GMFB Mouse

Glia Maturation Factor Beta Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

GMFB Gene Structure and Expression in Mice

Key Features of Mouse GMFB Models

FeatureDescriptionSource
Gene LocationChromosome 14 (conserved between humans and mice)
Protein Size17 kDa (identical to human GMFB except for two amino acids)
Expression PatternPredominantly in CNS; detectable in colon, thymus, and kidney
Developmental PeakHighest expression at 1 week post-birth in rats; stable serum levels

GMFB-KO mice, generated via CRISPR-mediated frame-shift mutations, produce truncated nonfunctional protein . These models enable direct analysis of GMFB’s role in health and disease.

Role in Neuroinflammation and Neurodegeneration

GMFB regulates immune responses and oxidative stress in the CNS.

Experimental Findings

  • Neuroinflammation:

    • GMFB-KO mice show reduced pro-inflammatory cytokines (e.g., TNF-α, GM-CSF) in experimental autoimmune encephalomyelitis (EAE) .

    • Overexpression of GMFB in astrocytes increases NF-κB activity and GM-CSF secretion, promoting microglial activation .

  • Neurodegeneration:

    • GMFB-KO mice exhibit reduced oxidative stress (lower MDA, NO levels) and neuroinflammation after MPP+ treatment, mimicking Parkinson’s disease (PD) .

    • In Alzheimer’s disease models, G9a inhibitors increase GMFB expression, enhancing neuronal plasticity and rescuing cognitive decline .

Table 1: GMFB-KO vs. WT Mice in Neurological Models

Disease ModelGMFB-KO OutcomeWT OutcomeSource
EAE20% incidence vs. 100% in WT; lower TNF-α/GM-CSFSevere inflammation and clinical scores
MPP+ ToxicityReduced lipid peroxidation (MDA ↓ 56%)Elevated oxidative stress
Traumatic Brain Injury (TBI)Attenuated reactive gliosis; shorter GFAP fibersPromoted radial glial proliferation

Role in Metabolic Diseases

GMFB contributes to complications in type 1 diabetes (T1D), particularly osteoporosis (T1D-OP).

Mechanistic Insights

  • T1D-OP Pathogenesis:

    • GMFB-KO rats show reduced osteoclast activity and bone resorption under high glucose conditions .

    • GMFB interacts with the Arp2/3 complex, regulating actin dynamics critical for osteoclast differentiation .

  • Therapeutic Targeting:

    • Small-molecule inhibitors (e.g., CCG-1423) targeting GMFB-Arp2/3 interactions suppress osteoclast formation in vitro .

Table 2: GMFB’s Role in T1D-OP

ParameterGMFB-KO RatsWT RatsSource
Osteoclast Activity↓ Bone resorption (↓ TRAP+ cells)↑ Bone resorption
Actin DynamicsReduced Arp2/3-mediated debranchingEnhanced lamellipodia formation

Mechanisms of Action: Signaling Pathways

GMFB modulates cellular responses through multiple pathways:

PathwayRole of GMFBKey OutcomesSource
p38 MAPKActivates NF-κB, driving GM-CSF secretion↑ Microglial proliferation
SUMOylationEnhances GMFB stability and nuclear translocation↑ Pro-inflammatory cytokine release
G9a/H3K9me2Epigenetic regulation via histone methylation↑ Neuronal plasticity in AD models

Therapeutic Implications

GMFB modulation offers potential for treating neuroinflammatory and metabolic disorders:

Strategies

  • Anti-GMFB Antibodies:

    • Intravenous administration reduces EAE severity and cytokine production .

  • Small-Molecule Inhibitors:

    • Virtual screening identified compounds disrupting GMFB-Arp2/3 binding, effective in T1D-OP models .

  • Epigenetic Modulators:

    • G9a inhibitors (e.g., UNC0642) increase GMFB expression, mitigating Alzheimer’s pathology .

Table 3: Therapeutic Approaches Targeting GMFB

ApproachTarget/MechanismEfficacy in ModelsSource
Anti-GMFB AntibodiesNeutralizes pro-inflammatory effects↓ EAE symptoms (clinical score ↓ 80%)
CCG-1423Inhibits MKL1/SRF pathway↓ Osteoclast differentiation
G9a Inhibitors↑ GMFB expression via H3K9me2 reduction↑ Cognitive function in SAMP8 mice

Product Specs

Introduction
Glia Maturation Factor-Beta (GMFB), a member of the GMF subfamily within the ADF family of actin-binding proteins, plays a critical role in the nervous system. Phosphorylated upon phorbol ester stimulation, GMFB is essential for brain cell differentiation, neural regeneration, and tumor cell proliferation inhibition. Notably, its overexpression in astrocytes enhances Brain-Derived Neurotrophic Factor (BDNF) production. Exercise-induced upregulation of GMFB highlights the significance of BDNF in this process.
Description
Recombinant Mouse Glia Maturation Factor-Beta (GMF-Beta), produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 141 amino acids, resulting in a molecular weight of 16.6 kDa. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
The lyophilized GMF-beta protein is prepared in a 0.2µm filtered concentrated solution using PBS at a pH of 7.4.
Solubility
For reconstitution, it is advised to dissolve the lyophilized GMFB in sterile 18MΩ-cm H2O to a minimum concentration of 100µg/ml. This solution can be further diluted with other aqueous solutions as needed.
Stability
While lyophilized GMF-B remains stable at room temperature for up to 3 weeks, storage in a desiccated state below -18°C is recommended. After reconstitution, GMF-beta should be kept at 4°C for 2-7 days. For extended storage, freezing below -18°C is advisable. To ensure optimal stability during long-term storage, adding a carrier protein like HSA or BSA (0.1%) is recommended. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeding 97.0% is confirmed through the following analyses: (a) RP-HPLC (b) SDS-PAGE
Synonyms
Glia maturation factor beta, GMFB, GMF-B, GMF-beta, GMF, C79176, AI851627, D14Ertd630e, 3110001H22Rik, 3110001O16Rik.
Source
Escherichia Coli.
Amino Acid Sequence
SESLVVCDVA EDLVEKLRKF RFRKETHNAA IIMKIDKDER LVVLDEELEG
VSPDELKDEL PERQPRFIVY SYKYQHDDGR VSYPLCFIFS SPVGCKPEQQ
MMYAGSKNKL VQTAELTKVF EIRNTEDLTE EWLREKLGFF H.

Q&A

Basic Question: What role does GMFB play in mouse physiology?

GMFB is a brain-specific protein that regulates the production of proinflammatory cytokines and chemokines in brain cells. Its activity can lead to the destruction of oligodendrocytes—cells responsible for myelin formation—and neurons. This makes GMFB critical for studying neurodegenerative disorders such as multiple sclerosis and Alzheimer's disease . The protein's structural similarity to human GMF-beta (with only two amino acid differences) further enhances its relevance for translational studies .

Advanced Question: How does GMFB influence neuroinflammatory pathways in experimental mouse models?

In experimental settings, GMFB has been shown to act upstream of inflammatory cascades by modulating cytokine release. Studies using knockout mice have demonstrated that the absence of GMFB can significantly alter immune responses, leading to either exacerbated or diminished neuroinflammation depending on the genetic background of the model . Researchers often use GMFB mice to investigate mechanisms underlying oligodendrocyte destruction and neuronal apoptosis induced by inflammatory mediators.

Basic Question: What are key considerations when designing experiments with GMFB mice?

When designing experiments involving GMFB mice, researchers must consider factors such as sample size, genetic background variability, and statistical power. For example, optimizing the number of mice per group ensures sufficient power to detect phenotypic differences while adhering to ethical guidelines under the 3Rs principle (Replacement, Reduction, Refinement) . Additionally, standardizing environmental conditions minimizes confounding variables that could affect experimental outcomes .

Advanced Question: What statistical methodologies are recommended for analyzing data from GMFB mouse experiments?

High-throughput phenotyping studies often employ nested ANOVA models to account for variance sources such as genetic background and environmental factors . Power analysis is crucial for determining sample sizes that balance statistical significance with ethical considerations. For example, false discovery rate (FDR) adjustments can mitigate risks associated with multiple testing problems in large datasets . In one case study involving non-invasive blood pressure measurements in mutant mice, nested ANOVA combined with FDR controls was used to ensure robust results while minimizing false positives .

Basic Question: Why is genetic background important in GMFB mouse studies?

The genetic background onto which a gene-targeted allele is placed can significantly affect phenotype expression. Variations may manifest as differences in penetrance or expressivity of traits related to neuroinflammation or neuronal damage . Mixed genetic backgrounds often provide a broader range of phenotypes but may introduce variability that complicates data interpretation .

Advanced Question: What strategies can be employed to minimize genetic variability in GMFB mouse experiments?

Researchers can use congenic strains to reduce genetic polymorphism and flanking gene effects that might confound results . Speed congenic techniques allow for rapid generation of strains with consistent genetic backgrounds within 1–2 years . Additionally, employing mixed-background knockouts initially can help identify modifier genes responsible for phenotypic variability before transitioning to more stable congenic lines .

Basic Question: How do researchers address conflicting data from GMFB mouse studies?

Conflicting data often arise from differences in experimental design or environmental conditions. For example, variations in housing temperature or diet can influence cytokine levels and immune responses in GMFB mice . To address these issues, researchers should standardize protocols and include control groups with similar genetic backgrounds.

Advanced Question: How can researchers validate findings from high-throughput phenotyping studies involving GMFB mice?

Validation requires follow-up experiments using specialized assays tailored to the observed phenotype. For instance, if initial screening suggests altered cytokine levels in GMFB knockout mice, targeted ELISA assays or flow cytometry can confirm these findings . Secondary phenotyping centers often play a complementary role by conducting more focused investigations based on primary screening results .

Basic Question: What experimental techniques are commonly employed with GMFB mice?

Techniques such as immunohistochemistry (IHC), Western blotting, and RNA sequencing are frequently used to assess protein expression levels and gene activity related to neuroinflammation in GMFB mice . Non-invasive imaging methods like MRI can also provide insights into structural changes within the brain caused by cytokine dysregulation.

Advanced Question: How do researchers model neurodegenerative diseases using GMFB mice?

GMFB knockout or transgenic mice serve as models for studying diseases characterized by neuroinflammation and neuronal loss. By exposing these mice to inflammatory stimuli such as lipopolysaccharides (LPS), researchers can mimic pathological conditions seen in human disorders like multiple sclerosis . Data from these models help elucidate pathways involved in oligodendrocyte destruction and axonal degeneration.

Product Science Overview

Introduction

Glia Maturation Factor Beta (GMF-β) is a protein that plays a crucial role in the development and maintenance of the nervous system. It belongs to the actin-binding proteins ADF family, specifically the GMF subfamily . This protein is involved in various cellular processes, including differentiation, maintenance, and regeneration of glial cells and neurons .

Structure and Properties

GMF-β contains an ADF-H (actin depolymerization factor homology) domain, which is essential for its function . The structures of mouse GMF-β, solved by both crystallography and NMR, reveal similarities and critical differences with ADF-H domains . The molecular weight of recombinant mouse GMF-β is approximately 16.6 kDa . It is typically produced in E. coli and is available as a sterile filtered white lyophilized (freeze-dried) powder .

Biological Functions

GMF-β is involved in several key biological processes:

  • Nervous System Development: GMF-β is crucial for the differentiation and maintenance of glial cells and neurons .
  • Immune Function: It plays a role in immune responses and has been implicated in angiogenesis .
  • Tumor Cell Proliferation: GMF-β has been shown to inhibit the proliferation of tumor cells .
Research and Applications

Research on GMF-β has revealed its potential therapeutic applications:

  • Diabetic Osteoporosis: GMF-β deficiency has been shown to protect against diabetic osteoporosis by suppressing osteoclast hyperactivity . This suggests that GMF-β inhibitors could be a potential therapeutic strategy for treating osteoporosis in diabetic patients.
  • Neurodegenerative Diseases: Given its role in nervous system development and maintenance, GMF-β is being studied for its potential in treating 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 2025 TheBiotek. All Rights Reserved.