GMF-Beta stimulates the differentiation of both neurons and glial cells. It exhibits inhibitory effects on the proliferation of N-18 neuroblastoma and C6 glioma cell lines while promoting their phenotypic expression.
GMFB enhances the phenotypic expression of glia and neurons, thereby inhibiting the proliferation of their respective tumor cells in cell culture. While astrocytes produce and store GMF-b intracellularly, they do not secrete it into the culture medium. Cell-surface GMFb acts locally on target cells through direct contact. Produced by thymic epithelial cells, GMF-Beta plays a crucial role in T cell development, particularly favoring CD4+ T cells.
As a brain-specific protein belonging to the actin-binding proteins (ADF) family, GMF-beta is implicated in the differentiation, maintenance, and regeneration of the nervous system. It may also contribute to the progression of certain autoimmune diseases, potentially through its ability to induce the production and secretion of pro-inflammatory cytokines.
The solution contains 1mg/ml of GMFB in a buffer of PBS at pH 7.4, supplemented with 10% Glycerol and 0.02% Sodium Azide.
The GMFB antibody has been validated using ELISA, Western blot, and ICC/IF techniques to confirm its specificity and reactivity. However, optimal working concentrations may vary depending on the specific application and should be determined empirically through titration.
Glia maturation factor beta, GMFB, GMF-B, GMF-beta, GMF.
GMFB antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
PAT44D8AT.
Anti-human GMFB mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human GMFB amino acids 1-142 purified from E. coli.
Mouse IgG2a heavy chain and Kappa light chain.
GMFB (Glia Maturation Factor Beta) is a protein that plays multiple critical roles in cellular function, particularly in the central nervous system. It causes differentiation of brain cells, stimulates neural regeneration, and inhibits the proliferation of tumor cells . GMFB functions as a regulatory factor in various cellular processes including inflammation, cellular growth, and differentiation. Research has demonstrated that GMFB can activate signaling pathways such as p38 MAPK and nuclear factor-κB (NF-κB), thereby inducing the expression and secretion of various cytokines including granulocyte-macrophage colony-stimulating factor (GM-CSF) . This activation suggests that GMFB plays a significant role in neuroinflammatory responses and potentially in neurodegenerative disease progression. Additionally, GMFB has been shown to upregulate neurotrophic factors such as brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF), indicating its importance in neural development and regeneration .
Research-grade GMFB antibodies are available in various formats, with recombinant monoclonal antibodies being particularly valued for their consistency and specificity. For example, rabbit recombinant monoclonal antibodies against GMFB are suitable for multiple applications including Western blotting (WB), immunocytochemistry/immunofluorescence (ICC/IF), flow cytometry (intracellular), and immunoprecipitation (IP) . These antibodies have been validated for reactivity with human, mouse, and rat samples. Polyclonal antibodies raised in rabbits have also been developed for GMFB detection and used in the development of sensitive enzyme immunoassays (EIA) . The choice between monoclonal and polyclonal antibodies depends on the specific experimental requirements, with monoclonal antibodies offering higher specificity for particular epitopes while polyclonal antibodies may provide stronger signals through recognition of multiple epitopes.
GMFB exhibits a distinctive tissue distribution pattern that has been characterized in both humans and rats. In rats, relatively high levels of GMFB are found throughout the central nervous system (with the exception of the spinal cord), as well as in the thymus and colon . Similarly, in humans, GMFB expression is highest in neural tissues, with significant expression also observed in the thymus and colon .
The expression of GMFB varies across different developmental stages. In rats, the serum concentration of GMFB remains relatively stable across different age groups, while the related protein GMFG (Glia Maturation Factor Gamma) shows peak concentration at 4 weeks of age followed by a rapid decrease within the first 30 days of life . In humans, a similar pattern exists, with GMFB levels remaining relatively constant across age groups while GMFG levels peak in the 21-30 year age group before rapidly declining . Interestingly, no significant sex differences in serum GMFB levels have been observed in either humans or rats .
For flow cytometry applications involving GMFB antibodies, researchers should follow a systematic protocol that ensures optimal detection of intracellular GMFB. Based on validated methodologies, cells should undergo fixation with 4% paraformaldehyde followed by permeabilization with 90% methanol to allow antibody access to intracellular GMFB . A recommended antibody dilution of 1/50 (equivalent to 1µg) has been established for optimal staining .
The following protocol has been validated for flow cytometric analysis of GMFB:
Fix cells with 4% paraformaldehyde for 10-15 minutes at room temperature
Permeabilize with 90% methanol for 15-30 minutes on ice or at -20°C
Wash cells in flow cytometry buffer (PBS containing 1-2% BSA)
Block with 5-10% normal serum from the same species as the secondary antibody
Incubate with primary GMFB antibody at 1/50 dilution
Wash to remove unbound primary antibody
Incubate with fluorophore-conjugated secondary antibody (e.g., Goat Anti-Rabbit IgG H&L Alexa Fluor® 488 at 1/2000 dilution)
Wash to remove unbound secondary antibody
Analyze by flow cytometry
This protocol has been successfully applied to human glioblastoma-astrocytoma epithelial cells (U-87 MG) and can be adapted for other cell types with appropriate controls .
Differentiating between GMFB and GMFG requires careful selection of antibodies and experimental methods due to potential cross-reactivity issues. Scientists have developed sensitive and specific two-site enzyme immunoassays (EIA) that allow for distinct identification of these closely related proteins .
For accurate differentiation:
Use antibodies specifically raised against unique epitopes of each protein
Implement two-site enzyme immunoassays that utilize two different antibodies recognizing distinct regions of the target protein
Include appropriate controls to verify specificity
Consider western blotting with antibodies verified for specificity to confirm results
The developed EIA systems have demonstrated sufficient sensitivity to detect GMFB and GMFG in both human and rat samples, enabling accurate analysis of tissue distribution and serum concentrations . These assays exhibit minimal cross-reactivity between the two proteins, making them suitable for studies requiring specific quantification of either GMFB or GMFG.
For optimal immunofluorescence visualization of GMFB in cultured cells, researchers should implement a protocol that balances fixation, permeabilization, and antibody incubation steps. Based on validated procedures:
Fix adherent cells with 4% paraformaldehyde for 10-15 minutes at room temperature
Permeabilize with 0.1% TritonX-100 for 5-10 minutes to allow antibody access to intracellular GMFB
Block with 5-10% normal serum (from the same species as the secondary antibody) for 1 hour
Incubate with primary GMFB antibody at optimized dilution (typically 1:100 to 1:500) overnight at 4°C
Wash thoroughly with PBS (3-5 washes)
Incubate with fluorophore-conjugated secondary antibody (e.g., Alexa Fluor® 488) at recommended dilution (typically 1:500 to 1:2000) for 1-2 hours at room temperature
Wash thoroughly with PBS (3-5 washes)
Counterstain nuclei with DAPI and mount with anti-fade mounting medium
This protocol has been successfully employed for immunofluorescent analysis of GMFB in various cell types, including U-87 MG human glioblastoma-astrocytoma epithelial cells .
Recent research has identified GMFB as a potential novel biomarker and therapeutic target for hepatocellular carcinoma (HCC). Analysis of GMFB expression in HCC using multiple databases, including Oncomine and The Cancer Genome Atlas (TCGA), has revealed that GMFB is significantly overexpressed in HCC tissues compared to normal liver tissues . This overexpression is associated with tumor stage and pathological grade in HCC patients .
At the molecular level, GMFB expression affects mitochondrial DNA copy number, mitochondrial membrane potential, and matrix metalloproteinase (MMP) expression in HCC cells . Functional studies have demonstrated that GMFB impacts cell migration, invasion, and adhesion in HCC cell lines, suggesting its potential role in HCC metastasis .
GMFB plays a significant role in neuroinflammation through several mechanisms, positioning it as a potential therapeutic target for neuroinflammatory and neurodegenerative diseases . The protein activates key inflammatory signaling pathways, notably the p38 MAPK pathway and nuclear factor-κB (NF-κB) . This activation leads to the upregulation and secretion of pro-inflammatory cytokines such as granulocyte-macrophage colony-stimulating factor (GM-CSF).
Experimental evidence demonstrates that overexpression of GMFB in mouse primary astrocytes leads to significant increases in GM-CSF secretion, with concentration levels reaching 78, 150, and 320 pg/mL at 12, 24, and 48 hours after GMFB transfection, compared to control groups that maintained low levels (5-8 pg/mL) . This effect can be blocked by inhibitors of p38 MAPK and NF-κB, confirming the pathway specificity .
In the central nervous system, GM-CSF functions as a pro-inflammatory cytokine and potent mitogen for microglia . Thus, GMFB's ability to induce GM-CSF secretion suggests it plays a role in microglial activation and subsequent inflammatory processes that contribute to neurodegeneration. This mechanistic understanding positions GMFB as a promising therapeutic target for conditions characterized by neuroinflammation, including Alzheimer's disease, Parkinson's disease, and multiple sclerosis.
To investigate GMFB's reported role in inhibiting tumor cell proliferation, researchers can employ several experimental approaches:
Gene Modulation Studies:
Overexpression of GMFB using viral vectors (such as adenovirus carrying GMFB cDNA) in tumor cell lines to assess effects on proliferation rates
GMFB knockdown using siRNA or CRISPR-Cas9 to evaluate whether reduced GMFB expression enhances proliferation
Creation of stable cell lines with controlled GMFB expression using inducible systems
Proliferation Assays:
MTT or WST-1 assays to measure metabolic activity as a surrogate for cell proliferation
BrdU incorporation assays to directly measure DNA synthesis
Colony formation assays to assess long-term proliferative potential
Cell cycle analysis using flow cytometry to determine if GMFB affects specific cell cycle phases
Signaling Pathway Analysis:
Western blotting to assess activation status of proliferation-related pathways (MAPK, PI3K/Akt)
Pharmacological inhibition of specific pathways to determine which are essential for GMFB's anti-proliferative effects
Co-immunoprecipitation studies to identify GMFB binding partners involved in proliferation regulation
In vivo Models:
Xenograft models with GMFB-modulated tumor cells to assess tumor growth rates
Patient-derived xenografts treated with GMFB or GMFB-targeting agents
Transgenic models with tissue-specific GMFB overexpression or knockout
These approaches can be combined with molecular analyses to elucidate the mechanisms through which GMFB inhibits tumor cell proliferation, potentially revealing new therapeutic strategies for cancer treatment.
For quantitative assessment of GMFB expression across tissues and developmental stages, researchers can employ multiple complementary approaches:
Enzyme Immunoassays (EIA):
Two-site EIAs using specific antibodies have been developed and validated for quantifying GMFB in both human and rat samples . These assays have demonstrated sufficient sensitivity for studying GMFB in various organs and serum samples. The following table summarizes comparative GMFB concentrations across tissues based on published EIA results:
Western Blotting:
Quantitative Western blotting using specific anti-GMFB antibodies (such as Proteintech #10690-1-AP at 1:2000 dilution) can be used to compare GMFB protein levels across different samples . This approach requires careful sample preparation, protein quantification using BCA assays, and appropriate loading controls (e.g., β-actin).
qRT-PCR:
Quantitative real-time PCR can measure GMFB mRNA expression levels, providing insight into transcriptional regulation across tissues and developmental stages. This method complements protein-level analyses and can reveal discrepancies between transcript and protein expression.
Immunohistochemistry with Digital Quantification:
IHC using specific antibodies (such as HPA053669) followed by digital image analysis enables spatial quantification of GMFB expression in tissue sections . This approach provides both quantitative data and information about cellular localization.
For developmental studies, these methods can be applied to samples collected at different time points, as has been done for serum GMFB levels in different age groups of humans and rats .
Validating the specificity of GMFB antibodies is crucial for obtaining reliable research results. Key considerations include:
Cross-reactivity Testing:
Test antibodies against both GMFB and the closely related GMFG to ensure specificity
Evaluate potential cross-reactivity with other proteins containing similar epitopes
Use samples from multiple species if working with antibodies claimed to be cross-reactive
Control Experiments:
Positive controls: Use samples or cell lines known to express GMFB
Negative controls: Use samples lacking GMFB expression or employ GMFB knockdown/knockout systems
Isotype controls: Include appropriate isotype controls in flow cytometry and immunostaining experiments
Blocking peptide controls: Pre-absorb antibodies with specific antigenic peptides to confirm binding specificity
Multiple Detection Methods:
Compare results across different techniques (Western blot, IHC, flow cytometry, ELISA)
Use antibodies recognizing different epitopes of GMFB to confirm findings
Employ orthogonal detection methods that do not rely on antibodies (e.g., mass spectrometry)
Antibody Characterization:
Genetic Validation:
Use GMFB-overexpressing systems to confirm signal increase
Use GMFB knockdown or knockout systems to confirm signal reduction
Employ rescue experiments to restore GMFB expression and antibody detection
Proper validation ensures that experimental observations attributed to GMFB are indeed specific to this protein and not due to antibody cross-reactivity or non-specific binding.
Optimizing Western blotting conditions for GMFB detection requires careful attention to multiple parameters across sample preparation, electrophoresis, transfer, and detection steps:
Sample Preparation:
Electrophoresis Conditions:
Transfer Parameters:
Blocking and Antibody Incubation:
Detection and Visualization:
Controls and Normalization:
These optimized conditions have been successfully applied for GMFB detection in various tissue and cell samples, including liver tissues and cell lines .
Recent research has illuminated GMFB's involvement in multiple signaling pathways, particularly those related to inflammation and cell growth. GMFB has been shown to activate the p38 MAPK pathway and nuclear factor-κB (NF-κB), leading to the upregulation and secretion of inflammatory cytokines such as granulocyte-macrophage colony-stimulating factor (GM-CSF) . This activation represents a key mechanism through which GMFB contributes to neuroinflammation.
To investigate these signaling mechanisms, researchers can employ several approaches:
Pathway Inhibition Studies:
Use specific inhibitors of p38 MAPK and NF-κB to block GMFB-induced effects
Measure downstream effects such as GM-CSF secretion (which can reach 78-320 pg/mL following GMFB overexpression, compared to 5-8 pg/mL in controls)
Employ genetic approaches (dominant negative constructs, CRISPR-Cas9) to disrupt specific pathway components
Phosphorylation Analysis:
Use phospho-specific antibodies to detect activation of pathway components
Implement kinase activity assays to measure functional activation
Perform time-course studies to determine the temporal dynamics of pathway activation
Protein-Protein Interaction Studies:
Conduct co-immunoprecipitation experiments to identify GMFB binding partners
Use proximity ligation assays to visualize protein interactions in situ
Employ yeast two-hybrid or BioID approaches to discover novel interactors
Transcriptomic and Proteomic Analyses:
These approaches can provide comprehensive insights into how GMFB influences cellular signaling networks and contributes to processes such as inflammation, cell proliferation, and neural differentiation.
GMFB has emerged as a promising therapeutic target for neuroinflammatory and neurodegenerative diseases . Antibody-based approaches targeting GMFB could be developed into effective therapeutic strategies through several mechanisms:
Direct Neutralization:
Develop neutralizing antibodies that block GMFB's ability to activate inflammatory signaling pathways
Target specific epitopes involved in GMFB's interaction with downstream effectors
Design antibodies that can cross the blood-brain barrier or be delivered intrathecally
Antibody-Drug Conjugates (ADCs):
Conjugate anti-GMFB antibodies with anti-inflammatory agents to deliver targeted therapy
Target GMFB-expressing cells such as activated astrocytes that contribute to neuroinflammation
Minimize systemic side effects through precise targeting
Cellular Targeting for Immunotherapy:
Develop bispecific antibodies that simultaneously engage GMFB and immune cells
Create chimeric antigen receptor (CAR) T-cells targeting GMFB-expressing cells
Use antibodies to selectively deplete GMFB-overexpressing cells contributing to pathology
Diagnostic and Therapeutic Monitoring:
Employ anti-GMFB antibodies for imaging neuroinflammation in vivo
Monitor treatment efficacy by measuring GMFB levels and localization
Stratify patients based on GMFB expression for personalized therapeutic approaches
The development of such approaches requires extensive preclinical validation in relevant models of neuroinflammatory and neurodegenerative diseases. Initial studies should focus on demonstrating target engagement, efficacy in reducing inflammatory markers (such as GM-CSF secretion), and improvements in functional outcomes in animal models before advancing to clinical studies.
To advance our understanding of GMFB's differential expression and function across cell types, several methodological innovations and approaches are needed:
Single-Cell Analysis Technologies:
Apply single-cell RNA-sequencing to characterize cell type-specific expression patterns of GMFB
Develop single-cell proteomics approaches to measure GMFB protein levels in individual cells
Use spatial transcriptomics to map GMFB expression within complex tissues while preserving spatial context
Advanced Imaging Techniques:
Implement multiplexed immunofluorescence to simultaneously visualize GMFB with cell type markers
Apply super-resolution microscopy to study subcellular localization of GMFB
Develop live-cell imaging approaches using fluorescently tagged GMFB to track dynamics in real-time
Cell Type-Specific Manipulation:
Create conditional knockout models using Cre-lox systems to delete GMFB in specific cell populations
Develop cell type-specific promoters for targeted overexpression of GMFB
Employ CRISPR-Cas9 with cell type-specific delivery for precise genome editing
3D Culture Systems and Organoids:
Investigate GMFB function in brain organoids that recapitulate aspects of neuronal-glial interactions
Develop co-culture systems to study GMFB's role in cellular communication
Use microfluidic devices to model complex tissue environments and cell-cell interactions
Systems Biology Approaches:
Integrate multi-omics data to create comprehensive models of GMFB regulation and function
Apply network analysis to identify cell type-specific GMFB interaction partners
Develop computational models predicting GMFB's differential effects across cell types
These methodological advances would enable researchers to answer key questions about GMFB biology, such as why its effects appear different in neurons versus glial cells, how its expression is regulated in different cellular contexts, and which cell types are most relevant to its roles in disease states like neuroinflammation and cancer.
Researchers working with GMFB antibodies may encounter several technical challenges that can affect experimental outcomes. These challenges and their solutions include:
Non-specific Binding:
Challenge: Background staining or multiple bands in Western blots
Solutions:
Inconsistent Detection:
Challenge: Variable results across experiments
Solutions:
Low Signal Strength:
Challenge: Weak or undetectable GMFB signal
Solutions:
Optimize fixation and permeabilization conditions for intracellular access
Use signal amplification methods (e.g., tyramide signal amplification)
Increase antibody concentration or incubation time
Employ more sensitive detection systems
Consider sample enrichment techniques
Cross-reactivity with GMFG:
Challenge: Difficulty distinguishing between closely related GMFB and GMFG
Solutions:
Fixation-Sensitive Epitopes:
Challenge: Loss of antibody recognition after certain fixation methods
Solutions:
Test multiple fixation protocols (PFA, methanol, acetone)
Optimize fixation duration and conditions
Consider antigen retrieval methods
Use antibodies raised against fixation-resistant epitopes
By systematically addressing these challenges, researchers can significantly improve the reliability and reproducibility of experiments utilizing GMFB antibodies.
When researchers encounter inconsistencies in GMFB expression data across different experimental platforms, a systematic approach can help reconcile these discrepancies:
Platform-Specific Validation:
Validate each detection method independently with appropriate controls
Establish platform-specific threshold values and dynamic ranges
Determine the detection limits for each method
Use recombinant GMFB protein standards to calibrate quantitative measurements
Sample Processing Harmonization:
Standardize sample collection, storage, and processing protocols
Use identical lysis buffers and protein extraction methods when possible
Implement consistent normalization strategies (housekeeping genes/proteins)
Process samples in parallel for cross-platform comparisons
Cross-Platform Correlation Analysis:
Generate correlation plots between methods (e.g., qPCR vs. Western blot)
Calculate correlation coefficients and determine if discrepancies are systematic
Develop conversion factors to harmonize data across platforms
Use rank-based comparisons when absolute values differ but trends are consistent
Orthogonal Validation:
Employ multiple independent techniques to measure GMFB
Compare protein (Western blot, immunoassay) and mRNA (qPCR, RNA-seq) measurements
Use genetic manipulation (overexpression, knockdown) to create internal validation standards
Consider mass spectrometry-based approaches as a reference method
Biological Context Analysis:
Evaluate whether discrepancies reflect genuine biological differences
Consider post-transcriptional regulation when mRNA and protein levels diverge
Examine protein localization and modification status
Account for cell type heterogeneity in complex samples
An example reconciliation approach might involve:
Measuring GMFB expression in the same samples using qPCR, Western blot, and immunoassay
Plotting correlations between methods and identifying systematic deviations
Developing a normalization formula that accounts for method-specific biases
Validating this formula with an independent sample set
Reporting both raw and harmonized data to maintain transparency
By systematically addressing these factors, researchers can improve data consistency and develop a more coherent understanding of GMFB expression across different experimental contexts.
GMFB research sits at a unique intersection between neuroinflammation and cancer biology, with emerging evidence suggesting common underlying mechanisms and potential for translational applications across both fields:
Inflammatory Signaling Pathway Activation:
In neuroinflammation: GMFB activates p38 MAPK and NF-κB pathways in glial cells, leading to increased production of pro-inflammatory cytokines like GM-CSF
In cancer: Similar inflammatory pathways contribute to tumor progression, with GMFB overexpression in HCC correlating with poor prognosis
Common mechanism: Activation of inflammatory signaling cascades that influence cell survival, proliferation, and migration
Cell Proliferation and Differentiation:
Neural context: GMFB influences neural cell differentiation and regeneration
Cancer context: GMFB affects tumor cell proliferation, with apparent inhibitory effects in some contexts but potential promotion of progression in others (as in HCC)
Integrative research opportunity: Understanding how GMFB's effects on proliferation are context-dependent
Migration and Invasion:
Mitochondrial Function:
Translational Applications:
Diagnostic biomarkers: GMFB as a potential biomarker in both neurological disorders and cancer (particularly HCC)
Therapeutic targeting: Developing strategies to modulate GMFB activity that could be applicable in both disease contexts
Risk stratification: Using GMFB expression for patient stratification, with particular relevance to gender-specific differences observed in HCC
This intersection creates opportunities for cross-disciplinary research approaches that leverage findings from both fields to accelerate understanding of GMFB biology and develop novel therapeutic strategies.
The future of GMFB antibody applications in precision medicine holds significant promise across several research directions:
Biomarker Development for Patient Stratification:
Utilize GMFB antibodies to develop immunohistochemical or liquid biopsy assays for cancer subtyping
Integrate GMFB expression with other biomarkers to create diagnostic and prognostic panels
Explore gender-specific roles of GMFB, particularly in HCC where significant differences in prognostic value between males and females have been observed
Develop standardized scoring systems for GMFB expression in tissue samples
Therapeutic Antibody Development:
Design neutralizing antibodies targeting GMFB for neuroinflammatory conditions
Develop antibody-drug conjugates directed against GMFB-expressing cells
Create bispecific antibodies linking GMFB recognition with immune cell engagement
Investigate intrabodies to modulate GMFB function within specific cellular compartments
Companion Diagnostics for Treatment Selection:
Develop GMFB antibody-based assays to predict response to specific therapies
Create immunoassays for monitoring treatment efficacy in real-time
Identify therapy-induced changes in GMFB expression or localization
Establish threshold values for GMFB expression that correlate with treatment outcomes
Theranostic Applications:
Design dual-purpose antibodies for simultaneous imaging and therapy
Develop imaging agents based on GMFB antibodies for visualization of neuroinflammation
Create radiolabeled GMFB antibodies for targeted radiotherapy of GMFB-overexpressing tumors
Implement PET/SPECT imaging of GMFB expression for disease monitoring
Cell-Specific Targeting in Complex Tissues:
Utilize antibodies for selective delivery of therapeutics to GMFB-expressing cells
Develop nanoparticle formulations conjugated with GMFB antibodies for targeted drug delivery
Create cell-selective gene therapy approaches using GMFB promoters or targeting
Design chimeric antigen receptor T-cells targeting GMFB-expressing pathogenic cells
These research directions have the potential to transform GMFB from a basic research focus to a clinically relevant target for precision medicine approaches across multiple diseases, particularly in neuroinflammatory conditions and cancer.
Developing a unified understanding of GMFB's biological functions requires systematic integration of findings across diverse model systems. Researchers can implement the following approaches to synthesize this knowledge effectively:
Cross-Species Comparative Analysis:
Systematically compare GMFB structure, expression, and function across species (human, mouse, rat)
Identify evolutionarily conserved domains and interaction partners
Correlate species-specific differences with functional outcomes
Create databases or repositories of GMFB functional data across species
Multi-Scale Integration:
Connect molecular mechanisms (e.g., p38 MAPK pathway activation) to cellular phenotypes (differentiation, migration)
Link cellular effects to tissue-level outcomes in different organ systems
Correlate tissue-specific observations with organismal phenotypes in animal models
Develop computational models that predict GMFB effects across biological scales
Disease-Specific Contextualization:
Compare GMFB's role across different pathological contexts (neurodegeneration, cancer, inflammation)
Identify common mechanisms versus context-specific functions
Create disease maps highlighting GMFB's position in different pathological processes
Establish whether GMFB functions as a driver or responder in various disease states
Methodological Harmonization:
Develop standardized protocols for GMFB detection and functional analysis
Create reference standards for quantitative comparisons across studies
Establish common ontologies and terminology for GMFB-related phenomena
Implement meta-analysis approaches to synthesize findings across multiple studies
Collaborative Research Networks:
Form interdisciplinary consortia spanning neuroscience, cancer biology, and immunology
Create shared resources such as validated antibodies, expression constructs, and animal models
Implement coordinated research agendas targeting key knowledge gaps
Develop centralized databases integrating GMFB-related findings
By implementing these integrative approaches, researchers can transcend the limitations of individual model systems and develop a comprehensive understanding of GMFB biology that spans from molecular mechanisms to physiological functions and pathological roles. This unified knowledge framework will facilitate more effective translation of GMFB research into clinical applications across multiple disease contexts.
GMFB is known to:
GMFB has been associated with various diseases and conditions:
Mouse anti-human GMFB antibodies are monoclonal antibodies produced in mice that specifically target human GMFB. These antibodies are used in various research applications, including:
Research on GMFB continues to uncover its potential therapeutic applications:
In conclusion, GMFB is a multifunctional protein with significant roles in glial cell maturation, neural regeneration, and disease modulation. Mouse anti-human GMFB antibodies are valuable tools in research, helping to elucidate the functions and therapeutic potential of GMFB.