LGMN Human

Legumain Human Recombinant
Shipped with Ice Packs
In Stock

Description

Lysosomal Protein Degradation

LGMN facilitates endosomal/lysosomal degradation of proteins, including pro-cathepsins B, H, and L. Deficiency leads to accumulation of these pro-enzymes, impairing lysosomal function .

Antigen Presentation

LGMN processes bacterial and endogenous proteins for MHC class II presentation in dendritic cells, critical for adaptive immunity .

Disease Mechanisms

  • Cancer: Overexpressed in breast, cervical, and ovarian tumors, LGMN promotes invasion/metastasis by activating MMP-2 and TGF-β1 pathways. Knockout reduces migration in cervical cancer cells .

  • Cardiovascular: In thoracic aortic dissection (TAD), LGMN binds integrin αvβ3 in vascular smooth muscle cells (VSMCs), inhibiting Rho GTPase and triggering extracellular matrix degradation. Lgmn knockout mice show reduced TAD incidence .

  • Pain Modulation: LGMN induces oral cancer pain via biased agonism of protease-activated receptor 2 (PAR2), activating nociceptors .

Cancer Biomarker

  • Elevated LGMN correlates with advanced tumor grades:

    • Cervical Cancer: 84% of tumors show high LGMN vs. 41% in healthy tissue .

    • Breast Cancer: High expression linked to aggressive subtypes .

Cardiovascular Disease

  • Serum LGMN levels are elevated in TAD patients. Macrophage-specific Lgmn deletion mitigates aortic degeneration in mice .

  • In hypertension, LGMN impairs regulatory T-cell (Treg) function, exacerbating blood pressure elevation .

Therapeutic Targets

  • Inhibitors: Aza-Asn epoxides (AEPi) block LGMN activity, reducing tumor growth and pain in preclinical models .

  • Recombinant LGMN: Used in research to study protease mechanisms and drug development .

Key Research Findings

StudyModelKey Outcome
BAPN-induced TAD Lgmn−/− miceReduced aortic dissection via suppressed VSMC dedifferentiation
OSCC pain mechanism Human/mouse modelsLGMN activates PAR2, causing hyperexcitability of nociceptors; inhibition relieves pain
TGF-β1/MMP-2 activation Lung cancerLGMN promotes extracellular matrix deposition and metastasis

Applications and Future Directions

  • Diagnostics: LGMN serves as a prognostic marker for tumor malignancy .

  • Therapeutics: Targeting LGMN with inhibitors (e.g., RR-11a) shows promise in TAD and cancer .

  • Structural Insights: Crystal structures (e.g., 7FQK) guide inhibitor design for precision therapies .

Product Specs

Introduction
Legumain, also known as LGMN, is a cysteine endopeptidase that exhibits strict specificity for hydrolyzing asparaginyl bonds. While it primarily targets asparaginyl bonds, LGMN can also cleave aspartyl bonds, albeit at a slower rate, particularly in acidic environments. This enzyme plays a critical role in the endosomal/lysosomal degradation pathway. Notably, Legumain deficiency leads to the accumulation of pro-cathepsins B, H, and L, which constitute another group of lysosomal cysteine proteases. Moreover, the overexpression of LGMN in tumors is implicated in invasion and metastasis.
Description
LGMN, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain encompassing amino acids 18 to 433. It is fused to a 6 amino acid His Tag at the C-terminus, resulting in a total of 422 amino acids and a molecular mass of 48.4 kDa. On SDS-PAGE, the molecular size will appear approximately between 40-57 kDa. The purification of LGMN is achieved through proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The LGMN protein solution is provided at a concentration of 1 mg/ml and 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 longer storage, it is recommended to freeze the product at -20°C. To further enhance long-term stability, adding a carrier protein (0.1% HSA or BSA) is advised. It's crucial to avoid repeated freeze-thaw cycles to maintain product integrity.
Purity
The purity of the LGMN protein is greater than 95%, as determined by SDS-PAGE analysis.
Synonyms
Legumain, PRSC1, Protease, Cysteine, 1 (Legumain), Asparaginyl Endopeptidase, Protease, Cysteine 1, EC 3.4.22.34, Cysteine Protease 1, LGMN1, AEP.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
VPIDDPEDGG KHWVVIVAGS NGWYNYRHQA DACHAYQIIH RNGIPDEQIV VMMYDDIAYS EDNPTPGIVI NRPNGTDVYQ GVPKDYTGED VTPQNFLAVL RGDAEAVKGI GSGKVLKSGP QDHVFIYFTD HGSTGILVFP NEDLHVKDLN ETIHYMYKHK MYRKMVFYIE ACESGSMMNH LPDNINVYAT TAANPRESSY ACYYDEKRST YLGDWYSVNW MEDSDVEDLT KETLHKQYHL VKSHTNTSHV MQYGNKTIST MKVMQFQGMK RKASSPVPLP PVTHLDLTPS PDVPLTIMKR KLMNTNDLEE SRQLTEEIQR HLDARHLIEK SVRKIVSLLA ASEAEVEQLL SERAPLTGHS CYPEALLHFR THCFNWHSPT YEYALRHLYV LVNLCEKPYP LHRIKLSMDH VCLGHYHHHH HH.

Q&A

What is LGMN and what are its basic biological functions?

LGMN (legumain), also known as asparaginyl endopeptidase (AEP) or protease cysteine 1 (PRSC1), is an in vivo-active cysteine protease that catalyzes the degradation of numerous proteins . The LGMN gene is located on human chromosome 14q32.1, consisting of 14 exons and 13 introns that encode a 433-amino acid polypeptide . Functionally, LGMN participates in several critical physiological processes:

  • Protein degradation and processing

  • Extracellular matrix remodeling

  • Immune response regulation

  • Cell proliferation and differentiation

  • Vascular smooth muscle cell phenotype regulation

Current research indicates LGMN may act upstream of or within negative regulation of neuron apoptotic processes, growth regulation, and cellular responses to acidic pH conditions . The enzyme's activity is highly pH-dependent, becoming activated under acidic conditions typical of lysosomes where it primarily resides.

What is the structure and processing of the LGMN protein?

LGMN exists in three principal forms with distinct molecular weights and functions:

LGMN FormMolecular WeightActivity StatusLocation/Characteristics
Progenitor56 kDaInactiveSynthesized with signal peptide
Mature form 146 kDaActiveProduced by autocatalytic processing
Mature form 236 kDaActiveGenerated by further carboxyl terminal cleavage

The LGMN precursor consists of three functionally distinct domains:

  • Signal peptide: Guides LGMN to the endoplasmic reticulum for processing

  • Catalytic functional region: Contains three critical active site residues (His148, Cys189, and Asn42) that stabilize enzyme activity

  • Legumain stabilization and activity modulation (LSAM) domain: Ensures stability of the LGMN precursor through an electrostatic coded stabilization switch near the catalytic domain

The activation of LGMN is a pH-dependent process. The 56 kDa progenitor undergoes autocatalytic processing in acidic environments to generate the 46 kDa mature enzyme. Further proteolytic processing in lysosomes can produce the 36 kDa form, with both mature forms exhibiting comparable enzymatic activities .

What techniques are available for measuring LGMN expression and activity?

Researchers investigating LGMN have access to a variety of methodological approaches:

TechniqueApplicationAdvantagesLimitations
ELISAQuantification in serum, plasma, cell culture supernatantsHigh sensitivity (down to 0.094 ng/ml); specific for human LGMNLimited to protein detection, not activity
Western blottingProtein detection and form differentiationCan distinguish between different LGMN forms (56, 46, 36 kDa)Semi-quantitative; requires careful optimization
RT-qPCRmRNA expression analysisHigh sensitivity for transcript detectionDoes not reflect post-translational modifications or activity
Activity assaysMeasuring enzymatic functionDirect assessment of functional capacityRequires specific pH conditions for optimal activity
ImmunohistochemistryTissue localizationVisualizes spatial distributionDependent on antibody specificity
Co-immunoprecipitationProtein-protein interactionsIdentifies binding partners (e.g., with integrin αvβ3) May detect non-physiological interactions

When designing experiments to measure LGMN, researchers should consider that expression levels may not directly correlate with enzymatic activity due to the protein's complex activation requirements. The Human LGMN ELISA Kit offers exceptional sensitivity with a detection range of 0.156-10 ng/ml, making it suitable for precise quantification across various sample types .

How does LGMN contribute to cardiovascular disease development?

LGMN plays a significant role in cardiovascular disease pathogenesis, particularly in thoracic aortic dissection (TAD) and related conditions:

  • LGMN expression is upregulated in aortic tissue from patients with TAD and in mouse models of angiotensin II-induced abdominal aortic aneurysm (AAA)

  • LGMN deficiency has been associated with lower prevalence of cardiovascular diseases, including myocardial infarction

  • Mechanistically, LGMN promotes extracellular matrix degradation either directly or by activating downstream signaling pathways

  • LGMN regulates vascular smooth muscle cell (VSMC) phenotypic transformation from a contractile to synthetic type, which is critical in TAD development

Experimental evidence demonstrates that LGMN knockout or inhibition significantly ameliorates disease progression in mouse models of TAD. Specifically:

  • Both global LGMN knockout mice (Lgmn–/–) and macrophage-specific LGMN knockout mice (LgmnF/F;LysM Cre) showed resistance to BAPN-induced TAD

  • Chemical inhibition using RR-11a in C57BL/6 mice similarly protected against TAD progression

  • Macrophage-specific deletion of LGMN alleviated extracellular matrix degradation in BAPN-treated mice

These findings suggest that targeting LGMN may represent a promising therapeutic strategy for cardiovascular diseases characterized by vascular remodeling and extracellular matrix degradation.

What is LGMN's role in cancer development and progression?

LGMN has emerged as an important factor in cancer biology, with significant implications for tumor development, progression, and patient outcomes:

Molecular analyses have revealed that LGMN-associated differentially expressed genes in osteosarcoma are enriched in immune response and antigen processing pathways, suggesting LGMN's role in modulating anti-tumor immunity . The correlation between high LGMN expression and increased abundance of M2 macrophages (which typically exhibit immunosuppressive properties) further supports LGMN's potential involvement in creating an immunosuppressive tumor microenvironment .

Consensus Clustering Analysis has identified two distinct molecular subtypes of osteosarcoma based on LGMN expression patterns, highlighting tumor heterogeneity and suggesting potential for personalized therapeutic approaches . These findings collectively position LGMN as both a potential biomarker and therapeutic target in cancer management.

How does LGMN affect cellular differentiation and phenotype transformation?

LGMN regulates cellular differentiation processes in multiple contexts:

In vascular smooth muscle cells:

  • Macrophage-derived LGMN inhibits VSMC differentiation as demonstrated in coculture systems

  • LGMN binds to integrin αvβ3 in VSMCs, potentially blocking integrin-mediated signaling pathways critical for cellular differentiation

  • Gene Ontology analysis has shown that LGMN regulates VSMC phenotype transformation from contractile to synthetic type, a key process in vascular pathologies

In bone marrow stromal/stem cells:

  • LGMN influences the differentiation fate of human bone marrow stromal/stem cells (hBMSCs)

  • LGMN knockdown in hBMSCs does not alter early osteoblast commitment markers (ALP, Col1a1) but significantly upregulates late osteoblast maturation markers (BGLAP and integrin-related genes)

  • LGMN transduction systems have been used to successfully overexpress legumain in hBMSCs to study its effects on differentiation

These findings suggest that LGMN functions as a cellular fate regulator, potentially through interaction with cell surface receptors and subsequent modulation of downstream signaling pathways that control differentiation programs.

What experimental models are most effective for studying LGMN function?

Researchers have developed several experimental models to investigate LGMN function across different contexts:

Model TypeDescriptionApplicationsKey Research Findings
Knockout miceLgmn–/– (global knockout)
LgmnF/F;LysM Cre (macrophage-specific)
Cardiovascular disease
Immune function studies
Protected against TAD development
Reduced extracellular matrix degradation
Chemical inhibitionRR-11a–treated C57BL/6 micePharmacological intervention testingAmeliorated BAPN-induced TAD progression
Disease inductionBAPN administration (1 g/kg/d) for 4 weeks
Angiotensin II infusion
TAD modeling
AAA modeling
Revealed LGMN's role in aortic dissection pathogenesis
Cell-based modelsLGMN-transduced hBMSCs
LGMN knockdown (shLGMN)
Macrophage-VSMC coculture
Cell differentiation studies
Protein interaction analysis
Demonstrated LGMN's effect on osteoblast maturation
Showed macrophage-derived LGMN inhibits VSMC differentiation
Bioinformatic analysisCIBERSORT algorithm with LM22 signatureImmune cell infiltration analysisIdentified correlation between LGMN expression and M2 macrophage abundance

When selecting an experimental model, researchers should consider several factors:

  • The specific aspect of LGMN biology under investigation (expression, activity, interactions)

  • The disease context or physiological process of interest

  • Whether cell-autonomous or non-cell-autonomous effects are being studied

  • The need for in vivo validation versus cellular mechanistic insights

Combining multiple model systems (e.g., in vitro cell culture with in vivo mouse models) often provides the most comprehensive understanding of LGMN function.

What are the methodological challenges in LGMN research?

Investigating LGMN presents several methodological challenges that researchers should address:

  • Post-translational processing complexity:

    • LGMN exists in multiple forms (56, 46, and 36 kDa) requiring careful experimental design to distinguish between inactive precursors and active enzymes

    • Different antibodies may recognize specific forms, potentially leading to inconsistent results across studies

  • Activity measurement considerations:

    • LGMN activation is highly pH-dependent, requiring acidic conditions (pH 4.5-5.5) for optimal activity assays

    • Standard cell lysis buffers may not maintain appropriate pH conditions for activity measurements

    • The enzyme's activity in neutral pH environments (e.g., extracellular space) may differ from its typical lysosomal function

  • Experimental model limitations:

    • Cell line-based studies may not fully recapitulate the complex microenvironment where LGMN functions

    • Mouse models may have species-specific differences in LGMN expression and function

    • Tissue-specific expression patterns necessitate careful selection of experimental systems

  • Technical considerations for protein-protein interaction studies:

    • LGMN's interactions with proteins like integrin αvβ3 require specialized approaches such as coimmunoprecipitation and colocalization analyses

    • The pH-dependent nature of LGMN may affect its binding properties under different experimental conditions

To address these challenges, researchers should employ complementary approaches, carefully optimize experimental conditions, and validate findings across multiple systems.

How can bioinformatic approaches enhance LGMN research?

Bioinformatic methodologies have significantly advanced LGMN research, offering powerful tools for data analysis and hypothesis generation:

  • Differential expression analysis:

    • Microarray and RNA-seq data from resources like GEO and TARGET databases enable identification of LGMN expression patterns across tissues and disease states

    • The limma package has been successfully used to obtain differential expression matrices and visualize results through volcano plots and heatmaps

  • Functional enrichment analysis:

    • Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) help identify biological pathways and functions associated with LGMN

    • These approaches have revealed LGMN's involvement in immune response and antigen processing pathways in osteosarcoma

  • Network analysis:

    • Protein-protein interaction (PPI) networks provide insights into LGMN's functional interactions

    • Competing endogenous RNA (ceRNA) networks help understand regulatory mechanisms involving LGMN

  • Immune infiltration analysis:

    • The CIBERSORT algorithm combined with the LM22 signature matrix can quantify immune cell infiltration and its correlation with LGMN expression

    • This approach identified a relationship between high LGMN expression and increased M2 macrophage abundance in osteosarcoma

  • Consensus Clustering Analysis:

    • This technique has successfully identified distinct subtypes within osteosarcoma samples based on LGMN expression patterns

    • Such stratification may guide personalized medicine approaches and therapeutic targeting strategies

For researchers conducting bioinformatic analyses of LGMN, it's advisable to:

  • Integrate multiple data types (transcriptomic, proteomic, clinical)

  • Validate computational findings with experimental approaches

  • Consider tissue-specific and context-dependent effects when interpreting results

How might LGMN be targeted therapeutically in disease contexts?

LGMN represents a promising therapeutic target in several disease contexts, with various targeting strategies:

  • Small molecule inhibitors:

    • RR-11a has demonstrated efficacy in reducing BAPN-induced TAD progression in mouse models

    • Development of selective, cell-permeable inhibitors that can access lysosomal LGMN may provide therapeutic benefits

  • Genetic modulation approaches:

    • siRNA or shRNA targeting LGMN has shown efficacy in cellular models

    • Tissue-specific delivery systems could enable targeted LGMN knockdown in disease-relevant tissues

  • Targeting LGMN-interaction partners:

    • Disrupting LGMN's interaction with integrin αvβ3 might prevent downstream effects on vascular smooth muscle cells

    • Combination approaches targeting both LGMN and its key effectors may provide synergistic benefits

  • Leveraging LGMN for targeted drug delivery:

    • LGMN's elevated expression in tumors and tumor-associated macrophages makes it potentially useful for tumor-specific drug activation

    • Prodrugs designed to be activated by LGMN's enzymatic activity could achieve tumor-selective effects

When developing LGMN-targeted therapies, researchers should consider:

  • Tissue-specific expression patterns to minimize off-target effects

  • The pH-dependent nature of LGMN activation

  • Potential differences between human and model organism LGMN

  • The need for biomarkers to identify patients most likely to benefit from LGMN-targeted approaches

What is the diagnostic and prognostic potential of LGMN in human diseases?

LGMN shows considerable promise as a diagnostic and prognostic biomarker:

In cancer:

In cardiovascular disease:

  • LGMN levels are elevated in aorta and sera from patients with thoracic aortic dissection (TAD)

  • LGMN deficiency is associated with lower prevalence of cardiovascular diseases, including myocardial infarction

For clinical implementation, researchers should consider:

  • Standardization of LGMN measurement methods across laboratories

  • Establishment of reference ranges in healthy populations

  • Validation in larger, diverse patient cohorts

  • Integration with other biomarkers for improved diagnostic and prognostic accuracy

The Human LGMN ELISA Kit offers exceptional sensitivity (0.094 ng/ml) and a detection range of 0.156-10 ng/ml, making it suitable for quantifying LGMN in clinical samples such as serum and plasma . This may facilitate translation of LGMN as a biomarker into clinical practice.

What are the emerging areas of LGMN research with clinical relevance?

Several emerging research directions show particular promise for clinical translation:

  • LGMN in immune modulation:

    • The correlation between LGMN expression and immune cell infiltration, particularly M2 macrophages, suggests a role in shaping the immune microenvironment

    • This could have implications for immunotherapy responsiveness in cancer patients

  • LGMN in biomarker panels:

    • Integration of LGMN with other biomarkers might improve diagnostic and prognostic accuracy

    • Multi-omics approaches combining LGMN protein levels with other molecular signatures could enhance patient stratification

  • LGMN in cellular differentiation:

    • LGMN's role in regulating differentiation of both vascular smooth muscle cells and bone marrow stromal cells suggests broader implications in tissue homeostasis and regeneration

    • This could inform regenerative medicine approaches for vascular and skeletal disorders

  • LGMN in prodrug activation:

    • LGMN's elevated expression in tumors makes it a candidate for tumor-selective prodrug activation

    • Development of LGMN-activated therapeutic agents could improve efficacy while reducing systemic toxicity

  • LGMN inhibition strategies:

    • Beyond RR-11a, development of novel, selective LGMN inhibitors could provide therapeutic options for cardiovascular diseases and cancer

    • Combination approaches targeting LGMN alongside standard therapies may enhance treatment efficacy

Researchers pursuing these directions should focus on translational aspects including:

  • Establishing clinically relevant endpoints

  • Developing companion diagnostics for LGMN-targeted therapies

  • Addressing delivery challenges for LGMN modulators

  • Conducting appropriately powered validation studies in relevant patient populations

Product Science Overview

Discovery and Structure

Legumain was first discovered in beans and blood fluke (Schistosoma mansoni) before its mammalian version was described by Chen and co-workers in 1997 . Human legumain is synthesized as a 433 amino acid precursor with a signal peptide (residues 1-17). The proenzyme (residues 18-433) is expressed with an N-terminal His tag .

Function and Activity

Legumain plays a pivotal role in the degradation system of cells. Its activity is found in several tissues and is essential for the proper functioning of the lysosomal degradation pathway. The deficiency of legumain causes the accumulation of pro-cathepsins B, H, and L, which are other lysosomal cysteine proteases .

Legumain has anti-apoptotic effects in mammals and plays an antigen-presenting role in the inflammatory response. It can activate multiple signal pathways to promote cell apoptosis and migration, inflammatory response, and the development of atherosclerosis .

Recombinant Human Legumain

Recombinant human legumain is produced using a mouse myeloma cell line, NS0-derived human legumain/asparaginyl endopeptidase protein (Ile18-Tyr433), with an N-terminal 7-His tag . The recombinant form is used in various research applications, including studying its role in diseases and potential as a biomarker.

Applications and Research

Legumain is being studied for its potential as a biomarker for atherosclerosis due to its involvement in vascular remodeling, inflammatory response, plaque stability, and the degradation of the extracellular matrix . It is also being researched for its role in colorectal cancer and other 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 2024 Thebiotek. All Rights Reserved.