MEP1A Human

Meprin A Subunit Alpha Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene Structure and Localization

The MEP1A gene is located on human chromosome 6p21 , spanning ~45 kb with 14 exons and 13 introns. Only 6.7% (3 kb) of the gene corresponds to exon sequences . Tissue-specific expression is prominent in the kidney, colon, small intestine, and appendix .

FeatureDetails
Chromosomal Location6p21
Exons/Introns14 exons, 13 introns
mRNA Expression SitesKidney, colon, small intestine, appendix

Protein Structure and Domains

MEP1A is a multidomain protein synthesized as a precursor (746 amino acids) containing:

  • Catalytic domain (residues 62–263): Mediates proteolytic activity.

  • MAM domain (residues 264–433): Facilitates protein-protein interactions.

  • MATH domain (residues 434–593): Implicated in oligomerization .

  • EGF-like and transmembrane domains: Enable membrane anchoring .

Meprin A exists as homomeric (α subunits) or heteromeric (α and β subunits) complexes, forming multimers >1 MDa . Recombinant human MEP1A (rhMEP1A) is enzymatically active after trypsin activation, cleaving substrates like fluorogenic peptides .

Biological Functions

  • Proteolytic Activity: Hydrolyzes collagen, fibronectin, and cytokines, influencing tissue repair and inflammation .

  • Inflammatory Regulation: Knockout mice exhibit exacerbated dextran sulfate sodium (DSS)-induced colitis, linking MEP1A to inflammatory bowel disease (IBD) .

  • Cancer Progression: Promotes cell migration, invasion, and epithelial-mesenchymal transition (EMT) in hepatocellular carcinoma (HCC) and colorectal cancer (CRC) .

Cancer

Cancer TypeRole of MEP1APrognostic Value
Hepatocellular CarcinomaOverexpression correlates with tumor size, metastasis, and poor survival (HR=2.05)Independent risk factor for post-resection mortality
Colorectal CancerEnhances proliferation and invasion; linked to advanced TNM stagingReduced survival in high-expression cohorts
Ovarian CancerLow expression distinguishes primary from secondary mucinous adenocarcinomaDiagnostic marker for tumor origin

Inflammatory Bowel Disease (IBD)

  • Polymorphisms in MEP1A (e.g., rs1059276) are associated with ulcerative colitis .

  • Meprin-α mRNA is reduced in inflamed IBD mucosa, exacerbating tissue damage .

Research Tools and Applications

Recombinant human MEP1A (rhMEP1A) is used to study enzymatic mechanisms and drug targeting. Key properties include:

  • Molecular Weight: ~90 kDa (reduced conditions) .

  • Activity Assay: Activated by trypsin, with specific activity measured via fluorogenic substrates (e.g., MCA-Tyr-Val-Ala-Asp-Ala-Pro-Lys(DNP)-OH) .

Therapeutic and Diagnostic Potential

  • Biomarker: Elevated plasma MEP1A levels correlate with acute coronary syndrome and cancer progression .

  • Therapeutic Target: Inhibition reduces HCC and CRC cell invasion in preclinical models .

Product Specs

Introduction
Meprin A subunit alpha (MEP1A), a member of the peptidase M12A family, is a single-pass type I membrane protein. Its expression is abundant in kidney and intestinal epithelial cells, and it is secreted into the urinary tract and intestinal lumen. Additionally, MEP1A is present in leukocytes and, under specific conditions, in cancer cells. This protein plays a role in inflammatory processes and possesses the ability to proteolytically degrade extracellular matrix proteins and process bioactive proteins.
Description
Produced in Sf9 Baculovirus cells, MEP1A is a single, glycosylated polypeptide chain with a molecular weight of 67.4 kDa. It comprises 589 amino acids (22-601 a.a.). On SDS-PAGE, its molecular size appears between 70-100 kDa. The protein is expressed with a 6 amino acid His tag at the C-terminus and undergoes purification using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The MEP1A protein solution (0.25 mg/ml) is prepared in Phosphate Buffered Saline (pH 7.4) with 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, freezing at -20°C is recommended. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Avoid repeated freezing and thawing cycles.
Purity
Purity exceeds 85.0% as assessed by SDS-PAGE.
Synonyms
Meprin A Subunit Alpha, PABA Peptide Hydrolase, N-Benzoyl-L-Tyrosyl-P-Amino-Benzoic Acid Hydrolase Subunit Alpha, Meprin A, Alpha (PABA Peptide Hydrolase), Endopeptidase-2, EC 3.4.24.18, PPH Alpha, BA268F1.1 (Meprin A Alpha (PABA Peptide Hydrolase)), EC 3.4.24, PPHA, MEP1A.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
ADPVPIKYLP EENVHDADFG EQKDISEINL AAGLDLFQGD ILLQKSRNGL RDPNTRWTFP IPYILADNLG LNAKGAILYA FEMFRLKSCV DFKPYEGESS YIIFQQFDGC WSEVGDQHVG QNISIGQGCA YKAIIEHEIL HALGFYHEQS RTDRDDYVNI WWDQILSGYQ HNFDTYDDSL ITDLNTPYDY ESLMHYQPFS FNKNASVPTI TAKIPEFNSI IGQRLDFSAI DLERLNRMYN CTTTHTLLDH CTFEKANICG MIQGTRDDTD WAHQDSAQAG EVDHTLLGQC TGAGYFMQFS TSSGSAEEAA LLESRILYPK RKQQCLQFFY KMTGSPSDRL VVWVRRDDST GNVRKLVKVQ TFQGDDDHNW KIAHVVLKEE QKFRYLFQGT KGDPQNSTGG IYLDDITLTE TPCPTGVWTV RNFSQVLENT SKGDKLQSPR FYNSEGYGFG VTLYPNSRES SGYLRLAFHV CSGENDAILE WPVENRQVII TILDQEPDVR NRMSSSMVFT TSKSHTSPAI NDTVIWDRPS RVGTYHTDCN CFRSIDLGWS GFISHQMLKR RSFLKNDDLI IFVDFEDITH LSQHHHHHH.

Q&A

What is the genomic structure and chromosomal location of the human MEP1A gene?

The human MEP1A gene maps to chromosome 6p21 and spans approximately 45 kilobases. The gene consists of 14 exons and 13 introns, with exon sequences constituting about 6.7% (3 kb) of the total gene . The zinc-catalytic center of MEP1A is encoded by exon 7, which is a critical region for its enzymatic function . This genomic organization is important for understanding regulatory mechanisms controlling MEP1A expression in different physiological and pathological contexts.

In which human tissues is MEP1A normally expressed?

MEP1A expression has been detected in several human tissues. Initially recognized in the kidney and gastrointestinal tract, dot blot analysis of poly(A) RNA from 50 different human tissues revealed MEP1A mRNA in kidney and appendix, in addition to previously identified expression in colon and small intestine . The protein is highly expressed on renal brush border cell membranes and intestinal epithelial cells . Understanding this tissue-specific expression pattern is essential for interpreting MEP1A's physiological roles and anticipating potential off-target effects when developing MEP1A-targeted therapeutics.

How does MEP1A contribute to abdominal aortic aneurysm (AAA) pathophysiology?

MEP1A plays a critical role in AAA formation through multiple mechanisms. Research demonstrates that MEP1A mediates AAA formation primarily by regulating mast cell secretion of TNF-α, which subsequently promotes matrix metalloproteinase (MMP) expression and apoptosis in smooth muscle cells (SMCs) . Increased MEP1A expression has been observed in both human AAA tissues and angiotensin II-induced mouse AAA tissues . Mechanistically, MEP1A is expressed mainly in mast cells where it mediates TNF-α expression and secretion. The released TNF-α enhances MMP2 expression in SMCs and promotes SMC apoptosis, leading to elastic lamina degradation and AAA progression .

What experimental approaches have been used to establish MEP1A as a susceptibility gene for atherosclerosis?

Researchers have employed a multi-faceted approach to establish MEP1A as an atherosclerosis susceptibility gene. This includes:

  • Bioinformatic analysis to prioritize MEP1A as a candidate gene for atherosclerosis susceptibility locus (Ath49) on mouse chromosome 17

  • Analysis of SNPs in both coding and regulatory regions of MEP1A between founder strains used to map Ath49

  • Generation of MEP1A knockout mice (MEP1A-/-) and subsequent crossing with ApoE-/- mice to create double-knockout models

  • Comparative assessment of early and advanced atherosclerotic lesion development between MEP1A-/- ApoE-/- mice and control ApoE-/- mice

  • Histological evaluation of plaque stability features (intraplaque necrosis, inflammatory cell content)

  • Measurement of circulating biomarkers including CXCL5 and markers of oxidative stress

These methodological approaches demonstrated that genetic ablation of MEP1A significantly decreased both early and advanced atherosclerotic lesion sizes in ApoE-/- mice and altered histological features associated with plaque stability .

How is MEP1A dysregulated in different types of human cancers?

MEP1A shows distinct expression patterns across various cancer types. In hepatocellular carcinoma (HCC), MEP1A mRNA expression is significantly increased in tumor tissue compared to paired adjacent non-neoplastic tissues and non-tumor liver tissues . Immunohistochemical analysis of tissue samples from 394 HCC patients revealed that positive expression of MEP1A in tumor cells serves as an important risk factor affecting survival after radical resection .

MEP1A expression has also been reported in human prostate cancer cell lines, where it promotes cell replication and invasion . This diverse expression pattern across multiple cancer types suggests MEP1A may serve as a valuable biomarker and potential therapeutic target.

What experimental methods can researchers use to study MEP1A's role in tumor progression?

To investigate MEP1A's role in tumor progression, researchers should consider multiple complementary approaches:

  • Expression analysis:

    • RT-PCR, western blotting, and immunohistochemistry to detect MEP1A in tumor tissues compared to normal tissues

    • Analysis of public cancer genomics databases to assess MEP1A expression across cancer types

  • Functional studies:

    • Gene knockdown or knockout using siRNA, shRNA, or CRISPR-Cas9 techniques to inhibit MEP1A expression in cancer cell lines

    • Overexpression studies using plasmid vectors containing MEP1A cDNA

    • Assays to measure cell proliferation, invasion, migration, and apoptosis following MEP1A manipulation

  • In vivo models:

    • Xenograft models using MEP1A-manipulated cancer cell lines

    • Genetic mouse models with tissue-specific MEP1A alterations

  • Mechanism investigation:

    • Substrate identification using proteomic approaches

    • Signaling pathway analysis to determine downstream effectors of MEP1A activity

    • Enzyme-linked immunosorbent assays to measure inflammatory cytokines regulated by MEP1A

  • Clinical correlation:

    • Analysis of MEP1A expression in relation to clinicopathological features and patient outcomes

    • Evaluation of MEP1A as a biomarker for tumor progression and treatment response

What are the recommended methods for detecting MEP1A expression in tissue samples?

For comprehensive MEP1A expression analysis in tissue samples, researchers should employ multiple complementary techniques:

  • mRNA expression analysis:

    • Real-time PCR (RT-PCR) for quantitative measurement of MEP1A mRNA levels

    • Dot blot analysis of poly(A) RNA for tissue-specific expression patterns

    • RNA sequencing for genome-wide expression profiling

  • Protein expression analysis:

    • Western blotting using specific antibodies against MEP1A and appropriate loading controls (e.g., β-actin)

    • Immunohistochemistry for spatial localization within tissues

    • Immunofluorescence microscopy for co-localization with other proteins of interest

  • Enzymatic activity assays:

    • Biochemical assays to measure MEP1A metalloendopeptidase activity

    • Zymography to assess proteolytic activity in tissue extracts

  • Sample preparation:

    • For protein extraction from tissues: mechanical disruption in liquid nitrogen followed by lysis buffer treatment and centrifugation

    • For histological analysis: proper fixation and sectioning of tissues

These methodologies should be selected based on specific research questions, available resources, and the nature of the tissues being examined.

How can researchers effectively generate and validate MEP1A knockout models for functional studies?

Generating and validating MEP1A knockout models requires systematic methodology:

  • Knockout strategy design:

    • Target critical functional regions (e.g., exon 7 encoding the zinc-catalytic center)

    • Consider potential compensatory mechanisms, particularly from related proteins like MEP1B

  • Generation techniques:

    • Traditional embryonic stem cell targeting using neomycin cassette insertion

    • CRISPR-Cas9 genome editing for more precise modifications

    • Conditional knockout systems (Cre-loxP) for tissue-specific or inducible deletion

  • Validation methods:

    • Genotyping using PCR to confirm genetic modifications

    • RT-PCR and western blotting to verify absence of MEP1A expression

    • Functional assays to confirm loss of MEP1A enzymatic activity

    • Short tandem repeat analysis to confirm genetic background (especially important for chromosome 17 where MEP1A is located near the H2 locus)

  • Phenotypic characterization:

    • Comprehensive histological analysis of tissues known to express MEP1A

    • Examination under both normal conditions and disease models (e.g., AAA, atherosclerosis, inflammatory conditions)

    • Measurement of relevant biomarkers such as inflammatory cytokines and oxidative stress indicators

  • Control considerations:

    • Use of littermate controls to minimize genetic background effects

    • Consider both heterozygous and homozygous knockout animals to assess gene dosage effects

    • Potential rescue experiments by reintroducing MEP1A to confirm specificity of observed phenotypes

How does MEP1A influence inflammatory pathways in disease processes?

MEP1A exerts complex effects on inflammatory pathways that vary by disease context:

  • Cardiovascular inflammation:

    • In AAA, MEP1A enhances TNF-α secretion by mast cells, promoting matrix metalloproteinase (MMP) expression and smooth muscle cell apoptosis

    • MEP1A deficiency reduces elastic lamina degradation and SMC apoptosis in AAA tissues

    • In atherosclerosis, MEP1A deficiency lowers circulating levels of proinflammatory chemokine CXCL5 and reduces oxidative stress

  • Intestinal inflammation:

    • MEP1A plays a protective role in inflammatory bowel disease, with MEP1A knockout mice showing more severe intestinal damage and inflammation than wild-type mice

    • Genetic association exists between MEP1A and ulcerative colitis in patient cohorts

  • Mechanistic considerations:

    • As a metalloendopeptidase, MEP1A likely influences inflammation through proteolytic processing of cytokines, chemokines, and their receptors

    • MEP1A may regulate extracellular matrix composition and remodeling through cleavage of matrix proteins including pro-collagen I, pro-collagen III, fibronectin, and collagen IV

    • Context-dependent effects suggest MEP1A may have both pro- and anti-inflammatory functions depending on tissue microenvironment

Understanding these diverse inflammatory mechanisms is crucial for targeting MEP1A therapeutically in different disease contexts.

What is the potential of MEP1A inhibitors as therapeutic agents for cardiovascular diseases and cancer?

MEP1A inhibitors show promising therapeutic potential across multiple disease contexts:

  • Cardiovascular applications:

    • Actinonin, a MEP1A inhibitor, significantly inhibits TNF-α secretion in mast cells

    • MEP1A deficiency reduces AAA formation and increases survival rates in mouse models

    • In atherosclerosis models, MEP1A deletion decreases both early and advanced atherosclerotic lesion sizes

    • MEP1A inhibition could potentially stabilize existing plaques by reducing intraplaque necrosis and modulating inflammatory cell content

  • Cancer applications:

    • Inhibition of MEP1A expression suppresses cell proliferation and invasion in colorectal cancer both in vitro and in vivo

    • MEP1A inhibitors could potentially target multiple cancer types including colorectal, hepatocellular, and prostate cancers where MEP1A overexpression correlates with poor prognosis

  • Therapeutic development considerations:

    • Selective inhibitors need to distinguish between MEP1A and related metalloproteinases

    • Tissue-specific delivery systems might help target MEP1A in disease-relevant tissues while minimizing off-target effects

    • Combination approaches with established therapies should be explored

    • Potential biomarkers (e.g., MEP1A expression levels) could help identify patients most likely to benefit from MEP1A-targeted therapies

  • Safety considerations:

    • MEP1A's role in normal physiology, particularly in kidney and intestinal function, necessitates careful assessment of inhibitor safety profiles

    • The protective role of MEP1A in intestinal inflammation suggests potential adverse effects of inhibition in patients with inflammatory bowel conditions

Research into MEP1A inhibitors is still in early stages, but preliminary evidence supports their continued development as potential therapeutic agents for both cardiovascular diseases and cancer.

What are the key unanswered questions regarding MEP1A's physiological and pathological roles?

Despite significant advances, several critical questions about MEP1A remain unresolved:

  • Substrate specificity:

    • What are the complete repertoire of physiological substrates for MEP1A across different tissues?

    • How does substrate specificity differ between MEP1A and MEP1B?

    • What determines substrate recognition and cleavage efficiency?

  • Regulation mechanisms:

    • What transcriptional, post-transcriptional, and post-translational mechanisms regulate MEP1A expression and activity?

    • How is MEP1A secretion and localization controlled in different cell types?

    • What signaling pathways modulate MEP1A function?

  • Interactome mapping:

    • What proteins interact with MEP1A to form functional complexes?

    • How do these interactions influence MEP1A activity and substrate specificity?

  • Tissue-specific functions:

    • Why does MEP1A have seemingly opposite effects in different inflammatory contexts?

    • What explains MEP1A's protective role in intestinal inflammation but pathogenic role in cardiovascular inflammation?

  • Therapeutic applications:

    • Can tissue-specific MEP1A inhibition be achieved to target pathological processes while preserving beneficial functions?

    • What are the long-term consequences of MEP1A inhibition?

Addressing these questions will require integrated approaches combining structural biology, proteomics, genetic models, and clinical studies.

What innovative methodologies might advance our understanding of MEP1A biology?

Emerging technologies offer promising avenues to address knowledge gaps in MEP1A research:

  • Advanced genetic models:

    • Tissue-specific and inducible MEP1A knockout/knockin models to dissect context-dependent functions

    • Humanized mouse models expressing human MEP1A variants to better translate findings to human disease

    • CRISPR-based screens to identify genetic modifiers of MEP1A function

  • Structural and biochemical approaches:

    • Cryo-electron microscopy to resolve MEP1A structure at atomic resolution

    • Activity-based protein profiling to identify MEP1A substrates in complex biological samples

    • Proteomic degradomics to systematically map MEP1A cleavage sites across the proteome

  • Single-cell technologies:

    • Single-cell RNA sequencing to map MEP1A expression at cellular resolution in normal and diseased tissues

    • Spatial transcriptomics to understand MEP1A expression in tissue context

    • Single-cell proteomics to correlate MEP1A activity with cell state

  • Translational methodologies:

    • Development of sensitive biomarkers for MEP1A activity in biological fluids

    • High-throughput screening platforms for identifying selective MEP1A inhibitors

    • Patient-derived organoids to test MEP1A-targeted interventions in personalized disease models

  • Systems biology approaches:

    • Integration of multi-omics data to position MEP1A within broader disease networks

    • Mathematical modeling of MEP1A-regulated processes in inflammation and tissue remodeling

    • Network pharmacology to predict optimal combination therapies involving MEP1A inhibition

These innovative methodologies, alone or in combination, could significantly accelerate our understanding of MEP1A biology and facilitate translation of findings into clinical applications.

Product Science Overview

Introduction

Meprin A subunit alpha, also known as MEP1A, is a protein-coding gene that plays a crucial role in various biological processes. It is part of the meprin A complex, which is involved in proteolysis, the breakdown of proteins into smaller polypeptides or amino acids. This article provides a detailed overview of the background, structure, function, and significance of Meprin A subunit alpha, particularly focusing on its human recombinant form.

Gene and Protein Structure

The MEP1A gene encodes the Meprin A subunit alpha protein. This protein is a single-pass type I membrane protein that belongs to the peptidase M12A family . The gene is located on chromosome 6 in humans and is known for its metalloendopeptidase activity, which is essential for the degradation of extracellular matrix proteins .

The Meprin A subunit alpha protein contains several important domains:

  • EGF-like domain: Involved in protein-protein interactions.
  • MAM domain: Plays a role in cell adhesion and signaling.
  • MATH domain: Involved in protein-protein interactions and ubiquitination processes .
Biological Functions

Meprin A subunit alpha is primarily expressed in kidney and intestinal epithelial cells. It is secreted into the urinary tract and intestinal lumen and can also be found in leukocytes and cancer cells under certain conditions . The protein is capable of proteolytically degrading extracellular matrix proteins and processing bioactive proteins, which makes it significant in various physiological and pathological processes.

Recombinant Form

The recombinant form of Meprin A subunit alpha is produced using DNA sequences encoding the human MEP1A extracellular domain. This recombinant protein is often fused with a polyhistidine tag at the C-terminus to facilitate purification . The recombinant human Meprin A subunit alpha is expressed in HEK293 cells and is typically purified to a high degree of purity (>90%) as determined by SDS-PAGE .

Applications and Significance

Recombinant Meprin A subunit alpha has several applications in research and medicine:

  • Proteolysis Studies: Used to study the mechanisms of protein degradation and processing.
  • Disease Research: Investigated for its role in diseases such as rectosigmoid junction neoplasm and sigmoid neoplasm .
  • Drug Development: Potential target for developing therapeutic agents aimed at modulating its activity in various diseases.

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.