GFAP Human

Glial Fibrillary Acidic Protein Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Domains

GFAP Human (UniProt: P14136) belongs to class III intermediate filament proteins, characterized by three domains:

  • Head domain: Contains phosphorylation sites (T7, S8, S13, S17, S38, S289) regulated by kinases like PKA, PKC, and CaMKII .

  • Rod domain: Central α-helical region with four coiled-coil segments (1A, 1B, 2A, 2B) essential for filament assembly .

  • Tail domain: Facilitates oligomerization and interacts with presenilins in isoforms like GFAP-δ .

Homology modeling based on vimentin predicts a conserved 3D structure (Figure 2B in ). GFAP monomers assemble into filaments via parallel dimers → antiparallel tetramers → octamers .

Isoforms and Expression Patterns

Ten GFAP isoforms arise from alternative splicing, with distinct tissue distributions:

IsoformResiduesKey FeaturesExpression Sites
GFAP-α432Dominant in CNS astrocytes Brain, spinal cord, PNS
GFAP-δ/ε431Binds presenilins; neural stem cellsSubventricular zone

GFAP-δ is a marker for quiescent neural stem cells in adult human subventricular zones but shows broader astrocyte expression in mice .

Post-Translational Modifications (PTMs)

GFAP undergoes dynamic PTMs influencing filament assembly and function:

ModificationSitesEnzymes InvolvedFunctional Impact
PhosphorylationS8, S13, S38PKA, PKC, CaMKII Regulates filament stability
ProteolysisMultipleCalpain Generates 38–48 kDa breakdown products (BDPs) in TBI

Calpain-cleaved GFAP-BDPs in cerebrospinal fluid (CSF) and serum serve as biomarkers for traumatic brain injury (TBI) severity .

Astrocyte Function

  • Maintains blood-brain barrier (BBB) integrity .

  • Supports neuronal communication via glutamate-mediated CaMKII signaling .

Disease Associations

  • Alexander disease: GFAP mutations (e.g., R239C) cause Rosenthal fiber accumulation .

  • Neurodegeneration: Elevated CSF/serum GFAP correlates with Alzheimer’s disease (AD), multiple sclerosis (MS), and TBI .

    • Plasma GFAP levels distinguish Aβ+ cognitively normal individuals from controls (AUC = 0.90) .

    • In AD, serum GFAP increases 2.5-fold compared to controls .

Biomarker Applications

Recent assays enable ultrasensitive GFAP detection:

Assay TypeSensitivity (LLOQ)Sample TypeClinical Utility
Microfluidic 7.21 pg/mLSerum/CSFDiscriminates AD from controls
ELISA 0.02 ng/mLSerumPredicts TBI mortality

GFAP-BDPs (38–50 kDa) in TBI CSF are captured non-preferentially by monoclonal antibodies (e.g., MAB25941) .

Research Tools and Reagents

  • Antibodies: MAB25941 detects GFAP in human brain lysates and CSF but shows low cross-reactivity with rodent GFAP .

  • Recombinant Protein: Used in ELISA standards (e.g., Prot-r-GFAP; 50 kDa) .

Future Directions

Ongoing studies focus on:

  • GFAP-δ’s role in neurogenesis and neural repair .

  • Targeting GFAP phosphorylation pathways to modulate astrogliosis .

Product Specs

Description
GFAP Human produced in E.coli is a single, non-glycosylated polypeptide chain (60-383 a.a.) with a molecular mass of 37906 Dalton.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
GFAP was lyophilized from 16mM NaHCO3, 0.05% CHAPS, and 0.05% Tween 20.
Solubility
It is recommended to reconstitute the lyophilized GFAP in sterile 18MΩ-cm H2O at a concentration not less than 100µg/ml. This solution can then be further diluted to other aqueous solutions.
Stability
Lyophilized GFAP, although stable at room temperature for 3 weeks, should be stored desiccated below -18°C. Upon reconstitution, Glial Fibrillary Acidic Protein should be stored at 4°C for between 2-7 days. For future use, store below -18°C. For long-term storage, adding a carrier protein (0.1% HSA or BSA) is recommended. Please prevent freeze-thaw cycles.
Purity
Greater than 90.0% as determined by SDS-PAGE.
Synonyms

Glial fibrillary acidic protein, GFAP

Source

Escherichia Coli.

Q&A

What is GFAP and where is it expressed in the human nervous system?

GFAP is an intermediate filament-III protein uniquely found in astrocytes in the central nervous system (CNS), non-myelinating Schwann cells in the peripheral nervous system (PNS), and enteric glial cells. It functions as a cytoskeletal protein that provides structural support to astrocytes, facilitates cell communication, and contributes to the formation of the blood-brain barrier . When studying GFAP expression, researchers typically employ immunohistochemistry with anti-GFAP antibodies (commonly at 1:2000 dilution) to visualize GFAP-positive cells in tissue sections. For protein quantification, western blotting using monoclonal or polyclonal antibodies against GFAP is the standard approach, with molecular weight markers identifying intact GFAP at approximately 50 kDa .

What regulates GFAP expression in human astrocytes?

GFAP expression is regulated through multiple mechanisms:

Regulatory FactorEffect on GFAP ExpressionMethodological Approach
Nuclear-receptor hormonesVariable regulationRT-qPCR, hormone treatment studies
Growth factorsGenerally upregulationCell culture with growth factor treatment
LipopolysaccharidesUpregulationIn vitro stimulation assays
Neurological injurySignificant upregulationAnimal models, human biopsy studies

For studying GFAP regulation, researchers should employ a combination of molecular techniques including RT-qPCR for mRNA expression analysis, western blotting for protein quantification, and reporter gene assays to identify promoter activity. Cell culture systems using human astrocytes or iPSC-derived astrocytes provide valuable experimental platforms for manipulating regulatory factors and observing effects on GFAP expression .

How can GFAP breakdown products be detected and quantified in human samples?

GFAP breakdown products (BDPs) can be detected in cerebrospinal fluid (CSF) and blood following traumatic brain injury, spinal cord injury, and stroke. These BDPs result from calpain-mediated truncation of GFAP, producing fragments ranging from 38 to 44 kDa compared to the 50 kDa intact protein .

Methodological approaches include:

  • Western blotting using antibodies specific to GFAP epitopes

  • Enzyme-linked immunosorbent assays (ELISA) developed for GFAP and its BDPs

  • Mass spectrometry for precise fragment identification and quantification

When analyzing GFAP BDPs, researchers should include a range of molecular weight markers (35-55 kDa) and use antibodies that recognize both intact GFAP and its fragments. Sample collection timing is critical, as BDPs appear rapidly after injury and may have different clearance rates in various biofluids .

What are the major post-translational modifications of GFAP?

GFAP undergoes several post-translational modifications that affect its assembly, stability, and function:

  • Phosphorylation: Multiple serine/threonine residues can be phosphorylated, affecting filament assembly

  • Calpain-mediated proteolysis: Results in characteristic breakdown products (38-44 kDa)

  • Citrullination: Modification of arginine residues, often increased in neuroinflammatory conditions

  • Glycosylation: O-GlcNAcylation can occur at specific sites

To study these modifications, researchers typically use phospho-specific antibodies, mass spectrometry techniques, and in vitro modification assays. When examining clinical samples, it's essential to incorporate protease inhibitors during sample preparation to prevent artificial degradation of GFAP .

How does GFAP contribute to astrogliosis in human CNS pathologies?

GFAP gene activation and protein induction play critical roles in astrocyte activation (astrogliosis) following CNS injuries and neurodegeneration. Methodologically, researchers can study this process through:

  • Immunohistochemical analysis of post-mortem brain tissue to visualize GFAP upregulation

  • In vitro astrocyte cultures subjected to inflammatory stimuli

  • Human iPSC-derived astrocyte models of injury response

  • Transgenic models with modified GFAP expression

When designing experiments to study astrogliosis, researchers should consider both acute and chronic timepoints, as GFAP expression patterns change throughout the progression of pathology. Additionally, co-staining with other astrocyte markers (S100β, glutamine synthetase) provides context for GFAP alterations .

What methodological approaches are used to study GFAP mutations in Alexander disease?

Alexander disease is predominantly caused by de novo heterozygous missense mutations in the GFAP gene, resulting in protein aggregates called Rosenthal fibers. Research methodologies include:

  • Genetic testing: Targeted GFAP sequencing for identifying variants in patients with:

    • Developmental delay

    • Abnormal brain MRI

    • Bulbar signs

    • Psychomotor regression

  • Functional validation of variants:

    • GFAP cDNA subcloning into expression vectors (e.g., pKTol2C-GFP)

    • Site-directed mutagenesis to introduce specific variants

    • Transfection into cell lines (SW13vim- cells are preferred as they lack endogenous cytoskeletal proteins)

    • Immunofluorescence analysis with anti-GFAP antibodies (typically 1:2000 dilution)

  • Phenotypic assessment:

    • Filament formation patterns (normal vs. aggregated)

    • Co-localization with vimentin or other intermediate filaments

    • Distribution patterns within cells

When assessing potential pathogenicity of GFAP variants, researchers should compare results with known pathogenic mutations (p.R239H, p.S247P) and benign variants as controls. Additionally, checking variant frequency in population databases (e.g., ExAC) is essential for interpretation .

How can iPSC-derived astrocyte models be used to study GFAP mutations?

Induced pluripotent stem cells (iPSCs) provide a powerful platform for studying GFAP mutations in human astrocytes:

  • Generation of patient-specific iPSCs:

    • Reprogram fibroblasts or blood cells from Alexander disease patients

    • Validate pluripotency markers and differentiation capacity

  • CRISPR/Cas9 gene editing:

    • Correct GFAP mutations in patient iPSCs to create isogenic controls

    • Alternatively, introduce mutations into control iPSCs

  • Astrocyte differentiation:

    • Follow established protocols (typically 4-8 weeks)

    • Verify astrocyte identity through marker expression (GFAP, S100β, ALDH1L1)

  • Phenotypic analysis:

    • Immunostaining for GFAP aggregates

    • Organelle morphology and distribution

    • Calcium signaling using fluorescent indicators

    • ATP release assays

This approach allows direct comparison between mutant and corrected human astrocytes, providing insights into disease mechanisms that may not be apparent in animal models. Human astrocytes are significantly larger, with more processes, and contact 100 times more synapses than rodent astrocytes, making iPSC models particularly valuable .

What are the methodological challenges in analyzing GFAP transcriptomes in disease models?

Transcriptomic analysis of GFAP mutations presents several methodological challenges:

  • Individual variation:

    • Patient-derived cells show divergent transcriptomes even after genetic correction

    • PCA analysis reveals that different GFAP mutations can cause distinct transcriptional responses

  • Data analysis approaches:

    • Spearman's correlation of total transcriptomes

    • Principal component analysis of most variable transcripts

    • Circle of correlation to compare transcriptional profiles

  • Pathway identification:

    • Common pathways affected by GFAP mutations include ER function, vesicle regulation, and cellular metabolism

    • Different mutations may affect overlapping functional pathways through different gene expression changes

When conducting transcriptomic studies, researchers should include isogenic controls whenever possible and apply multiple analytical approaches to identify both mutation-specific and shared transcriptional changes. Additionally, validation of key findings through protein expression analysis and functional assays is essential .

Analysis ApproachPurposeConsiderations
RNA sequencingComprehensive transcriptomeHigh depth (>30M reads) needed
PCAIdentify major sources of variationTop 1000 variable transcripts typically used
Pathway analysisIdentify affected cellular processesMultiple algorithms recommended
qRT-PCRValidate key findingsReference genes must be carefully selected

How should researchers approach the study of anti-GFAP antibodies in autoimmune disorders?

Anti-GFAP antibodies have been identified in a novel meningoencephalomyelitis known as autoimmune GFAP astrocytopathy (GFAP-A). Methodological approaches include:

  • Patient classification:

    • Age-based grouping (pediatric <18 years, adult ≥18 years)

    • Clinical phenotyping (encephalitis, myelitis, meningitis, combinations)

  • Antibody detection:

    • Sample selection: CSF testing is preferred (93.2% positive in CSF vs. 6.8% positive only in serum)

    • Comparative titers between CSF and serum

    • Testing for overlapping autoantibodies

  • Imaging correlations:

    • MRI features (paraventricular linear radial enhancement is characteristic)

    • Timing of imaging (normal findings or delayed abnormalities may occur)

  • Outcome assessment:

    • Monophasic vs. relapsing course tracking

    • Disability scoring

    • Treatment response evaluation

When designing studies on anti-GFAP antibodies, researchers should consider both pediatric and adult populations, as clinical characteristics may differ between age groups. Additionally, follow-up periods should be sufficiently long (median 9 months in recent studies) to capture relapse patterns .

What methodological approaches can differentiate between pathogenic and non-pathogenic GFAP variants?

Distinguishing pathogenic from benign GFAP variants requires a multi-faceted approach:

  • Genetic evidence:

    • De novo status (most pathogenic GFAP variants arise de novo)

    • Absence from population databases (e.g., ExAC)

    • Segregation with disease in familial cases

  • Functional validation:

    • Cellular assays examining filament formation

    • Co-expression with vimentin (negative screen)

    • Imaging of GFAP distribution patterns

  • Structural prediction:

    • In silico modeling of variant effects on protein structure

    • Conservation analysis across species

    • Domain-specific impact assessment

  • Clinical correlation:

    • Age of onset (infantile, juvenile, adult)

    • MRI abnormalities

    • Symptom presentation and progression

Importantly, researchers should note that some GFAP variants (e.g., p.R376W) can present with atypical clinical manifestations or variable onset ages, necessitating functional validation even when genetic evidence is strong. Cell-based assays have demonstrated that some variants initially considered pathogenic actually exhibit wild-type phenotypes, emphasizing the importance of functional studies .

What are the key methodological considerations when using GFAP as a biomarker in neurological disorders?

GFAP and its breakdown products have emerged as promising biomarkers for traumatic brain injury, spinal cord injury, and stroke. Key methodological considerations include:

  • Sample type selection:

    • CSF provides higher sensitivity but requires lumbar puncture

    • Serum/plasma is more accessible but may have lower concentrations

    • Consideration of blood-brain barrier integrity

  • Timing of sample collection:

    • GFAP release occurs rapidly after injury

    • Different clearance kinetics between CSF and blood

    • Temporal profiles vary by injury type and severity

  • Assay selection:

    • ELISA-based methods for clinical applications

    • Western blot for research distinguishing intact vs. fragmented GFAP

    • Mass spectrometry for detailed characterization of BDPs

  • Data interpretation challenges:

    • Establishing appropriate reference ranges

    • Accounting for age, sex, and comorbidity effects

    • Correlation with imaging and clinical outcomes

When designing biomarker studies, researchers should include appropriate control groups and consider serial sampling to capture dynamic changes in GFAP levels. Additionally, sample handling and storage conditions must be standardized to prevent artificial degradation of GFAP .

How can calcium signaling disruptions in GFAP-mutant astrocytes be effectively studied?

GFAP mutations can disrupt calcium wave propagation in astrocytes through reduced ATP release. Methodological approaches include:

  • Calcium imaging techniques:

    • Fluorescent calcium indicators (Fluo-4, Fura-2)

    • Genetically encoded calcium indicators (GCaMP variants)

    • High-speed confocal or two-photon microscopy

  • Stimulation protocols:

    • Mechanical stimulation (micropipette touch)

    • Pharmacological agents (ATP, glutamate)

    • Optogenetic activation

  • Analysis parameters:

    • Wave propagation velocity (5x faster in human vs. rodent astrocytes)

    • Amplitude of calcium response

    • Duration of signaling

    • Intercellular coordination

  • ATP release quantification:

    • Luciferase-based assays

    • Biosensor cells expressing purinergic receptors

    • Correlation with calcium signaling defects

When comparing wild-type and GFAP-mutant astrocytes, researchers should use isogenic controls whenever possible and standardize cell density and culture conditions, as these factors significantly impact calcium signaling properties. Additionally, examining calcium handling in different cellular compartments (cytosol, ER, mitochondria) can provide mechanistic insights into signaling disruptions .

What experimental approaches can evaluate the impact of GFAP on the blood-brain barrier?

GFAP plays a role in astrocyte endfeet that contact blood vessels and contribute to blood-brain barrier (BBB) function. Research methodologies include:

  • In vitro BBB models:

    • Co-culture systems with human astrocytes and brain microvascular endothelial cells

    • Transwell permeability assays with fluorescent tracers

    • Electrical resistance measurements (TEER)

  • GFAP manipulation approaches:

    • siRNA knockdown of GFAP in astrocytes

    • Astrocytes derived from GFAP-mutant iPSCs

    • Comparison of wildtype and corrected isogenic lines

  • Endpoint measurements:

    • Tight junction protein expression and localization

    • Paracellular permeability to various sized molecules

    • Expression of transporters and efflux pumps

    • Inflammatory response under challenge conditions

When studying GFAP's role in BBB function, researchers should consider using human cells whenever possible, as species differences in astrocyte-endothelial interactions are significant. Additionally, three-dimensional models incorporating flow conditions more accurately recapitulate the in vivo BBB environment compared to static cultures .

Product Science Overview

Introduction

Glial Fibrillary Acidic Protein (GFAP) is a type III intermediate filament protein that is predominantly expressed in astrocytes, which are a major type of glial cell in the central nervous system (CNS). GFAP plays a crucial role in maintaining the structural integrity and function of astrocytes. The human recombinant form of GFAP is produced using recombinant DNA technology, which allows for the expression of the protein in a host organism, such as bacteria or yeast.

Structure and Function

GFAP is composed of a central rod domain flanked by non-helical head and tail domains. The rod domain is responsible for the formation of coiled-coil dimers, which further assemble into higher-order structures, forming the intermediate filaments. These filaments provide mechanical support to astrocytes and are involved in various cellular processes, including cell shape maintenance, motility, and response to injury.

Expression and Regulation

GFAP expression begins during embryonic development in radial glia and continues throughout life in astrocytes. The expression of GFAP is highly sensitive to various pathological conditions, such as acute brain injury (e.g., stroke, trauma), chronic neurodegenerative diseases (e.g., Alzheimer’s and Parkinson’s disease), and aging . The human GFAP promoter has been extensively used to drive the expression of transgenes in astrocytes, facilitating the study of astrocyte function in health and disease .

Recombinant Production

The recombinant production of GFAP involves the insertion of the human GFAP gene into an expression vector, which is then introduced into a host organism. Commonly used hosts include Escherichia coli (E. coli) and yeast. The host organism expresses the GFAP protein, which can then be purified using various chromatographic techniques. The recombinant GFAP protein is often tagged with a histidine (His) tag to facilitate purification .

Applications

Recombinant GFAP has several applications in research and clinical settings:

  1. Astrocyte Marker: GFAP is widely used as a marker to identify and study astrocytes in the CNS. It helps in distinguishing astrocytes from other glial cells.
  2. Disease Research: GFAP is used to study the role of astrocytes in various neurological diseases, including Alzheimer’s disease, Parkinson’s disease, and multiple sclerosis.
  3. Injury Response: GFAP is a key marker for astrocyte activation in response to CNS injuries, such as traumatic brain injury and stroke.
  4. Biomarker: GFAP levels in blood and cerebrospinal fluid can serve as biomarkers for brain injury and neurodegenerative diseases .

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.