GDNF Human

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

Description

Overview of GDNF Human

GDNF Human is a 134-amino acid glycosylated homodimer protein (30.4 kDa) belonging to the transforming growth factor-β (TGF-β) superfamily . It signals through a receptor complex involving GDNF family receptor alpha (GFRα1/2) and the RET tyrosine kinase proto-oncogene . Key functions include:

  • Neuroprotection: Supports dopaminergic, motor, and enteric neurons .

  • Spermatogenesis: Regulates spermatogonia differentiation via RET and SCF gradients .

  • Organ Development: Critical for kidney morphogenesis and hair follicle formation .

Mechanisms of Action

GDNF activates multiple pathways:

  • RET-Dependent: Promotes dimerization of RET, triggering PI3K/AKT and MAPK/ERK cascades for survival and neurite outgrowth .

  • RET-Independent: Binds neural cell adhesion molecule (NCAM) to modulate synaptic plasticity .

  • Dopaminergic Support: Enhances dopamine uptake and prevents apoptosis in Parkinson’s disease (PD) models .

Parkinson’s Disease

  • Clinical Trials: Intraputaminal GDNF infusion in PD patients showed:

    • 100% improvement in dopamine transporter PET signals in targeted brain regions .

    • 5–50% symptom improvement in movement, mood, and daily activities, though statistical significance was not achieved .

  • Biomarker Potential: Serum GDNF levels correlate with executive function in PD patients (e.g., working memory, cognitive flexibility) .

Amyotrophic Lateral Sclerosis (ALS)

  • Neural Progenitor Transplants: GDNF-secreting human neural progenitors survived for 42 months post-transplantation in ALS patients, demonstrating safety and sustained protein delivery .

Table 2: Key Clinical Trial Outcomes (GDNF in PD)

Trial PhaseParticipantsDelivery MethodOutcomeSource
Phase II41Intraputaminal portPET scan improvement (+100%), no statistical symptom benefit
PreclinicalRodent/PrimateViral vector (AAV5-GDNF)Rescue of dopaminergic neurons, reduced motor deficits

Research Challenges

  • Delivery Barriers: Poor blood-brain barrier penetration necessitates invasive methods (e.g., intracranial ports) .

  • Variable Efficacy: Inconsistent results in human trials vs. robust preclinical data suggest dosage or timing limitations .

  • Transcript Variants: Alternative splicing produces isoforms (e.g., astrocyte-derived trophic factors) with unclear biological roles .

Future Directions

  • Gene Therapy: AAV vectors with GFAP promoters show promise for targeted astrocytic GDNF expression .

  • Biomarker Development: Correlating serum GDNF levels with cognitive decline could enable early PD diagnosis .

  • Combination Therapies: Pairing GDNF with dopamine agonists or anti-inflammatory agents may enhance efficacy .

Product Specs

Introduction

GDNF is a protein that plays a crucial role in the survival, differentiation, and protection of specific nerve cells. It helps these neurons develop and function correctly. GDNF works by binding to a receptor called RET, which signals within the cell to promote its survival. This protein is important for the development of the nervous system, and alterations in its gene may be linked to certain developmental disorders. Furthermore, GDNF has demonstrated the ability to enhance the survival and specialized development of a specific type of nerve cell and boost their ability to absorb substances.

Description
Recombinant Human Glial Derived Neurotrophic Factor, a protein produced in E. coli, is a non-glycosylated homodimer with two identical chains connected by a disulfide bond. Each chain contains 135 amino acids, totaling a molecular weight of approximately 30kDa. The protein is purified using advanced chromatographic methods to ensure its high quality and purity.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
GDNF was lyophilized from a 0.2µm filtered concentrated solution in 1xPBS, pH 7.4.
Solubility
To reconstitute lyophilized Glial Derived Neurotrophic Factor, it is recommended to dissolve it in sterile 18MΩ-cm H2O at a concentration of at least 100µg/ml. This solution can be further diluted in other aqueous solutions as needed.
Stability
Lyophilized Glial-derived Neurotrophic Factor remains stable at room temperature for up to 3 weeks. However, for long-term storage, it is recommended to store it desiccated below -18°C. Once reconstituted, GDNF should be stored at 4°C for 2-7 days or below -18°C for future use. Adding a carrier protein like HSA or BSA (0.1%) is recommended for extended storage. Avoid repeated freeze-thaw cycles to maintain protein stability.
Purity
The purity of the protein is greater than 95.0%, as determined by: (a) Analysis by RP-HPLC. (b) Analysis by SDS-PAGE.
Biological Activity
The ED50, determined by the proliferation of rat C6 cells, is < 0.1 ng/ml, which corresponds to a specific activity of > 1.0x107 units/mg.
Synonyms
ATF1, ATF2, HFB1-GDNF, GDNF.
Source
Escherichia Coli.
Amino Acid Sequence
MSPDKQMAVL PRRERNRQAA AANPENSRGK GRRGQRGKNR GCVLTAIHLN VTDLGLGYET KEELIFRYCS GSCDAAETTY DKILKNLSRN RRLVSDKVGQ ACCRPIAFDD DLSFLDDNLV YHILRKHSAK RCGCI.

Q&A

What is the structural composition of human GDNF?

Human GDNF is a glycosylated, disulfide-bonded homodimer molecule consisting of two 15 kDa polypeptide chains (monomers). Each monomer contains seven conserved cysteine residues, with Cys-101 specifically used for inter-chain disulfide bridging. The other cysteine residues are involved in intramolecular ring formation creating a cysteine-knot configuration. The calculated molecular weight of recombinant human GDNF is approximately 30.4 kDa . The mature form of GDNF exists as a secreted homodimer that functions as a ligand for the RET (rearranged during transfection) protooncogene .

How is GDNF normally expressed in the healthy human brain?

In the healthy adult brain, GDNF expression is primarily restricted to neurons rather than glial cells, despite its name suggesting glial origin. GDNF expression is highest during development and decreases in adulthood, becoming limited to specific regions including the cortex, hippocampus, striatum, substantia nigra, thalamus, cerebellum, and spinal cord . Interestingly, in some regions like the striatum, GDNF is expressed by specific neuronal subpopulations, particularly parvalbumin-expressing interneurons . This neuronal-derived GDNF acts in a paracrine fashion by forming a complex with the GDNF family receptor α1 (GFRα1), which then activates downstream signaling pathways .

What are the primary signaling mechanisms of GDNF in neuronal cells?

GDNF signals through multiple pathways:

  • Primary pathway: GDNF forms a complex with GDNF family receptor α1 (GFRα1), which then signals through the RET receptor tyrosine kinase .

  • Alternative pathways:

    • GDNF/GFRα1 can signal through neural cell adhesion molecule (NCAM) with lower affinity

    • GDNF can signal independently from GFRα1 by interacting with syndecan-3

GFRα1 can act in two modes:

  • In cis as a membrane-bound factor

  • In trans as a soluble factor

The specificity of GDNF's pro-survival effect appears to be determined mainly by RET expression, which is found in neurons involved in several pathways (nigro-striatal dopaminergic neurons, motor neurons, enteric neurons, sensory neurons, etc.) .

How does GDNF expression change during neuroinflammation and neurodegeneration?

During brain injury or neuroinflammation, GDNF expression patterns change dramatically. Unlike in the healthy brain where GDNF is primarily neuron-derived, de novo expression occurs in glial cells in the diseased brain . Neuroinflammation induces GDNF expression in:

  • Activated astrocytes

  • Activated microglia

  • Infiltrating macrophages

  • Nestin-positive reactive astrocytes

  • Neuron/glia (NG2) positive microglia-like cells

This disease-related GDNF overexpression can be either beneficial or detrimental depending on the brain region affected and the level and duration of glial cell activation . Some studies have also described the upregulation of RET and GFRα1 in glial cells, suggesting that GDNF could play a role in modulating neuroinflammation .

What is the relationship between GDNF and dopamine signaling in schizophrenia?

Recent research has identified a critical relationship between GDNF and dopaminergic abnormalities in schizophrenia:

  • A 2-3 fold increase in endogenous GDNF in the brain is sufficient to induce molecular, cellular, and functional changes in dopamine signaling in both the striatum and prefrontal cortex

  • These changes include increased striatal presynaptic dopamine levels and reduction of dopamine in the prefrontal cortex, which resemble dopaminergic abnormalities seen in schizophrenia

  • Adenosine A2a receptor (A2AR) has been identified as a possible mediator of GDNF-driven dopaminergic abnormalities

  • Pharmacological inhibition of A2AR with istradefylline partially normalized striatal GDNF and striatal and cortical dopamine levels in mice

  • Significantly, GDNF levels are increased in the cerebrospinal fluid of first-episode psychosis patients and in post-mortem striatum of schizophrenia patients

These findings suggest GDNF-A2AR crosstalk may regulate dopamine function in a therapeutically targetable manner for schizophrenia treatment .

How effective is GDNF as a therapeutic agent in Parkinson's disease models?

GDNF has shown promising therapeutic potential in Parkinson's disease models:

  • GDNF promotes the survival and differentiation of dopaminergic neurons in culture and prevents apoptosis of motor neurons induced by axotomy

  • It enhances survival and morphological differentiation of dopaminergic neurons and increases their high-affinity dopamine uptake

  • In animal models, GDNF delivery to the striatum protects, regenerates, and improves the metabolism of substantia nigra pars compacta neurons, a key neuronal population that degenerates during PD pathogenesis

  • GDNF can reverse motor deficits and nigrostriatal pathology, but the timing of administration is critical. It is effective if administered during ongoing neurodegeneration, but ineffective if administered after the nigrostriatal system has been fully lesioned

The efficacy of GDNF therapy appears to be dependent on the stage of neurodegeneration, with early intervention showing the most promise .

What are the optimal delivery methods for GDNF in experimental models?

Several delivery methods have been developed for GDNF administration in experimental models:

  • Viral Vector-Mediated Delivery:

    • Lentiviral vectors expressing regulated GDNF delivered to the striatum allow controlled GDNF expression

    • This approach enables temporal control, allowing researchers to turn GDNF expression on or off at specific timepoints during disease progression

  • Cell-Based Delivery Systems:

    • Human neural progenitor cells (hNPC) isolated from the cortex and expanded in culture can be modified using lentivirus to secrete GDNF (hNPC-GDNF)

    • This approach overcomes challenges of direct administration of large proteins to the brain

    • Cell-based delivery provides sustained, localized release of GDNF

  • Regulated Expression Systems:

    • Inducible expression systems allow for controlled temporal expression of GDNF

    • This is particularly important for studying the effects of GDNF at different stages of neurodegenerative diseases

The selection of delivery method depends on research goals, with viral vector approaches offering good targeting and regulation, while cell-based approaches provide sustained delivery in a more physiological context.

How can GDNF-mediated neuroprotection be objectively measured in experimental models?

Assessment MethodParameters MeasuredAdvantagesLimitations
Behavioral TestingMotor function (rotational behavior, limb use)Functional relevance, non-invasiveIndirect measure, affected by compensatory mechanisms
Neurochemical AnalysisDopamine and metabolite levelsDirect measure of neurotransmitter functionTerminal procedure, snapshot measurement
Histological AssessmentTH+ neuron counts, fiber densityDirect visualization of neuroanatomyTerminal procedure, labor-intensive
PET/SPECT ImagingIn vivo tracer uptake (e.g., F-DOPA)Longitudinal assessment possibleExpensive, limited resolution
ElectrophysiologyNeuronal firing patternsFunctional assessment at cellular levelTechnically demanding, limited sampling

Effective evaluation requires a combination of these approaches to comprehensively assess GDNF effects on behavior, neurochemistry, and neuroanatomy .

What factors influence the efficacy of GDNF therapy in neurodegenerative diseases?

The efficacy of GDNF therapy is influenced by several critical factors:

  • Stage of Neurodegeneration:

    • GDNF is effective if administered while the nigrostriatal system is still degenerating

    • It is ineffective if administered after the nigrostriatal system has been fully lesioned

    • A sufficient number of remaining neurons must be present to respond to the therapy

  • Delivery Method and Location:

    • Direct delivery to target regions (e.g., striatum) is crucial

    • The distribution volume within target tissue affects efficacy

    • Continuous vs. pulsatile delivery may yield different results

  • Dose and Duration:

    • Optimal dosing is critical - both insufficient and excessive GDNF can limit efficacy

    • The duration of treatment must be sufficient to allow for neuronal recovery

  • Presence of Co-factors:

    • Expression of GDNF receptors (RET, GFRα1) is necessary for response

    • The inflammatory environment may alter receptor expression and signaling

These factors help explain the discrepancy between promising preclinical results and the challenges encountered in clinical trials with GDNF .

How do findings from animal models of GDNF therapy translate to human clinical trials?

The translation of GDNF therapy from animal models to human trials has faced several challenges:

  • Efficacy Discrepancies:

    • Despite excellent results in experimental settings, clinical trials testing GDNF have often failed to meet primary endpoints

    • One theory explaining these negative outcomes is that clinical trials were conducted in late-stage patients with advanced nigrostriatal degeneration who may not respond to neurotrophic factor therapy

  • Delivery Challenges:

    • The blood-brain barrier limits systemic delivery

    • Direct intraparenchymal delivery is invasive

    • Achieving adequate distribution throughout affected brain regions is difficult

  • Timing Considerations:

    • Animal research suggests that GDNF efficacy depends on having sufficient remaining neurons to respond

    • Most clinical trials have enrolled patients with advanced disease

  • Species Differences:

    • Differences in GDNF signaling, distribution of receptors, and neuroanatomy between rodents and humans

    • The rate of neurodegeneration differs substantially between acute toxin models and the chronic progressive nature of human neurodegenerative diseases

Future clinical trials may benefit from targeting earlier disease stages and employing advanced delivery techniques based on these translational insights .

What novel approaches are being developed to enhance GDNF efficacy in clinical applications?

Several innovative approaches are being developed to overcome challenges and enhance GDNF efficacy:

  • Conditional Expression Systems:

    • Development of regulatable gene therapy systems that allow controlled expression of GDNF

    • These systems permit adjustment of GDNF levels based on clinical response and potential side effects

  • Cell-Based Delivery:

    • Human neural progenitor cells modified to secrete GDNF (hNPC-GDNF) offer a biological delivery system

    • These cells can potentially integrate into host tissue and provide sustained, regulated GDNF delivery

  • Combination Therapies:

    • Combining GDNF with factors targeting complementary pathways

    • For example, studies have examined delivery of sonic hedgehog or GDNF to dopamine-rich grafts

  • Receptor-Targeted Approaches:

    • Development of small molecules that enhance GDNF signaling through RET or alternative pathways

    • Manipulation of adenosine A2A receptor (A2AR) to modulate GDNF effects, as shown in schizophrenia research

  • Patient Stratification:

    • Identification of patient subgroups most likely to respond to GDNF therapy

    • Development of biomarkers to guide patient selection and treatment timing

These approaches aim to address the limitations encountered in previous clinical trials while leveraging mechanistic insights from basic research.

What are emerging areas of GDNF research beyond neurodegenerative disorders?

GDNF research is expanding beyond traditional neurodegenerative applications:

  • Psychiatric Disorders:

    • Recent evidence suggests GDNF dysregulation may contribute to dopaminergic abnormalities in schizophrenia

    • GDNF levels are increased in cerebrospinal fluid of first-episode psychosis patients and post-mortem striatum of schizophrenia patients

    • The GDNF-A2AR interaction represents a potential therapeutic target for schizophrenia

  • Neuroinflammatory Modulation:

    • GDNF may play a role in modulating neuroinflammation

    • Upregulation of RET and GFRα1 in glial cells during neuroinflammation suggests GDNF could influence inflammatory processes

    • This presents opportunities for targeting inflammatory aspects of neurological disorders

  • Tissue Engineering Applications:

    • GDNF is being utilized in 2.5D actuating substrates to study mechanical and biochemical effects of muscle exercise on motor neurons

    • This has applications in developing advanced in vitro models of neuromuscular systems

  • Biomarker Development:

    • GDNF levels in cerebrospinal fluid could serve as biomarkers for certain neurological and psychiatric conditions

    • Changes in GDNF signaling components might indicate disease progression or treatment response

These emerging areas highlight GDNF's broader potential beyond traditional applications in Parkinson's disease.

What methodological advances are needed to overcome current limitations in GDNF research?

Several methodological advances would significantly advance GDNF research:

  • Improved Delivery Technologies:

    • Development of non-invasive delivery methods that cross the blood-brain barrier

    • Creation of smart delivery systems that respond to local tissue conditions

    • Advancement of targeted delivery to specific neural circuits

  • Better Disease Models:

    • Development of progressive neurodegenerative models that better recapitulate human disease

    • Creation of patient-derived models using iPSC technology to account for genetic background

    • Models that incorporate both neurodegeneration and neuroinflammation

  • Advanced Imaging Techniques:

    • Development of PET ligands for GDNF receptors to enable in vivo monitoring of receptor availability

    • Improved imaging methods to track GDNF distribution and effects non-invasively

    • Functional imaging that correlates GDNF action with neural circuit activity

  • Systems Biology Approaches:

    • Integration of multi-omics data to understand GDNF effects at system level

    • Computational modeling of GDNF signaling networks

    • Machine learning approaches to predict responders vs. non-responders

  • Standardized Assessment Protocols:

    • Development of standardized protocols to assess GDNF efficacy across laboratories

    • Establishment of relevant biomarkers that translate between animal models and human patients

These methodological advances would address key obstacles in current GDNF research and facilitate clinical translation.

Product Science Overview

Structure and Function

GDNF is a disulfide-bonded homodimeric protein, meaning it consists of two identical subunits linked by disulfide bonds . Each subunit is approximately 15 kDa in size and contains seven conserved cysteine residues, which are essential for the formation of the protein’s stable dimeric structure . The protein signals through a multicomponent receptor system composed of the receptor tyrosine kinase RET and one of the four GFRα (α1-α4) receptors .

Biological Activity

GDNF has been shown to promote the survival and morphological differentiation of midbrain neurons, enhance dopamine uptake, and prevent apoptosis of motor neurons . It is also involved in the development and maintenance of the central and peripheral nervous systems, renal morphogenesis, and spermatogenesis . In addition, GDNF supports the survival and differentiation of various peripheral neurons, including sympathetic, parasympathetic, sensory, and enteric neurons .

Recombinant Human GDNF

Recombinant human GDNF is produced using Escherichia coli (E. coli) expression systems, ensuring that the protein is free from animal-derived components . This is particularly important for applications where the transmission of animal viruses and contamination must be avoided. Recombinant GDNF is optimized for use in cell culture, differentiation studies, and functional assays .

Applications

Human recombinant GDNF is widely used in research to study neuronal growth, differentiation, and survival. It is also employed in various assays to analyze apoptosis and signaling pathways . In clinical research, GDNF has shown promise in improving conditions such as Parkinson’s disease by alleviating symptoms like bradykinesia, rigidity, and postural instability .

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.