ASPH Human

Aspartate Beta-Hydroxylase Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to ASPH Human

ASPH Human (Aspartyl/asparaginyl beta-hydroxylase), encoded by the ASPH gene, is a type II transmembrane enzyme belonging to the α-ketoglutarate-dependent dioxygenase family. It catalyzes the β-hydroxylation of aspartic acid (Asp) and asparagine (Asn) residues within epidermal growth factor-like domains (EGFDs) of proteins, including Notch signaling components . This post-translational modification regulates calcium storage, cell migration, and signaling pathways critical to development and disease .

Oncology

ASPH overexpression is strongly linked to malignancy:

  • Solid Tumors: Elevated in hepatocellular carcinoma (HCC), pancreatic, colorectal, lung, and breast cancers. Correlates with metastasis and poor prognosis .

  • Hematologic Malignancies: Expressed on blasts in ~40% of acute myeloid leukemia (AML) cases, with potential as a therapeutic target .

  • Chondrosarcoma: High ASPH expression predicts metastasis and death. Inhibition reduces tumor growth and lung metastasis in preclinical models .

Genetic Disorders

  • Traboulsi Syndrome: Mutations in ASPH cause facial dysmorphism, lens dislocation, and anterior-segment abnormalities. ASPH knockout mice replicate these phenotypes .

Bone Metabolism

  • Osteogenic Differentiation: Regulates bone formation via Runx2 and Col1a1 expression. Downregulation impairs osteogenesis in bone marrow stromal cells (BMSCs) .

  • Aging and Senescence: Reduced ASPH expression in elderly individuals correlates with age-related bone loss and cellular senescence .

Small-Molecule Inhibitors

High-throughput screens identified potent ASPH inhibitors, including natural products and repurposed therapeutics (e.g., vadadustat) . A synthetic thioether substrate mimic enabled robust inhibition assays with Z’-factors >0.7 .

Inhibitor TypeExamplesMechanismPreclinical Efficacy
Natural ProductsCompetitive EGFD binding
SyntheticVadadustat, 2,4-PDCAFe(II)-chelation, substrate mimicryReduced tumor growth, MMP activity

Immunotherapies

  • Nanoparticle Vaccines: SNS-301 (anti-ASPH) is in Phase 1 trials for prostate cancer, inducing CD4+/CD8+ T-cell responses .

  • Antibody-Drug Conjugates (ADCs): Radiolabeled or cytotoxin-linked anti-ASPH antibodies target AML blasts .

Cancer Biomarker Potential

ASPH expression predicts metastatic risk in chondrosarcoma and serves as a surface antigen for immunotherapies . In AML, higher expression occurs in monoblasts vs. myeloblasts, with racial disparities noted in African American patients .

Preclinical Models

  • Chondrosarcoma Xenografts: ASPH inhibition reduces tumor growth by 50% and lung metastasis burden .

  • BMSCs: Overexpression enhances osteogenesis, while knockdown reduces Runx2 and Col1a1 expression .

Product Specs

Introduction
ASPH, an enzyme involved in calcium homeostasis, hydroxylates Asp or Asn residues within EGF domains. The expression of ASPH is regulated by two promoters and undergoes extensive alternative splicing, resulting in a variety of ASPH protein isoforms. These isoforms share similar N-terminal regions but exhibit significant variations in their C-terminal domains, leading to distinct functional properties. The longest isoforms (a and f) possess a C-terminal Aspartyl/Asparaginyl beta-hydroxylase domain responsible for hydroxylating aspartic acid or asparagine residues in EGF domains of various proteins, including protein C, coagulation factors VII, IX, and X, and complement factors C1R and C1S. Other isoforms differ primarily in their C-terminal sequences, lack the hydroxylase domain, and some localize to the endoplasmic reticulum and sarcoplasmic reticulum.
Description
Recombinant human ASPH, expressed in E. coli, is a single, non-glycosylated polypeptide chain. This protein consists of 217 amino acids (residues 75-270), has a molecular weight of 24.5 kDa, and includes a 20 amino acid His Tag. Purification is achieved through standard chromatographic techniques.
Physical Appearance
A sterile, colorless solution.
Formulation
The ASPH protein solution is formulated in 20mM Tris-HCl (pH 8.0), 1mM DTT, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product should be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure optimal long-term stability, adding a carrier protein (0.1% HSA or BSA) is advised. Repeated freezing and thawing cycles should be avoided.
Purity
Purity is determined to be greater than 90.0% using SDS-PAGE analysis.
Synonyms
AAH, BAH, CASQ2BP1, HAAH, JCTN, Junctin, EC 1.14.11.16, Aspartyl/asparaginyl beta-hydroxylase, Aspartate beta-hydroxylase, Peptide-aspartate beta-dioxygenase, ASP beta-hydroxylase, ASPH.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MFDLVDYEEV LGKLGIYDAD GDGDFDVDDA KVLLGLKERS TSEPAVPPEE AEPHTEPEEQ VPVEAEPQNI EDEAKEQIQS LLHEMVHAEH ETEHSYHVEE TVSQDCNQDM EEMMSEQENP DSSEPVVEDE RLHHDTDDVT YQVYEEQAVY EPLENEGIEI TEVTAPPEDN PVEDSQVIVE EVSIFPVEEQ QEVPPDT.

Q&A

What is ASPH and what is its normal expression pattern in human tissues?

Aspartate β-hydroxylase (ASPH) is a transmembrane protein that catalyzes the hydroxylation of aspartate and asparagine residues in epidermal growth factor-like domains (EGFDs) of various substrates . In normal adult tissues, ASPH is rarely expressed, with the notable exception of placental trophoblastic cells . This limited expression pattern in healthy adult tissues makes ASPH particularly interesting as a potential biomarker and therapeutic target in pathological conditions where its expression becomes dysregulated.

The protein's normal function appears to be developmentally regulated, with higher expression during embryogenesis followed by significant downregulation in most adult tissues. This expression pattern suggests ASPH plays important roles in cellular differentiation and tissue development that are largely unnecessary in fully differentiated adult tissues.

How is ASPH expression regulated in human cells?

ASPH expression is regulated through several signaling cascades, with insulin/insulin-like growth factor 1 (IGF-1) signaling pathways playing a central role. Research has demonstrated that stimulation with insulin and IGF-1 increases ASPH mRNA and protein expression in human cell lines . This upregulation is mediated primarily through:

  • The ERK/MAPK (extracellular signal-regulated kinase/mitogen-activated protein kinase) pathway

  • The PI3K/Akt (phosphatidylinositol 3-kinase/protein kinase B) pathway

Specifically, insulin receptor substrate 1 (IRS1)-mediated signaling has been identified as an upstream regulator of ASPH expression . These findings highlight how growth factor signaling can drive ASPH expression, which becomes particularly relevant in understanding its pathological overexpression in cancer contexts.

What is the relationship between ASPH and human malignancies?

ASPH overexpression has been documented in numerous human malignancies, occurring in approximately 70-90% of human tumors . Specifically, elevated ASPH expression has been observed in:

  • Hepatocellular carcinoma (HCC)

  • Cholangiocarcinoma

  • Pancreatic cancer

  • Lung cancer

  • Breast cancer

  • Colon cancer

  • Neoplasms of the nervous system

In hepatocellular carcinoma patients, ASPH overexpression significantly correlates with higher recurrence rates and lower survival rates following surgical intervention . This association is particularly pronounced in early-stage HCC patients, suggesting ASPH may serve as a prognostic biomarker. In pancreatic cancer, ASPH overexpression promotes proliferation, migration, invasion, and malignant transformation through multiple signaling pathways .

What techniques are currently used to measure ASPH activity in research settings?

Recent methodological advances have significantly improved our ability to assess ASPH activity in research settings. A notable development is the solid-phase extraction mass spectrometry-based high-throughput ASPH inhibition assay . This technique employs:

  • A stable synthetic thioether mimic of ASPH substrates

  • Solid-phase extraction methodology

  • Mass spectrometry detection

This assay demonstrates excellent robustness with high Z'-factors and good signal-to-noise/background ratios . The approach allows for efficient screening of potential ASPH inhibitors and has been successfully applied to screen approximately 1,500 bioactive small molecules, including natural products and active pharmaceutical ingredients .

Prior to this development, researchers faced significant challenges in efficiently monitoring the activity of isolated ASPH, which limited progress in identifying small-molecule ASPH inhibitors.

How does ASPH mechanistically affect the Notch signaling pathway in human cancer progression?

ASPH has been demonstrated to activate the Notch signaling pathway, which represents a key mechanism driving malignant transformation, particularly in pancreatic cancer cells . The Notch signaling cascade is a highly conserved pathway critical for cell-cell signaling and cell fate determination during embryogenesis.

The mechanistic relationship between ASPH and Notch activation in pancreatic ductal adenocarcinoma (PDAC) involves:

  • Overexpression of activated Notch1 and hairy and enhancer of split-1 (HES1, a Notch-responsive gene) in PDAC cells compared to adjacent normal tissues

  • ASPH-mediated hydroxylation of EGF-like repeats in Notch receptors, enhancing their interactions with ligands

  • Subsequent activation of downstream transcriptional programs that promote cellular proliferation, survival, and invasiveness

While Notch receptors and ligands are typically downregulated in normal adult pancreatic tissue, they become activated during pancreatic tumorigenesis. Notch activation promotes epithelial-to-mesenchymal transition (EMT), facilitating invasion and metastasis of pancreatic cancer cells . This ASPH-Notch signaling axis represents a critical pathway that can be targeted therapeutically to potentially inhibit cancer progression.

What is the role of ASPH in mitochondrial DNA integrity and how does this contribute to oncogenesis?

ASPH has been implicated in disrupting mitochondrial DNA (mtDNA) integrity, which contributes to oncogenesis through multiple mechanisms. Somatic mtDNA mutations have been detected in various tumor types, including pancreatic cancer . Research in hepatocellular carcinoma has demonstrated that ASPH overexpression significantly correlates with:

  • Decreased copy numbers of the displacement loop (D-loop) region of mtDNA

  • Reduced NADH dehydrogenase subunit 1

  • Increased somatic mutations in the D-loop region

The mechanistic basis for these effects involves ASPH disruption of the H2AX-mtTFA signaling pathway:

  • Under normal conditions, H2AX (H2A histone family member X) functions as a shuttle protein transporter that carries mitochondrial transcription factor A (mtTFA) from the cytoplasm to mitochondria

  • mtTFA plays a crucial role in mtDNA replication, transcription, and repair

  • Overexpressed ASPH competes with mtTFA for binding to H2AX, blocking mtTFA translocation into mitochondria

  • This competition reduces mtTFA binding to the D-loop, disrupting mtDNA maintenance

These alterations ultimately lead to:

  • Increased mutations in mtDNA

  • Reduced mtDNA copy number

  • Decreased expression of mitochondrial respiratory chain enzymes

  • Aberrant mitochondrial membrane potential

  • Decreased ATP production

  • Increased reactive oxygen species (ROS)

This cascade of mitochondrial dysfunction contributes to the metabolic reprogramming characteristic of cancer cells and potentially enhances their invasive capabilities.

What therapeutic approaches targeting ASPH are under development for human cancers?

ASPH has emerged as a promising therapeutic target due to its overexpression in 70-90% of human tumors and its cell surface localization in cancer cells . Several therapeutic approaches are being investigated:

Immunotherapeutic Approaches

ASPH-based immunotherapy leverages the protein's translocation from the endoplasmic reticulum to the plasma membrane in cancer cells, where it is exposed to the extracellular environment and can function as a tumor-associated antigen (TAA) .

Research has demonstrated that:

  • ASPH-loaded dendritic cells (DCs) exhibit substantial anti-tumor effects in hepatocellular carcinoma models

  • Both CD4+ T cells and CD8+ cytotoxic T cells contribute to these anti-tumor effects

  • ASPH protein-loaded DCs can activate CD4+ T cells and CD8+ CTLs via ASPH-derived HLA class I- and class II-restricted peptides

Similar effects have been observed in cholangiocarcinoma models, where ASPH-loaded DCs exhibited cytotoxic effects in vitro, suppressed intrahepatic tumor growth and metastasis in rats, and increased infiltration of CD3+ lymphocytes into tumors .

Small Molecule Inhibitors

Several small molecule inhibitors targeting ASPH have shown promise:

  • MO-I-1100: This β-hydroxylase inhibitor reduced ASPH activity by 80% and inhibited ASPH-induced proliferation, migration, invasion, and colony formation in pancreatic cancer models, while suppressing Notch signaling

  • MO-I-1151: Demonstrated significant reduction in viability and directional motility of glioblastoma multiforme cells

  • Vadadustat: Originally developed as a hypoxia-inducible factor prolyl-hydroxylase inhibitor, this compound was found to inhibit ASPH with moderate potency

Antibody-Based Approaches

Monoclonal antibodies targeting ASPH show potential for both imaging and therapeutic applications:

  • Radiolabeled human monoclonal antibody PAN-622 targeting cell surface ASPH shows promise for imaging and potentially treating metastatic breast cancer

  • Monoclonal antibodies against the ASPH C-terminal (ASPH-C) increase antibody-dependent cellular cytotoxicity of NK cells against various cancer cell lines

Nucleic Acid-Based Therapies

RNA interference approaches have demonstrated efficacy in preclinical models:

  • Antisense oligodeoxynucleotide inhibition of ASPH expression reduced 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

What methodological advances have enabled high-throughput screening for ASPH inhibitors?

Recent methodological advances have overcome previous limitations in ASPH inhibitor screening. A key breakthrough came with the development of a stable synthetic thioether mimic of ASPH substrates that can be employed in solid-phase extraction mass spectrometry-based high-throughput ASPH inhibition assays .

The development process involved:

  • Understanding that ASPH substrates have a non-canonical EGFD disulfide pattern

  • Creating a stable synthetic thioether mimic that retains appropriate substrate properties

  • Incorporating this mimic into a solid-phase extraction methodology

  • Coupling with mass spectrometry detection for high-throughput analysis

This assay system demonstrates:

  • Excellent robustness (high Z'-factors)

  • Good signal-to-noise and signal-to-background ratios

  • Suitability for screening large compound libraries

Using this methodology, researchers successfully screened approximately 1,500 bioactive small molecules, including natural products and active pharmaceutical ingredients of approved human therapeutics . This screening identified potent ASPH inhibitors from both compound classes, demonstrating the utility of the approach for drug discovery efforts.

How does ASPH expression correlate with specific cancer subtypes and patient outcomes?

ASPH expression patterns vary across cancer subtypes and correlate with specific clinical outcomes. In hepatocellular carcinoma, Wang et al. demonstrated a significant association between ASPH overexpression and higher recurrence rates as well as lower survival rates following surgical intervention . This correlation is particularly pronounced in early-stage HCC patients, suggesting ASPH may serve as an early prognostic biomarker.

Research has established correlations between ASPH expression and:

These correlations highlight the potential utility of ASPH as both a prognostic biomarker and a therapeutic target.

Product Science Overview

Introduction

Aspartate Beta-Hydroxylase (ASPH) is a type II transmembrane protein that plays a crucial role in the post-translational modification of specific aspartyl and asparaginyl residues in epidermal growth factor-like domains (EGFDs) of target proteins. This enzyme is highly conserved and is part of the alpha-ketoglutarate-dependent dioxygenase family, which is essential for collagen biosynthesis .

Structure and Function

ASPH is an approximately 86 kDa protein located on the luminal side of the endoplasmic reticulum (ER). It hydroxylates the β-carbons of specific aspartyl and asparaginyl residues in the presence of ferrous iron. Unlike canonical EGFD disulfide patterns, ASPH catalyzes noncanonical EGFD substrates . The gene encoding ASPH is located at position q12.1 on human chromosome 8 .

Biological Role

ASPH is widely expressed in proliferating placenta trophoblastic cells and is almost undetectable in normal adult tissues . However, its expression is significantly upregulated in various human malignancies, where it is associated with poor survival and prognosis . ASPH contributes to tumor cell migration, infiltration, and metastasis by enhancing cell proliferation, migration, and invasion. It also promotes tumor growth by stimulating angiogenesis and immunosuppression, primarily through the activation of Notch and SRC signaling pathways .

Clinical Significance

Due to its role in malignant transformation, ASPH has become a target for cancer therapy. Small molecule inhibitors of ASPH enzymatic activity have shown anti-metastatic effects in preclinical mouse models . Additionally, ASPH can be targeted by monoclonal antibodies and has been used as a tumor-associated antigen to induce both CD8+ and CD4+ T cells in mice . The PAN-301-1 vaccine against ASPH has already been tested in a phase 1 clinical trial in patients with prostate cancer .

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.