GDNF Human, Sf9

Glial-Derived Neurotrophic Factor Human Recombinant, Sf9
Shipped with Ice Packs
In Stock

Description

Production and Formulation

GDNF Human, Sf9 is manufactured using a baculovirus expression system in Sf9 insect cells, ensuring proper folding and glycosylation. Key production parameters include:

ParameterDetail
Source CellsSpodoptera frugiperda (Sf9) insect cells
FormulationLyophilized in 10% glycerol, PBS (pH 7.4)
Storage-20°C for long-term; 4°C for short-term (avoid freeze-thaw cycles)
Stability Additive0.1% HSA/BSA recommended for long-term storage

This system avoids the limitations of bacterial (E. coli) production, which lacks glycosylation capacity and often yields less bioactive GDNF .

Functional Mechanisms and Applications

GDNF Human, Sf9 promotes neuronal survival through RET receptor signaling (via GFRα1 co-receptor) and potentially RET-independent pathways (e.g., NCAM interactions) .

Key Functions

  • Neuronal Survival: Protects dopaminergic neurons in Parkinson’s disease and motor neurons in ALS .

  • Axonal Growth: Stimulates axon outgrowth in hippocampal and spinal neurons .

  • Developmental Roles: Critical for kidney development, spermatogenesis, and enteric nervous system formation .

Research Findings

Study FocusOutcomeSource
Neuroinductive EfficacySf9-produced GDNF shows higher bioactivity than E. coli-derived variants in vitro
Parkinson’s Disease ModelsPromotes dopaminergic neuron survival in rodent models; clinical trials show mixed results
Heparin BindingHeparin affinity influences GDNF distribution and therapeutic efficacy

Comparative Analysis of Production Systems

The choice of producer cells significantly impacts GDNF’s bioactivity:

SystemAdvantagesLimitations
Sf9 (Baculovirus)Glycosylation, proper folding, high purity (>90%)Less efficient than mammalian systems
HEK293 (Mammalian)Optimal post-translational modifications, maximal bioactivityHigher production costs
E. coli (Bacterial)Low-cost, high-yield productionNo glycosylation, reduced bioactivity

Sf9-produced GDNF bridges the gap between bacterial and mammalian systems, offering a balance of cost and functionality .

Clinical and Therapeutic Implications

While GDNF has shown promise in preclinical models, clinical trials for Parkinson’s disease have been inconclusive. Challenges include:

  • Delivery Limitations: Poor penetration of the blood-brain barrier .

  • Immunogenicity: Potential immune responses to recombinant GDNF .

Emerging strategies focus on targeted delivery (e.g., intrastriatal infusion) and combination therapies (e.g., with growth factors like BDNF) .

Product Specs

Introduction

Glial cell-derived neurotrophic factor (GDNF) is a protein that belongs to the GDNF family of ligands. It plays a critical role in various cellular processes, including neurite outgrowth, cell differentiation, cell survival, and cell migration. GDNF promotes the survival of neurons, particularly those affected by Parkinson's disease or amyotrophic lateral sclerosis (ALS), by interacting with GFRa receptors, primarily GFRa1. Additionally, GDNF contributes to spermatogenesis, kidney development, and has been implicated in alcohol metabolism regulation.

Description

Recombinant Human GDNF, produced in Baculovirus, is a single glycosylated polypeptide chain consisting of 113 amino acids (109-211 aa). It has a molecular weight of 12.8 kDa. The protein includes a 10 amino acid His tag at the C-terminus and is purified using proprietary chromatographic techniques.

Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation

The GDNF protein solution has a concentration of 0.25 mg/ml and is supplied in a buffer containing 10% glycerol and Phosphate-Buffered Saline (pH 7.4).

Stability

For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.

Purity

The purity of the GDNF protein is greater than 90.0% as determined by SDS-PAGE analysis.

Synonyms

Glial cell line-derived neurotrophic factor, hGDNF, Astrocyte-derived trophic factor, ATF, ATF1, ATF2, HFB1-GDNF, HSCR3.

Source

Sf9, Baculovirus cells.

Amino Acid Sequence

ADPMRGQRGK NRGCVLTAIH LNVTDLGLGY ETKEELIFRY CSGSCDAAET TYDKILKNLS RNRRLVSDKV GQACCRPIAF DDDLSFLDDN LVYHILRKHS AKRCGCIHHH HHH

Q&A

What is GDNF and why is it produced in Sf9 insect cells?

GDNF (Glial cell line-derived neurotrophic factor) is a member of the neurotrophin family and GDNF family of ligands (GFL). It plays a crucial role in the development, growth, and survival of neurons, particularly midbrain dopaminergic neurons. GDNF enhances neuron survival primarily through interaction with GFRα receptors (mainly GFRα1) .

Sf9 insect cells are used as an expression system for recombinant human GDNF protein production because they provide several advantages:

  • They enable proper folding and post-translational modifications of complex proteins

  • The baculovirus expression system in Sf9 cells can produce higher yields of bioactive proteins compared to some mammalian systems

  • This system allows for production of glycosylated polypeptides that more closely resemble native human proteins

How should recombinant human GDNF from Sf9 cells be stored and handled?

For optimal stability and activity of recombinant human GDNF from Sf9 cells, follow these research-validated handling guidelines:

  • Short-term storage (2-4 weeks): Store at 4°C

  • Long-term storage: Store frozen at -20°C

  • For extended storage, it is recommended to add a carrier protein (0.1% HSA or BSA)

  • Avoid multiple freeze-thaw cycles as they may decrease bioactivity

  • The protein is typically supplied in a formulation containing PBS (pH 7.4) and 10% glycerol

  • Typical concentration is 0.25 mg/ml in commercial preparations

What are the basic applications of human GDNF in neuroscience research?

Human GDNF has several fundamental applications in neuroscience research:

  • Maintenance of neurons and cortical organoids in vitro

  • Differentiation of dopaminergic neurons from human pluripotent stem cell-derived neural progenitors

  • Facilitation of neural progenitor differentiation to astrocytes

  • Investigation of neuronal survival mechanisms

  • Study of neurite outgrowth, cell differentiation, and cell migration

  • Research on neurodegenerative conditions, particularly Parkinson's disease and ALS

How do experimental conditions affect GDNF bioactivity assays?

The bioactivity of GDNF is significantly influenced by experimental conditions, with several critical factors to consider:

GDNF bioactivity can be determined using SH-SY5Y cell proliferation assays. In a typical protocol:

  • Cells are incubated with different GDNF concentrations

  • Co-factors such as retinoic acid and recombinant GFRα1 must be present

  • Incubation typically occurs for 3 days before viable cell measurement

  • Cell viability is measured using MTS assays

  • The EC50 value for high-quality preparations is approximately 18 ng/ml

For reproducible results across experiments:

  • The presence of GFRα receptors is essential as GDNF signaling occurs through these co-receptors

  • The cell type chosen affects response magnitude (SH-SY5Y, primary neurons, etc.)

  • The physiological state of cells (passage number, confluence) impacts reproducibility

  • Duration of treatment must be optimized for the specific outcome measure being assessed

What are the molecular mechanisms of GDNF signaling through GFRα receptors?

GDNF signaling operates through a complex receptor system with diverse downstream effects:

Primary signaling pathway:

  • GDNF binds preferentially to GFRα1 (though it can interact with other GFRα receptors including GFRα3)

  • This complex then recruits and activates the RET receptor tyrosine kinase

  • Activated RET initiates multiple downstream signaling cascades including:

    • PI3K/Akt pathway (promoting cell survival)

    • MAPK/ERK pathway (regulating differentiation and growth)

    • PLCγ pathway (modulating synaptic plasticity)

Alternative signaling mechanisms:

  • GDNF can signal through GFRα1 in a RET-independent manner via NCAM (neural cell adhesion molecule)

  • This alternative pathway activates Fyn and FAK kinases

  • The NCAM-mediated pathway is particularly important for neuritogenesis in the cochlear spiral ganglion

Receptor specificity considerations:

  • While GDNF preferentially binds GFRα1, cross-talk with GFRα3 receptors occurs at higher concentrations

  • This receptor promiscuity should be considered when designing experiments targeting specific neuronal populations

How does Sf9-produced GDNF compare with other expression systems in terms of research applications?

Comparative analysis of GDNF from different expression systems reveals important differences relevant to research applications:

Expression SystemAdvantagesLimitationsOptimal Applications
Sf9 Baculovirus- Higher yield
- Glycosylation pattern
- Proper folding
- >85-90% purity
- Insect-type glycosylation
- Potential for endotoxin
- In vitro assays
- Animal models
- Structural studies
E. coli (bacterial)- Cost-effective
- High yield
- Simpler purification
- Lacks glycosylation
- Refolding often required
- Endotoxin concerns
- Structural studies
- Applications where glycosylation is not critical
Mammalian cells- Human-like glycosylation
- Native conformation
- Highest bioactivity
- Lower yield
- Higher cost
- More complex production
- Clinical applications
- Studies sensitive to glycosylation patterns

Key considerations for research applications:

  • For most in vitro neuronal culture systems, Sf9-produced GDNF provides sufficient bioactivity

  • For complex 3D organoid systems or in vivo studies where glycosylation patterns may affect distribution or immunogenicity, mammalian-expressed GDNF may be preferable

  • When protein folding and bioactivity are critical, Sf9 and mammalian systems consistently outperform bacterial expression systems

What are the current challenges in translating GDNF research to therapeutic applications?

Despite promising preclinical results, several challenges have hindered the translation of GDNF research into clinical therapies:

Delivery challenges:

  • GDNF has poor blood-brain barrier (BBB) penetration

  • Direct intraventricular administration caused adverse effects without reaching target tissues

  • Intraputamenal infusion showed mixed results in clinical trials

  • Development of reliable, long-term delivery systems remains challenging

Clinical trial outcomes:

  • Early phase 1 studies demonstrated therapeutic potential with direct infusion into caudate putamen

  • Subsequent phase 2 trials showed no clinical benefit

  • Development of neutralizing antibodies against GDNF was observed in some patients

  • These mixed results led to termination of some clinical trials

Current research directions:

  • Development of small molecules that mimic GDNF activity

  • Investigation of related neurotrophic factors like CDNF and MANF

  • Exploration of alternative delivery methods including:

    • Encapsulated cell biodelivery (ECB)

    • Gene therapy approaches

    • BBB-penetrating fusion proteins or peptide mimetics

How should researchers design experiments to assess GDNF neuroprotective effects?

When designing experiments to assess GDNF neuroprotective effects, researchers should consider these methodological approaches:

For in vitro models:

  • Primary midbrain dopaminergic neurons provide the most physiologically relevant system

  • SH-SY5Y neuroblastoma cells offer a more accessible alternative

  • Always include GFRα1 co-receptors in the experimental system

  • Use neurotoxins specific to the pathway being studied (e.g., 6-OHDA, MPP+, or rotenone for dopaminergic models)

  • Include both preventive (GDNF added before toxin) and restorative (GDNF added after toxin) treatment paradigms

  • Measure multiple endpoints: cell viability, neurite outgrowth, dopamine production, gene expression

For in vivo models:

  • The rat 6-OHDA model of Parkinson's disease is well-established for testing GDNF efficacy

  • Assess both neuroprotective effects (when GDNF is administered before lesion) and neurorestorative effects (when administered after lesion)

  • Evaluate not only dopamine neuron survival in substantia nigra but also integrity of TH-positive striatal fibers

  • Include behavioral assessments (e.g., rotational behavior, motor function tests)

  • Consider long-term studies to address potential development of neutralizing antibodies

Controls and validations:

  • Include appropriate negative controls (denatured GDNF, unrelated proteins)

  • Use positive controls (known neuroprotective agents)

  • Validate GDNF bioactivity before experiments using established assays

  • Test multiple concentrations to establish dose-response relationships

What methodological approaches can resolve conflicting data on GDNF efficacy?

Researchers encountering conflicting data on GDNF efficacy should implement these methodological approaches:

Standardization of experimental protocols:

  • Use consistent sources of GDNF with documented bioactivity

  • Standardize cell types, animal strains, and lesion models across studies

  • Implement blinded assessment of outcomes to reduce experimenter bias

  • Develop and adhere to reporting guidelines similar to ARRIVE guidelines for animal studies

Analysis of contributing variables:

  • Examine GDNF preparation differences (expression system, purification method, presence of tags)

  • Assess storage conditions and potential degradation

  • Evaluate the influence of co-factors and receptor expression in experimental systems

  • Consider timing of intervention as critical (preventive vs. restorative approaches)

  • Account for age, sex, and species differences in GDNF responsiveness

Advanced analytical approaches:

  • Perform meta-analyses of published studies to identify sources of variability

  • Use systems biology approaches to model GDNF signaling networks

  • Implement multiparametric outcome measures rather than single endpoints

  • Consider pharmacokinetic/pharmacodynamic modeling to optimize dosing regimens

  • Utilize transgenic animals with altered GDNF signaling components to clarify mechanism-specific effects

How can researchers optimize Sf9-based expression systems for human GDNF production?

To optimize Sf9-based expression systems for high-quality human GDNF production, researchers should consider these critical parameters:

Vector design considerations:

  • Include appropriate signal sequences for secretion

  • Optimize codon usage for insect cell expression

  • Consider fusion tags that facilitate purification but don't interfere with folding

  • Design constructs with removable tags when native protein is required

  • Include appropriate promoters (polyhedrin or p10) for high-level expression

Culture and infection optimization:

  • Maintain Sf9 cells in mid-logarithmic growth phase for infection

  • Optimize multiplicity of infection (MOI) - typically between 1-10

  • Determine optimal harvest time (usually 48-72 hours post-infection)

  • Consider co-expression of chaperones for complex proteins

  • Optimize culture medium and supplements for maximum yield

Purification strategy development:

  • Implement two-step purification protocols for >90% purity

  • Consider affinity chromatography (using His-tags) followed by size exclusion

  • Validate proper dimerization using non-reducing SDS-PAGE

  • Confirm glycosylation status using glycosidase treatments

  • Verify biological activity using cell-based assays

Quality control measures:

  • Assess batch-to-batch consistency using bioactivity assays

  • Implement endotoxin testing protocols

  • Verify protein identity using mass spectrometry

  • Confirm proper folding using circular dichroism

  • Establish storage conditions that maintain long-term stability

What are the most sensitive techniques for measuring GDNF-induced signaling in neuronal models?

To effectively measure GDNF-induced signaling in neuronal models, researchers should employ these sensitive techniques:

Protein phosphorylation detection:

  • Phospho-specific Western blotting for key signaling nodes:

    • pRET (Y905, Y1015, Y1062)

    • pAkt (S473)

    • pERK1/2 (T202/Y204)

    • pCREB (S133)

  • Phospho-proteomics using mass spectrometry for unbiased assessment

  • Kinase activity assays to measure downstream enzymatic activation

  • Proximity ligation assays to detect protein-protein interactions in situ

Transcriptional response analysis:

  • RNA-seq for genome-wide transcriptional changes

  • qRT-PCR for targeted gene expression analysis of known GDNF-responsive genes

  • Single-cell RNA-seq to capture heterogeneity in neuronal responses

  • ChIP-seq to identify transcription factor binding events downstream of GDNF signaling

Functional readouts:

  • High-content imaging for neurite outgrowth, branching, and morphology

  • Calcium imaging to assess neuronal activity changes

  • Electrophysiology to measure functional changes in neuronal excitability

  • Neurotransmitter release assays (HPLC or ELISA) for dopamine production

  • Metabolic flux analysis to detect changes in energy metabolism

Live-cell signaling dynamics:

  • FRET-based biosensors for real-time monitoring of signaling events

  • Optogenetic tools combined with GDNF signaling

  • Live-cell receptor trafficking assays

  • Fluorescently tagged GDNF to track binding and internalization dynamics

How does GDNF compare with newer neurotrophic factors like CDNF and MANF?

A comparative analysis of GDNF, CDNF, and MANF reveals important differences and complementary properties:

PropertyGDNFCDNFMANF
StructureGFL family member, forms homodimerNew NTF family, monomericNew NTF family, monomeric
Receptor SystemGFRα1/RET or NCAMNot fully characterizedNot fully characterized
Expression SystemsBacterial, Sf9, mammalianBacterial, Sf9Bacterial, Sf9
Tissue SpecificityPNS and CNS effectsMore CNS-specificCNS and ER stress response
In vitro PNS EffectsPromotes survival of sympathetic and sensory neuronsLimited effect on PNS neuronsLimited effect on PNS neurons
NeuroprotectionProtects dopaminergic neuronsProtects dopaminergic neuronsProtects dopaminergic neurons
NeurorestorationRestores damaged dopaminergic neuronsRepairs TH-positive striatal fibersProtective against ER stress
BBB PenetrationPoorBetter than GDNFBetter than GDNF

Research implications:

  • CDNF appears to be more CNS-specific than GDNF, with virtually no in vitro effects on PNS neurons

  • Both CDNF and MANF may offer advantages for certain CNS applications due to potentially better BBB penetration

  • The different receptor systems suggest potential for complementary or synergistic effects

  • The newer factors (CDNF/MANF) may have unique mechanisms that could overcome limitations encountered with GDNF in clinical trials

What are emerging research directions for GDNF in non-neurological applications?

Beyond its established role in neurological research, GDNF shows promising applications in several other fields:

Reproductive biology:

  • GDNF plays a critical role in spermatogenesis and testicular development

  • Research opportunities exist in:

    • Male fertility treatments

    • Understanding spermatogonial stem cell maintenance

    • Development of in vitro spermatogenesis systems

    • Potential contraceptive approaches targeting GDNF signaling

Kidney development and disease:

  • GDNF is essential for kidney development and may have roles in kidney regeneration

  • Emerging research areas include:

    • Kidney organoid development using GDNF

    • Potential therapeutic applications in acute kidney injury

    • Understanding GDNF's role in renal progenitor cell biology

    • Development of kidney tissue engineering approaches

Addiction research:

  • GDNF has an ameliorating role in alcohol metabolism and addiction

  • New research directions include:

    • GDNF's potential in reducing alcohol cravings and consumption

    • Application in other substance use disorders

    • Understanding the neurobiological mechanisms of addiction that involve GDNF

    • Development of GDNF mimetics for addiction treatment

Cancer biology:

  • GDNF signaling through RET is implicated in several cancers

  • Emerging research opportunities include:

    • Understanding GDNF's role in tumor microenvironments

    • Development of RET inhibitors for cancer therapy

    • Exploration of GDNF as a biomarker in certain cancers

    • Investigation of GDNF's role in cancer cell migration and invasion

What methodological innovations are advancing GDNF delivery for research and potential therapeutic applications?

Recent methodological innovations are addressing the challenges of GDNF delivery for both research and therapeutic applications:

Advanced delivery technologies:

  • Encapsulated cell biodelivery (ECB) systems:

    • Living cells encapsulated in a semipermeable, immunoisolatory membrane

    • Provides continuous GDNF production with stable genetic expression

    • Removable if necessary, offering better control than traditional methods

    • Bypasses the need for repeated injections or permanent genetic modification

  • Nanoparticle-based delivery systems:

    • Biodegradable polymeric nanoparticles for sustained release

    • Lipid nanoparticles with targeting moieties for enhanced BBB penetration

    • Exosome-based delivery of GDNF or GDNF mRNA

    • Magnetic nanoparticles allowing external control of GDNF delivery

Genetic engineering approaches:

  • AAV-based gene therapy vectors with:

    • Neuron-specific promoters for targeted expression

    • Inducible promoter systems for controlled GDNF production

    • Cell-type specific targeting through engineered capsids

    • Regulated secretion systems to prevent GDNF overexpression

Molecular engineering strategies:

  • Development of small molecule GDNF mimetics

  • Creation of BBB-penetrating GDNF fusion proteins

  • Design of GDNF variants with enhanced stability or receptor specificity

  • Receptor-targeted approaches activating GDNF signaling pathways without the protein itself

How can researchers address translational challenges in moving GDNF from bench to bedside?

To overcome translational challenges for GDNF therapies, researchers should implement these strategic approaches:

Improved preclinical models:

  • Develop more predictive animal models that better recapitulate human disease pathophysiology

  • Utilize patient-derived iPSCs and organoids to test GDNF efficacy in human cellular contexts

  • Implement longitudinal imaging to track GDNF distribution and effects over time

  • Design studies that account for disease heterogeneity seen in human patients

Optimized delivery approaches:

  • Address BBB penetration issues through:

    • Development of shuttle peptides for enhanced transport

    • Focused ultrasound techniques for temporary BBB disruption

    • Intranasal delivery approaches for CNS targeting

    • Implantable devices for controlled, site-specific release

Biomarker development:

  • Identify and validate biomarkers that:

    • Predict GDNF responsiveness in patient subgroups

    • Monitor treatment efficacy in real-time

    • Detect development of neutralizing antibodies early

    • Guide optimal dosing and administration schedules

Clinical trial design optimization:

  • Implement adaptive trial designs to adjust for emerging data

  • Use patient stratification based on genetic or biomarker profiles

  • Develop more sensitive clinical outcome measures

  • Consider combinatorial approaches (GDNF with other neuroprotective agents)

  • Address potential immunogenicity through modified GDNF variants or immunomodulatory approaches

Product Science Overview

Introduction

Glial-Derived Neurotrophic Factor (GDNF) is a highly conserved neurotrophic factor that plays a crucial role in the survival and differentiation of various types of neurons. The human recombinant form of GDNF, produced in Sf9 insect cells, is particularly significant in research and therapeutic applications due to its ability to mimic the natural protein’s functions.

Structure and Function

GDNF is a small protein encoded by the GDNF gene in humans. It is known for its ability to promote the survival and morphological differentiation of dopaminergic neurons, which are critical for motor control and are affected in diseases such as Parkinson’s disease . GDNF functions by binding to the GDNF family receptor alpha (GFRα) and the RET proto-oncogene, initiating intracellular signaling pathways that prevent apoptosis and promote neuronal survival .

Production and Purification

The human recombinant form of GDNF is produced using the Sf9 insect cell expression system. This system is advantageous for producing complex proteins with proper folding and post-translational modifications. The recombinant GDNF is typically purified using chromatographic techniques to achieve high purity levels, ensuring its efficacy in research and therapeutic applications .

Biological Activity

Recombinant GDNF has been shown to enhance the survival and differentiation of dopaminergic neurons in culture. It also prevents apoptosis of motor neurons induced by axotomy, a process where neurons are damaged or severed . The protein’s biological activity is measured by its ability to promote the proliferation of specific cell lines, such as rat C6 cells, with an effective dose (ED50) of less than 0.1 ng/ml .

Applications

GDNF has significant therapeutic potential, particularly in the treatment of neurodegenerative diseases like Parkinson’s disease. Its ability to support neuronal survival and differentiation makes it a promising candidate for developing treatments aimed at halting or reversing neuronal damage. Additionally, GDNF is used in various research applications to study neuronal development, survival mechanisms, and neuroprotection .

Stability and Storage

The lyophilized form of recombinant GDNF is stable at room temperature for up to three weeks but should be stored desiccated below -18°C for long-term storage. Upon reconstitution, it should be stored at 4°C for short-term use and below -18°C for long-term use, with the addition of a carrier protein to prevent freeze-thaw cycles .

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.