HEPACAM Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze / thaw cycles.
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery times.
Synonyms
FLJ25530 antibody; GlialCAM antibody; HECAM_HUMAN antibody; HEPACAM antibody; Hepatocyte cell adhesion molecule antibody; Protein hepaCAM antibody
Target Names
HEPACAM
Uniprot No.

Target Background

Function
HEPACAM is involved in the regulation of cell motility and cell-matrix interactions. It may inhibit cell growth by suppressing cell proliferation.
Gene References Into Functions
  1. This study indicates that HepaCAM potentially represents a therapeutic target and PF3084014 might be a promising agent for the treatment of refractory prostate cancer. PMID: 29658567
  2. HepaCAM associates with connexin 43, a key component of gap junctions, and enhances connexin 43 localization to the plasma membrane at cellular junctions. PMID: 27819278
  3. This study revealed that hepaCAM was downregulated in CRC tissues and cell lines. Overexpression of hepaCAM inhibited CRC cell proliferation, migration, and invasion in vitro. Furthermore, the tumorigenesis assay showed that increased expression of hepaCAM suppressed CRC tumor growth and metastasis in vivo. PMID: 28244854
  4. Novel mutations in HEPACAM were identified in a group of Egyptian patients with megalencephalic leukoencephalopathy. PMID: 27389245
  5. DNMT1 up-regulation induced by IL-6/STAT3 signaling was essential for IL-6-mediated hepaCAM loss in renal cell carcinoma (RCC) cell lines ACHN and 769-P, while DNMT3b up-regulation was crucial for hepaCAM loss in A498. PMID: 28093267
  6. Out of 20 patients, macrocephaly, classic MRI features, motor development delay, and cognitive impairment were detected in 20 (100%), 20 (100%), 17 (85%), and 4 (20%) patients, respectively. 20 (100%) were clinically diagnosed with MLC. 19 (95%) were genetically diagnosed with 10 novel mutations in MLC1. MLC1 and GlialCAM mutations were identified in 15 and 4 patients, respectively. PMID: 27322623
  7. HepaCAM depletion was discovered in bladder cancer tissues compared with adjacent normal tissues, and the decreased level was associated with the degradation of FoxO3. PMID: 28229220
  8. HepaCAM proteins were significantly decreased in bladder carcinoma. Low hepaCAM was not statistically associated with clinicopathological characteristics of the patients. HepaCAM overexpression activated caspase 3/8/9, downregulated poly-ADP ribose polymerase and p-SMAD2/3, and decreased apoptosis. PMID: 26873485
  9. The suppressive roles of HEPACAM in NSCLC. PMID: 26392113
  10. Due to the ability to reactivate expression of hepaCAM and inhibit growth of bladder cancer cells, AZAC may represent an effective treatment for bladder cancer. PMID: 26677113
  11. The extracellular domain of GlialCAM is necessary for cell junction targeting and for mediating interactions with itself or with MLC1 and ClC-2. PMID: 26033718
  12. HepaCAM may prevent the translocation of PKCepsilon from cytosolic to particulate fractions, resulting in the inhibition of 786-0 cell proliferation. PMID: 24515280
  13. GlialCAM is able to interact with all CLC channels tested in this study, targeting them to cell junctions and activating them by stabilizing the open configuration of the common gate. PMID: 25185546
  14. Results allow classifying the effect of HEPACAM gene mutations in different subtypes and authors indicate different cellular mechanisms that lead to megalencephalic leukoencephalopathy pathogenesis. PMID: 25044933
  15. We demonstrate an evolutionary conserved role for MLC1 in regulating glial surface levels of GLIALCAM, and this interrelationship explains why patients with mutations in either gene (MLC1 or GLIALCAM) share the same clinical phenotype. PMID: 24824219
  16. High expression of hepaCAM is associated with renal carcinoma. PMID: 24645843
  17. Analysis of mutations in GLIALCAM in patients with megalencephalic leukoencephalopathy with subcortical cysts [case report]. PMID: 24202401
  18. Results suggested that HepaCAM acted as a tumor suppressor in prostate cancer. PMID: 24811146
  19. Re-activation of hepaCAM gene by 5-aza-CdR can inhibit growth of cancer cells and arrest cells at the G0/G1 phase. PMID: 24324362
  20. Results indicate GlialCAM is necessary for MLC1 protein expression, and its reduction affects the activity of volume-regulated anion currents (VRAC) which may cause astrocyte vacuolation; work extends the role of GlialCAM as a chaperone of MLC1 needed for proper VRAC activation. PMID: 23793458
  21. No clear association between GLIALCAM mutations and an autism-epilepsy phenotype. PMID: 24580998
  22. Results suggest an important connection between HEPACAM and interferon-gamma, which may inhibit BIU-87 proliferation through HEPACAM re-expression and p21(WAF1) up-regulation to arrest cells at the G(0)/G(1) phase. PMID: 22906662
  23. Research implies that the decrease in c-Myc protein expression, resulting from ectopic expression of hepaCAM, may contribute to the inhibition of proliferation in these cells. PMID: 21618595
  24. The study presents a more detailed characterization of the effect of mutations found in MLC1 and GLIALCAM in megalencephalic leukoencephalopathy with subcortical cysts. PMID: 21624973
  25. Dominant HEPACAM mutations can cause either macrocephaly and mental retardation with or without autism or benign familial macrocephaly. PMID: 21419380
  26. Downregulation of hepaCAM expression plays a significant role in the tumorigenesis and development of bladder cancer. PMID: 20628239
  27. There is a close relationship between hepaCAM and VEGF in urothelial carcinoma. PMID: 20593288
  28. The truncation mutant of hepaCAM failed to promote cell-ECM adhesion and migration, and lost the inhibitory effects on cell growth, suggesting a regulatory role of the cleavage in hepaCAM functions. PMID: 20514407
  29. HepaCAM is involved in cell adhesion and growth control, and its expression is frequently silenced in TCCB. The extracellular domain of hepaCAM is essential to its physiological and biological functions. PMID: 20205955
  30. Observational study of gene-disease association. (HuGE Navigator) PMID: 20237496
  31. HEPN1 is frequently silenced in HCC, and exogenous HEPN1 exhibits an antiproliferative effect on HepG2 cells, suggesting that silencing of HEPN1 may be associated with carcinogenesis of hepatocytes. PMID: 12971969
  32. Encodes an Ig-like transmembrane glycoprotein and is involved in cell adhesion and growth control. PMID: 15885354
  33. The cytoplasmic domain of hepaCAM is essential to its function on cell-matrix interaction and cell motility. PMID: 15917256
  34. The study examined the glycosylation of the GlialCAM (hepaCAM) extracellular domain expressed in HEK and CHO cells. PMID: 18082421
  35. GlialCAM, an immunoglobulin-like cell adhesion molecule, is expressed in glial cells of the central nervous system. PMID: 18293412
  36. The expression of hepaCAM in MCF7 cells not only inhibits cell growth but also induces cellular senescence through the p53/21 pathway. PMID: 18845560
  37. HepaCAM is partially localized in the lipid rafts/caveolae and interacts with Cav-1 through its first immunoglobulin domain. PMID: 19059381
  38. The data suggest that an intact hepaCAM protein is critical for establishing a stable physical association with the actin cytoskeleton; and such association is important for modulating hepaCAM-mediated cell adhesion and motility. PMID: 19142852
  39. High expression of hepaCAM significantly accelerated cell adhesion but inhibited cell proliferation and migration; cell differentiation was noticeably less apparent in cells expressing low-level hepaCAM. PMID: 19507233

Show More

Hide All

Database Links

HGNC: 26361

OMIM: 611642

KEGG: hsa:220296

STRING: 9606.ENSP00000298251

UniGene: Hs.745294

Involvement In Disease
Leukoencephalopathy, megalencephalic, with subcortical cysts, 2A (MLC2A); Leukoencephalopathy, megalencephalic, with subcortical cysts, 2B (MLC2B)
Subcellular Location
Cytoplasm. Membrane; Single-pass type I membrane protein; Cytoplasmic side. Note=In MCF-7 breast carcinoma and hepatic Hep 3B2.1-7 and Hep-G2 cell lines, localization of HEPACAM is cell density-dependent. In well spread cells, localized to punctate structures in the perinuclear membrane, cytoplasm, and at cell surface of protusions. In confluent cells, localized predominantly to the cytoplasmic membrane, particularly in areas of cell-cell contacts. Colocalizes with CDH1.

Q&A

What is HepaCAM and what role does it play in cancer research?

HepaCAM (Hepatocyte Cell Adhesion Molecule) is a member of the immunoglobulin superfamily primarily localized at the cell membrane. It consists of three domains: extracellular, transmembrane, and cytoplasmic, with the cytoplasmic domain being fundamental to its biological function . In cancer research, HepaCAM has emerged as a significant tumor suppressor that exhibits reduced expression or complete absence in various cancer tissues and cell lines . Research methodologies typically focus on examining HepaCAM's ability to inhibit cell proliferation, promote apoptosis, and suppress cancer cell invasion and migration .

How is HepaCAM expression regulated in different cancer types?

HepaCAM expression patterns vary significantly between cancer types and require specific methodological approaches for proper characterization. In prostate cancer, HepaCAM shows reduced expression compared to normal tissue . Conversely, in glioblastoma (GBM), TCGA database analysis has revealed significantly increased expression of HEPACAM mRNA compared to non-cancerous brain tissue . When analyzing HepaCAM regulation, researchers should employ both transcriptional (RT-PCR) and translational (Western blot) approaches, while considering tissue-specific expression patterns. Immunohistochemical analysis of normal human cortex shows enriched HepaCAM protein in astrocytes surrounding cerebral blood vessels, while GBM samples show robust expression throughout the tumor core with enrichment in perivascular tumor cells .

What experimental models are suitable for studying HepaCAM function?

Multiple experimental models have proven effective for investigating HepaCAM function:

Model SystemApplicationValidated By
Cancer cell lines (U373 MG, LNCaP, HEK293T)Protein interaction studies, cell proliferationWestern blot
Primary GBM stem cells (GSCs)Cancer stem cell biology, invasion studiesImmunoblotting, sphere formation assays
Xenograft mouse modelsIn vivo invasion and tumor growthImmunolabeling of fixed brain sections
Transfection modelsFunctional domain studiesCycloheximide chase assays

For knockdown studies, lentiviral shRNA approaches with appropriate controls have demonstrated efficiency between 30-70% reduction in HepaCAM expression, as validated by both immunoblotting and immunofluorescence labeling .

What are the optimal conditions for HepaCAM detection by Western blotting?

For optimal HepaCAM detection via Western blot, researchers should consider the following methodological parameters:

  • Sample preparation: Use PVDF membrane with appropriate reducing conditions and Immunoblot Buffer Group 1

  • Antibody concentration: 0.25 μg/mL of Anti-Human HepaCAM Monoclonal Antibody has demonstrated specific detection

  • Detection system: HRP-conjugated Secondary Antibody followed by appropriate chemiluminescent detection

  • Expected band pattern: HepaCAM typically appears as a specific band at approximately 45-70 kDa under reducing conditions

  • Loading controls: GAPDH serves as an effective loading control for normalizing HepaCAM expression

For quantification, densitometric analysis using ImageJ with normalization to housekeeping proteins yields reliable results for comparing expression levels across experimental conditions .

How can HepaCAM antibodies be effectively used in immunoprecipitation studies?

Successful immunoprecipitation of HepaCAM requires specific methodology:

  • Antibody selection: Use antibodies targeting the HepaCAM extracellular domain for efficient precipitation

  • Negative controls: Include mouse IgG1 immunoprecipitation as a negative control to evaluate non-specific binding

  • Validation approach: Evaluate immunoprecipitation efficiency using HRP-conjugated FLAG antibody when using tagged constructs

  • Loading controls: Detect IgG heavy chain with HRP-conjugated anti-mouse antibody as a procedural control

  • Co-immunoprecipitation: For protein interaction studies, use 2% input as reference and perform Western blot analysis on immunoprecipitates using antibodies against potential interacting proteins

This approach has successfully demonstrated HepaCAM's interaction with connexin 43 in multiple experimental systems .

What protocols are recommended for HepaCAM immunofluorescence studies?

For immunofluorescence detection of HepaCAM:

  • Fixation: Standard paraformaldehyde fixation is effective for most cell types

  • Antibody concentration: Anti-HepaCAM extracellular domain antibodies perform well at 10 μg/ml for both soluble treatment and immunostaining

  • Visualization: Confocal microscopy under a 60× objective provides optimal resolution for subcellular localization

  • Co-localization studies: For detecting protein interactions, dual-labeling with antibodies against HepaCAM (visualized in green) and potential interacting proteins like connexin 43 (visualized in red) effectively reveals co-localization (yellow fluorescence)

  • Nuclear counterstaining: DAPI effectively complements HepaCAM staining for determining relative subcellular localization

For in vivo studies, anti-GFP antibodies can be used for immunolabeling fixed brain sections when studying GFP-tagged HepaCAM-expressing cells .

How can researchers investigate HepaCAM's interaction with connexin 43?

Multiple complementary approaches can rigorously establish HepaCAM-connexin 43 interactions:

  • Co-immunoprecipitation: Using antibodies against HepaCAM's extracellular domain to precipitate protein complexes, followed by Western blot analysis with connexin 43 antibodies. Include appropriate IgG controls to confirm specificity .

  • Co-localization by immunofluorescence: Double-labeling with HepaCAM (green) and connexin 43 (red) antibodies to visualize co-localization at cell-cell contacts via confocal microscopy. Yellow fluorescence indicates protein interaction zones .

  • Expression correlation studies: Transfection of HepaCAM constructs followed by quantification of connexin 43 protein levels via Western blot. Data shows HepaCAM expression increases connexin 43 protein levels by approximately 2-fold (p<0.01) based on densitometric analysis .

  • Protein stability assays: Cycloheximide chase experiments comparing connexin 43 stability in cells with and without HepaCAM expression. This approach reveals HepaCAM's role in regulating connexin 43 stability rather than transcription, as confirmed by RT-PCR showing unchanged mRNA levels .

  • Functional perturbation: Treating HepaCAM-expressing cells with antibodies against the HepaCAM extracellular domain prevents the association of HepaCAM with connexin 43 at cell-cell contacts, providing further evidence of their interaction .

How can the cytoplasmic domain of HepaCAM be specifically studied?

The cytoplasmic domain of HepaCAM is critical for its biological function and can be studied through:

  • Domain-specific mutations: Creating constructs with targeted mutations or deletions in the cytoplasmic domain allows for functional characterization. In prostate cancer research, cytoplasmic domain studies have revealed its crucial role in cell proliferation, migration, and invasion .

  • Chimeric protein approaches: Generating fusion proteins containing only the cytoplasmic domain linked to reporter proteins can isolate domain-specific functions.

  • Interaction mapping: Identifying specific cytoplasmic domain residues that interact with other proteins (like AR or Ran) through site-directed mutagenesis followed by co-immunoprecipitation or pull-down assays .

  • Subcellular localization: Comparing wild-type HepaCAM with cytoplasmic domain mutants using immunofluorescence to determine how this domain affects protein localization and trafficking .

  • Pathway analysis: Investigating how the cytoplasmic domain specifically affects downstream signaling, such as the suppression of AR nuclear translocation via Ran in prostate cancer cells .

How does HepaCAM influence cancer cell proliferation and invasion?

HepaCAM demonstrates complex and sometimes context-dependent effects on cancer cell behavior that require multifaceted experimental approaches:

In Prostate Cancer:

  • HepaCAM functions as a tumor suppressor with reduced expression in PCa tissues

  • The cytoplasmic domain inhibits cell proliferation, migration and invasion as demonstrated through MTT, wound healing, and Transwell assays

  • Mechanistically, HepaCAM suppresses nuclear translocation of androgen receptor (AR) via interaction with Ran

In Glioblastoma:

  • Paradoxically, HEPACAM mRNA shows increased expression in GBM compared to non-cancerous brain

  • Knockdown approaches (shRNA with 70% reduction) reveal a dual role:

    • Reduced GSC growth and spheroid formation in vitro (40% smaller cross-sectional areas)

    • Enhanced invasion both in vitro and in vivo, with significantly increased numbers of invading cells in the corpus callosum (254 vs. 101 cells) and contralateral hemisphere (182 vs. 48 cells)

  • HepaCAM suppresses focal adhesion signaling as revealed by RPPA analysis, with knockdown leading to elevated levels of focal adhesion proteins like paxillin, p130Cas, and β1 integrin

These findings highlight the importance of employing multiple experimental approaches and considering tissue-specific contexts when studying HepaCAM's complex biological roles.

How can researchers assess HepaCAM's tumor suppressor functions?

To comprehensively assess HepaCAM's tumor suppressor functions, researchers should implement multiple complementary approaches:

  • Cell proliferation assays: MTT assays to measure cell viability and proliferation in cells with manipulated HepaCAM expression .

  • Migration analysis: Wound healing assays to quantify the rate of cell migration in response to HepaCAM expression or knockdown .

  • Invasion studies: Transwell assays with extracellular matrix components to evaluate invasive capacity .

  • In vivo models: Intracranial injection of cells with controlled HepaCAM expression into mice, followed by analysis of tumor growth and invasive patterns using immunofluorescence labeling .

  • Sphere formation assays: For cancer stem cells, quantifying spheroid formation and size over time (e.g., 7-day period) in response to HepaCAM manipulation .

  • Molecular pathway analysis: Western blotting to identify changes in relevant signaling pathways, such as focal adhesion proteins (paxillin, p130Cas) or nuclear translocation factors (AR, Ran) .

This multi-modal approach provides a comprehensive understanding of HepaCAM's tumor suppressor activities across different cancer contexts.

How can HEPACAM gene knockdown experiments be optimized?

Optimizing HEPACAM knockdown requires careful consideration of several methodological factors:

  • Vector selection: pGIPZ lentiviruses expressing GFP and shRNAs targeting HEPACAM provide efficient knockdown with the added benefit of tracking via GFP expression .

  • Multiple shRNA designs: Test multiple shRNA sequences targeting different regions of HEPACAM. In published research, three different shRNAs produced variable knockdown efficiencies (30-70% reduction) .

  • Validation methods: Confirm knockdown through both:

    • Immunoblotting of detergent-soluble cell lysates (protein-level validation)

    • Immunofluorescence labeling (provides cellular distribution information)

  • Quantification approach: Use densitometric analysis to precisely measure knockdown efficiency relative to non-targeting controls .

  • Appropriate controls: Include non-targeting (NT) shRNAs expressed in the same vector system to control for non-specific effects of viral infection and shRNA expression .

  • Functional validation: Confirm biological relevance of knockdown through functional assays specific to the research question (e.g., sphere formation assays for stem cell properties, invasion assays for metastatic potential) .

The highest reported knockdown efficiency (70% reduction with shRNA #3) provides a benchmark for successful HEPACAM silencing in research applications .

How should researchers approach apparent contradictions in HepaCAM's roles across different cancer types?

Reconciling contradictory findings regarding HepaCAM requires systematic methodological approaches:

  • Context-specific analysis: Compare HepaCAM expression and function across multiple cancer types using consistent methodologies. For example, HepaCAM shows reduced expression in prostate cancer but increased expression in GBM .

  • Microenvironment considerations: Analyze HepaCAM in relation to the tumor microenvironment. In GBM, HepaCAM knockdown leads to increased numbers of Iba1+ microglial cells and GFAP+ astrocytes in contralateral lesions, suggesting microenvironment influences .

  • Pathway comparison: Use techniques like RPPA to identify signaling pathway differences that might explain context-dependent roles. In GBM, HepaCAM suppresses focal adhesion signaling, while in prostate cancer it regulates AR nuclear translocation .

  • Domain-specific functions: Determine if different functional domains mediate different effects. The cytoplasmic domain is crucial for HepaCAM's tumor suppressor role in prostate cancer .

  • Cell-type specific effects: Compare HepaCAM function in differentiated cancer cells versus cancer stem cells, as they may respond differently. In GBM, HepaCAM knockdown reduces GSC growth but enhances invasion .

By systematically addressing these aspects, researchers can develop more nuanced models of HepaCAM function that account for its seemingly contradictory roles in different cancer contexts.

What emerging techniques can advance HepaCAM research in the tumor microenvironment?

Advanced techniques for studying HepaCAM in the tumor microenvironment include:

  • Multi-marker immunofluorescence: Combining HepaCAM antibodies with markers for microenvironmental components (CD31 for vessels, Iba1 for microglia, GFAP for astrocytes) to analyze spatial relationships within the tumor context .

  • Reverse-phase protein arrays (RPPA): This antibody-based high-throughput system can identify HepaCAM-mediated changes in multiple signaling cascades simultaneously, providing a comprehensive view of pathway alterations .

  • In vivo imaging techniques: Using GFP-labeled cells with HepaCAM manipulation to track invasion patterns and interactions with microenvironmental components in real-time .

  • 3D organoid cultures: Developing organoid models that incorporate both tumor cells and microenvironmental components to study HepaCAM function in a more physiologically relevant context.

  • Single-cell analysis approaches: Applying single-cell RNA sequencing or proteomics to characterize heterogeneous HepaCAM expression patterns and functions within tumor populations.

These emerging approaches can provide more nuanced insights into HepaCAM's complex roles within the tumor microenvironment context, potentially resolving contradictory findings observed in different experimental systems.

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.