DUSP18 Human, Active

Dual Specificity Phosphatase 18 Human Recombinant, Active
Shipped with Ice Packs
In Stock

Description

Enzymatic Activity and Substrates

DUSP18 exhibits phosphatase activity toward synthetic MAPK peptides, preferring phosphotyrosine and diphosphorylated forms over phosphothreonine . Key findings include:

  • Substrate specificity: Dephosphorylates ERBB receptors (e.g., EGFR, ERBB2), insulin receptor (INSR), and MAPK8IP2, modulating downstream signaling .

  • In vitro activity: Efficiently hydrolyzes p-nitrophenyl phosphate (pNPP), a common phosphatase substrate .

  • Regulatory role: Stabilizes transcription factors like USF1, which activates cholesterol biosynthesis pathways .

Table 2: Validated Substrates of DUSP18

SubstrateBiological RoleInteraction Evidence
USF1Transcriptional regulation of cholesterol genesCo-immunoprecipitation
SREBP2Cholesterol biosynthesis regulationRNA-seq, proteomics
MAPK14 (p38α)Stress response signalingDephosphorylation assay
EGFR/ERBB receptorsGrowth factor signalingYeast two-hybrid

Cancer and Immune Evasion

Recent studies implicate DUSP18 in colorectal cancer (CRC) immune evasion:

  • Mechanism: DUSP18 stabilizes USF1, which upregulates SREBF2 (cholesterol biosynthesis) and increases lanosterol production. Lanosterol suppresses CD8+ T-cell activation in the tumor microenvironment (TME), enabling immune escape .

  • Therapeutic targeting: Inhibition of DUSP18 with Lumacaftor (FDA-approved) synergizes with anti-PD-1 therapy, reducing CRC growth in mice by enhancing T-cell cytotoxicity .

Metabolic Regulation

  • Cholesterol biosynthesis: DUSP18 knockdown reduces lanosterol and cholesterol intermediates, impairing tumor cell proliferation .

  • Proteomic profiling: DUSP18 loss downregulates 440 proteins involved in cholesterol metabolism while upregulating antigen presentation pathways .

Recombinant DUSP18 Human, Active

The recombinant human DUSP18 protein (e.g., ab123154) is produced in Escherichia coli with >95% purity . Specifications include:

  • Amino acid range: 1–188 (full-length).

  • Activity: Validated via dephosphorylation assays using pNPP and MAPK peptides .

  • Applications: Used in enzymology studies, drug screening, and structural biology .

Product Specs

Introduction
Dual specificity phosphatase 18 (DUSP18), a member of the dual-specificity phosphatase (DSP) family, plays a crucial role in dephosphorylating phosphotyrosine and phosphothreonine residues. Its enzymatic activity is biased towards phosphorylated tyrosine residues over threonine residues. Moreover, DUSP18 exhibits dephosphorylation activity against p-nitrophenyl phosphate (pNPP) in vitro. Its activity is influenced by certain factors, being inhibited by iodoarectic acid and activated by manganese ions. DUSP18 is widely expressed, with notably high levels found in the liver, brain, ovary, and testis.
Description
Produced in E.Coli, DUSP18 is a single, non-glycosylated polypeptide chain comprising 212 amino acids (1-188a.a.) with a molecular weight of 23.6kDa. It features a 24 amino acid His-tag fused at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear solution, sterile filtered.
Formulation
The DUSP18 protein solution (0.5mg/ml) is supplied in a buffer containing 20mM Tris-HCl (pH 8.0), 0.1mM PMSF, 1mM DTT, 40% glycerol, and 1mM EDTA.
Stability
For short-term storage (up to 2-4 weeks), keep at 4°C. For extended periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity
Purity exceeds 95.0% as assessed by SDS-PAGE.
Biological Activity
The specific activity, exceeding 300 units/mg, represents the enzyme quantity required to hydrolyze 1.0 nanomole of p-nitrophenyl phosphate (pNPP) per minute at pH 7.5 and 37°C.
Synonyms

Dual specificity protein phosphatase 18, Low molecular weight dual specificity phosphatase 20, LMW-DSP20, DUSP18, LMWDSP20, VHP, DUSP26.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSHMTAPSC AFPVQFRQPS VSGLSQITKS LYISNGVAAN NKLMLSSNQI TMVINVSVEV VNTLYEDIQY MQVPVADSPN SRLCDFFDPI ADHIHSVEMK QGRTLLHCAA GVSRSAALCL AYLMKYHAMS LLDAHTWTKS CRPIIRPNSG FWEQLIHYEF QLFGKNTVHM VSSPVGMIPD IYEKEVRLMI PL

Q&A

What is DUSP18 and what are its primary functions?

DUSP18 (also known as Low molecular weight dual specificity phosphatase 20, LMW-DSP20, VHP, or DUSP26) is a member of the dual-specificity protein tyrosine phosphatase family . Like most dual-specificity phosphatases, DUSP18 displays dephosphorylating activity towards both phosphotyrosine and phosphothreonine residues . Recent research has revealed that DUSP18 dephosphorylates and stabilizes the USF1 bHLH-ZIP transcription factor, which subsequently induces the SREBF2 gene in colorectal cancer cells . At the molecular level, DUSP18 also interacts with and dephosphorylates SAPK (Stress-Activated Protein Kinase), inhibiting the SAPK/JNK signal pathway .

How does DUSP18 differ from other dual-specificity phosphatases?

DUSP18 is distinct from other known DSPs in several important ways. Structurally, it possesses approximately 30 additional residues at the C-terminus of the catalytic domain, which fold into two antiparallel β-strands and participate in extensive interactions with the catalytic domain . These interactions likely contribute to its unusual thermostability, with DUSP18 exhibiting optimum activity at 328 K (55°C) . Additionally, while the catalytic domain adopts a fold similar to other DSPs, substantial differences exist in the regions surrounding the active site, suggesting that DUSP18 has unique substrate specificity . These distinctive characteristics indicate that DUSP18 constitutes a specialized phosphatase with potentially unique biological functions.

What is known about the crystal structure of DUSP18?

The crystal structure of human DUSP18 has been determined at 2.0 Å resolution . The structural analysis reveals that while DUSP18's catalytic domain shares similarities with other dual-specificity phosphatases, it possesses unique features, particularly in regions surrounding the active site . The C-terminal extension of approximately 30 residues forms two antiparallel β-strands that interact extensively with the catalytic domain . These structural characteristics likely explain DUSP18's thermostability and provide insights into its substrate specificity. The resolved structure (PDB: 2ESB) serves as a valuable resource for structure-based drug design targeting DUSP18 .

What are the biochemical properties of active DUSP18 protein?

Active recombinant human DUSP18 protein typically has a specific activity greater than 300 units/mg, where one unit is defined as the amount of enzyme that hydrolyzes 1.0 nmole of p-nitrophenyl phosphate (pNPP) per minute at pH 7.5 and 37°C . The protein is typically formulated in 20mM Tris-HCl buffer (pH 8.0) containing 0.1mM PMSF, 1mM DTT, 40% glycerol, and 1mM EDTA . Commercially available DUSP18 is often produced in Escherichia coli expression systems and purified to >95% purity as determined by SDS-PAGE . For optimal storage stability, it is recommended to store the protein at 4°C if using within 2-4 weeks, or at -20°C for longer periods, preferably with a carrier protein such as 0.1% HSA or BSA to prevent activity loss .

What are the most effective methods for studying DUSP18 activity in vitro?

For in vitro analysis of DUSP18 phosphatase activity, the p-nitrophenyl phosphate (pNPP) assay remains the gold standard for initial characterization . This colorimetric assay measures the hydrolysis of pNPP to p-nitrophenol, which can be detected spectrophotometrically at 405 nm. For more specific substrate analyses, researchers should consider using phosphopeptide substrates corresponding to physiological targets like SAPK/JNK or USF1 .

Phosphatase assays should be conducted under optimal conditions for DUSP18: pH 7.5 and temperatures between 37°C (for standard assays) and 55°C (where DUSP18 shows maximum activity) . When designing inhibitor screening assays, it's important to include appropriate controls to account for potential interference with the detection method. Fluorescence-based assays using fluorescein diphosphate or methylumbelliferyl phosphate offer increased sensitivity for detecting subtle changes in phosphatase activity.

How can researchers effectively study DUSP18 in cellular contexts?

Studying DUSP18 in cellular contexts requires a multifaceted approach. Antibody-based immunofluorescence and spatial proteomics techniques can effectively map DUSP18's subcellular localization and potential colocalization with substrate proteins . CRISPR-Cas9 gene editing has proven valuable for investigating DUSP18 function, as demonstrated in colorectal cancer models .

For analyzing protein-protein interactions, co-immunoprecipitation followed by western blotting or mass spectrometry can identify binding partners. Proximity ligation assays provide an alternative for detecting protein interactions in situ. To assess the functional impact of DUSP18, researchers should measure phosphorylation levels of known substrates (e.g., USF1, SAPK) following DUSP18 manipulation . RNA-seq and proteomics approaches can further elucidate downstream pathways affected by DUSP18 activity, as seen in studies identifying connections to SREBF2 and cholesterol biosynthesis pathways .

What role does DUSP18 play in colorectal cancer development and progression?

Recent CRISPR screens in mouse models of colorectal cancer (CRC) have implicated DUSP18 in establishing tumor-directed immune evasion . Mechanistically, DUSP18 dephosphorylates and stabilizes the USF1 bHLH-ZIP transcription factor, which in turn induces SREBF2 gene expression . This pathway allows cancer cells to accumulate the cholesterol biosynthesis intermediate lanosterol and release it into the tumor microenvironment .

The released lanosterol is taken up by CD8+ T cells, where it suppresses the mevalonate pathway, reduces KRAS protein prenylation and function, and inhibits T cell activation—establishing a molecular basis for tumor cell immune escape . Inhibition of DUSP18 reduces CRC growth rates, which correlates with high levels of CD8+ T cell activation . This mechanism represents a novel metabolic pathway through which cancer cells can modulate the immune response, suggesting that targeting DUSP18 may have dual benefits of directly affecting cancer cell metabolism while enhancing anti-tumor immunity.

How is DUSP18 involved in hepatocellular carcinoma?

In hepatocellular carcinoma (HCC), DUSP18 has been identified as a key mediator in hypoxia-induced cancer progression . Under hypoxic conditions, HIF1A (Hypoxia-Inducible Factor 1-Alpha) activates DUSP18-mediated MAPK14 (p38 MAPK) dephosphorylation . This mechanism promotes HCC cell migration and invasion, contributing to the aggressive phenotype and metastatic potential of liver cancer cells .

The HIF1A-DUSP18-MAPK14 axis represents a distinct pathway from DUSP18's role in colorectal cancer, highlighting the context-dependent functions of this phosphatase in different cancer types. Understanding these tissue-specific mechanisms is crucial for developing targeted therapeutic approaches. Researchers investigating DUSP18 in HCC should consider hypoxic culture conditions and examine interactions with HIF1A and MAPK14 phosphorylation status to comprehensively evaluate its role in this cancer type.

How can DUSP18 inhibition be leveraged for cancer immunotherapy?

Emerging research indicates that DUSP18 inhibition may enhance cancer immunotherapy efficacy, particularly in colorectal cancer. Inhibition of DUSP18 disrupts the metabolic immune evasion pathway involving lanosterol release, which normally suppresses CD8+ T cell activation . Experimentally, the combination of an anti-PD-1 antibody and Lumacaftor (an FDA-approved small molecule inhibitor of DUSP18) synergistically inhibits CRC growth in mice and enhances anti-tumor immunity .

This approach represents a promising strategy of combining immune checkpoint and metabolic blockade as a rationally-designed, mechanistically-based therapy. Researchers exploring this avenue should consider several methodological approaches: (1) testing various DUSP18 inhibitors in combination with different immunotherapy agents; (2) monitoring changes in tumor-infiltrating lymphocyte activity; (3) analyzing metabolomic profiles of the tumor microenvironment; and (4) assessing changes in cholesterol metabolites, particularly lanosterol levels in both tumor cells and immune cells. Such comprehensive analyses will provide insights into the translational potential of DUSP18 inhibition as an immunotherapy-enhancing strategy.

What are the challenges in developing specific inhibitors for DUSP18?

Structure-based drug design approaches utilizing the crystal structure of DUSP18 (PDB: 2ESB) can guide rational inhibitor development . Researchers should focus on compounds that interact with DUSP18-specific structural features rather than the conserved catalytic pocket. High-throughput screening methods should incorporate counterscreens against related phosphatases to identify truly selective compounds. Additionally, the unusual thermostability of DUSP18 (optimum activity at 55°C) presents both a challenge for assay standardization and an opportunity for developing inhibitors that specifically disrupt the structural elements contributing to this thermostability .

Lumacaftor has been identified as an FDA-approved DUSP18 inhibitor, but its mechanism of action and specificity require further characterization . Researchers aiming to develop DUSP18 inhibitors should consider multiple methods including in silico screening, fragment-based approaches, and allosteric inhibitor development that targets the unique C-terminal region of DUSP18.

What are common pitfalls in DUSP18 activity assays and how can they be addressed?

Several challenges can affect the reliability of DUSP18 activity assays. First, DUSP18's unusual temperature optimum at 55°C may lead to underestimation of activity when standard assay temperatures (37°C) are used . Researchers should consider performing activity measurements across a temperature range to determine optimal conditions.

Second, DUSP18's susceptibility to oxidation can lead to inconsistent results. Including reducing agents such as DTT or β-mercaptoethanol in assay buffers is essential . Third, commercially available recombinant DUSP18 preparations may vary in specific activity; therefore, each lot should be characterized before use in critical experiments.

For substrate specificity studies, researchers should be cautious about extrapolating from artificial substrates (like pNPP) to physiological substrates. Validation with phosphopeptides derived from known substrates such as USF1 or SAPK provides more physiologically relevant information . Additionally, when using cell-based assays, the expression level of DUSP18 relative to its substrates can significantly impact results, necessitating careful consideration of stoichiometry in overexpression studies.

How should researchers approach contradictory findings in DUSP18 research across different cancer types?

The seemingly contradictory roles of DUSP18 across different cancer types highlight the context-dependent function of this phosphatase. To address these contradictions methodologically, researchers should:

  • Carefully document the cancer cell type, experimental conditions, and genetic background in all studies, as these factors may explain discrepancies in findings.

  • Perform comprehensive pathway analyses to determine whether DUSP18 engages different substrates or signaling pathways in different cellular contexts. In colorectal cancer, DUSP18 primarily affects the USF1-SREBF2-lanosterol pathway , while in hepatocellular carcinoma, it influences the HIF1A-MAPK14 axis .

  • Consider the tumor microenvironment, particularly the role of hypoxia, which has been shown to regulate DUSP18 in hepatocellular carcinoma . Different oxygen tensions in various tumor types may explain differential regulation and function.

  • Evaluate the immune component in different models, as DUSP18's role in immune evasion in colorectal cancer may not be equally relevant in all cancer types .

  • Use multiple model systems (cell lines, organoids, and animal models) to confirm findings and ensure they are not artifacts of a particular experimental system.

By systematically addressing these factors, researchers can reconcile contradictory findings and develop a more nuanced understanding of DUSP18's role in cancer biology.

Product Science Overview

Introduction

Dual Specificity Phosphatase 18 (DUSP18) is a member of the dual-specificity phosphatase (DSP) family, which is part of the larger type I cysteine-based protein-tyrosine phosphatase superfamily. These enzymes are characterized by their ability to dephosphorylate both tyrosine and serine/threonine residues, playing a crucial role in modulating various signaling pathways .

Gene and Protein Structure

The DUSP18 gene is located on chromosome 22 and encodes a protein consisting of 212 amino acids with a molecular mass of approximately 23.6 kDa . The protein contains a consensus DUSP C-terminal catalytic domain but lacks the N-terminal CH2 domain found in the mitogen-activated protein kinase phosphatase (MKP) class of DUSPs .

Enzymatic Activity

DUSP18 exhibits a preferential enzymatic activity for phosphorylated tyrosine residues over threonine residues. It can dephosphorylate single and diphosphorylated synthetic MAPK peptides, with a preference for the phosphotyrosine and diphosphorylated forms . Additionally, DUSP18 dephosphorylates p-nitrophenyl phosphate (pNPP) in vitro and is inhibited by iodoacetic acid while being activated by manganese ions .

Expression and Localization

DUSP18 is extensively expressed in various tissues, with the highest levels observed in the liver, brain, ovary, and testis . The recombinant form of DUSP18 is produced in E. coli as a single, non-glycosylated polypeptide chain and is purified using proprietary chromatographic techniques .

Biological Functions

As a dual-specificity phosphatase, DUSP18 plays a significant role in regulating critical signaling pathways by dephosphorylating key proteins involved in these pathways. This regulation is essential for maintaining cellular homeostasis and responding to various physiological stimuli .

Clinical Relevance

Mutations or dysregulation of DUSP18 have been associated with certain diseases, including spastic paraplegia 26, an autosomal recessive disorder . Understanding the function and regulation of DUSP18 can provide insights into the pathogenesis of such diseases and potentially lead to the development of targeted therapies.

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 2024 Thebiotek. All Rights Reserved.