GPM6B Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to GPM6B and Its Antibody

GPM6B (Glycoprotein M6B) is a member of the proteolipid protein family, encoded by the GPM6B gene (NCBI Gene ID: 2824) located on human chromosome Xp22.2 . It is expressed in neurons, oligodendrocytes, and osteoblasts, with roles in cellular trafficking, serotonin uptake regulation, and bone formation . GPM6B antibodies are polyclonal or monoclonal reagents designed to detect this protein in research and diagnostic applications, such as Western blotting (WB), immunohistochemistry (IHC), and enzyme-linked immunosorbent assays (ELISA) .

Functional Insights

  • Neural Differentiation: GPM6B expression correlates with neural markers (GFAP, TUBB3, MAP2) and is upregulated during retinoic acid-induced differentiation of NT2 cells . CRISPR/Cas9 deletion of a GA-repeat in the GPM6B promoter reduced its expression by 49%, impairing neural differentiation .

  • Cancer Prognosis:

    • Glioblastoma: Low GPM6B expression correlates with aggressive glioma subtypes and poor survival (HR = 0.44, p < 0.001) .

    • Prostate Cancer: GPM6B acts as a tumor suppressor by inhibiting serotonin uptake (p < 0.05 in proliferation assays) .

    • Osteoblast Function: Regulates matrix vesicle release and cytoskeleton maintenance, critical for bone mineralization .

Glioma and Glioblastoma

  • Survival Analysis: High GPM6B expression predicts prolonged survival in glioma patients (5-year survival rate: 65% vs. 28% in low-expression groups) .

  • Mechanistic Role: Overexpression in mesenchymal glioblastoma stem cells (GSCs) inhibits Wnt/β-catenin signaling, reducing self-renewal and tumorigenicity (p < 0.01 in neurosphere assays) .

Psychiatric and Neurological Disorders

  • Serotonin Regulation: GPM6B modulates serotonin transporter (SERT) trafficking, impacting neuropsychiatric traits like depression and anxiety .

  • Mouse Models: CRISPR-edited Gpm6b mutants exhibit altered sensorimotor gating, supporting its role in neurodevelopmental disorders .

Therapeutic Potential

  • Prostate Cancer: GPM6B overexpression reduces cell proliferation by 40% in vitro and inhibits xenograft growth in vivo .

  • Bone Diseases: Dysregulation linked to osteoporosis; GPM6B knockout mice show defective bone mineralization .

Future Directions

  • Clinical Translation: Validate GPM6B as a biomarker for glioma subtyping and prostate cancer prognosis .

  • Therapeutic Targeting: Explore small-molecule modulators of GPM6B for neurodegenerative and bone disorders .

  • Mechanistic Studies: Elucidate its role in serotonin signaling and cross-talk with pathways like Wnt/β-catenin .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days of receiving your order. Delivery times may vary depending on the shipping method and destination. For specific delivery times, please consult your local distributor.
Synonyms
GPM6B; M6B; Neuronal membrane glycoprotein M6-b; M6b
Target Names
GPM6B
Uniprot No.

Target Background

Function
GPM6B is implicated in neural development. It plays a role in regulating osteoblast function and bone formation. GPM6B is involved in matrix vesicle release by osteoblasts, likely through its influence on the actin cytoskeleton. It may also contribute to the cellular trafficking of the serotonin transporter (SERT) and thereby the regulation of serotonin uptake.
Gene References Into Functions
  1. Elevated mRNA and protein expression levels of GPM6B are associated with various types of gynecological malignancies. PMID: 25113253
  2. Circulating levels of DR6 and GPM6B correlate with breast cancer tumor grade. PMID: 24696529
  3. High resolution melting analysis in promoters of tumor markers neuronal membrane glycoprotein M6-B, melanoma antigen family A12, and immunoglobulin superfamily Fc receptor indicated invasiveness of hepatocellular carcinoma. PMID: 23950870
  4. Microarray expression analysis of GPM6B-depleted osteogenic human mesenchymal stem cells revealed significant changes in genes involved in cytoskeleton organization and biogenesis. PMID: 21638316
  5. Mutations in M6B are unlikely to play a role in Pelizaeus-Merzbacher-like syndrome, as examined in eight patients. PMID: 15214007
  6. Research suggests that M6B regulates serotonin uptake by influencing the cellular trafficking of the serotonin transporter. PMID: 18581270

Show More

Hide All

Database Links

HGNC: 4461

OMIM: 300051

KEGG: hsa:2824

UniGene: Hs.495710

Protein Families
Myelin proteolipid protein family
Subcellular Location
Cell membrane; Multi-pass membrane protein.
Tissue Specificity
Neurons and glia; cerebellar Bergmann glia, in glia within white matter tracts of the cerebellum and cerebrum, and in embryonic dorsal root ganglia.

Q&A

What is GPM6B and why is it significant for research?

GPM6B is a four-transmembrane domain protein belonging to the proteolipid protein family. In humans, the canonical protein consists of 265 amino acid residues with a molecular mass of 29 kDa, primarily localized to the cell membrane . Up to four different isoforms have been reported, with notable expression in the cerebral cortex and colon .

As a member of the Myelin proteolipid protein family, GPM6B plays crucial roles in neural development and has been implicated in several cellular processes . Recent research has demonstrated its involvement in:

  • Neural cell functionality and differentiation

  • Smooth muscle cell (SMC) differentiation through TGF-β-Smad2/3 signaling

  • Interactions with other transmembrane proteins and signaling pathways

This multifunctionality makes GPM6B a valuable target for studies on neural development, vascular biology, and cellular differentiation mechanisms.

What applications are most suitable for GPM6B antibodies?

GPM6B antibodies can be utilized in multiple research applications, each with specific advantages for investigating different aspects of GPM6B biology:

ApplicationCommon UsageTechnical Considerations
Western Blot (WB)Protein expression quantificationMost widely used; enables detection of multiple isoforms
ELISAQuantitative protein detectionUseful for high-throughput screening
Flow Cytometry (FCM)Single-cell analysisAllows sorting of GPM6B-expressing populations
Immunohistochemistry (IHC)Tissue localizationParticularly valuable for brain tissue studies
Immunofluorescence (IF)Subcellular localizationEnables co-localization studies with neural markers

When selecting an antibody, consider the specific isoforms you wish to detect and the species compatibility required for your experimental system .

How should I select the appropriate GPM6B antibody for my research?

Selection of GPM6B antibodies should be guided by several critical factors:

  • Target epitope: Antibodies targeting different regions (N-terminal vs. full-length) may detect distinct isoforms or conformations

  • Species reactivity: Available antibodies show varying cross-reactivity with human, mouse, rat, and other species

  • Validation status: Check if the antibody has been validated in your specific application (WB, IHC, FCM, etc.)

  • Conjugation requirements: Consider whether unconjugated antibodies or those with specific tags (FITC, HRP, biotin) are needed for your experimental design

  • Isoform specificity: Determine whether your research requires detection of all GPM6B isoforms or specific variants

For neural differentiation studies, antibodies validated in immunofluorescence applications with demonstrated reactivity against your species of interest would be most suitable .

How should I design experiments to study GPM6B in neural differentiation?

When investigating GPM6B's role in neural differentiation, a comprehensive experimental design should include:

Cell Models:

  • NT2 cells treated with retinoic acid (RA) represent an established model for studying neural differentiation

  • Primary neural cultures from appropriate species based on antibody reactivity

  • Neural stem cells or induced pluripotent stem cells undergoing differentiation

Experimental Approaches:

  • Time-course studies: Monitor GPM6B expression throughout the differentiation process

  • Loss-of-function experiments:

    • CRISPR/Cas9-mediated deletion of GPM6B or regulatory elements

    • shRNA knockdown approaches

  • Gain-of-function experiments: Overexpression systems to evaluate effects on differentiation

  • Marker co-expression analysis: Correlate GPM6B with neural markers like GFAP, TUBB3, MAP2, and NES

Critical Controls:

  • Include appropriate isotype controls for antibody specificity

  • Use CRISPR/Cas9 or shRNA knockdown controls to validate antibody signals

  • Include positive control tissues known to express GPM6B (e.g., cerebral cortex)

This experimental framework enables comprehensive characterization of GPM6B's functional role in neural differentiation processes.

What protocols are recommended for Western blotting with GPM6B antibodies?

For effective detection of GPM6B via Western blotting, the following optimized protocol is recommended:

Sample Preparation:

  • Harvest cells or tissue samples

  • Lyse in membrane protein-compatible buffer (containing appropriate detergents like NP-40 or Triton X-100)

  • Do not boil membrane protein samples; instead, incubate at 37°C for 30 minutes

  • Centrifuge lysates and quantify protein concentration

Gel Electrophoresis:

  • Load 20-50 μg protein per lane

  • Use 10-12% SDS-PAGE gels for standard separation

  • Consider gradient gels (4-20%) for better resolution of multiple isoforms

  • Include molecular weight markers covering the 25-35 kDa range

Transfer and Detection:

  • Transfer to PVDF membrane (preferred for membrane proteins)

  • Block with 5% non-fat milk or BSA in TBST

  • Incubate with anti-GPM6B primary antibody at manufacturer's recommended dilution (typically 1:500-1:1000)

  • Wash thoroughly and incubate with appropriate HRP-conjugated secondary antibody

  • Develop using chemiluminescence detection

Special Considerations:

  • GPM6B shows multiple isoforms; the canonical form appears at approximately 29 kDa

  • Post-translational modifications, particularly glycosylation, may cause shifts in apparent molecular weight

  • When studying TGF-β1-induced expression, consider time-course experiments to capture peak expression at day 3

This protocol has been effectively employed to demonstrate GPM6B upregulation during TGF-β1-induced SMC differentiation .

How can I optimize immunofluorescence staining for GPM6B?

Immunofluorescence staining for GPM6B requires careful optimization given its membrane localization and multiple isoforms:

Sample Preparation:

  • Culture cells on coverslips or prepare tissue sections

  • Fix with 4% paraformaldehyde for 15-20 minutes

  • For membrane proteins like GPM6B, use gentle permeabilization:

    • 0.1-0.2% Triton X-100 for 5-10 minutes, or

    • 0.1% saponin (milder for membrane proteins)

Staining Procedure:

  • Block with 5% normal serum in PBS containing 0.1% Triton X-100

  • Incubate with primary anti-GPM6B antibody overnight at 4°C

  • Wash thoroughly with PBS

  • Apply appropriate fluorophore-conjugated secondary antibody

  • Counterstain nuclei with DAPI

  • Mount with anti-fade medium

Co-staining Strategies:
For neural differentiation studies, combine GPM6B staining with established markers:

  • GFAP for astrocyte identification

  • TUBB3 for neurons

  • MAP2 for mature neurons

  • NES for neural progenitors

Image Acquisition:

  • Use confocal microscopy for precise membrane localization

  • Capture z-stacks to fully visualize membrane distribution

  • Apply consistent exposure settings across experimental conditions

This approach has successfully demonstrated the relationship between GPM6B expression and neural differentiation markers in studies utilizing CRISPR/Cas9-mediated deletion of GPM6B regulatory elements .

How does GPM6B interact with TGF-β signaling pathways?

GPM6B has been identified as a critical component of TGF-β signaling, particularly in the context of smooth muscle cell differentiation. This interaction involves several sophisticated molecular mechanisms:

Direct Protein Interactions:

  • GPM6B directly binds to TGF-β type I receptor (TβRI) but not to TGF-β type II receptor (TβRII) or Smad2/3

  • This interaction can be confirmed through co-immunoprecipitation experiments

  • GPM6B does not affect TβRI or TβRII expression levels at either protein or mRNA level

Signaling Regulation Mechanism:

  • GPM6B modulates the interactions between TβRI, TβRII, and Smad2/3

  • Knockdown of GPM6B inhibits the association between TβRI and Smad2/3, as well as between TβRI and TβRII

  • This regulatory function facilitates Smad2/3 phosphorylation and subsequent signaling activation

Feedback Mechanism:

  • Activation of TGF-β signaling upregulates GPM6B expression

  • This creates a positive feedback loop that enhances SMC differentiation efficiency

These findings establish GPM6B as a novel regulator of TGF-β-Smad2/3 signaling, with potential implications for various cellular differentiation processes and tissue development.

What is the role of GPM6B in cellular differentiation processes?

Research has revealed GPM6B's involvement in multiple differentiation pathways, with particularly well-characterized roles in:

Smooth Muscle Cell (SMC) Differentiation:

  • GPM6B expression is significantly upregulated during TGF-β1-induced SMC differentiation

  • Both protein and mRNA levels increase, peaking at approximately day 3 of differentiation

  • Multiple GPM6B isoforms show similar upregulation patterns

  • Knockdown of GPM6B significantly inhibits SMC marker expression, including:

    • α-SMA (smooth muscle α-actin)

    • MYH11 (smooth muscle myosin heavy chain)

    • h1-calponin

Neural Cell Differentiation:

  • GPM6B is considered a key gene in neural cell functionality

  • CRISPR/Cas9-mediated deletion of a regulatory GA-repeat in GPM6B significantly decreases its expression

  • This reduction leads to disrupted differentiation of NT2 cells into neural lineages

  • Decreased expression of neural markers is observed, including:

    • GFAP (0.77-fold reduction)

    • TUBB3 (0.57-fold reduction)

    • MAP2 (0.2-fold reduction)

These findings establish GPM6B as a multifunctional regulator of cellular differentiation across different tissue contexts, potentially through its interactions with fundamental signaling pathways.

How can CRISPR/Cas9 technology be utilized to study GPM6B function?

CRISPR/Cas9 provides powerful approaches for investigating GPM6B's functional roles through precise genetic modifications:

Targeting Strategies:

  • Complete gene knockout:

    • Design sgRNAs targeting early exons to create frameshift mutations

    • Target conserved regions essential for protein function

  • Regulatory element modification:

    • Target non-coding regulatory sequences, such as the GA-repeat region

    • This approach has successfully demonstrated the regulatory importance of this element for GPM6B expression

  • Domain-specific editing:

    • Create specific modifications in transmembrane domains or protein interaction regions

Experimental Design Examples:
In published research, CRISPR/Cas9-mediated deletion of a GA-repeat in human GPM6B led to:

  • Significantly decreased GPM6B expression at both RNA (p < 0.05) and protein (40% reduction) levels

  • Disrupted differentiation of NT2 cells into neural lineages

  • Dramatic decreases in neural marker expression

  • Increased numbers of NES-positive cells (p < 0.01)

Technical Considerations:

  • Design sgRNAs with minimal off-target effects

  • For regulatory element targeting, design sgRNAs flanking the target region

  • Validate editing efficiency through sequencing and expression analysis

  • Include appropriate controls (non-targeting sgRNAs)

This approach enables precise dissection of GPM6B's regulatory mechanisms and functional roles in various cellular contexts.

How should I address specificity issues with GPM6B antibodies?

Ensuring antibody specificity is critical for reliable research outcomes. For GPM6B antibodies, consider these strategies:

Common Specificity Issues:

  • Cross-reactivity with related proteins:

    • GPM6B belongs to the proteolipid protein family, which includes similar proteins like GPM6A

    • Potential cross-reactivity may occur due to sequence homology

  • Isoform-specific detection:

    • Up to four different GPM6B isoforms have been reported

    • Different antibodies may recognize specific isoforms or all variants

Validation Approaches:

  • Genetic validation:

    • Use CRISPR/Cas9 knockout or shRNA knockdown samples as negative controls

    • Signal should be significantly reduced in these samples

  • Multiple antibody validation:

    • Use antibodies targeting different epitopes of GPM6B

    • Consistent results across different antibodies increase confidence in specificity

  • Peptide competition assay:

    • Pre-incubate antibody with immunizing peptide

    • This should abolish specific signals while non-specific binding remains

  • Positive controls:

    • Include tissues with known GPM6B expression (cerebral cortex, colon)

    • Use recombinant GPM6B protein where appropriate

These validation approaches should be implemented across all applications (WB, IF, FCM) to ensure consistent and reliable detection of GPM6B.

What factors influence GPM6B detection in experimental systems?

Several factors can significantly impact GPM6B detection and should be considered when designing experiments:

Biological Factors:

  • Expression levels and patterns:

    • GPM6B expression varies by tissue type (high in cerebral cortex and colon)

    • Expression changes dynamically during differentiation processes

    • Peak expression may occur at specific timepoints (e.g., day 3 in TGF-β1-induced differentiation)

  • Post-translational modifications:

    • GPM6B undergoes glycosylation

    • These modifications may affect antibody binding or protein migration patterns

  • Protein interactions:

    • GPM6B interacts with other proteins like TβRI

    • These interactions may mask epitopes in certain applications

Technical Factors:

  • Sample preparation:

    • As a membrane protein, GPM6B requires appropriate extraction methods

    • Avoid boiling samples for Western blotting

    • Use detergents compatible with membrane protein extraction

  • Fixation methods:

    • For immunofluorescence, fixation conditions affect membrane protein detection

    • Cryofixation may preserve native conformation better than chemical fixation

  • Antibody selection:

    • Different antibodies target distinct epitopes

    • N-terminal antibodies may detect different isoforms than C-terminal antibodies

These considerations should guide experimental design and help interpret variable results across different detection methods or experimental conditions.

How can I analyze contradictory data when studying GPM6B function?

When faced with seemingly contradictory results in GPM6B research, a systematic analytical approach is essential:

Sources of Experimental Variability:

  • Cell type and context:

    • GPM6B functions differently in neural cells versus smooth muscle cells

    • TGF-β signaling context significantly affects GPM6B function and expression

  • Temporal dynamics:

    • Expression and function change during differentiation time courses

    • Different sampling timepoints may yield apparently contradictory results

  • Isoform-specific effects:

    • The multiple isoforms of GPM6B may have distinct functions

    • Different detection methods may preferentially identify specific isoforms

Reconciliation Strategies:

  • Comprehensive experimental design:

    • Include detailed time courses and multiple cell types

    • Assess all GPM6B isoforms when possible

    • Examine both overexpression and knockdown/knockout effects

  • Pathway analysis:

    • Evaluate GPM6B in the context of relevant signaling pathways (TGF-β)

    • Consider interaction partners (TβRI) that may modulate function

  • Integration of multiple techniques:

    • Combine protein-level (WB, IF) and RNA-level (qPCR) analyses

    • Correlate functional outcomes with expression patterns

    • Use genetic approaches (CRISPR) to validate antibody-based findings

By integrating these approaches, researchers can develop a more nuanced understanding of GPM6B's context-dependent functions and reconcile apparently contradictory experimental results.

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.