΩ-cm H2O to a concentration of at least 100 μg/ml. This solution can then be further diluted in other aqueous solutions as needed.
Stability
Lyophilized Beta-NGF remains stable at room temperature for up to 3 weeks; however, it is recommended to store it desiccated at a temperature below -18°C. After reconstitution, NGF-Beta should be stored at 4°C for a period of 2 to 7 days. For long-term storage, it is advisable to freeze it at -18°C. To enhance stability during long-term storage, consider adding a carrier protein (0.1% HSA or BSA). Avoid repeated freeze-thaw cycles.
Purity
The purity of the protein is determined by SDS-PAGE and is greater than 95.0%.
Biological Activity
The ED50, which represents the effective concentration required to achieve 50% of the maximum response, is determined by measuring the protein's ability to stimulate TF-1 cell proliferation. The typical ED50 is 1.31 ng/ml, which corresponds to a specific activity of 7.6 x 10^5 units/mg.
Synonyms
Beta Polypeptide, NGF, NGFB, HSAN5, Beta-NGF, MGC161426, MGC161428.
Source
Escherichia Coli.
Amino Acid Sequence
MSSSHPIFHRG EFSVCDSVSV WVGDKTTATD IKGKEVMVLG EVNINNSVFK QYFFETKCRD PNPVDSGCRG IDSKHWNSYC TTTHTFVKAL TMDGKQAAWR FIRIDTACVC VLSRKAVRRA.

Q&A

What is human β-NGF and what distinguishes it from proNGF?

Human β-NGF (beta-Nerve Growth Factor) is a secreted dimeric neurotrophin approximately 13 kDa in size that plays a critical role in the development and maintenance of sympathetic and some sensory neurons in the peripheral nervous system. It functions as a trophic factor for basal forebrain cholinergic neurons in the central nervous system and additionally promotes mast cell and basophil proliferation and activation while inducing the differentiation of B cells . β-NGF represents the mature, processed form derived from its precursor, proNGF (approximately 32 kDa), through proteolytic cleavage. The relationship between these two forms is complex, as proNGF is not merely an inactive precursor but possesses distinct biological activities and receptor-binding properties . While β-NGF primarily binds with high affinity to tropomyosin receptor kinase A (TrkA) to promote neuronal survival and growth, proNGF preferentially binds to the p75 neurotrophin receptor, which can trigger apoptotic pathways under certain conditions . These distinct signaling mechanisms result in different, sometimes opposing biological effects, making the balance between proNGF and mature NGF critically important in both developmental processes and pathological conditions.

What methodologies are most reliable for measuring β-NGF levels in human biological samples?

Accurate quantification of human β-NGF in biological samples requires specialized methodologies that address its relatively low abundance and the potential interference from other proteins. Immunoaffinity liquid chromatography-tandem mass spectrometry (LC-MS/MS) represents one of the most sensitive and selective approaches, capable of simultaneously measuring total NGF (tNGF; sum of mature and proNGF) and proNGF specifically using full and relative quantification strategies . This technique offers advantages in distinguishing between the mature and precursor forms, which is crucial given their distinct biological activities. Other commonly employed methods include enzyme-linked immunosorbent assays (ELISA), which provide high sensitivity but may vary in their ability to discriminate between NGF forms depending on the antibodies used. The Simple Plex™ platform, a microfluidic-based immunoassay system, offers an alternative with high precision and reproducibility for β-NGF quantification, particularly useful for samples with limited volume . When selecting a measurement approach, researchers should consider factors such as required sensitivity (typically pg/mL range for β-NGF), specificity for mature versus precursor forms, sample matrix effects, and the need to measure other neurotrophins simultaneously.

How do β-NGF levels fluctuate during normal human physiology, particularly in pregnancy?

β-NGF demonstrates significant physiological fluctuations throughout the human lifespan, with particularly notable changes during pregnancy that suggest important developmental and regulatory roles. Research using qualified immunoaffinity LC-MS/MS assays has revealed that serum total NGF (tNGF) and proNGF levels vary significantly across the three gestational trimesters compared to non-pregnant female controls, indicating regulatory roles in both implantation and maternal vascular adaptation . These fluctuations appear to parallel observations in animal models, where cynomolgus monkeys demonstrate a large and continuous increase in NGF expression (up to approximately 78-fold) during gestation . The balance between mature β-NGF and proNGF is particularly significant, as studies suggest proNGF levels increase during gestation possibly due to impaired processing of the pro form to the mature form . Beyond pregnancy, NGF levels also show dynamic patterns in neonatal development, with levels elevated in human neonates at day 4 compared to umbilical cord blood (after an initial decrease at day 1), while other neurotrophins decreased significantly . These physiological fluctuations underscore the importance of considering reproductive status and developmental stage when designing human studies involving NGF measurement.

What are the primary signaling pathways activated by human β-NGF and how do they differ across cell types?

Human β-NGF activates a complex network of intracellular signaling cascades primarily initiated through binding to the tropomyosin receptor kinase A (TrkA) and the p75 neurotrophin receptor (p75NTR). Upon binding to TrkA, β-NGF triggers receptor dimerization and autophosphorylation, activating three major downstream pathways: the phospholipase C (PLC), mitogen-activated protein kinase (MAPK), and phosphoinositide 3-kinase (PI3K) pathways, which collectively regulate neuronal survival, differentiation, growth and synaptic plasticity . The PLC pathway generates inositol trisphosphate and diacylglycerol, leading to calcium mobilization and protein kinase C activation, while the MAPK pathway stimulates gene transcription critical for neuronal differentiation and survival. Simultaneously, the PI3K pathway activates Akt (protein kinase B), promoting cell survival through inhibition of apoptotic mechanisms . These signaling mechanisms exhibit notable cell-type specificity, with sympathetic neurons demonstrating robust survival responses to NGF, while sensory neurons show pronounced sensitization and sprouting effects that contribute to hyperalgesia and allodynia under inflammatory conditions . In non-neuronal cells like mast cells and basophils, NGF signaling promotes proliferation and degranulation through partially overlapping pathways, contributing to inflammatory responses . This cellular heterogeneity in signaling outcomes highlights the importance of context-specific experimental design when investigating NGF mechanisms.

How can researchers effectively distinguish between direct and indirect effects of β-NGF in complex biological systems?

Distinguishing direct from indirect effects of β-NGF requires multi-faceted experimental approaches that combine temporal analysis, cell-specific manipulations, and molecular pathway dissection. Researchers should implement time-course studies to establish the sequence of events following β-NGF administration or manipulation, as immediate responses (minutes to hours) are more likely to represent direct effects, while delayed responses (days) may involve intermediary signals . Cell-type specific knockdown or knockout of TrkA receptors using CRISPR-Cas9 or conditional genetic approaches can help isolate direct β-NGF responders from secondary signal propagation. Pharmacological approaches using selective TrkA inhibitors or function-blocking antibodies with appropriate controls can complement genetic approaches, particularly in human samples where genetic manipulation is not feasible . Co-culture systems with physical barriers permeable only to soluble factors can determine whether cell-to-cell contact is required for observed effects or if they are mediated by secreted molecules induced by β-NGF. Molecular pathway analysis through phosphoproteomics or targeted western blotting for activated signaling molecules immediately after β-NGF exposure can identify direct signaling nodes versus secondary activation waves. Additionally, transcriptomic analysis comparing immediate early gene responses to later transcriptional changes can help separate primary from secondary effects in the complex biological cascade initiated by β-NGF.

What methodological considerations are crucial when investigating contradictory findings about β-NGF's role in human disease states?

When addressing contradictory findings in β-NGF research, investigators must carefully evaluate several critical methodological factors that might contribute to discrepancies. Assay selection and validation represent paramount considerations, as different immunoassays for β-NGF may detect different epitopes or cross-react with proNGF, potentially measuring different molecular species . Researchers should specifically determine whether their assays measure mature β-NGF, proNGF, or total NGF, as these forms have distinct and sometimes opposing biological effects. Sample handling introduces another significant variable, as NGF is relatively unstable and sensitive to freeze-thaw cycles, proteolytic degradation, and binding to plasma proteins, necessitating standardized collection, processing, and storage protocols . Population heterogeneity contributes substantially to contradictory findings, with factors such as age, sex, comorbidities, medication use, and genetic background potentially influencing NGF levels and responses independently of the disease state under investigation. Temporal considerations are equally important, as NGF levels fluctuate throughout the day and in response to various physiological conditions, including stress and inflammatory status . For clinical studies, disease definition and staging require precise standardization, as different studies may use varying diagnostic criteria or include patients at different disease stages, leading to apparent contradictions in findings about NGF's role in conditions like neuropathic pain or neurodegenerative disorders .

What are the best experimental approaches for studying β-NGF's role in neurodegeneration and potential therapeutic applications?

Investigating β-NGF's role in neurodegeneration requires an integrated experimental approach spanning multiple model systems with complementary advantages. In vitro models using human induced pluripotent stem cell (iPSC)-derived neurons offer a disease-relevant human cellular context for examining NGF's direct effects on neuronal survival, neurite outgrowth, and protection against toxic insults such as amyloid-β or oxidative stress . These systems allow precise manipulation of NGF signaling through genetic or pharmacological approaches while maintaining human-specific protein interactions. For examining circuit-level effects, ex vivo brain slice preparations preserve neural architecture while allowing controlled NGF delivery and electrophysiological assessment of functional outcomes. Animal models of neurodegeneration provide the critical in vivo context, with transgenic mice overexpressing or lacking NGF components revealing developmental and maintenance roles of NGF signaling pathways . When selecting delivery methods for exogenous NGF in these models, researchers should consider alternative administration routes such as intranasal or eye-topical application, which have demonstrated ability to reach damaged brain neurons while avoiding adverse effects associated with direct intracerebral administration . Combining these approaches with state-of-the-art imaging techniques like two-photon microscopy for tracking neuronal morphology or positron emission tomography (PET) with radiolabeled NGF or TrkA ligands can provide dynamic information about NGF's interactions with degenerating neurons. Finally, correlative studies in human biospecimens from well-characterized patient cohorts, including post-mortem brain tissue and cerebrospinal fluid, are essential for validating findings from model systems and establishing clinical relevance.

What explains the discrepancies between promising preclinical findings and clinical trial outcomes for NGF-based therapies?

The translation of promising preclinical findings to effective clinical NGF-based therapies has encountered significant challenges that highlight important considerations for future research. Delivery challenges represent a primary factor, as NGF is a large protein that does not readily cross the blood-brain barrier, necessitating either invasive delivery methods for central nervous system applications or the development of novel delivery strategies such as intranasal or eye-topical administration routes that have shown promise in preclinical studies . Dose-finding complexities further complicate translation, as the therapeutic window for NGF may be narrow, with insufficient doses failing to activate neuroprotective pathways while excessive doses potentially triggering pain hypersensitivity or other adverse effects . Target engagement verification has been inadequate in many trials, making it difficult to determine whether failed outcomes resulted from insufficient NGF delivery to target tissues or true lack of efficacy at the target site. Species differences in NGF signaling and neural circuit architecture may limit the predictive value of rodent models, which often cannot fully recapitulate the complexity of human neurodegenerative conditions or pain states . Patient heterogeneity introduces additional variability, as clinical trials sometimes enroll patients at different disease stages or with varying comorbidities, potentially diluting treatment effects visible only in specific patient subgroups—exemplified by the conflicting outcomes between phase II and phase III trials of NGF for diabetic neuropathy . Additionally, inadequate biological preparation quality has been specifically cited as a potential explanation for undesired side-effects in some clinical trials, suggesting that highly purified human NGF preparations may be required for therapeutic success .

How should researchers design experiments to assess β-NGF's role in pain sensitization and chronic pain conditions?

Designing robust experiments to investigate β-NGF's role in pain requires multi-level approaches addressing both peripheral and central mechanisms. Researchers should implement a comprehensive behavioral assessment battery that includes measures of both evoked pain (mechanical, thermal, and chemical stimuli) and spontaneous pain behaviors, as these may involve different NGF-dependent mechanisms . Species selection requires careful consideration, as rodent models may not fully recapitulate the complexity of human pain conditions, particularly for chronic states like osteoarthritis or chronic low back pain where NGF antibodies have shown clinical promise . Cell-specific genetic manipulation using Cre-lox systems targeting TrkA or p75NTR in specific neuronal populations can help dissect the relative contributions of different NGF-responsive cells to pain states. Temporal considerations are critical, as NGF induces both rapid sensitization through post-translational modifications and longer-term effects via transcriptional changes and structural neuronal remodeling, necessitating both acute and chronic experimental timelines . When assessing potential therapeutic interventions, dose-response relationships should be carefully established, as the therapeutic window for NGF modulation may be narrow, with complete NGF blockade potentially compromising normal neuronal function while insufficient blockade fails to alleviate pain . Molecular phenotyping of dorsal root ganglia and dorsal horn neurons using immunohistochemistry, in situ hybridization, or single-cell RNA sequencing should complement behavioral assessments to reveal mechanisms underlying observed pain behaviors, such as changes in ion channel expression, neuropeptide content, or neuronal sprouting induced by NGF .

What are the optimal storage and handling conditions for maintaining β-NGF stability in experimental settings?

Maintaining β-NGF stability throughout experimental procedures requires strict adherence to optimized handling protocols that preserve its biological activity. Recombinant human β-NGF should be stored at -80°C in single-use aliquots to avoid repeated freeze-thaw cycles, which can significantly reduce bioactivity through protein denaturation and aggregation . When preparing working solutions, researchers should use low-binding microcentrifuge tubes and pipette tips to minimize protein loss through adsorption to plastic surfaces, a common issue with neurotrophic factors due to their hydrophobic regions. Carrier proteins such as bovine serum albumin (0.1-1%) should be added to dilute NGF solutions to prevent adsorption and increase stability, particularly for solutions below 10 μg/mL. The recommended buffer for β-NGF reconstitution is phosphate-buffered saline (pH 7.4) containing 0.1% human serum albumin or bovine serum albumin, as acidic or strongly basic conditions accelerate NGF degradation . Temperature management is crucial during experiments, with NGF solutions kept on ice when possible and exposure to room temperature minimized to less than 2 hours to prevent activity loss. For long-term storage of reconstituted NGF, sterile filtration through 0.22 μm low-protein-binding filters helps prevent microbial contamination while minimizing protein loss. Researchers should regularly validate NGF bioactivity using functional assays such as TrkA phosphorylation or neurite outgrowth in PC12 cells or dorsal root ganglion neurons to ensure that storage conditions are preserving functional integrity.

What considerations are important when selecting between different sources of recombinant human β-NGF for research applications?

The selection of recombinant human β-NGF sources requires careful evaluation of multiple factors that can significantly impact experimental outcomes. Expression system differences represent a primary consideration, as human β-NGF produced in bacterial systems (E. coli) typically lacks post-translational modifications such as glycosylation, potentially affecting protein folding, stability, and receptor interactions compared to mammalian cell-derived NGF . Animal-free production systems have become increasingly important for researchers concerned about potential immune responses or ethical considerations, with dedicated animal-free recombinant NGF now available that is manufactured using non-animal reagents throughout the production process . Purity specifications vary significantly between suppliers, with research-grade preparations typically offering >95% purity while higher-grade preparations may exceed 98% purity with reduced endotoxin levels (<0.1 EU/μg protein), a critical factor for immunology or in vivo studies where endotoxin contamination can confound results . Formulation composition, including buffer components, preservatives, and carrier proteins, requires evaluation for compatibility with intended experimental systems, particularly for electrophysiology, cell culture, or in vivo applications where additives may have independent effects. Functional validation data provided by manufacturers should be scrutinized to ensure the preparation demonstrates bioactivity in relevant assay systems, ideally including dose-response data for TrkA phosphorylation, neurite outgrowth, or survival promotion in appropriate neuronal models . Batch-to-batch consistency represents a final critical consideration, with researchers advised to maintain records of specific lots used for critical experiments and consider bulk purchases of single lots for extended study series to minimize variability.

How can researchers effectively measure and interpret changes in β-NGF receptor expression and signaling?

Comprehensive assessment of β-NGF receptor expression and signaling dynamics requires integration of multiple analytical approaches targeted at different levels of the signaling cascade. For receptor quantification, flow cytometry offers single-cell resolution for measuring surface TrkA and p75NTR levels in heterogeneous cell populations, while quantitative PCR provides information on receptor mRNA expression but may not reflect functional receptor availability at the cell surface . Western blotting for total and phosphorylated forms of TrkA and downstream signaling proteins (MAPK, PI3K, PLC-γ) can reveal activation patterns, with temporal analysis (5-60 minutes post-stimulation) distinguishing between early and late signaling events. Receptor trafficking analysis using fluorescently tagged receptors or antibodies against extracellular domains combined with markers for different cellular compartments can reveal internalization, recycling, or degradation dynamics that regulate NGF signal duration and intensity . Proximity ligation assays or fluorescence resonance energy transfer (FRET) techniques can detect protein-protein interactions within the signaling complex, revealing adaptor protein recruitment patterns specific to different cell types or pathological states. For functional assessment, calcium imaging provides real-time information about NGF-induced calcium transients, while electrophysiological recordings can detect changes in neuronal excitability resulting from NGF-mediated ion channel modulation . When interpreting signaling data, researchers should consider factors such as receptor cross-talk (particularly between Trk and p75NTR pathways), parallel activation by other neurotrophins or growth factors, and differences between acute versus chronic NGF exposure, which can lead to receptor desensitization or compensatory changes in expression patterns.

What are the emerging alternative delivery strategies for β-NGF to target neurological tissues while minimizing adverse effects?

Innovative delivery strategies for β-NGF have emerged to overcome the limitations of conventional administration routes, particularly for targeting the central nervous system while minimizing systemic adverse effects. The nose-to-brain pathway represents a promising non-invasive route that bypasses the blood-brain barrier through the olfactory epithelium, with studies in animal models of Alzheimer's disease demonstrating that nasally administered NGF can reach and protect damaged basal forebrain cholinergic neurons and improve behavioral performance . Detailed mechanistic studies have confirmed this delivery pathway in mice, rats, primates, and humans, offering a means for repeated NGF administration without the invasiveness of direct intracerebral injection or the adverse effects associated with systemic delivery . Similarly, the eye-to-brain pathway has been validated as an alternative route, with topical ocular NGF administration capable of reaching damaged brain neurons, exerting neuroprotective effects, and improving behavioral outcomes in rodent models . This approach has additional benefits for treating concurrent ocular conditions, as demonstrated by NGF's efficacy in resolving corneal ulcers and enhancing tear production in dry eye conditions . Engineered delivery systems such as nanoparticles, liposomes, or polymeric microspheres offer controlled release options that can protect NGF from degradation while providing sustained delivery to target tissues. Gene therapy approaches using viral vectors for ex-vivo NGF gene delivery have reached clinical trials for Alzheimer's disease, demonstrating the potential for spatially restricted, long-term NGF production directly within affected brain regions . These alternative delivery strategies collectively represent promising approaches to overcome the delivery challenges that have limited the clinical translation of NGF's therapeutic potential.

What explains the therapeutic potential of β-NGF antibodies in pain conditions despite NGF's role in neuronal health?

The therapeutic utility of β-NGF antibodies in pain conditions despite NGF's crucial role in neuronal health reflects the specialized role of NGF in pain hypersensitivity that can be targeted without compromising essential neuronal maintenance functions. NGF functions as a key pronociceptive factor in inflammatory and chronic pain states through multiple mechanisms: it sensitizes peripheral nociceptors by enhancing ion channel activity (particularly TRPV1), induces local neuronal sprouting at injury sites, and contributes to both peripheral and central sensitization through TrkA activation . These pain-promoting effects represent inducible, context-specific NGF actions that can be distinguished from its constitutive roles in neuronal survival, which are more critical during development than in the adult nervous system under normal conditions . Anti-NGF antibodies have demonstrated efficacy in clinical trials for osteoarthritis and chronic low-back pain by specifically interrupting these pathological pain-promoting mechanisms without affecting baseline neuronal function . The safety profile of NGF antibodies supports this therapeutic concept, as common adverse events primarily include peripheral edema, paresthesia, hypoesthesia, and arthralgias rather than overt neurodegeneration, although joint safety concerns emerged in some studies where NGF antibodies were combined with NSAIDs . This therapeutic approach is supported by evidence that NGF levels are elevated in multiple chronic pain conditions, and that administration of exogenous NGF can induce hyperalgesia in otherwise normal experimental animals and healthy human subjects . The successful targeting of NGF for pain relief without compromising neuronal health underscores the context-dependent nature of neurotrophin signaling and demonstrates how careful modulation of these pathways can yield therapeutic benefits for specific conditions.

How should researchers design clinical biomarker studies involving β-NGF measurements in human subjects?

Designing robust clinical biomarker studies for β-NGF requires meticulous attention to pre-analytical, analytical, and post-analytical variables to generate reliable and interpretable results. Pre-analytical considerations should include standardized collection protocols specifying time of day (to control for diurnal variations), fasting status, physical activity levels, and medication restrictions, as these factors can significantly influence NGF levels . Sample processing must be strictly controlled, with immediate centrifugation and aliquoting of serum or plasma samples followed by storage at -80°C with documentation of freeze-thaw cycles, as NGF is sensitive to degradation . The selection of appropriate analytical methods is critical, with immunoaffinity LC-MS/MS offering advantages in distinguishing between mature NGF and proNGF, an important consideration given their distinct biological activities . Assay validation for the specific biological matrix (serum, plasma, cerebrospinal fluid, tissue homogenates) must be thoroughly documented, including limits of detection and quantification, linearity ranges, and potential interfering substances. Study design should incorporate longitudinal sampling where possible to capture intra-individual variations and disease progression effects, with careful matching of case and control subjects for confounding factors such as age, sex, BMI, and comorbidities . Sample size calculations must account for the typically high variability in NGF measurements, with pilot studies recommended to establish expected effect sizes and variability in the specific clinical population. Statistical analysis plans should pre-specify approaches for handling values below detection limits, outliers, and multiple testing corrections, with consideration of both absolute NGF values and ratios between NGF forms (e.g., mature/proNGF ratio) as potentially informative biomarkers .

What are the key differences in β-NGF biology across species that may impact translational research?

Species-specific differences in β-NGF biology represent critical considerations for translational research, potentially explaining discrepancies between promising preclinical findings and clinical outcomes. Sequence variations between human and rodent NGF, though modest (approximately 90% homology), result in structural differences that can affect receptor binding affinity and downstream signaling efficiency, potentially altering both therapeutic efficacy and side effect profiles when translating between species . Expression patterns of NGF and its receptors show notable species differences, with variations in the distribution and density of TrkA receptors across neural populations and in target tissues such as skin, joints, and visceral organs, requiring careful consideration when selecting animal models for specific conditions . Developmental regulation of NGF and its receptors also varies between species, with humans showing more prolonged expression of TrkA receptors in certain neuronal populations compared to rodents, potentially affecting the interpretation of developmental studies or age-dependent interventions . Pharmacokinetic differences further complicate translation, as plasma protein binding, tissue distribution, metabolism, and clearance of both endogenous NGF and therapeutic NGF or NGF-targeted antibodies can vary substantially between species, necessitating careful dose adjustment rather than simple weight-based scaling . Finally, pain processing and perception mechanisms show significant species differences, with humans having more complex pain modulation systems and affective pain components that cannot be fully modeled in rodents, potentially explaining why NGF-targeted therapies might show different efficacy or side effect profiles when translated to human pain conditions . These species differences collectively underscore the importance of incorporating multiple model systems, including human tissue and cell-based assays, when developing and evaluating NGF-targeted therapeutics.

How does β-NGF contribute to the pathophysiology of neurodegenerative disorders and what measurement approaches are most informative?

β-NGF plays a complex role in neurodegenerative disorders, particularly Alzheimer's disease (AD), where alterations in NGF metabolism and signaling contribute to the selective vulnerability of basal forebrain cholinergic neurons (BFCN). Multiple lines of evidence indicate that NGF synthesis and/or brain NGF signaling are markedly affected in neurodegenerative disorders, while exogenous administration of NGF demonstrates potential to protect degenerating neurons . In AD, the primary issue appears to be disrupted NGF transport and processing rather than reduced production, with studies showing accumulation of proNGF in the affected brain regions alongside reduced mature NGF levels, suggesting impaired conversion of proNGF to mature NGF . This imbalance is particularly significant given the contrasting effects of these forms, with mature NGF promoting neuronal survival while proNGF can trigger apoptosis through p75NTR when TrkA levels are reduced, as occurs in AD . Measurement approaches for investigating NGF's role in neurodegeneration should include quantification of both mature NGF and proNGF, ideally using methods like immunoaffinity LC-MS/MS that can distinguish between these forms in brain tissue or cerebrospinal fluid . Additionally, assessment of TrkA and p75NTR receptor expression and activation state in affected neuronal populations provides crucial information about NGF signaling capacity, best achieved through immunohistochemistry on post-mortem tissue or PET imaging with radiolabeled ligands in living subjects. Functional evaluation of NGF's neuroprotective potential requires monitoring downstream signaling pathways including MAPK/ERK and PI3K/Akt activation, neuronal survival markers, and behavioral outcomes in experimental models or clinical trials of NGF-based interventions .

What is the current understanding of β-NGF's role in inflammatory conditions and immune regulation?

β-NGF functions as a significant neuro-immune mediator that orchestrates bidirectional communication between the nervous and immune systems in both physiological and pathological inflammatory states. During inflammatory responses, immune cells including mast cells, basophils, and macrophages produce and release NGF, which then acts on sensory neurons to enhance nociceptor sensitivity and promote neurogenic inflammation through neuropeptide release, creating a feed-forward inflammatory cycle . Simultaneously, NGF directly modulates immune cell functions, promoting mast cell and basophil proliferation and activation while influencing B cell differentiation, further amplifying inflammatory cascades . The enhanced neuronal sensitivity induced by NGF involves multiple mechanisms, including post-translational modification of ion channels (particularly TRPV1), increased expression of nociceptive neuropeptides like Substance P and Calcitonin Gene-Related Peptide, and structural neuronal remodeling through local sprouting at inflammatory sites . These processes contribute to pain hypersensitivity in conditions such as osteoarthritis, where NGF levels are elevated in affected tissues and correlate with disease severity, providing the rationale for therapeutic targeting of NGF with antibodies that have shown efficacy in clinical trials . Beyond localized inflammation, systemic inflammatory states like diabetes mellitus show altered NGF regulation, with elevated NGF serving as both a protective factor against diabetic neuropathy and vasculopathy and a contributor to insulin resistance in gestational diabetes . These complex and sometimes contradictory roles of NGF in inflammatory regulation highlight the context-dependent nature of neurotrophin signaling and underscore the importance of precisely targeted therapeutic approaches that modulate specific aspects of NGF function rather than completely blocking all NGF signaling.

How should researchers interpret β-NGF measurements in pregnancy-related conditions and reproductive disorders?

Interpretation of β-NGF measurements in pregnancy and reproductive disorders requires careful consideration of normal physiological fluctuations, methodological factors, and the complex interplay between maternal and fetal systems. Researchers should first establish trimester-specific reference ranges for both total NGF and proNGF using qualified assays, as studies indicate significant variations across the three gestational trimesters compared to non-pregnant female controls . When evaluating results, the relative balance between mature NGF and proNGF may be more informative than absolute levels of either form alone, as pregnancy appears to involve changes in NGF processing with potentially increased proNGF levels due to impaired conversion to the mature form . Sample type selection significantly impacts findings, with studies reporting different NGF patterns in maternal serum versus placental tissue or amniotic fluid, necessitating consistent sampling approaches when comparing across studies or patient groups . In pregnancy complications such as gestational diabetes mellitus, elevated NGF has been specifically linked to glucose metabolism, insulin resistance, and pancreatic β cell function, suggesting potential utility as a biomarker for metabolic dysregulation in pregnancy when appropriately normalized and compared to matched controls . For implantation failure or early pregnancy loss, assessment of NGF distribution in placental tissue rather than circulating levels may be more informative, as unbalanced NGF distribution in placental tissue has been associated with miscarriages in humans . Longitudinal sampling designs offer advantages over cross-sectional approaches by allowing each subject to serve as her own control, thereby reducing the impact of inter-individual variability in baseline NGF levels and improving the ability to detect clinically meaningful changes associated with pregnancy complications or reproductive disorders.

What is the evidence for β-NGF's therapeutic potential in ocular conditions and what are the optimal delivery methods?

β-NGF has demonstrated significant therapeutic potential for various ocular conditions through its multifaceted effects on corneal, retinal, and optic nerve tissues, with several delivery approaches showing promise in both experimental and clinical settings. For corneal conditions, topical eye NGF administration has proven particularly effective for promoting epithelial healing, with clinical studies confirming that NGF restores corneal integrity in human patients with immune or neurotrophic corneal ulcers by stimulating migration and proliferation of corneal epithelial cells . The efficacy of this approach stems from NGF's dual action on both the epithelial cells themselves and on the dense network of sensory nerves that innervate the cornea, which play a critical role in maintaining corneal health and proper tear film dynamics . For dry eye syndrome, NGF eye drops have enhanced tear release in both human patients and animal models by restoring the neuronal component of lacrimation regulation, addressing an underlying mechanism of the condition rather than merely providing symptomatic relief . In retinal pathologies, studies in animal models of retinitis pigmentosa have shown that topical NGF administration can delay photoreceptor degeneration, with mechanistic studies demonstrating enhanced gene and receptor expression in visual cortex and geniculate nucleus following ocular NGF treatment . The optimal delivery method depends on the target ocular tissue, with topical eye drops providing excellent access to corneal and anterior segment tissues while having the additional advantage of potential transport to central visual pathways through the eye-to-brain route . For posterior segment conditions affecting the retina and optic nerve, intravitreal injection or sustained-release systems may provide more direct access, though the potential for topical NGF to reach these tissues through transcleral diffusion or neural transport pathways continues to be investigated.

How can researchers address data inconsistencies in β-NGF measurements across different laboratories and assay platforms?

Resolving data inconsistencies in β-NGF measurements requires a systematic approach to identify and mitigate sources of variability across assay platforms and laboratory settings. Implementation of standard reference materials represents an essential first step, with recombinant human β-NGF calibrated against international standards serving as a common calibrator across laboratories to enable direct comparison of absolute values . Method harmonization through detailed standard operating procedures that address critical pre-analytical variables (sample collection, processing, storage conditions) and analytical parameters (incubation times, temperatures, washing procedures) can significantly reduce method-dependent variability. Interlaboratory comparison studies, where identical samples are analyzed across multiple laboratories and platforms, can quantify systematic biases between methods and establish conversion factors when necessary, similar to approaches used for standardizing other biomarkers. Method validation parameters should be comprehensively reported in publications, including limits of detection and quantification, linearity ranges, recovery rates, matrix effects, and both intra- and inter-assay coefficients of variation to enable proper interpretation of reported values and assessment of method suitability for specific applications . Epitope mapping of antibodies used in immunoassays is particularly important for NGF measurements, as antibodies may recognize different forms (mature NGF versus proNGF) or be affected by post-translational modifications or protein-protein interactions that alter epitope accessibility . When comparing data across studies, researchers should employ statistical approaches that account for method-dependent biases, such as standardization to method-specific reference ranges or the use of ratios or percent changes rather than absolute values when methods cannot be directly harmonized. Additionally, integration of orthogonal measurement approaches, such as combining immunoassays with functional bioactivity assays, can provide complementary information and increase confidence in findings despite methodological differences.

What statistical approaches are most appropriate for analyzing complex β-NGF pathway interactions and regulatory networks?

Table 1: Comparison of Methodological Approaches for β-NGF Measurement in Human Samples
Method
Immunoaffinity LC-MS/MS
ELISA
Simple Plex™ Platform
Western Blot
Gene Expression Analysis

How can researchers determine biologically significant changes in β-NGF levels versus technical or physiological variability?

Distinguishing biologically meaningful changes in β-NGF levels from technical or physiological variability requires multifaceted approaches that incorporate appropriate experimental controls, statistical methods, and biological validation. Establishing assay-specific technical variability through repeated measurements of identical samples under controlled conditions allows researchers to calculate the minimum detectable difference that exceeds technical noise, typically requiring changes of at least 2-3 times the coefficient of variation for reliable detection . Physiological variability assessment through longitudinal sampling of healthy individuals under standardized conditions (controlling for time of day, physical activity, fasting status) provides critical information about normal fluctuation ranges, with studies suggesting that intra-individual variability in NGF levels may range from 15-30% depending on the biological matrix and measurement method . Defining effect sizes that constitute biological significance should incorporate known physiological changes observed in well-characterized conditions, such as the documented increase in NGF during pregnancy or inflammatory states, providing benchmarks for evaluating novel findings . Correlation with functional outcomes strengthens the biological significance interpretation, particularly when NGF level changes correspond to alterations in downstream signaling markers (TrkA phosphorylation, MAPK activation), target tissue responses (neuronal survival, sprouting), or clinical parameters (pain scores, disease progression markers) . Power analysis incorporating both technical and biological variability should guide sample size determination, with larger samples needed when investigating subtle NGF changes or heterogeneous populations. Multiple testing correction using methods appropriate to the study design (Bonferroni, false discovery rate) prevents inflation of false positive findings when examining NGF levels across multiple timepoints or in relation to numerous clinical parameters. Finally, independent validation in separate cohorts or through orthogonal measurement methods provides the strongest evidence that observed NGF changes represent genuine biological phenomena rather than methodological artifacts or random variation.

What are the best practices for integrating β-NGF data with other neurotrophin measurements and broader -omics datasets?

Integration of β-NGF data with other neurotrophin measurements and broader -omics datasets requires systematic approaches that address technical heterogeneity while maximizing biological insights from complementary data types. Data normalization techniques specific to each platform should be applied before integration attempts, with methods such as quantile normalization for transcriptomic data, total ion current normalization for proteomics, and standardization to reference ranges for NGF and other neurotrophin measurements ensuring compatibility across diverse data types . Batch effect correction using methods such as ComBat or surrogate variable analysis is essential when integrating data from different experimental runs or laboratories, particularly for sensitive measurements like NGF levels that may be affected by technical factors. Multi-omics integration frameworks such as similarity network fusion, DIABLO (Data Integration Analysis for Biomarker discovery using Latent cOmponents), or joint matrix factorization approaches can identify coordinated patterns across data types, revealing how NGF signaling cascades influence gene expression, protein abundance, and metabolic pathways . Pathway enrichment analysis incorporating NGF and other neurotrophins as well as their downstream targets can identify biological processes and molecular functions coordinately regulated by these factors, with tools like Ingenuity Pathway Analysis or MetaCore providing curated knowledge bases of neurotrophin signaling networks. Visualization approaches that simultaneously display multiple data types, such as multi-omics heatmaps, Circos plots, or network visualizations with nodes colored by data type, facilitate interpretation of complex relationships and generate testable hypotheses about NGF's role within broader biological systems. Longitudinal integration strategies that track changes in NGF alongside other molecular and clinical measures over time can reveal temporal relationships and potential causal sequences, particularly important when studying developmental processes or disease progression. Lastly, predictive modeling approaches that incorporate NGF data with other -omics measures to forecast biological outcomes or treatment responses can prioritize NGF-related features for further investigation while establishing their relative importance within broader molecular landscapes.

Product Science Overview

Structure and Characteristics

β-NGF is a protein that belongs to the cysteine-knot family of growth factors, characterized by their stable dimeric structures . The molecular weight of human recombinant β-NGF is approximately 13.6 kDa . It is a potent neurotrophic factor that signals through its high-affinity receptor, tyrosine kinase A (trkA), and low-affinity receptor, p75 neurotrophin receptor (p75NTR) .

Biological Function

β-NGF is essential for the survival and maintenance of sympathetic and sensory neurons . It regulates cell growth, promotes differentiation into neurons, and facilitates neuron migration . Additionally, β-NGF acts as a growth and differentiation factor for B lymphocytes and enhances B-cell survival . It also possesses pro-inflammatory and pro-fibrogenic properties .

Mechanism of Action

The biological activity of β-NGF is mediated through its binding to the trkA receptor with high affinity and to the p75NTR with low affinity . This binding activates several intracellular signaling pathways, including the AKT and MAPK pathways, which are crucial for cell survival, differentiation, and proliferation .

Applications

Human recombinant β-NGF has several applications in research and medicine:

  1. Neuroscience Research: It is used to study the mechanisms of neuronal survival, differentiation, and growth.
  2. Stem Cell Research: β-NGF promotes the differentiation of bone marrow mesenchymal stem cells into neurons .
  3. Immunology: It plays a role in the regulation of the immune system and may function as an intracellular messenger to regulate the body’s response to stress .
  4. Therapeutic Potential: Due to its neurotrophic properties, β-NGF is being explored for potential therapeutic applications in neurodegenerative diseases and nerve injuries.
Production and Purity

Human recombinant β-NGF is typically produced in expression systems such as HEK293 cells or E. coli . The protein is purified to a high degree, with purity levels exceeding 95% . It is available in various formulations, including animal component-free versions, to meet the needs of different research applications .

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.