The ASPH antibody is a laboratory-generated immunoglobulin targeting aspartate β-hydroxylase (ASPH), an enzyme encoded by the ASPH gene on human chromosome 8. ASPH is a member of the α-ketoglutarate-dependent hydroxylase family and plays roles in post-translational modification of proteins, including hydroxylation of aspartyl/asparaginyl residues in epidermal growth factor-like domains . Overexpression of ASPH is strongly associated with malignant transformation, metastasis, and poor prognosis in over 20 solid and hematologic cancers, making it a critical biomarker and therapeutic target . ASPH antibodies enable researchers to detect, quantify, and study the functional and clinical implications of ASPH expression in disease models and patient samples .
ASPH antibodies are utilized in diverse experimental workflows:
Western Blot (WB): Detects ASPH at 1:200–1:1000 dilution in tissue lysates (e.g., human brain, mouse liver) .
Immunohistochemistry (IHC): Identifies ASPH overexpression in tumor biopsies at 1:50–1:500 dilution, with antigen retrieval using TE/citrate buffer .
Enzyme-Linked Immunosorbent Assay (ELISA): Quantifies ASPH levels in serum or cell supernatants for cancer biomarker studies .
Immunotherapy Development: Guides ADC (antibody-drug conjugate) and CAR T-cell therapy design by confirming cell-surface ASPH expression .
ASPH overexpression is a hallmark of aggressive malignancies:
ASPH antibodies are critical for stratifying patients eligible for ASPH-targeted therapies and monitoring treatment response .
ASPH antibodies underpin multiple therapeutic strategies:
Vaccines: Anti-ASPH nanoparticle vaccines (e.g., SNS-301) induce T-cell responses in prostate cancer trials .
Antibody-Drug Conjugates (ADCs): Radiolabeled or toxin-linked ASPH antibodies show efficacy in preclinical AML and breast cancer models .
Small-Molecule Inhibitors (SMIs): MO-I-1100 inhibits ASPH enzymatic activity, reducing tumor cell motility in pancreatic cancer and glioblastoma .
Current research focuses on:
ASPH antibodies are commonly used in several research applications:
Western Blotting (WB): For detection of ASPH protein expression levels in tissue and cell lysates.
Immunohistochemistry (IHC): For visualization of ASPH expression and localization in tissue sections.
ELISA: For quantitative measurement of ASPH in biological samples.
Knockout/Knockdown validation: For confirming the specificity of antibody and evaluating functional changes.
The recommended dilution ranges for different applications are:
| Application | Dilution |
|---|---|
| Western Blot (WB) | 1:200-1:1000 |
| Immunohistochemistry (IHC) | 1:50-1:500 |
It is recommended that researchers titrate the antibody in each testing system to obtain optimal results, as optimal conditions can be sample-dependent .
ASPH antibodies show different reactivity profiles depending on their design and production. For example, the Proteintech antibody 14116-1-AP has been tested and confirmed to react with human and mouse samples in Western blot and immunohistochemistry applications. Literature also cites reactivity with rat samples . When selecting an ASPH antibody for your research, it's important to verify that it has been validated in your species of interest and application.
ASPH antibodies, like the Proteintech 14116-1-AP, are typically stored in PBS with 0.02% sodium azide and 50% glycerol at pH 7.3. The recommended storage temperature is -20°C, where the antibody remains stable for one year after shipment. For 20 μl size preparations, they often contain 0.1% BSA. Aliquoting is generally unnecessary for -20°C storage . Proper storage ensures antibody stability and consistent performance across experiments.
ASPH hydroxylase activity plays a critical role in promoting cancer cell migration and invasion. Research has shown that specific blockade of ASPH hydroxylase inhibits hepatocellular carcinoma (HCC) cell migration. A polyclonal antibody (FE1) against the Fe-binding His-2 motif at the C-terminal of ASPH, which is a key region for hydroxylase activity, was able to neutralize the hydroxylase activity of ASPH .
In experimental studies, FE1 displayed a dose-dependent inhibitory effect on cell migration in Huh-7 cells that overexpressed wild-type ASPH, with an IC50 of approximately 100 μg/ml resulting in 80% enzymatic inhibition. Importantly, FE1 did not block cell migration in Huh-7 cells overexpressing the enzymatic mutant H679A variant of ASPH, demonstrating the specific dependence on hydroxylase activity .
The relationship between hydroxylase activity and cell migration was further confirmed by the observation that FE1 inhibited cell migration in EHBC-512 cells (which express membrane ASPH) in a dose-dependent manner but was ineffective in MHCC-97L cells (which have low levels of membrane ASPH) .
ASPH has been shown to activate the Notch signaling pathway, which plays a crucial role in tumor development and progression. In pancreatic cancer (PC), ASPH overexpression promotes proliferation, migration, invasion, and malignant transformation of cancer cells through multiple signaling pathways, including Notch .
The relationship between ASPH and Notch signaling was highlighted in a study by Dong et al., which reported that MO-I-1100, a small molecule inhibitor (SMI) of β-hydroxylase, reduced ASPH activity by 80%, inhibited ASPH-induced proliferation, migration, invasion, and colony formation, and suppressed Notch signaling in pancreatic cancer .
Similarly, Sturla et al. demonstrated that SMIs MO-I-1100 and MO-I-1151 significantly reduced viability and directional motility of glioblastoma multiforme (GBM) cells while suppressing Notch activation, confirming the role of ASPH in these processes .
Mass spectrometry and immunoprecipitation studies have revealed that ASPH interacts directly with vimentin, an intermediate filament protein involved in cell migration. In pull-down assays, vimentin peptides were detected in protein complexes precipitated by anti-Flag antibodies in cells overexpressing FLAG-tagged ASPH. Conversely, ASPH peptides were observed in protein complexes from pull-down assays of HA-tagged vimentin .
The interaction between endogenous ASPH and vimentin was further confirmed by reciprocal immunoprecipitation and immunoblot using vimentin and ASPH antibodies in MHCC-97L cells .
Functional studies demonstrated that:
Overexpression of ASPH enhanced the expression of vimentin and vice versa
Downregulation of ASPH slightly decreased vimentin expression
Overexpression of vimentin greatly promoted HCC cell migration
Knockdown of vimentin inhibited cell migration
Overexpression of vimentin effectively reversed the inhibitory effect on cell migration caused by ASPH knockdown
ASPH overexpression failed to enhance cell migration when vimentin was silenced
These findings suggest that vimentin is a critical mediator of ASPH-induced cell migration in HCC.
ASPH has emerged as an important biological target for controlling tumor cell migration and invasion, as its overexpression has been observed in 70-90% of human tumors. Several therapeutic approaches targeting ASPH are being investigated:
Immunotherapy: ASPH can be used as a tumor-associated antigen (TAA) as it transfers from the endoplasmic reticulum to the plasma membrane in tumor cells, exposing it to the extracellular environment. ASPH-loaded dendritic cells (DCs) have shown substantial anti-tumor effects in HCC models, activating both CD4+ T cells and CD8+ cytotoxic T cells .
Small Molecule Inhibitors (SMIs): MO-I-1100, an SMI of β-hydroxylase, reduced ASPH activity by 80% and inhibited ASPH-induced proliferation, migration, invasion, and colony formation in pancreatic cancer. Similarly, SMIs MO-I-1100 and MO-I-1151 significantly reduced viability and directional motility of glioblastoma multiforme cells .
Monoclonal Antibodies: Radiolabeled human monoclonal antibody (mAb) PAN-622 targeting ASPH on the surface of cancer cells has shown promise in imaging and potentially treating metastatic breast cancer. Furthermore, mAb against the ASPH C-terminal (ASPH-C) increased antibody-dependent cellular cytotoxicity of NK cells against HeLa, MCF-7, and HepG2 cells .
RNA Interference: Antisense oligodeoxynucleotides inhibiting ASPH expression significantly reduced the motility of cholangiocarcinoma cells. Small interfering RNAs (siRNAs) targeting exon 2 of the ASPH gene inhibited ASPH expression and reduced directional motility in HCC cells .
For optimal Western blotting results with ASPH antibody:
Sample Preparation:
Total protein extraction from tissues (brain, kidney) or cell lines (A549, HCC cell lines)
Include protease inhibitors to prevent degradation
Denature samples in loading buffer with reducing agent
Gel Electrophoresis and Transfer:
Use 8-10% SDS-PAGE gels to effectively separate proteins in the 26-141 kDa range
Transfer to PVDF or nitrocellulose membrane at 100V for 60-90 minutes
Blocking and Antibody Incubation:
Block with 5% non-fat milk or BSA in TBST for 1 hour at room temperature
Incubate with primary ASPH antibody (e.g., 14116-1-AP) at 1:200-1:1000 dilution in blocking buffer overnight at 4°C
Wash 3 times with TBST, 5 minutes each
Incubate with HRP-conjugated secondary antibody at appropriate dilution for 1 hour at room temperature
Wash 3 times with TBST, 5 minutes each
Detection:
Controls:
Include positive controls such as mouse brain tissue, A549 cells, or human brain tissue
Consider using ASPH knockdown/knockout cells as negative controls
For optimal immunohistochemistry with ASPH antibody:
Tissue Preparation:
Fix tissues in 10% neutral buffered formalin
Embed in paraffin and section at 4-5 μm thickness
Antigen Retrieval:
Blocking and Antibody Incubation:
Block endogenous peroxidase with 3% H₂O₂
Block non-specific binding with 5-10% normal serum
Incubate with ASPH antibody at 1:50-1:500 dilution overnight at 4°C
Wash with PBS
Incubate with appropriate secondary antibody
Develop with DAB substrate and counterstain with hematoxylin
Controls and Validation:
Analysis:
Evaluate staining pattern, intensity, and localization
ASPH is typically localized to the cytoplasm or membrane in cancer cells
Distinguishing between membrane and cytoplasmic ASPH expression is important in cancer research since ASPH's translocation to the cell membrane is associated with malignancy. Here are methodological approaches:
Immunofluorescence without permeabilization:
Flow cytometry:
Cell fractionation and Western blot:
Separate membrane and cytoplasmic fractions
Perform Western blot with ASPH antibody on each fraction
Use appropriate markers for each fraction (e.g., Na+/K+-ATPase for membrane, GAPDH for cytoplasm)
Confocal microscopy:
Use Z-stack imaging to distinguish membrane from cytoplasmic localization
Co-stain with membrane markers for colocalization analysis
To ensure the specificity of ASPH antibodies and validate experimental results:
ASPH Knockdown/Knockout Controls:
Use RNA interference (siRNA or shRNA) to knockdown ASPH
The search results indicate that siRNAs targeting exon 2 of the ASPH gene effectively inhibited ASPH expression
CRISPR/Cas9-mediated knockout can provide complete absence of the target protein
Perform Western blot or immunostaining to confirm absence or reduction of signal
Peptide Competition Assays:
Domain-Specific Antibody Testing:
Recombinant Protein Expression:
Overexpress tagged ASPH in cell lines with low endogenous expression
Confirm antibody detection of the overexpressed protein
Multi-technique Validation:
Verify consistent results across different techniques (Western blot, IHC, IF)
Confirm expected molecular weight and subcellular localization
Measuring ASPH hydroxylase activity is crucial for studies investigating its role in cancer progression. Several approaches can be employed:
α-Ketoglutarate (α-KG) Consumption Assay:
Site-directed Mutagenesis:
Inhibitor Studies:
Mass Spectrometry:
Detect hydroxylated versus non-hydroxylated peptides containing aspartate or asparagine residues
Functional Readouts:
Cell migration assays as indirect measures of hydroxylase activity
Notch signaling activation as a downstream consequence
When designing experiments to study ASPH in cancer models, researchers should consider the following:
Cell Line Selection:
Expression System Design:
Functional Assays:
Migration assays to assess metastatic potential
Proliferation and colony formation assays
Invasion assays using Matrigel
Signaling pathway analysis (especially Notch pathway components)
Interaction Studies:
In Vivo Models:
Understanding the correlation between ASPH expression and clinical outcomes is important for translational research. The available research indicates:
Hepatocellular Carcinoma (HCC):
Pancreatic Cancer:
Other Cancers:
When designing studies to investigate clinical correlations, researchers should:
Use tissue microarrays with adequate sample sizes
Employ multiple antibodies or detection methods
Correlate expression with established clinical parameters and outcomes
Consider both expression levels and subcellular localization
Use multivariate analysis to account for confounding factors
ASPH can appear at different molecular weights in Western blots due to several factors:
Multiple Isoforms and Post-translational Modifications:
Technical Factors:
Incomplete denaturation can cause abnormal migration
Different gel concentrations can affect apparent molecular weight
Insufficient blocking can lead to non-specific bands
Antibody Specificity:
Different antibodies may recognize different domains or isoforms
For example, antibodies targeting the N-terminal versus C-terminal of ASPH may detect different forms
Tissue/Cell Type Variations:
Different tissues or cell lines may express different ASPH isoforms
Cancer cells often have altered expression patterns compared to normal cells
To address these variations, researchers should:
Run appropriate molecular weight markers
Include positive control samples with known ASPH expression
Consider using multiple antibodies targeting different epitopes
Validate bands using ASPH knockdown or knockout controls
When faced with discrepancies between different ASPH antibodies:
Review Antibody Specifications:
Check the epitope/immunogen for each antibody
Determine if antibodies target different domains (N-terminal vs. C-terminal)
The research results show that different antibodies can recognize different forms of ASPH; for example, FE1 (targeting the Fe-binding domain) only recognized wild-type ASPH, while antibodies targeting the N-terminal recognized both wild-type and mutant forms
Validate with Genetic Approaches:
Use ASPH knockdown or knockout controls
Overexpress ASPH in null backgrounds
Use domain-specific mutants to map epitopes
Employ Multiple Detection Methods:
Compare results from Western blot, IHC, and immunofluorescence
Different techniques may provide complementary information
Consider Post-translational Modifications:
Investigate if discrepancies might be due to detection of different modified forms
Use phosphatase treatment to remove phosphorylation if relevant
Consult Literature and Technical Support:
Review published validations for each antibody
Contact antibody manufacturers for technical assistance
Share your experimental conditions for more targeted advice