MPPED2 Human

Metallophosphoesterase Domain Containing 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

WAGR Syndrome

  • Deletions in 11p13 (including MPPED2) are linked to Wilms tumor, aniridia, genitourinary anomalies, and intellectual disability .

Cancer

Cancer TypeMPPED2 RoleKey FindingsSource
Breast CancerTumor suppressor- Hypermethylation silences MPPED2 in 98% of tumors .
- Restoring MPPED2 reduces proliferation, migration, and invasion in vitro .
GlioblastomaDownregulatedAssociated with poor prognosis .
Oral Squamous CellTumor suppressormiR-448 downregulates MPPED2 to promote growth .

Trauma and Inflammation

  • rs2065418 TT genotype in MPPED2 correlates with:

    • Elevated organ dysfunction (Marshall MODScore: p = 0.01) .

    • Prolonged hospitalization (21.8 vs. 15.9 days; p = 0.03) and mechanical ventilation (9.2 vs. 5.7 days; p = 0.04) .

    • Dysregulated inflammatory mediators (e.g., IL-6, IL-10) .

Dental Caries

  • rs12797813 in MPPED2 associates with susceptibility (p < 0.05) and higher DMFT scores (p = 0.025) .

Neurological Roles

Model SystemFindingsSource
Drosophila- dMpped KO reduces lifespan; rescued by human MPPED2 .
- Modulates odorant response and neuronal connectivity .
Mouse NeuronsExpressed in cortical neurons; co-localizes with Tuj1 .

Epigenetic Regulation

  • Hypermethylation of MPPED2 promoter drives downregulation in breast cancer .

  • Long non-coding RNA MPPED2-AS1 regulates MPPED2 expression via DNA methylation .

Inflammatory Modulation

  • MPPED2 SNPs alter systemic inflammation post-trauma, increasing IL-6 (p < 0.01) and IL-10 (p = 0.002) .

Genetic Variants and Clinical Impact

SNP IDAssociationClinical OutcomeSource
rs2065418Trauma severity↑ Organ failure, ↑ hospital stay
rs12797813Dental caries risk↑ DMFT scores

Therapeutic Implications

  • Breast Cancer: Demethylating agents (e.g., 5-Aza-dC) restore MPPED2 expression, suppressing tumor growth .

  • Neurodegeneration: MPPED2 overexpression in Drosophila rescues lifespan deficits, suggesting conserved neuroprotective roles .

Unresolved Questions

  • Mechanistic links between MPPED2 and immune modulation.

  • Tissue-specific roles in development vs. disease.

Product Specs

Introduction
Metallophosphoesterase domain containing 2 (MPPED2) is a member of the UPF0046 family. It exhibits weak metallophosphoesterase activity in laboratory settings. Additionally, MPPED2 codes for a metallophosphoesterase and may play a role in nervous system and brain development. Associations with MPPED2 include Wagr syndrome and aniridia.
Description
MPPED2 Human Recombinant, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 317 amino acids (1-294 a.a.) with a molecular weight of 35.7 kDa. It is fused to a 23 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
A clear, sterile solution that has been filtered.
Formulation
The MPPED2 protein solution (0.25 mg/ml) contains 20mM Tris-HCl buffer with a pH of 8.0, 0.15M NaCl, 10% glycerol, and 1mM DTT.
Stability
For optimal storage, keep at 4°C if the entire vial will be used within 2-4 weeks. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
SDS-PAGE analysis indicates a purity greater than 85%.
Synonyms
Metallophosphoesterase Domain Containing 2, C11orf8, 239FB, Fetal Brain Protein 239, Chromosome 11 Open Reading Frame 8, Metallophosphoesterase MPPED2, EC 3.1., FAM1B, D11S302E, Metallophosphoesterase Domain-Containing Protein 2.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMAHGIPS QGKVTITVDE YSSNPTQAFT HYNINQSRFQ PPHVHMVDPI PYDTPKPAGH TRFVCISDTH SRTDGIQMPY GDILLHTGDF TELGLPSEVK KFNDWLGNLP YEYKIVIAGN HELTFDKEFM ADLVKQDYYR FPSVSKLKPE DFDNVQSLLT NSIYLQDSEV TVKGFRIYGA PWTPWFNGWG FNLPRGQSLL DKWNLIPEGI DILMTHGPPL GFRDWVPKEL QRVGCVELLN TVQRRVRPKL HVFGGIHEGY GIMTDGYTTY INASTCTVSF QPTNPPIIFD LPNPQGS

Q&A

What is MPPED2 and where is it located in the human genome?

MPPED2 (metallophosphoesterase domain containing protein 2) is a highly conserved protein with orthologs found from worms to humans. In humans, the MPPED2 gene is located on chromosome 11p13, positioned between the FSHB and PAX6 genes . This region is particularly significant as it is deleted in patients with WAGR syndrome (Wilms tumor, aniridia, genitourinary anomalies, and mental retardation) . The gene has several alternative names including 239FB and C11orf8 .

At the molecular level, MPPED2 is predicted to enable manganese ion binding activity, phosphoric diester hydrolase activity, and purine ribonucleotide binding activity . The protein contains a metallophosphoesterase domain that is structurally similar to phosphoprotein phosphatases, though with distinct functional characteristics.

What is the normal biological function of MPPED2 in human tissues?

MPPED2 demonstrates tissue-specific expression patterns, with notably high expression in the fetal brain where it is positively associated with neurodevelopment . The protein functions as a metallophosphodiesterase that cleaves 3',5'-cyclic phosphate nucleosides into 5'-phosphate nucleosides, thereby regulating levels of cyclic second messengers and their degradation rates .

Despite its structural similarity to phosphodiesterases, MPPED2 shows relatively low enzymatic activity compared to other members of this family. This reduced activity is attributed to two key factors: the substitution of an active-site histidine residue by glycine and the binding of AMP or GMP to the active site . This unique structural arrangement suggests that substrate hydrolysis may not be the primary function of MPPED2, and the protein may instead serve as a scaffolding or adaptor protein in cellular signaling networks .

How does MPPED2 expression vary across different tissue types and developmental stages?

MPPED2 exhibits variable expression across tissues, with particularly high levels observed in neural tissues during development. Expression data from various studies indicates the following pattern:

Tissue TypeMPPED2 Expression LevelDevelopmental Stage
Fetal BrainHighDevelopmental
Adult BrainModeratePost-developmental
Cervical Tissue (normal)ModerateAdult
Cancerous TissuesLow/AbsentPathological

The high expression in fetal brain strongly suggests a role in neurodevelopment . The downregulation of MPPED2 in various cancers, including neuroblastoma, cervical carcinoma, oral squamous carcinomas, and breast cancer, further indicates its potential tumor suppressor function . This expression pattern makes MPPED2 a valuable marker for both developmental processes and pathological states.

What is the relationship between MPPED2 expression and cancer prognosis?

Research has established a significant correlation between MPPED2 expression levels and cancer prognosis across multiple tumor types. In glioblastoma (GBM), the most aggressive and lethal neoplasia of the central nervous system in adults, MPPED2 expression is consistently downregulated, and this downregulation shows a positive correlation with reduced patient survival . TCGA and Gravendeel databases analyses reveal that MPPED2 expression negatively correlates with the most aggressive mesenchymal subtype of GBM .

Similar prognostic relationships have been observed in other cancers, including cervical carcinoma, where MPPED2 expression is inversely correlated with the expression of p16INK4A, a well-established marker of high-risk HPV integration . This pattern of downregulation across multiple cancer types supports the hypothesis that MPPED2 functions as a tumor suppressor, and its loss contributes to more aggressive disease phenotypes.

For researchers investigating cancer prognosis, MPPED2 expression analysis should be considered alongside established prognostic markers to develop more comprehensive predictive models for patient outcomes.

How does MPPED2 restoration affect cancer cell behavior in experimental models?

Experimental restoration of MPPED2 expression in cancer cell lines has revealed significant anti-tumor effects. In GBM cell lines (U251 and GLI36), restoration of MPPED2 expression produces three key effects:

  • Decreased cell growth and proliferation

  • Reduced cell migration capacity

  • Enhanced sensitivity to temozolomide (the standard chemotherapeutic agent for GBM), inducing apoptotic cell death

These findings suggest multiple mechanisms through which MPPED2 may exert its tumor-suppressive effects, including regulation of cell cycle progression, modulation of cell motility pathways, and sensitization to apoptotic signals. The enhanced chemosensitivity is particularly significant, as it suggests MPPED2 restoration could potentially improve the efficacy of existing therapeutic regimens.

For researchers designing experiments to study MPPED2's effects on cancer cells, it is essential to include appropriate controls and measure multiple parameters of cell behavior, including proliferation, migration, invasion, and response to therapeutic agents.

What molecular mechanisms underlie MPPED2's tumor suppressor function?

The tumor suppressor role of MPPED2 appears to involve multiple molecular mechanisms, though complete elucidation requires further research. Current evidence suggests:

  • Cell Cycle Regulation: MPPED2 likely influences cell cycle progression, as evidenced by reduced cell growth following its restoration in cancer cell lines .

  • Migration Pathway Modulation: The decreased migration observed in MPPED2-restored cells suggests interactions with cytoskeletal regulators and cell adhesion molecules .

  • Apoptotic Pathway Sensitization: MPPED2 enhances cellular sensitivity to apoptotic stimuli, particularly in response to chemotherapeutic agents like temozolomide .

  • Cyclic Nucleotide Signaling: As a phosphodiesterase that cleaves 3',5'-cyclic phosphate nucleosides, MPPED2 may regulate second messenger levels that control cellular proliferation and differentiation .

  • Protein Scaffolding: The unique structural elements of MPPED2 suggest it may function as a scaffolding or adaptor protein within signaling complexes, potentially influencing multiple downstream pathways .

The downregulation of MPPED2 in cancers is potentially linked to epigenetic mechanisms, particularly promoter hypermethylation, which has been observed in several malignant neoplasias .

What are the optimal techniques for measuring MPPED2 expression in clinical samples?

For accurate measurement of MPPED2 expression in clinical samples, researchers should consider a multi-technique approach:

  • RT-qPCR (Reverse Transcription Quantitative PCR):

    • Highly sensitive for detecting MPPED2 mRNA expression

    • Commercial assays available (e.g., Bio-Rad's PrimePCR assay with unique ID qHsaCEP0049498)

    • Recommended parameters: amplicon length of 100bp, primer efficiency >97%, R² >0.999

    • Use appropriate reference genes for normalization

  • Immunohistochemistry (IHC):

    • Allows visualization of MPPED2 protein expression with spatial context

    • Semi-quantitative scoring of protein intensity can be performed (0-3+)

    • Enables correlation with histopathological features

    • Can be paired with other markers (e.g., p16INK4A) for co-expression studies

  • Western Blotting:

    • Provides quantitative assessment of MPPED2 protein levels

    • Allows detection of potential isoforms or post-translational modifications

  • In Situ Hybridization:

    • Alternative to IHC for detecting MPPED2 mRNA in intact tissues

    • Useful when antibodies show cross-reactivity issues

For clinical correlation studies, researchers should collect comprehensive patient data including age, tumor grade, histological subtype, and treatment history, as significant correlations have been observed between MPPED2 expression, age, and other clinical parameters .

What experimental approaches are effective for studying MPPED2 function in cellular models?

To investigate MPPED2 function in cellular models, researchers can employ several complementary approaches:

  • Gene Expression Modulation:

    • Overexpression: Transfection of MPPED2 expression vectors in cancer cell lines with low endogenous expression (e.g., U251 and GLI36 GBM lines)

    • Knockdown: siRNA or shRNA targeting MPPED2 in cells with high endogenous expression

    • CRISPR-Cas9: For stable knockout or knock-in models

  • Functional Assays:

    • Proliferation: MTT/XTT assays, BrdU incorporation, or real-time cell analysis

    • Migration: Wound healing assays, transwell migration

    • Apoptosis: Annexin V/PI staining, caspase activity assays

    • Drug sensitivity: Dose-response curves for chemotherapeutics like temozolomide

  • Protein Interaction Studies:

    • Co-immunoprecipitation to identify binding partners

    • Proximity ligation assays for in situ detection of protein interactions

    • Yeast two-hybrid screening for novel interactors

  • Enzymatic Activity Assessment:

    • Phosphodiesterase activity assays with various potential substrates

    • Structural studies to explore the effects of active site mutations

  • Signaling Pathway Analysis:

    • Western blotting for downstream effectors

    • Phosphoproteomics to identify signaling changes

    • Reporter assays for transcriptional activity

When designing these experiments, researchers should include appropriate controls and consider the potential scaffolding function of MPPED2 beyond its enzymatic activity .

How can researchers effectively analyze MPPED2 promoter methylation status?

Given that MPPED2 downregulation in cancers may be linked to promoter hypermethylation , analyzing methylation status is crucial. Researchers can employ these methodologies:

  • Bisulfite Sequencing:

    • Gold standard for site-specific CpG methylation analysis

    • Requires bisulfite conversion of unmethylated cytosines to uracil

    • Allows single-nucleotide resolution of methylation patterns

    • Can be applied to the entire MPPED2 promoter region

  • Methylation-Specific PCR (MSP):

    • Rapid screening method using primers specific for methylated or unmethylated sequences

    • Less labor-intensive than bisulfite sequencing

    • Useful for screening large sample cohorts

  • Pyrosequencing:

    • Quantitative assessment of methylation at multiple CpG sites

    • Higher throughput than traditional bisulfite sequencing

    • Provides percentage methylation at each analyzed site

  • Array-Based Methylation Analysis:

    • Platforms like Illumina's Infinium MethylationEPIC allow genome-wide methylation profiling

    • Can reveal MPPED2 methylation in context of broader epigenetic changes

    • Useful for identifying methylation signatures across cancer types

  • Chromatin Immunoprecipitation (ChIP):

    • Assess binding of methylation-related proteins (e.g., MeCP2, MBDs) to the MPPED2 promoter

    • Can be combined with transcription factor ChIP to understand regulatory mechanisms

For comprehensive analysis, researchers should examine both promoter methylation and expression levels in the same samples to establish direct correlations between these parameters.

How does the unique structural configuration of MPPED2 contribute to its functional properties?

MPPED2 exhibits a distinctive structural configuration that differs from typical metallophosphoesterases, which significantly influences its functional properties. Structural studies reveal that MPPED2's poor enzymatic activity stems from two key features:

  • The substitution of an active-site histidine residue by glycine, which alters the catalytic core functionality

  • The binding of AMP or GMP to the active site, which may act as regulatory elements

These structural peculiarities suggest that MPPED2 has evolved to utilize the conserved phosphoprotein-phosphatase-like fold in a unique manner. This adaptation allows MPPED2 to combine limited enzymatic activity with potential scaffolding or adaptor protein functions . The evolutionary conservation of these structural features across species indicates their functional importance.

For researchers investigating MPPED2's structure-function relationship, site-directed mutagenesis experiments targeting the glycine residue (reverting it to histidine) could provide insights into the evolutionary pressure that led to this seemingly detrimental substitution. Additionally, crystallographic studies with various nucleotide ligands could illuminate how binding affects protein conformation and potential interaction surfaces.

How does MPPED2 interact with the tumor microenvironment in different cancer types?

The interaction between MPPED2 and the tumor microenvironment (TME) represents a complex and understudied aspect of cancer biology. Based on MPPED2's functional properties and expression patterns, several potential interactions with the TME can be hypothesized:

  • Immune Cell Regulation: As a potential regulator of cyclic nucleotide signaling, MPPED2 might influence immune cell function within the TME, particularly given that cAMP and cGMP are important second messengers in immune responses.

  • Extracellular Matrix Remodeling: The reduced migration observed in MPPED2-restored cancer cells suggests potential effects on extracellular matrix interactions , which are critical components of the TME.

  • Angiogenesis Modulation: If MPPED2 influences signaling pathways relevant to angiogenesis, its loss in tumors could contribute to altered vascularization patterns.

  • Stromal Cell Communication: As a potential scaffolding protein , MPPED2 might mediate interactions between tumor cells and surrounding stromal cells.

Research approaches to explore these interactions could include:

  • Co-culture experiments with cancer cells and stromal components (fibroblasts, immune cells)

  • Analysis of secreted factors in MPPED2-modulated cancer cells

  • In vivo studies using xenograft models with MPPED2-restored cancer cells

  • Single-cell RNA sequencing to identify cell-specific effects in heterogeneous tumors

Understanding these interactions could reveal additional mechanisms through which MPPED2 loss contributes to cancer progression and potentially identify new therapeutic strategies targeting the TME.

What is the potential of MPPED2 as a therapeutic target in cancer treatment?

MPPED2 shows considerable promise as a therapeutic target in cancer treatment, particularly given its tumor suppressor properties and the effects observed upon its restoration in cancer cell lines. The therapeutic potential of MPPED2 can be considered from multiple angles:

  • Gene Therapy Approaches: Restoring MPPED2 expression in tumors where it is downregulated could potentially reduce cell growth, inhibit migration, and enhance sensitivity to conventional chemotherapeutics . This strategy could employ viral vectors or non-viral delivery systems to introduce functional MPPED2 into tumor cells.

  • Epigenetic Modulation: Since MPPED2 downregulation appears linked to promoter hypermethylation in several cancer types , DNA methyltransferase inhibitors (DNMTi) or other epigenetic modifiers could potentially restore endogenous MPPED2 expression. This approach would need to be carefully evaluated for specificity and off-target effects.

  • Small Molecule Mimetics: Developing small molecules that mimic MPPED2's tumor-suppressive functions could circumvent the challenges of protein delivery and expression.

  • Combination Therapies: The enhanced sensitivity to temozolomide observed in GBM cells with restored MPPED2 expression suggests particular value in combination approaches. Similarly, in cervical carcinoma, MPPED2-based therapies might complement existing HPV-targeted treatments .

Research findings from cervical carcinoma studies specifically note the potential of MPPED2 protein "to prevent cervical carcinoma progression in the near future" , highlighting clinical researcher optimism about therapeutic applications.

How can MPPED2 expression analysis be integrated into clinical diagnostic workflows?

Integrating MPPED2 expression analysis into clinical diagnostic workflows could enhance cancer classification, prognostication, and treatment planning. Implementation considerations include:

  • Diagnostic Platform Selection:

    • IHC-based detection could be readily incorporated into existing pathology workflows

    • RT-qPCR assays with validated primers (such as Bio-Rad's PrimePCR assay) offer quantitative assessment

    • Next-generation sequencing panels could include MPPED2 alongside other prognostic markers

  • Standardization and Interpretation:

    • Establish clear cutoff values for MPPED2 expression levels that correlate with clinical outcomes

    • Develop scoring systems that incorporate MPPED2 with existing diagnostic markers

    • For cervical cancer, combined analysis with p16INK4A expression could provide more comprehensive assessment

  • Clinical Context Integration:

    • In GBM, MPPED2 expression correlates with survival and molecular subtypes

    • In cervical carcinoma, MPPED2 expression inversely correlates with p16INK4A and high-risk HPV integration

    • These associations should inform the interpretation of MPPED2 expression data

  • Quality Control Measures:

    • Include appropriate positive and negative controls

    • Regularly validate assay performance across different laboratories

    • Participate in external quality assessment programs

Implementation would require close collaboration between research laboratories, pathology departments, and clinical oncology teams to ensure appropriate interpretation and clinical utility of MPPED2 expression data.

What are the potential biomarker applications of MPPED2 across different cancer types?

MPPED2 shows significant potential as a biomarker across multiple cancer types, with applications spanning diagnosis, prognosis, and treatment response prediction:

  • Diagnostic Biomarker:

    • Differential expression between normal and cancerous tissues makes MPPED2 a potential diagnostic marker

    • In cervical tissue, MPPED2 expression patterns differ between normal epithelium and carcinoma

    • Could be used in conjunction with established markers for improved diagnostic accuracy

  • Prognostic Biomarker:

    • In GBM, MPPED2 expression positively correlates with patient survival

    • MPPED2 expression negatively correlates with the aggressive mesenchymal subtype of GBM

    • These associations suggest value in stratifying patients according to risk

  • Predictive Biomarker:

    • The enhanced sensitivity to temozolomide in MPPED2-restored GBM cells suggests MPPED2 status might predict treatment response

    • Could guide selection of patients for specific therapeutic regimens

  • Monitoring Biomarker:

    • Changes in MPPED2 expression during treatment could potentially serve as an indicator of response

    • Particularly relevant in conjunction with epigenetic therapies that might restore MPPED2 expression

A comprehensive table comparing MPPED2 biomarker potential across cancer types would be valuable:

Cancer TypeDiagnostic ValuePrognostic ValuePredictive ValueCurrent Evidence Strength
GlioblastomaModerateHighHigh (for temozolomide)Strong
Cervical CarcinomaModerateUnder investigationPotentialEmerging
NeuroblastomaUnder investigationUnder investigationUnknownLimited
Oral Squamous CarcinomaUnder investigationUnder investigationUnknownLimited
Breast CancerUnder investigationUnder investigationUnknownLimited

Validating these biomarker applications will require large-scale, prospective clinical studies with standardized measurement techniques and comprehensive clinical annotation.

Product Science Overview

Introduction

Metallophosphoesterase Domain Containing 2 (MPPED2) is a protein encoded by the MPPED2 gene in humans. This protein is a member of the UPF0046 family and is known for its low metallophosphoesterase activity in vitro. MPPED2 is believed to play a role in the development of the nervous system and brain development .

Gene and Protein Structure

The MPPED2 gene is located on chromosome 11 at the 11p13 region, situated between the FSHB and PAX6 genes . The gene encodes a protein that consists of 317 amino acids and has a molecular mass of approximately 35.7 kDa . The recombinant form of this protein is produced in Escherichia coli and is purified using proprietary chromatographic techniques .

Function and Activity

MPPED2 displays low metallophosphoesterase activity, which means it has the ability to hydrolyze phosphoric diester bonds, although this activity is not very pronounced in vitro . The protein is predicted to enable manganese ion binding activity, phosphoric diester hydrolase activity, and purine ribonucleotide binding activity . These functions suggest that MPPED2 may be involved in various biochemical pathways, particularly those related to the nervous system.

Clinical Significance

MPPED2 has been associated with several diseases, including WAGR syndrome, diabetic cataract, and common warts . WAGR syndrome is a pathological condition characterized by the presence of Wilms tumor, aniridia, genitourinary anomalies, and mental retardation . The deletion of the chromosomal region containing the MPPED2 gene has been linked to this syndrome .

Research and Applications

Recombinant MPPED2 is used primarily for research purposes. It is not intended for use in diagnostic or therapeutic procedures . Researchers utilize this protein to study its role in the nervous system and its potential involvement in various diseases. The recombinant protein is available in a purified form and is typically stored at -20°C for long-term use .

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.