EMCN Human

Endomucin Human Recombinant
Shipped with Ice Packs
In Stock

Description

Protein Characteristics of EMCN Human

Molecular Composition

  • Recombinant EMCN Human (PRO-1156) contains 197 amino acids (residues 19-190) with a 25-amino acid His-tag at the N-terminus .

  • Molecular mass: 20.5 kDa .

  • Expressed in E. coli, purified via chromatographic techniques .

PropertySpecification
Purity>90% (SDS-PAGE)
Storage4°C (short-term), -20°C (long-term)
Formulation0.25 mg/mL in Tris-HCl (pH 8.0)
StabilitySensitive to freeze-thaw cycles; carrier proteins (HSA/BSA) recommended for storage

Angiogenesis Regulation

  • Modulates VEGF-induced signaling by interacting with VEGFR2. Truncation mutants lacking N-glycosylation fail to bind VEGFR2, impairing endothelial cell migration and tube formation .

  • Knockdown reduces endothelial cell growth by 85–93% and suppresses VEGF-driven angiogenesis .

Immune Modulation

  • Inhibits leukocyte-endothelial adhesion by disrupting LFA-1/ICAM-1 interactions. EMCN-deficient mice exhibit increased CD45+ leukocyte infiltration in kidneys .

  • Acts as a ligand for L-selectin, regulating leukocyte rolling under shear stress .

Vascular Permeability

  • EMCN knockout mice develop albuminuria and disrupted glomerular filtration barriers due to podocyte foot process effacement and endothelial fenestration disorganization .

Non-Endothelial Expression

  • Single-cell RNA sequencing identified EMCN expression in choroidal pericytes and fibroblasts, alongside endothelial cells. These fibroblasts exhibit distinct chemokine signaling profiles compared to EMCN-negative counterparts .

  • EMCN+ stromal cells are conserved in human choroid and mouse brain, suggesting broader roles in tissue homeostasis .

Knockout Phenotypes

  • Vascular Development: EMCN-deficient mice show delayed retinal vascularization and impaired choroidal endothelial cell proliferation .

  • Kidney Dysfunction: Global EMCN knockout leads to glomerular hypercellularity, podocin downregulation, and MAPK/ERK pathway suppression .

Clinical Implications

Kidney Disease

  • Loss of EMCN correlates with proteinuria and glomerulosclerosis, highlighting its role in maintaining endothelial-podocyte crosstalk .

Inflammatory Disorders

  • Overexpression suppresses TNF-α-induced leukocyte adhesion, suggesting therapeutic potential in conditions like uveitis .

Cancer

  • EMCN’s anti-adhesive properties may influence metastasis by modulating focal adhesion complexes .

Product Specs

Introduction
Endomucin, also known as endothelial sialomucin, plays a role in cell-cell and cell-extracellular matrix interactions. It is found in various organs including the heart, kidney, and lung.
Description
This recombinant EMCN protein, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 197 amino acids (residues 19-190), with a molecular weight of 20.5 kDa. The protein includes a 25 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The EMCN protein is supplied in a solution at a concentration of 0.25 mg/ml. The solution contains 20mM Tris-HCl buffer (pH 8.0), 10% glycerol, 1mM DTT, and 100mM NaCl.
Stability
For short-term storage (up to 4 weeks), the product can be kept at 4°C. For extended storage, it is recommended to freeze the protein at -20°C. Adding a carrier protein such as HSA or BSA (0.1%) is advisable for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity of the EMCN protein is greater than 90%, as assessed by SDS-PAGE.
Synonyms
Endomucin, Endomucin-2, Gastric cancer antigen Ga34, Mucin-14, MUC-14, EMCN, EMCN2, MUC14.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMNSTGV LEAANNSLVV TTTKPSITTP NTESLQKNVV TPTTGTTPKG TITNELLKMS LMSTATFLTS KDEGLKATTT DVRKNDSIIS NVTVTSVTLP NAVSTLQSSK PKTETQSSIK TTEIPGSVLQ PDASPSKTGT LTSIPVTIPE NTSQSQVIGT EGGKNASTSA TSRSYSS.

Q&A

What is human EMCN and what is its basic molecular structure?

Human Endomucin (EMCN) is a 261 amino acid transmembrane sialomucin glycoprotein with a molecular mass of approximately 20.5 kDa in its non-glycosylated form . The protein contains a single transmembrane domain and undergoes extensive post-translational modifications, particularly glycosylation, resulting in an apparent molecular mass of 55-100 kDa in its mature form . Methodologically, researchers can assess EMCN's structure through techniques such as SDS-PAGE, which reveals its glycosylation-dependent migration pattern, and western blot analysis using validated anti-EMCN antibodies such as SDBC0008N-G or L6H10 . For structural studies, recombinant EMCN can be produced in expression systems like E. coli (for non-glycosylated protein) or HEK293 cells (for glycosylated variants) .

How does glycosylation affect EMCN function and detection methods?

Glycosylation substantially impacts EMCN's molecular weight, causing it to migrate at 55-100 kDa despite its 20.5 kDa core protein mass . This post-translational modification is crucial for EMCN's biological functions in cell adhesion and extracellular matrix interactions . For experimental approaches, researchers should be aware that detection methods may be affected by glycosylation status. When performing western blots, glycosidase treatments can help distinguish glycosylation-dependent changes. For functional studies, comparing glycosylated forms (from mammalian expression systems like HEK293) with non-glycosylated variants (from bacterial expression) can isolate glycosylation-dependent functions . Flow cytometric analysis should account for potential epitope masking by glycans, and antibody selection should consider recognition of glycosylated versus core protein epitopes .

What are the validated antibodies and detection methods for human EMCN?

For human EMCN detection, multiple validated antibodies have been confirmed through cross-validation studies. Specifically, two independent rat anti-human EMCN monoclonal antibodies (SDBC0008N-G and L6H10) have been tested against both human bone marrow cells and recombinant human EMCN protein . For flow cytometry applications, these antibodies effectively stain immunophenotypic HSCs, while for western blot analyses, they recognize recombinant human EMCN protein . Methodologically, researchers should include appropriate positive controls (recombinant EMCN protein) and negative controls when establishing new detection protocols. For immunohistochemistry applications, optimization of antigen retrieval methods is particularly important given EMCN's glycosylation status. When designing experiments, researchers should consider that the apparent molecular weight of EMCN on SDS-PAGE will be 55-100 kDa for glycosylated forms rather than the core protein's 20.5 kDa .

How should recombinant human EMCN be handled and stored for experimental use?

Recombinant human EMCN is typically supplied as either a lyophilized powder or a sterile filtered colorless solution . For lyophilized preparations, reconstitution should be performed in sterile PBS (pH 7.4), with some preparations containing 5-8% trehalose as a protective agent . Once reconstituted, if not used within a month, the protein should be aliquoted and stored at -80°C to avoid repeated freeze-thaw cycles that can compromise protein integrity . For short-term storage (2-4 weeks), recombinant EMCN can be kept at 4°C . To enhance stability during long-term storage, addition of carrier proteins such as 0.1% human serum albumin (HSA) or bovine serum albumin (BSA) is recommended . When working with recombinant EMCN, researchers should verify protein quality through SDS-PAGE analysis, with expected purity of >90% . For functional studies, it's critical to verify that the recombinant protein maintains biological activity after storage and handling.

What experimental controls should be included when studying EMCN expression in human tissues?

Robust experimental design for EMCN expression studies requires multiple controls. Positive tissue controls should include samples known to express EMCN, such as venous and capillary endothelial cells or purified HSCs . Negative controls should include lineage-committed HPCs, which have been demonstrated to lack EMCN expression . For antibody validation, testing against recombinant human EMCN protein is essential, and using multiple independent antibodies (such as SDBC0008N-G and L6H10) can help confirm specificity . To address potential false positives from non-specific binding, isotype control antibodies matched to the EMCN-specific antibodies should be included . When performing transcriptomic analyses, appropriate housekeeping genes must be selected for normalization. For single-cell studies, careful attention to potential cell doublets is critical, as apparent co-expression of EMCN with other markers could represent technical artifacts rather than biological reality .

How does EMCN expression differ between HSCs and HPCs, and what methodologies best capture these differences?

EMCN has emerged as a discriminating marker between hematopoietic stem cells (HSCs) and lineage-committed hematopoietic progenitor cells (HPCs) . While both cell populations express CD34, only HSCs demonstrate significant EMCN expression . This differential expression provides a more specific marker for HSCs than CD34 alone. Methodologically, flow cytometry using validated anti-EMCN antibodies (SDBC0008N-G or L6H10) combined with traditional HSC markers can identify these populations . Gene expression profiling through microarray or RNA-sequencing across sorted human cord blood (CB), fetal liver (FL), and bone marrow (BM) HSCs (lineage-negative, CD34+, CD38-, CD45RA-, CD90+) and HPCs (lineage-negative, CD34+, CD38+) has consistently demonstrated high EMCN transcript levels in HSCs compared to HPCs . For functional validation of EMCN as an HSC marker, transplantation assays using NOG mice have shown that lineage-negative, EMCN-positive (L-E+) cells demonstrate significant multilineage engraftment capacity, whereas lineage-negative, EMCN-negative (L-E-) cells do not, even when transplanted at 25-fold higher cell numbers .

What experimental approaches can confirm EMCN's functional role in human HSCs beyond correlation studies?

While correlative studies have established EMCN as a marker of human HSCs, determining its functional significance requires additional experimental approaches. Researchers should employ gene knockdown or knockout strategies using siRNA, shRNA, or CRISPR-Cas9 technologies in primary human HSCs or HSC-like cell lines to assess effects on stem cell properties. Phenotypic analyses should include colony-forming unit assays, cell proliferation, apoptosis assessment, and differentiation potential following EMCN manipulation . For in vivo functional studies, xenotransplantation of EMCN-manipulated human HSCs into immunocompromised mice (such as NOG mice) enables assessment of long-term multilineage engraftment capacity . Rescue experiments reintroducing wild-type or mutant EMCN into knockout cells can establish structure-function relationships. Additionally, protein interaction studies using techniques such as co-immunoprecipitation or proximity ligation assays can identify binding partners of EMCN in HSCs, potentially revealing mechanistic insights into its role in stem cell biology .

How might EMCN be incorporated into HSC purification protocols for research and clinical applications?

EMCN represents a promising marker for improved HSC isolation strategies, potentially offering greater specificity than traditional markers like CD34 alone . For research applications, EMCN can be incorporated into flow cytometry-based sorting protocols using a simplified two-color approach (lineage markers and EMCN) . This could replace or complement more complex protocols that rely on multiple markers (CD34, CD38, CD45RA, CD90). For potential clinical applications, EMCN-based isolation could be integrated into existing automated cell separation technologies already employed in clinical settings . Methodologically, researchers should compare the purity and functional capacity of HSCs isolated using traditional markers versus EMCN-incorporated protocols through in vitro colony-forming assays and in vivo xenotransplantation models. Importantly, for autologous transplantation scenarios, EMCN-based sorting could potentially improve removal of CD34-expressing malignant cells, while for allogeneic transplantation, it could facilitate more effective T-cell depletion . Development of clinical-grade anti-EMCN antibodies would be necessary for translation to therapeutic applications.

What are the non-endothelial EMCN-expressing cell populations and how can they be experimentally distinguished?

Recent research has expanded our understanding of EMCN expression beyond endothelial cells. Single-cell RNA sequencing of choroidal tissue from adult wild-type mice has identified EMCN expression in choroidal pericytes and a subset of fibroblasts, in addition to endothelial cells . These non-endothelial EMCN-expressing cells do not express other endothelial markers such as PECAM1 or PODXL, distinguishing them from endothelial populations . Methodologically, researchers investigating non-endothelial EMCN expression should employ multi-parameter approaches combining EMCN detection with established markers for candidate cell types. For fibroblast populations, comparative gene expression analysis has revealed that EMCN-expressing fibroblasts exhibit high levels of chemokine and interferon signaling genes, while EMCN-negative fibroblasts are enriched in genes encoding extracellular matrix proteins . Similar non-endothelial EMCN expression patterns have been detected in published datasets from mouse brain and human choroid, suggesting evolutionary conservation of this expression pattern . To exclude technical artifacts such as cell doublets in single-cell sequencing data, researchers should verify absence of other endothelial markers in these populations and employ computational approaches to identify and remove potential doublets.

How does EMCN expression and function change during development and across different pathological conditions?

Developmental regulation of EMCN expression has been observed in multiple systems. During embryonic development, EMCN is expressed on murine HSCs but not erythroid HPCs, a pattern consistent with observations in human systems . To methodologically address developmental changes, researchers should design time-course studies across developmental stages, employing both transcriptomic and protein-level analyses. For pathological contexts, comparative studies between healthy tissues and disease models can reveal context-dependent alterations in EMCN biology. Since EMCN plays roles in angiogenesis and immune cell recruitment , researchers should particularly focus on vascular pathologies and inflammatory conditions. Experimental approaches should include immunohistochemical analyses of tissue samples, flow cytometric assessment of EMCN expression levels in isolated cell populations, and functional assays to determine whether EMCN's biological activities are altered in disease states. Correlation of EMCN expression patterns with clinical outcomes in patient samples can provide insights into potential prognostic significance.

What contradictions exist in current EMCN research literature and how might they be experimentally addressed?

While EMCN has traditionally been characterized as an endothelial marker, recent single-cell RNA sequencing has revealed expression in non-endothelial populations, including certain fibroblasts and pericytes . This apparent contradiction can be experimentally addressed through comprehensive co-localization studies using multiple independent EMCN antibodies alongside established markers for endothelial cells, fibroblasts, and pericytes. Another potential contradiction relates to EMCN's molecular weight, with the core protein predicted at 20.5 kDa but observed migration at 55-100 kDa due to glycosylation . Researchers can resolve this through glycosidase treatments prior to western blotting. For functional roles, EMCN has been described both as interfering with focal adhesion complexes and inhibiting cell-extracellular matrix interactions , while also playing a role in VEGF-mediated angiogenesis . These potentially conflicting functions could be reconciled through context-dependent studies across different cell types and conditions. Methodologically, side-by-side comparisons using standardized experimental approaches across multiple laboratories could help resolve contradictions in the literature.

How can single-cell technologies advance our understanding of EMCN biology?

Single-cell RNA sequencing has already revealed previously unrecognized EMCN-expressing cell populations, including choroidal pericytes and subsets of fibroblasts . Future research should expand these approaches across diverse tissue types and developmental stages to comprehensively map EMCN expression at single-cell resolution. Methodologically, integration of single-cell transcriptomics with single-cell proteomics, such as CyTOF or CODEX imaging, would provide multi-omic characterization of EMCN-expressing cells. For functional insights, single-cell ATAC-seq could identify regulatory elements controlling EMCN expression in different cellular contexts. Spatial transcriptomics approaches would preserve crucial information about the anatomical localization of EMCN-expressing cells relative to tissue structures and other cell types. Additionally, single-cell clonal tracing methods could reveal the developmental relationships between different EMCN-expressing populations. These advanced technologies should be complemented by computational approaches that can integrate multi-modal single-cell data to generate comprehensive models of EMCN biology across tissues and conditions.

What therapeutic applications might emerge from advanced understanding of EMCN biology?

EMCN's role as a specific marker for HSCs opens significant therapeutic possibilities, particularly in hematopoietic stem cell transplantation . Methodologically, developing clinical-grade anti-EMCN antibodies for immunomagnetic cell sorting could improve HSC purification for both autologous and allogeneic transplantation . For autologous transplantation in malignancy treatment, EMCN-based sorting could potentially reduce contamination with CD34-expressing malignant cells, while for allogeneic transplantation, improved HSC purification could reduce graft-versus-host disease through more effective T-cell depletion . Beyond transplantation, EMCN's roles in angiogenesis and immune cell recruitment suggest potential therapeutic targets for vascular and inflammatory disorders . Experimental approaches should include testing EMCN-targeting strategies in relevant disease models, such as pathological angiogenesis or chronic inflammation. Development of biologics that modulate EMCN function, such as function-blocking antibodies or recombinant EMCN fragments, could provide novel therapeutic tools. Additionally, gene therapy approaches targeting EMCN or its regulatory pathways might offer new treatment strategies for disorders involving dysregulated angiogenesis or inflammation.

Product Science Overview

Structure and Expression

Endomucin is a membrane-bound glycoprotein expressed luminally by endothelial cells that line postcapillary venules, which are primary sites of leukocyte recruitment during inflammation . The human recombinant form of Endomucin is produced in E. coli and is a single, non-glycosylated polypeptide chain containing 197 amino acids with a molecular mass of 20.5 kDa . It is often fused to a His-tag at the N-terminus for purification purposes .

Functional Role

Endomucin interferes with the assembly of focal adhesion complexes and inhibits interactions between cells and the extracellular matrix . It is essential for vascular endothelial growth factor (VEGF)-induced clathrin-mediated endocytosis and signaling of VEGF receptor 2 (VEGFR2) . This process is crucial for angiogenesis, the formation of new blood vessels from pre-existing ones, which is necessary for various homeostatic and pathological processes such as embryonic development, wound healing, and tumor growth .

Mechanism of Action

Endomucin interacts with the AP2 complex, which is essential for clathrin-mediated endocytosis . It does not affect clathrin recruitment to the AP2 complex following VEGF stimulation but is necessary for the interaction between VEGFR2 and the AP2 complex during endocytosis . This specificity is highlighted by the fact that Endomucin does not inhibit VEGFR1 and FGFR1 internalization or their downstream activities .

Research and Applications

Human recombinant Endomucin is used in various research applications, including blocking assays and control experiments . It is also studied for its role in preventing leukocyte-endothelial cell adhesion, which has implications for inflammatory responses .

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.