GFAP Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Structure and Production of Recombinant GFAP Monoclonal Antibodies

Recombinant GFAP antibodies are generated by cloning immunoglobulin DNA sequences from immunized animals (e.g., rabbits or mice) into expression vectors. This process bypasses traditional hybridoma methods, enabling precise control over antibody design and production. Key features include:

  • Host Species: Produced in rabbits (e.g., RM246 , ASTRO/1974R ) or mice (e.g., rASTRO/789 , MAB25941 ).

  • Epitope Specificity: Target conserved regions of GFAP, avoiding cross-reactivity with other intermediate filaments (e.g., vimentin, cytokeratins) .

  • Advantages Over Traditional Antibodies:

    FeatureTraditional AntibodiesRecombinant Antibodies
    SpecificityVariableHigh (engineered epitope binding)
    Lot ConsistencyVariableConsistent
    Animal-Free ProductionNoYes (in vitro systems)
    ImmunoreactivityLimited species reactivityBroader reactivity (e.g., human, mouse, rat)

Applications in Research and Diagnostics

Recombinant GFAP antibodies are validated for diverse techniques, including:

Western Blotting (WB)

  • Detection Range: 35–50 kDa bands corresponding to GFAP isoforms .

  • Protocol Example:

    • Sample Preparation: Human brain lysates (cerebellum, motor cortex) .

    • Primary Antibody: Mouse anti-GFAP (0.25 µg/mL) .

    • Secondary Antibody: HRP-conjugated anti-mouse IgG .

Immunohistochemistry (IHC)

Antibody CloneHost SpeciesTested TissuesDilutionDetection Method
RM246 RabbitHuman brain cortex0.25 µg/mLAlexa Fluor™ 555
MAB25941 MouseHuman caudate nucleus0.2 µg/mLDAB (brown) + hematoxylin
ASTRO/1974R RabbitParaffin-embedded CNS tumors1:500–1:1000IHC with counterstaining

Immunofluorescence (IF) and Flow Cytometry

  • Cell Line Studies:

    • U-251MG Glioblastoma Cells: GFAP detected in cytoplasm using mouse anti-GFAP (1.7 µg/mL) with NL007 secondary antibody .

    • Knockout Validation: GFAP absent in U937 KO cells, confirming antibody specificity .

Diagnostic Utility

  • Tumor Differentiation: Distinguishes primary gliomas (GFAP+) from metastatic lesions (GFAP−) .

  • Astrocytic Markers: Identifies astrocytic differentiation in non-CNS tumors (e.g., salivary gland neoplasms) .

Specificity and Cross-Reactivity

Recombinant antibodies exhibit stringent specificity for GFAP:

  • Epitope Mapping:

    • SMI-23 Antibody: Binds residues 312–340 of GFAP .

    • Clone 2A5 (Mouse): Specific for GFAP in bovine, human, mouse, pig, and rat .

  • Cross-Reactivity:

    • Negative Reactivity: No binding to vimentin, cytokeratin, or neurofilament proteins .

    • Species Reactivity:

      Antibody CloneHost SpeciesReactivity
      RM246 RabbitHuman, Mouse
      MAB25941 MouseHuman, Rat
      GA5 MouseHuman, Mouse, Rat

Neurodegenerative Diseases

  • Alexander Disease: Linked to GFAP gene mutations; antibodies aid in studying astrocyte dysfunction .

  • Neuroinflammation: GFAP upregulation in astrocytes correlates with conditions like Alzheimer’s disease .

Tumor Biology

  • Glioblastoma: GFAP expression in U-251MG cells confirms astrocytic origin .

  • Metastasis Detection: GFAP negativity in metastatic CNS tumors helps rule out primary gliomas .

Epigenetic and Epitope Studies

  • Knockout Models: GFAP KO U937 cells validate antibody specificity in flow cytometry .

  • Epitope Overlap: SMI-23, -24, -25, and 6F2 antibodies target overlapping regions (residues 312–340) .

Advantages in Modern Research

Recombinant GFAP antibodies address limitations of traditional methods:

  • Lot-to-Lot Consistency: Critical for longitudinal studies or clinical trials .

  • Broad Immunoreactivity: Detects GFAP in human, mouse, rat, bovine, and pig tissues .

  • Animal-Free Production: Aligns with ethical and regulatory trends .

Product Specs

Buffer
Rabbit IgG in phosphate-buffered saline (PBS), pH 7.4, containing 150 mM NaCl, 0.02% sodium azide, and 50% glycerol.
Description

The GFAP recombinant monoclonal antibody is produced using a robust process involving in vitro cloning. Genes encoding both the heavy and light chains of the GFAP antibody are inserted into expression vectors and subsequently transfected into host cells for recombinant antibody expression in cell culture. Following expression, the GFAP recombinant monoclonal antibody is purified from the cell culture supernatant via affinity chromatography. This purified antibody exhibits specific reactivity with the human GFAP protein and is suitable for use in ELISA and flow cytometry (FC) applications.

GFAP, a class III intermediate filament protein, is primarily associated with astrocytes and plays a critical role in maintaining astrocyte structure, morphology, and function within the central nervous system (CNS). Its functions include supporting the blood-brain barrier, regulating ion and water homeostasis, facilitating neurotransmitter uptake, and contributing to neuroprotection. Furthermore, GFAP expression is upregulated during reactive gliosis in response to CNS injury or disease, participating in glial scar formation and the overall CNS response to damage.

Form
Liquid
Lead Time
Orders are typically dispatched within 1-3 business days of receipt. Delivery times may vary depending on the purchasing method and location. Please consult your local distributor for precise delivery timelines.
Synonyms
Glial fibrillary acidic protein (GFAP), GFAP
Target Names
Uniprot No.

Target Background

Function

GFAP, a class III intermediate filament, serves as a cell-specific marker distinguishing astrocytes from other glial cells during CNS development.

Gene References Into Functions

Studies highlighting the role of GFAP in various neurological conditions:

  1. Perivascular inflammation and loss of astrocytes and neurons in GFAP astrocytopathies. PMID: 29193473
  2. Amniotic fluid GFAP levels differentiate between myelomeningocele and myeloschisis. PMID: 28768252
  3. Review of type III intermediate filaments (desmin, GFAP, vimentin, and peripherin) and their roles in health and disease. PMID: 29196434
  4. Plasma GFAP concentrations and their association with stroke occurrence (but not severity or mortality) in a West African cohort. PMID: 29074065
  5. Elevated levels of GFAP and AQP4-expressing microparticles in traumatic brain injury. PMID: 28972406
  6. Higher serum levels of GFAP and UCH-L1 in brain-injured children, correlating with injury severity and predicting poor outcomes. PMID: 27319802
  7. QKI6B expression as a predictor of GFAP expression in the prefrontal cortex of schizophrenic individuals. PMID: 28552414
  8. Elevated plasma GFAP, tau, and AmyloidBeta42 levels up to 90 days post-traumatic brain injury. PMID: 27312416
  9. Decreased expression of nestin, tyrosine hydroxylase (TH), GFAP, and IL-17, and increased Foxp3 and Foxp3/IL-17 ratio in bone marrow of acute myeloid leukemia (AML) patients. PMID: 27016413
  10. GFAP levels >0.29 ng/ml specifically identified intracerebral hemorrhage (ICH) during prehospital care. PMID: 27951536
  11. Autoantibodies against GFAP as a potential predictive marker for autoimmune diabetes. PMID: 28546444
  12. Higher median plasma GFAP values in intracerebral hemorrhage compared to acute ischemic stroke, stroke mimics, and controls. PMID: 28751552
  13. GFAP's pivotal role in the mechanical stress resistance of auricular chondrocytes. PMID: 28063220
  14. Bevacizumab treatment's association with altered GFAP and vimentin content and mRNA expression. PMID: 28419863
  15. Slower exchange of GFP-GFAPdelta compared to GFP-GFAPalpha within the intermediate filament network. PMID: 27141937
  16. GFAP aggregation, unfolded protein response (UPR), and endoplasmic reticulum (ER) stress induction in astrocytes due to Tat or GFAP expression. PMID: 27609520
  17. Distinct temporal profiles of GFAP expression over 7 days in traumatic brain injury patients. PMID: 27018834
  18. Potential association between serum GFAP levels and autism spectrum disorder severity in Chinese children. PMID: 28088366
  19. Association between high GFAP expression and retinoblastoma. PMID: 27488116
  20. Lack of significant association between GFAP levels and peripartum brain injury in neonates with congenital heart defects, with a trend towards elevation on postnatal day 4 in neonates with left heart obstruction. PMID: 26786018
  21. Significantly lower serum GFAP levels in autism spectrum disorders compared to controls. PMID: 27097671
  22. Downregulation of GFAP mRNA and protein in the mediodorsal thalamus and caudate nucleus of depressed suicide victims. PMID: 26033239
  23. No difference in cord blood GFAP concentration between hypoxic-ischemic encephalopathy neonates and controls. PMID: 26135781
  24. GFAP upregulation as an indicator of CNS pathology following brain insult or injury. PMID: 25846779
  25. Decreased density of GFAP-immunoreactive astrocytes in the left hippocampus in major depressive disorder. PMID: 26742791
  26. GFAP as a promising biomarker to distinguish ischemic stroke from intracerebral hemorrhage. PMID: 26526443
  27. Significantly higher GFAP levels in Alzheimer's disease, dementia with Lewy bodies, and frontotemporal lobar degeneration patients compared to healthy controls. PMID: 26485083
  28. GFAP's association with outcome in traumatic brain injury (TBI), but not adding predictive power beyond commonly used prognostic variables. PMID: 26547005
  29. Absence of GFAP coding-region rearrangements in Alexander disease or Alexander-related leukoencephalopathies. PMID: 26208460
  30. Human vitreous body GFAP as a biomarker for glial activation in retinal pathologies. PMID: 26279003
  31. Diagnostic value of serum GFAP, pNF-H, and NSE levels in assessing the severity of human traumatic spinal cord injury. PMID: 25341992
  32. Temporal differences in GFAP levels in hemorrhagic versus ischemic brain lesions. PMID: 26081945
  33. Absence of GFAP in astrocytes during early fetal spinal cord development until 9 months of gestation. PMID: 25904356
  34. Potential clinical relevance of GFAP expression in cerebral astrocytoma invasiveness. PMID: 25178519
  35. Essential role of tight histone acetylation regulation in astrocytes to prevent GFAP aggregation, as seen in Alexander's disease. PMID: 25128567
  36. Identification of a novel nonsense mutation in the rod domain of GFAP associated with Alexander disease. PMID: 24755947
  37. Role of S100B protein, neuron-specific enolase, and GFAP in evaluating hypoxic brain injury in acute carbon monoxide poisoning. PMID: 24505052
  38. Review of GFAP's involvement in physiological and pathophysiological astrocyte functions, particularly in astrocyte activation and reactive gliosis. PMID: 25726916
  39. GFAP expression changes in Alexander disease reflecting primary astrocyte pathology. PMID: 25859599
  40. Potential use of preoperative plasma IGFBP-2, GFAP, and YKL-40 levels as an additional diagnostic tool for inoperable brain lesions suggestive of glioblastoma multiforme. PMID: 25139333
  41. Significant increases in GFAP levels in children undergoing cardiopulmonary bypass for congenital heart disease repair, with highest values during rewarming. PMID: 23845562
  42. Decreased fibrillary astrocytes in the subgenual cingulate in schizophrenia. PMID: 24374936
  43. 3.77-fold average increase in anti-GFAP autoantibody levels in TBI patients from early (0-1 days) to late (7-10 days) post-injury. PMID: 24667434
  44. Overexpression and hypophosphorylation of GFAP in enteric glial cells of Parkinson's disease patients. PMID: 24749759
  45. Association between GFAP expression and plaque load-related astrogliosis in Alzheimer's disease. PMID: 24269023
  46. Caspase-mediated GFAP proteolysis as a common event in GFAP mutation and excess. PMID: 24102621
  47. Increased GFAP expression in the prefrontal cortex in psychotic illness. PMID: 23911257
  48. Gfapdelta expression in developing mouse brain subventricular zones. PMID: 23991052
  49. Reliable distinction of traumatic brain injury severity using GFAP breakdown product blood levels. PMID: 23489259
  50. Association between the C/C genotype at rs2070935 of the GFAP promoter and earlier onset and faster progression of ambulatory disability in late-onset Alexander disease. PMID: 23903069
Database Links

HGNC: 4235

OMIM: 137780

KEGG: hsa:2670

STRING: 9606.ENSP00000253408

UniGene: Hs.514227

Involvement In Disease
Alexander disease (ALXDRD)
Protein Families
Intermediate filament family
Subcellular Location
Cytoplasm.
Tissue Specificity
Expressed in cells lacking fibronectin.

Q&A

What is GFAP and why is it a significant target for antibody development?

GFAP (Glial fibrillary acidic protein) is a member of the class III intermediate filament protein family that serves as a defining cytoskeletal component in astrocytes. It is heavily and specifically expressed in astrocytes and certain astroglia of the central nervous system, satellite cells of peripheral ganglia, and non-myelinating Schwann cells of peripheral nerves . GFAP has become an invaluable target for antibody development because it functions as a highly specific marker for astrocytes, allowing researchers to distinguish these cells from other glial populations during development and in mature tissue . Additionally, neural stem cells strongly express GFAP, making it useful for studying neurogenesis and neural progenitor dynamics .

The significance of GFAP extends to pathological conditions as well. Many types of brain tumors, presumably derived from astrocytic cells, heavily express GFAP, making these antibodies essential diagnostic tools in neuropathology . Furthermore, mutations in the GFAP gene cause Alexander disease, a rare disorder of astrocytes in the central nervous system, highlighting its importance in understanding neurological disease mechanisms .

How do recombinant monoclonal antibodies differ from traditional monoclonal antibodies for GFAP detection?

Recombinant monoclonal antibodies represent a significant technological advancement over traditional monoclonal antibodies in several important ways:

  • Production methodology: Recombinant GFAP antibodies are produced using in vitro expression systems by cloning specific antibody DNA sequences from immunoreactive rabbits, followed by screening individual clones to select optimal candidates for production . This contrasts with traditional monoclonal antibodies, which typically involve immunizing animals and harvesting antibodies from hybridoma cells.

  • Reproducibility and consistency: Traditional antibodies face challenges with reproducibility between batches. Recombinant antibodies offer superior lot-to-lot consistency because they're produced from defined genetic sequences rather than biological systems with inherent variability .

  • Ethical considerations: Recombinant antibody production substantially reduces animal use and associated ethical concerns that accompany traditional monoclonal antibody production .

  • Specificity and sensitivity: Recombinant rabbit monoclonal antibodies typically demonstrate better specificity and sensitivity than traditional antibodies, improving experimental reliability .

  • Formulation advantages: Recombinant antibodies can be produced in animal origin-free formulations, which reduces potential contaminants and immunogenic components that might interfere with experimental systems .

What applications are most suitable for GFAP recombinant monoclonal antibodies?

GFAP recombinant monoclonal antibodies excel in multiple research applications:

ApplicationSuitabilityKey Advantages
ImmunohistochemistryExcellentSpecific localization of GFAP in formalin-fixed paraffin-embedded tissue sections
ImmunofluorescenceExcellentHigh signal-to-noise ratio for imaging applications
Western BlottingVery GoodDetection of GFAP protein in tissue lysates, as demonstrated with human brain tissues
MicroarrayGoodSuitable for high-throughput screening applications
Neural cell identificationExcellentSpecifically stains glial cells (Bergmann glia) and astrocytes

These antibodies have been extensively tested for immunohistochemistry in human, pig, and rat tissues, making them versatile tools for comparative studies across species . For western blot applications, GFAP antibodies successfully detect the protein in various human brain regions including motor cortex, cerebellum, and hypothalamus .

What are the critical considerations for validating GFAP recombinant monoclonal antibody specificity?

Validating antibody specificity is essential for generating reliable research data. For GFAP recombinant monoclonal antibodies, researchers should implement a multi-faceted validation approach:

  • Positive and negative tissue controls: Compare staining patterns in tissues known to express GFAP at high levels (astrocyte-rich regions of CNS) versus tissues that lack GFAP expression. The antibody should demonstrate reactivity to human and mouse GFAP while showing appropriate tissue specificity .

  • Western blot analysis: Verify that the antibody detects a band of appropriate molecular weight (~50 kDa for GFAP) in brain tissue lysates. Cross-reference results across different brain regions that variably express GFAP, such as motor cortex, cerebellum, and hypothalamus .

  • Knockout/knockdown validation: When possible, compare staining in GFAP knockout/knockdown models versus wild-type controls to confirm specificity.

  • Peptide competition assays: Pre-incubate the antibody with purified GFAP protein to demonstrate that this blocks subsequent tissue staining.

  • Cross-reactivity assessment: Test reactivity against related intermediate filament proteins to ensure specificity for GFAP over similar structural proteins.

  • Epitope mapping: Understanding the precise epitope recognized by the antibody helps predict potential cross-reactivity issues and interpret experimental outcomes. Some GFAP antibodies have been characterized using well-defined GFAP fragments to pinpoint their binding regions .

How can researchers optimize protocols for GFAP immunohistochemistry in different CNS tissue preparations?

Optimizing GFAP immunohistochemistry requires consideration of several technical variables:

  • Fixation parameters:

    • For formalin-fixed paraffin-embedded (FFPE) sections: Optimal fixation time in 10% neutral buffered formalin is typically 24-48 hours. Extended fixation can mask epitopes.

    • For frozen sections: Brief fixation (10-20 minutes) with 4% paraformaldehyde is usually sufficient.

  • Antigen retrieval methods:

    • Heat-induced epitope retrieval (HIER): Use citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) for 20 minutes.

    • Enzymatic retrieval: Proteinase K treatment (10-20 μg/mL for 10-15 minutes) can be effective for some tissue preparations.

  • Blocking conditions:

    • Use 5-10% normal serum from the same species as the secondary antibody.

    • Include 0.1-0.3% Triton X-100 for improved penetration in thicker sections.

    • For sections with high background, add 1% BSA to reduce non-specific binding .

  • Antibody dilution:

    • Titrate antibody concentrations; typical working dilutions for GFAP monoclonal antibodies are 1:300 .

    • Extend primary antibody incubation to overnight at 4°C for improved signal-to-noise ratio.

  • Detection systems:

    • For fluorescence: Use secondary antibodies with minimal cross-reactivity and appropriate spectral properties.

    • For chromogenic detection: HRP-conjugated secondary antibodies with DAB substrate produce robust signals.

  • Species considerations:

    • Human and mouse tissues typically work well with recombinant rabbit monoclonal GFAP antibodies .

    • For other species, preliminary validation is recommended.

How can GFAP recombinant monoclonal antibodies be utilized in studying neurodegenerative diseases?

GFAP recombinant monoclonal antibodies serve as powerful tools for investigating neurodegenerative diseases through several methodological approaches:

  • Astrogliosis quantification: Neurodegenerative diseases typically feature reactive astrogliosis, characterized by increased GFAP expression. Quantitative analysis of GFAP immunoreactivity can assess disease progression and therapeutic responses.

  • Regional vulnerability mapping: Different brain regions show variable vulnerability in neurodegenerative conditions. GFAP antibodies help map astrocytic responses across brain regions to understand disease-specific patterns.

  • Morphological analysis: Beyond measuring expression levels, detailed morphological analysis of GFAP-positive astrocytes (process length, complexity, and polarization) provides insights into functional changes in astrocytes during disease progression.

  • Co-localization studies: Combining GFAP staining with markers for disease-specific proteins (e.g., amyloid-β, tau, α-synuclein) helps analyze astrocyte-pathology relationships in conditions like Alzheimer's and Parkinson's diseases.

  • Alexander disease research: As mutations in the GFAP gene cause Alexander disease, these antibodies are essential for studying this rare neurodegenerative disorder . They allow assessment of mutant GFAP aggregation and subsequent cellular responses.

  • Blood-brain barrier studies: Astrocytic end-feet expressing GFAP contribute to blood-brain barrier function, which is often compromised in neurodegenerative diseases. GFAP antibodies help visualize these structures and their alterations.

What strategies can address common technical challenges when using GFAP recombinant monoclonal antibodies?

Researchers frequently encounter technical challenges when working with GFAP antibodies. Below are methodological solutions to common issues:

  • High background staining:

    • Increase blocking time to 2 hours with 5% normal serum plus 1% BSA.

    • Reduce primary antibody concentration; try 1:500 dilution rather than 1:300.

    • Include 0.1% Tween-20 in wash buffers to reduce non-specific binding.

    • For sections with high endogenous peroxidase activity, incorporate an additional quenching step (3% H₂O₂ for 10 minutes).

  • Weak or absent signal:

    • Ensure appropriate antigen retrieval; for difficult samples, extend HIER time to 30 minutes.

    • For FFPE tissues with extensive fixation, try combining HIER with enzymatic retrieval.

    • Increase antibody concentration and extend incubation time to overnight at 4°C.

    • Switch to a more sensitive detection system (e.g., polymer-based detection or tyramide signal amplification).

  • Non-specific binding:

    • Pre-adsorb the primary antibody with tissue powder from a species different from your experimental tissue.

    • Use secondary antibodies specifically adsorbed against potentially cross-reactive species.

    • Include 0.1-0.3% Triton X-100 in blocking buffer to reduce membrane-associated non-specific binding.

  • Inconsistent results between experiments:

    • Standardize tissue processing protocols, particularly fixation time.

    • Prepare larger volumes of antibody dilutions to use across multiple experiments.

    • Consider using automated staining platforms for improved reproducibility.

    • Take advantage of the lot-to-lot consistency that is a hallmark advantage of recombinant antibodies .

How can researchers design appropriate positive and negative controls for GFAP antibody experiments?

Designing robust controls is essential for validating GFAP antibody experiments:

Positive Controls:

  • Tissue selection: Include sections from regions with known high GFAP expression:

    • Cerebellum (Bergmann glia are strongly GFAP-positive)

    • Astrocyte-rich white matter tracts

    • Reactive astrocytes in injured brain tissue

  • Cell line controls: Use astrocytoma cell lines or primary astrocyte cultures with verified GFAP expression.

  • Recombinant protein controls: For western blot and ELISA applications, include purified recombinant GFAP protein. Recombinant human GFAP from E. coli can serve as an excellent positive control .

Negative Controls:

  • Antibody omission: Process sections without primary antibody to assess secondary antibody specificity.

  • Isotype controls: Use an irrelevant antibody of the same isotype (IgG1 for many monoclonal GFAP antibodies) and host species at the same concentration.

  • Non-expressing tissues: Include neural tissue types with minimal GFAP expression (e.g., mature oligodendrocytes) or non-neural tissues as negative controls.

  • Absorption controls: Pre-absorb the antibody with excess purified GFAP antigen to demonstrate specificity.

  • Genetic controls: When available, GFAP knockout tissues provide the most definitive negative control.

What considerations should researchers take when using GFAP antibodies in multiplexed immunofluorescence studies?

Multiplexed immunofluorescence experiments require careful planning when incorporating GFAP antibodies:

  • Antibody compatibility:

    • Select primary antibodies raised in different host species to avoid cross-reactivity.

    • If using multiple rabbit-derived antibodies (including recombinant rabbit monoclonals), consider sequential staining with thorough blocking between rounds or use directly conjugated antibodies.

  • Spectral considerations:

    • Choose fluorophores with minimal spectral overlap.

    • When analyzing cells with high GFAP expression, assign GFAP to a channel with lower quantum yield to prevent bleed-through.

    • Consider using spectral unmixing algorithms for closely overlapping fluorophores.

  • Signal balancing:

    • Titrate each antibody separately before multiplexing to determine optimal concentrations.

    • GFAP typically produces strong signals; other markers may require amplification to achieve comparable intensity.

  • Staining sequence optimization:

    • For multi-round staining, apply the GFAP antibody in earlier rounds to take advantage of its robust epitope recognition.

    • If using tyramide signal amplification, apply it to weaker signals rather than GFAP detection.

  • Validation strategies:

    • Always include single-stained controls for each marker to confirm specificity and assess bleed-through.

    • Consider computational approaches like linear unmixing to resolve spectral overlap issues.

How can GFAP recombinant monoclonal antibodies facilitate studies of astrocyte heterogeneity?

Astrocyte heterogeneity represents a frontier in neuroscience research, and GFAP antibodies provide crucial methodological approaches to explore this diversity:

  • Regional heterogeneity analysis:

    • GFAP expression varies across brain regions, with some astrocyte populations showing higher expression than others.

    • Quantitative immunohistochemistry with GFAP antibodies can map this regional heterogeneity precisely.

    • Combining GFAP with region-specific markers enables classification of astrocyte subtypes.

  • Developmental trajectory studies:

    • Neural stem cells strongly express GFAP, and recombinant antibodies can track the temporal changes in expression during differentiation into mature astrocytes .

    • Time-course experiments can reveal how GFAP expression patterns evolve during development.

  • Single-cell analysis approaches:

    • GFAP antibodies compatible with flow cytometry enable isolation of GFAP-expressing cells for single-cell RNA sequencing.

    • This approach has revealed substantial transcriptional heterogeneity among GFAP-positive cells.

  • Alternative splicing detection:

    • The GFAP gene undergoes alternative splicing, resulting in multiple transcript variants encoding distinct isoforms .

    • Epitope-specific antibodies can distinguish between these isoforms, revealing functional diversity.

  • Reactive astrocyte subpopulation identification:

    • During pathological conditions, astrocytes become reactive and upregulate GFAP.

    • Co-staining with GFAP and markers of specific reactive phenotypes (neurotoxic vs. neuroprotective) helps classify astrocyte responses.

What are the best practices for quantifying GFAP expression in experimental models of neuroinflammation?

Quantifying GFAP expression in neuroinflammation models requires rigorous methodological approaches:

  • Standardized tissue processing:

    • Maintain consistent fixation protocols across experimental groups.

    • Process control and experimental tissues simultaneously to minimize technical variables.

  • Systematic sampling approaches:

    • Use unbiased stereological methods for quantification.

    • Define anatomical regions of interest based on consistent landmarks.

    • Analyze multiple sections per animal (typically 4-6 sections spaced at regular intervals).

  • Quantification metrics:

    • Area fraction: Percentage of tissue area with GFAP immunoreactivity

    • Mean optical density: Average intensity of GFAP staining

    • Cell counts: Number of GFAP-positive cells per unit area

    • Morphological parameters: Process length, branching complexity, soma size

  • Western blot quantification:

    • Include loading controls (β-actin, GAPDH) for normalization.

    • Generate standard curves using recombinant GFAP for absolute quantification.

    • Use human brain tissue lysates from relevant regions as positive controls .

  • Statistical considerations:

    • Account for biological and technical replicates in experimental design.

    • Use appropriate statistical tests for the data distribution (parametric vs. non-parametric).

    • Consider power analysis to determine adequate sample sizes.

How do alternative GFAP splice variants impact antibody selection for specific research questions?

GFAP exists in multiple splice variants that play distinct functional roles, necessitating careful antibody selection:

  • Common GFAP isoforms:

    • GFAPα: The canonical isoform and most abundant in adult CNS

    • GFAPδ (also known as GFAPε): Enriched in neurogenic regions and subpial astrocytes

    • GFAPκ: Found in Alexander disease models

    • Other variants: GFAPβ, GFAPγ, GFAPζ

  • Epitope considerations:

    • The C-terminal region differs between GFAP isoforms; antibodies targeting this region may not detect all variants.

    • Antibodies against the conserved rod domain detect most GFAP isoforms.

    • Recombinant antibodies with well-characterized epitopes provide clarity about which isoforms will be detected .

  • Research question alignment:

    • For studies of adult astrocytes: Antibodies detecting GFAPα are typically sufficient.

    • For neurogenesis research: Select antibodies that recognize GFAPδ, which is enriched in neural stem cells.

    • For Alexander disease: Consider antibodies that detect disease-associated isoforms.

  • Validation approaches:

    • When isoform specificity is critical, validate antibodies using cells transfected with specific GFAP variants.

    • Western blotting can distinguish some isoforms based on molecular weight differences.

    • Consider complementing protein studies with RT-PCR to detect specific transcripts.

What are the key steps in producing high-quality recombinant monoclonal antibodies against GFAP?

The production of recombinant monoclonal antibodies against GFAP involves several sophisticated steps:

  • Sequence identification and optimization:

    • Start with antibody heavy and light chain sequences, either from existing hybridomas or from immunized rabbits showing high affinity .

    • Optimize codon usage for expression in human cells to maximize production efficiency .

    • Design gene fragments containing the entire heavy chain sequence and the entire light chain sequence .

  • Expression vector construction:

    • Clone heavy and light chain DNA into separate expression plasmids using methods like Gibson assembly .

    • Use plasmids designed for high-level protein expression in mammalian cells, typically driven by a CMV promoter .

    • For optimal results, maintain a 1:2 molar ratio of plasmid digest to gene fragment during cloning .

  • Cell culture and transfection:

    • Transfect the plasmids into HEK293 suspension culture cells, which provide a suitable environment for proper antibody folding and post-translational modifications .

    • This approach offers high yield and relatively low cost compared to traditional hybridoma methods .

  • Purification process:

    • Harvest cell culture supernatant containing secreted antibodies.

    • Purify using protein A or protein G affinity chromatography.

    • Perform additional purification steps as needed (size exclusion chromatography, ion exchange).

  • Quality control assessment:

    • Verify antibody purity by SDS-PAGE and size exclusion chromatography.

    • Confirm target binding using ELISA with recombinant GFAP.

    • Validate specificity with western blot analysis using brain tissue lysates .

    • Perform functional testing in relevant applications (immunohistochemistry, flow cytometry).

How does the choice between different expression systems impact recombinant GFAP antibody performance?

The expression system selected for recombinant GFAP antibody production significantly influences antibody characteristics:

  • Mammalian expression systems (HEK293, CHO cells):

    • Advantages: Proper folding and mammalian glycosylation patterns that enhance stability and effector functions.

    • These systems are ideal for producing recombinant rabbit monoclonal antibodies with characteristics closely matching naturally produced antibodies .

    • HEK293 suspension culture cells offer high yield with relatively low cost .

  • Bacterial expression systems (E. coli):

    • Advantages: Lower cost, higher yield, and simpler purification.

    • Limitations: Lack proper glycosylation and may have folding issues with full-length antibodies.

    • Better suited for antibody fragments (Fabs, scFvs) rather than complete antibodies.

    • Can be useful for producing recombinant GFAP antigen for antibody validation .

  • Insect cell systems (Sf9, High Five):

    • Intermediate option with some post-translational modifications.

    • Provides higher yields than mammalian systems but with simplified glycosylation.

  • Yeast expression systems:

    • Advantages: Higher yields than mammalian cells with some eukaryotic processing.

    • Limitations: Non-human glycosylation patterns may affect antibody clearance and effector functions.

  • Cell-free expression systems:

    • Emerging option for rapid production of antibody fragments.

    • Limitations: Generally not suitable for complete glycosylated antibodies.

The choice of expression system should align with the intended application. For research applications requiring high specificity and sensitivity, mammalian expression systems like those used for recombinant rabbit monoclonal antibodies offer superior performance characteristics .

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.