CDC42EP4 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
We typically dispatch products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery time estimates.
Synonyms
CDC42EP4 antibody; BORG4 antibody; CEP4 antibody; Cdc42 effector protein 4 antibody; Binder of Rho GTPases 4 antibody
Target Names
CDC42EP4
Uniprot No.

Target Background

Function
CDC42EP4 is likely involved in the organization of the actin cytoskeleton. It may function downstream of CDC42 to induce actin filament assembly, leading to cell shape changes. Overexpression of CDC42EP4 in fibroblasts induces pseudopodia formation.
Gene References Into Functions
  1. A study found that CDC42EP4 messenger RNA levels were significantly upregulated in individuals with schizophrenia in both laminar and cellular samples. PMID: 25981171
Database Links

HGNC: 17147

OMIM: 605468

KEGG: hsa:23580

STRING: 9606.ENSP00000338258

UniGene: Hs.3903

Protein Families
BORG/CEP family
Subcellular Location
Endomembrane system; Peripheral membrane protein. Cytoplasm, cytoskeleton.
Tissue Specificity
Not detected in any of the adult tissues tested. May be expressed only in fetal or embryonic tissues.

Q&A

What is CDC42EP4 and what cellular functions does it regulate?

CDC42EP4 functions as a downstream effector protein of CDC42 that mediates critical cellular processes in multiple tissues. In the brain, CDC42EP4 is highly expressed in Bergmann glial cells of the cerebellum where it forms a unique perisynaptic scaffold with septin proteins . This scaffold facilitates the perisynaptic localization of GLAST (glutamate transporter) and optimizes glutamate buffering and clearance around synapses. In prostate tissue, CDC42EP4 acts as a tumor suppressor gene, inhibiting cancer cell proliferation, migration, and invasion through regulation of the ERK signaling pathway . The protein contains specific domains, including CRIB and BD3 (septin-binding) regions, which enable its interactions with binding partners and downstream effectors.

What are the key specificity considerations when selecting a CDC42EP4 antibody?

When selecting a CDC42EP4 antibody, researchers should consider epitope location within the protein structure. High-specificity antibodies targeting regions between the CRIB and BD3 (septin-binding) domains have been successfully employed in research applications . Researchers should verify antibody specificity through multiple validation techniques including western blotting against CDC42EP4-null tissues as negative controls, immunofluorescence with appropriate blocking controls, and immunoprecipitation followed by mass spectrometry to confirm target binding. The antibody should demonstrate minimal cross-reactivity with other CDC42EP family members (CDC42EP1/2/3/5) which share structural similarities but have distinct functions.

How does CDC42EP4 expression vary across different tissue types?

CDC42EP4 expression demonstrates significant tissue specificity based on immunoblot profiling. The protein shows highest expression in the cerebellum compared to other brain regions and non-neural tissues . Within the cerebellum, CDC42EP4 exhibits highly selective expression in Bergmann glial cells, with minimal expression in Purkinje cells as demonstrated by fluorescence in situ hybridization (FISH) and immunofluorescence studies . In prostate tissue, CDC42EP4 expression is significantly lower in cancerous tissues compared to normal prostate tissues, with expression levels inversely correlating with Gleason grade, suggesting its potential utility as a diagnostic marker . This differential expression pattern underscores the importance of selecting appropriate positive and negative control tissues when validating CDC42EP4 antibody specificity.

What are the optimal protocols for immunofluorescence staining using CDC42EP4 antibodies?

For optimal immunofluorescence staining with CDC42EP4 antibodies, tissue preparation and fixation methods significantly impact results. Paraformaldehyde fixation (4%) for 24 hours followed by proper antigen retrieval is recommended for brain tissue sections. For visualization in Bergmann glia, lower gain settings during microscopy are essential to observe the characteristic "hotspot" pattern of CDC42EP4 immunoreactivity, which appears as clusters interspersed along Purkinje cell dendrites and tightly apposed to dendritic spines . Double-labeling with calbindin (Purkinje cell marker) and GLAST (Bergmann glia marker) allows for precise localization of CDC42EP4 within specific cell types. For prostate tissue, optimization may require modified antigen retrieval methods to overcome potentially lower expression levels. Researchers should test several antibody dilutions (typically 1:100 to 1:1000) to determine optimal signal-to-noise ratio for their specific tissue and fixation method.

How can CDC42EP4 antibodies be effectively utilized in co-immunoprecipitation experiments?

For successful co-immunoprecipitation experiments with CDC42EP4 antibodies, researchers should begin with careful lysate preparation using detergent compositions that preserve protein-protein interactions. A mild lysis buffer containing 1% NP-40 or Triton X-100, 150mM NaCl, 50mM Tris-HCl (pH 7.5), and protease inhibitors is recommended. Immunoaffinity chromatography techniques have successfully identified CDC42EP4's binding partners, including nine septin subunits (SEPT2/3/4/5/6/7/8/10/11) . To validate results, reciprocal co-immunoprecipitation using antibodies against binding partners (such as SEPT4) to pull down CDC42EP4 provides stronger evidence of interaction. Both direct antibody coupling to beads and protein A/G approaches are effective, but direct coupling may reduce background from heavy chain interference in subsequent immunoblotting. Validation of results using CDC42EP4-null tissues as negative controls is essential to confirm specificity .

What techniques are recommended for quantifying CDC42EP4 protein levels in research samples?

Quantification of CDC42EP4 protein levels requires careful selection of detection methods and normalization strategies. Western blotting using fluorescent secondary antibodies rather than chemiluminescence enables more accurate quantification across a broader dynamic range. When analyzing CDC42EP4 in brain samples, normalization to a Bergmann glia-specific housekeeping protein rather than global housekeeping proteins provides more accurate relative quantification due to the cell-specific expression pattern . For prostate cancer samples, where CDC42EP4 expression varies significantly between normal and malignant tissues, researchers should establish a standard curve using recombinant CDC42EP4 protein for absolute quantification . ELISA-based methods can provide higher throughput quantification but require validation against western blot results to ensure comparable sensitivity and specificity. Careful sample preparation including phosphatase inhibitors is essential if analyzing the relationship between CDC42EP4 and phosphorylation-dependent pathways like ERK.

How can researchers address non-specific binding issues with CDC42EP4 antibodies?

Non-specific binding with CDC42EP4 antibodies can be minimized through several optimization strategies. Increasing blocking stringency by using 5-10% normal serum matched to the secondary antibody host species, combined with 1-3% BSA in PBS-T (0.1% Tween-20) for 1-2 hours at room temperature can reduce background. If non-specific bands persist in western blots, pre-adsorption of the primary antibody with recombinant CDC42EP4 protein can help determine which bands represent true CDC42EP4 signal. For immunohistochemistry applications, including an avidin/biotin blocking step if using biotinylated secondary antibodies reduces endogenous biotin-related background. The use of CDC42EP4-null tissues as negative controls provides the most definitive validation of antibody specificity . If cross-reactivity with other CDC42EP family members (CDC42EP1/2/3/5) is suspected, competitive binding assays with recombinant proteins can help identify the extent of cross-reactivity.

What are the key considerations when interpreting CDC42EP4 subcellular localization data?

Interpreting CDC42EP4 subcellular localization requires attention to several methodological considerations. In Bergmann glial cells, CDC42EP4 appears as submembranous clusters particularly concentrated around spine necks of Purkinje cells, with a decreasing gradient from the base (neck) to the apex (head) of spines . This distinctive localization pattern can only be accurately observed using high-resolution techniques such as silver-enhanced immunoelectron microscopy or super-resolution optical microscopy. Researchers should be cautious about fixation artifacts that can disrupt the native CDC42EP4 distribution pattern, particularly when examining membrane-associated pools. Co-localization studies with septin markers such as SEPT4/H5 can confirm proper detection of CDC42EP4 given their similar distribution patterns. For quantitative analysis of CDC42EP4 distribution, measuring the distance from synaptic contact sites provides meaningful data about its gradient distribution in perisynaptic processes .

How should researchers validate CDC42EP4 knockdown or overexpression experiments?

Validation of CDC42EP4 knockdown or overexpression requires multi-level confirmation using both protein and functional readouts. For knockdown validation, western blotting should demonstrate reduction of CDC42EP4 protein with off-target effects assessed by measuring other CDC42EP family members. Functional validation should include examination of known downstream pathways - in prostate cancer cells, ERK pathway activation (p-ERK1/2 levels) should increase with CDC42EP4 knockdown , while in glial cells, GLAST-septin interactions should decrease . For overexpression systems, researchers should verify that the expressed protein localizes correctly (perisynaptic localization in Bergmann glia or appropriate distribution in cancer cells) and interacts with expected binding partners (septins in brain tissue). Dose-dependent effects should be carefully assessed, as extremely high overexpression may lead to aggregation or non-physiological interactions. Rescue experiments in knockout/knockdown models provide the strongest validation that phenotypic effects are specifically due to CDC42EP4 modulation.

How does CDC42EP4 interact with the septin cytoskeleton, and how can these interactions be studied?

CDC42EP4 serves as a critical molecular bridge between CDC42 signaling and the septin cytoskeleton, particularly in Bergmann glial cells. Proteomic analysis of CDC42EP4 binding partners identified nine septin subunits (SEPT2/3/4/5/6/7/8/10/11) as major interaction partners . These interactions can be studied through multiple complementary approaches. Immunoaffinity chromatography coupled with mass spectrometry provides unbiased identification of septin binding partners as shown in Table 1:

Identified proteinsPeptide numberSequence coverage (%)
KO IgGKO ep4WT IgGWT ep4KO IgGKO ep4WT IgGWT ep4
CDC42EP40002000063
SEPT70102102.1045
SEPT40101901.9043
SEPT110001400034
SEPT50001200042
SEPT20001100047

Co-immunoprecipitation experiments followed by western blotting can validate these interactions and assess how they are affected by CDC42EP4 manipulations. Additionally, fluorescence resonance energy transfer (FRET) or proximity ligation assays (PLA) provide powerful tools to study CDC42EP4-septin interactions in intact cells with spatial resolution. Mutation studies targeting the septin-binding domain (BD3) can help determine which regions of CDC42EP4 are essential for septin binding. Interestingly, while CDC42EP4 interacts strongly with septins, CDC42 itself was not detected in pull-down experiments, suggesting that CDC42EP4 may function independently of continuous CDC42 binding after initial activation .

What are the emerging roles of CDC42EP4 in cancer biology beyond prostate cancer?

While CDC42EP4 has been well-characterized as a tumor suppressor in prostate cancer , emerging evidence suggests broader roles in cancer biology that merit investigation using CDC42EP4 antibodies. In prostate cancer, CDC42EP4 inhibits cell proliferation, migration, and invasion through suppression of the ERK pathway, with CDC42EP4 expression negatively correlating with cancer progression . Researchers investigating CDC42EP4 in other cancer types should examine its effects on epithelial-mesenchymal transition (EMT) markers, as CDC42EP4 overexpression upregulates E-cadherin while downregulating N-cadherin in prostate cancer cells . The relationship between CDC42EP4 and the tumor microenvironment represents a promising research direction, particularly given its role in organizing cell-cell interfaces in normal tissues. CDC42EP4 antibodies can facilitate investigation of potential associations between CDC42EP4 expression patterns and clinical outcomes, cancer stem cell populations, or therapy resistance mechanisms across diverse cancer types. Comparative analysis of CDC42EP4 function between cancer types may reveal tissue-specific regulatory mechanisms.

How does phosphorylation affect CDC42EP4 function, and what antibodies can detect these modifications?

Phosphorylation likely plays a critical role in regulating CDC42EP4 function, though this area remains underexplored. While standard CDC42EP4 antibodies detect total protein levels, phospho-specific antibodies would be required to detect post-translational modifications that may regulate its activity or interactions. Researchers investigating CDC42EP4 phosphorylation should first conduct in silico analysis to identify potential phosphorylation sites, followed by mass spectrometry validation of these modifications in vivo. The relationship between CDC42EP4 and the ERK pathway in prostate cancer cells suggests potential regulatory phosphorylation events, as CDC42EP4 inhibits ERK phosphorylation without altering total ERK levels . Development of phospho-specific CDC42EP4 antibodies would enable investigation of how these modifications affect septin binding, GLAST interaction, or subcellular localization. Kinase inhibitor screens coupled with phospho-specific antibodies could identify upstream regulators of CDC42EP4 phosphorylation, providing new insights into its regulation. Mutation of key phosphorylation sites to phosphomimetic or non-phosphorylatable residues, coupled with antibody detection, would help establish the functional significance of these modifications.

How do CDC42EP4 antibodies facilitate research into glutamate transport and excitotoxicity?

CDC42EP4 antibodies provide essential tools for investigating the protein's role in glutamate homeostasis, particularly in cerebellar function. In CDC42EP4-null mice, GLAST (glutamate transporter) dissociates from septins and becomes delocalized away from parallel fiber-Purkinje cell synapses, leading to impaired glutamate clearance . Researchers can use CDC42EP4 antibodies in combination with GLAST antibodies to study their co-localization in normal and pathological conditions. For quantitative analysis of CDC42EP4-dependent glutamate transport, antibody-based proximity ligation assays can measure direct interactions between CDC42EP4, septins, and GLAST in tissue sections or primary cell cultures. The role of CDC42EP4 in protecting against excitotoxicity can be assessed using antibodies to measure its expression changes following ischemic or excitotoxic insults. Additionally, CDC42EP4 antibodies enable investigation of potential dysregulation in neurological disorders associated with glutamate transport abnormalities, such as spinocerebellar ataxias, where altered CDC42EP4-septin-GLAST interactions might contribute to pathogenesis.

What approaches can be used to study CDC42EP4's role in prostate cancer progression?

CDC42EP4 antibodies enable multifaceted investigation of its tumor suppressor functions in prostate cancer. Immunohistochemical analysis of tissue microarrays using validated CDC42EP4 antibodies can quantify expression across different Gleason grades and correlate with clinical outcomes . For mechanistic studies, researchers can combine CDC42EP4 antibodies with antibodies against ERK pathway components to analyze how CDC42EP4 regulates this signaling cascade. Co-immunoprecipitation experiments can identify novel CDC42EP4 binding partners in prostate cells that might mediate its tumor suppressive functions. Live-cell imaging studies incorporating fluorescently-tagged CDC42EP4 antibody fragments can track dynamic changes in CDC42EP4 localization during cell migration and invasion processes. For translational applications, CDC42EP4 antibodies could help evaluate whether restoring CDC42EP4 expression sensitizes prostate cancer cells to existing therapies, particularly those targeting the ERK pathway. Animal studies using CDC42EP4 antibodies for tissue analysis can validate findings from xenograft models showing that CDC42EP4 knockdown promotes tumor growth in vivo .

How can researchers distinguish between CDC42EP4 and other CDC42 effector proteins in experimental systems?

Distinguishing CDC42EP4 from other CDC42 effector proteins (CDC42EP1/2/3/5) requires careful antibody selection and experimental design. Antibodies targeting unique regions outside the conserved CRIB domain provide the highest specificity. Western blotting with recombinant CDC42EP family proteins can establish cross-reactivity profiles for each antibody. For immunoprecipitation experiments, researchers should validate pull-downs by mass spectrometry to confirm protein identity rather than relying solely on molecular weight. In tissue-specific studies, knowledge of differential expression patterns helps interpretation - CDC42EP4 shows highest expression in cerebellum with selective localization to Bergmann glia , while other family members may predominate in different cell types. Knockout or knockdown validation experiments are essential when introducing CDC42EP4 antibodies to new experimental systems. When studying CDC42EP4's unique functions in septin organization or ERK pathway regulation, researchers should include appropriate positive controls demonstrating these specific interactions rather than general CDC42 binding. To confidently attribute phenotypes to CDC42EP4 specifically, rescue experiments should demonstrate that reintroduction of CDC42EP4, but not other family members, restores normal function.

How might CDC42EP4 antibodies contribute to therapeutic development for neurological disorders?

CDC42EP4 antibodies could accelerate therapeutic development for neurological disorders through several research approaches. Given CDC42EP4's role in maintaining proper glutamate transport through septin-GLAST interactions in Bergmann glia , therapeutic strategies targeting this pathway might improve glutamate clearance in conditions characterized by excitotoxicity. Researchers can use CDC42EP4 antibodies to screen compound libraries for molecules that enhance CDC42EP4-septin interactions or stabilize the CDC42EP4/septin/GLAST complex. For disorders involving cerebellar dysfunction, such as spinocerebellar ataxias, monitoring CDC42EP4 expression and localization using specific antibodies could identify early pathological changes before symptom onset. Development of conformation-specific antibodies that distinguish active versus inactive CDC42EP4 states would enable more nuanced understanding of its dysfunction in disease states. CDC42EP4 antibodies conjugated to nanoparticles or cell-penetrating peptides might eventually be developed into therapeutic agents themselves, potentially helping deliver corrective molecules to restore CDC42EP4 function in affected tissues.

What novel insights might be gained from studying CDC42EP4 across different model organisms?

Comparative studies of CDC42EP4 across model organisms could reveal evolutionarily conserved functions and species-specific adaptations. CDC42EP4 antibodies that recognize conserved epitopes would enable cross-species analysis of expression patterns, subcellular localization, and binding partners. In zebrafish models, where real-time imaging of developing neural circuits is possible, CDC42EP4 antibodies could track its expression during cerebellar development and glial-neuronal interaction establishment. Drosophila models might reveal previously unknown functions of CDC42EP4 homologs in non-mammalian septins or glutamate signaling pathways. For larger animal models more closely resembling human neuroanatomy, CDC42EP4 antibodies could help determine whether the protein's role in glutamate homeostasis extends beyond cerebellum to other brain regions. Comparative oncology studies using CDC42EP4 antibodies might identify whether its tumor suppressor function in prostate cancer is conserved in naturally occurring canine prostate tumors. This cross-species approach would provide broader context for understanding CDC42EP4's fundamental biological functions and how they may have been adapted during evolution.

How can advanced microscopy techniques enhance CDC42EP4 antibody-based research?

Advanced microscopy techniques can significantly enhance CDC42EP4 antibody-based research by providing unprecedented spatial and temporal resolution. Super-resolution microscopy approaches such as STORM or PALM, combined with CDC42EP4 antibodies, can reveal nanoscale organization of CDC42EP4 clusters at perisynaptic membranes that conventional microscopy cannot resolve. The characteristic gradient distribution of CDC42EP4 from spine neck to head could be precisely quantified using these techniques. Multi-color STED microscopy with CDC42EP4 and septin antibodies would provide detailed visualization of their co-assembly structures without diffraction limitations. For live-cell applications, development of recombinant antibody fragments (Fabs) labeled with bright, photostable fluorophores could track CDC42EP4 dynamics during cellular processes like septin filament assembly or glutamate transporter redistribution. Correlative light and electron microscopy (CLEM) using CDC42EP4 antibodies would bridge the resolution gap between optical and ultrastructural data, enabling precise localization of CDC42EP4 relative to synaptic structures and glial processes. These advanced imaging approaches would provide mechanistic insights into CDC42EP4 function that conventional antibody applications cannot achieve.

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.