MECOM Human

MDS1 And EVI1 Complex Locus Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene Structure

The MECOM gene is located on chromosome 3q26.2, spanning 60–65 kb with 16 exons (10 protein-coding). It produces multiple mRNA variants through alternative splicing, including EVI1, MDS1/EVI1, and fusion transcripts like AML1/MDS1/EVI1 .

Protein Isoforms

The primary MECOM protein isoforms include:

IsoformSize (kDa)Key Features
EVI1145Contains 7 zinc finger motifs, repression domain, and acidic C-terminus
MDS1/EVI1180Fusion product with MDS1; retains transcription activation/repression domains
Δ32488Lacks zinc fingers 6–7 due to alternative splicing
−Rp9~135Missing 9 amino acids in the repression domain

These isoforms exhibit distinct DNA-binding and regulatory properties, influencing hematopoietic differentiation and oncogenesis .

Developmental Regulation

MECOM is essential for embryogenesis, particularly in cardiovascular and neural development. Knockout mice exhibit embryonic lethality with defects in heart, kidney, and neural tube formation. In humans, MECOM is highly expressed in fetal tissues (kidney, lung, heart) but minimally in adults .

Hematopoiesis

  • Stem Cell Maintenance: Sustains hematopoietic stem cell (HSC) self-renewal via interactions with SMAD3, CREBBP, and MAPK pathways .

  • Differentiation Regulation:

    • Inhibits granulocyte/erythroid differentiation.

    • Promotes megakaryocyte lineage commitment .

Epigenetic and Transcriptional Control

Recent studies identify MECOM as a master regulator of endothelial cell (EC) identity. It binds enhancers forming chromatin loops to activate EC-specific genes (e.g., VEGF) and suppress non-arterial markers .

MECOM-Associated Syndromes

Germline MECOM mutations cause radioulnar synostosis with amegakaryocytic thrombocytopenia 2 (RUSAT2), characterized by:

  • Hematologic Defects: Congenital thrombocytopenia, progressing to pancytopenia and bone marrow failure .

  • Skeletal Abnormalities: Radioulnar synostosis, clinodactyly .

  • Somatic Genetic Rescue: Some patients exhibit clonal hematopoiesis via copy-neutral loss of heterozygosity at 3q, restoring wild-type MECOM expression .

Oncogenic Role

MECOM overexpression is linked to aggressive myeloid malignancies (e.g., AML, MDS) and solid tumors. Key mechanisms include:

  • Chromosomal translocations (e.g., t(3;21)(q26;q22) forming AML1-MDS1-EVI1 fusions) .

  • Transcriptional repression of tumor suppressors (e.g., TGF-β pathway genes) .

Endothelial Lineage Regulation

A 2023 study demonstrated that MECOM depletion disrupts human EC differentiation and zebrafish angiogenesis. Hi-C/ChIP-seq analyses revealed MECOM-bound enhancers loop to EC identity genes like VEGF .

Mutation Spectrum in Disease

Mutation TypeClinical PhenotypeExample Variants
Zinc finger missenseRUSAT2 with skeletal defectsp.Arg938Gln, p.Arg1036*
Frameshift/nonsenseIsolated thrombocytopenia or BMFp.Glu566*, exon 4 deletions
Somatic rescueAsymptomatic carriers or late-onset disease3q LOH

Protein Engineering and Tools

Recombinant MECOM proteins (e.g., AA 1-169 with His tag) are used for functional studies. Key characteristics:

  • Source: E. coli-expressed, >85% purity .

  • Applications: DNA-binding assays, antibody development .

Future Directions

  • Targeting MECOM-driven enhancer dysregulation in leukemia.

  • Exploring somatic rescue mechanisms to mitigate bone marrow failure .

Product Specs

Introduction
MDS1, a transcriptional regulator and oncoprotein, plays a crucial role in various cellular processes such as hematopoiesis, development, cell differentiation, apoptosis, and proliferation. It interacts with several proteins, including CTBP1, SMAD3, CREBBP, KAT2B, MAPK8, and MAPK9. Notably, MDS1 overexpression due to translocation with the AML1 gene has been linked to leukemia development. Multiple transcript variants encoding different isoforms of MDS1 have been identified.
Description
Recombinant MECOM protein, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 192 amino acids (specifically, amino acids 1-169) and exhibits a molecular weight of 21.1 kDa. The MECOM protein is fused to a 23 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear solution that has undergone sterile filtration.
Formulation
The MECOM protein solution has a concentration of 0.25 mg/ml and is prepared in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.4 M Urea, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the MECOM protein should be kept at 4°C. For extended storage, it is recommended to freeze the protein at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Avoid repeated freeze-thaw cycles to maintain protein integrity.
Purity
The purity of the MECOM protein is greater than 85%, as assessed by SDS-PAGE analysis.
Synonyms
MDS1 And EVI1 Complex Locus, Ecotropic Virus Integration Site 1 Protein Homolog, Myelodysplasia Syndrome-Associated Protein 1, AML1-EVI-1, MDS1-EVI1, EVI1, MDS1, PRDM3, AML1-EVI-1 Fusion Protein, MDS1 And EVI1 Complex Locus Protein EVI1, MDS1 And EVI1 Complex Locus Protein MDS1, Oncogene EVI1, Zinc Finger Protein Evi1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMRSKGRA RKLATNNECV YGNYPEIPLE EMPDADGVAS TPSLNIQEPC SPATSSEAFT PKEGSPYKAP IYIPDDIPIP AEFELRESNM PGAGLGIWTK RKIEVGEKFG PYVGEQRSNL KDPSYGWEVH LPRSRRVSVH SWLYLGKRSS DVGIAFSQAD VYMPGLQCAF LS

Q&A

What is MECOM and what is its primary biological function in humans?

MECOM (MDS1 and EVI1 complex locus) functions as a critical endothelial cell lineage regulator that plays essential roles in vascular development and function. Research demonstrates that MECOM binds to enhancers that form chromatin loops to regulate endothelial cell identity genes . Through integrative analysis of multiple genomic datasets, MECOM has been identified as a key regulator of the VEGF signaling pathway, which is central to endothelial cell development . Beyond endothelial regulation, MECOM also serves crucial functions in hematopoietic stem cells (HSCs), with its depletion resulting in profound HSC loss . The gene's evolutionary conservation across species suggests its involvement in fundamental biological processes .

How is MECOM expression distributed across normal human tissues?

MECOM expression analysis across multiple datasets (HPA, GTEx, and FANTOM5) reveals a broad tissue distribution pattern with varying intensity:

Tissue/Cell TypeRelative MECOM ExpressionData Source
StomachHighest mRNA expressionHPA, GTEx, FANTOM5
KidneysHigh expressionHPA, GTEx, FANTOM5
LungsHigh expressionHPA, GTEx, FANTOM5
Alveolar cell type 2Highest cellular expressionSingle-cell analysis
Collecting duct cellsHigh cellular expressionSingle-cell analysis

Immunohistochemistry (IHC) results from the Human Protein Atlas generally corroborate the mRNA expression patterns, showing strong MECOM staining in normal prostate, kidney, lung, and breast tissues compared to their tumor counterparts . This broad but differential tissue expression pattern suggests context-specific roles for MECOM in various human tissues.

What experimental techniques are most effective for studying MECOM function?

Multiple complementary approaches have proven effective for investigating MECOM:

  • Genomic and Epigenomic Methods:

    • Single-cell RNA sequencing to analyze expression at cellular resolution

    • ChIP-Seq to identify MECOM binding sites in the genome

    • Hi-C analysis to study chromatin interactions mediated by MECOM

    • DNase-Seq to assess chromatin accessibility at MECOM-regulated regions

  • Functional Manipulation:

    • siRNA-mediated knockdown in human embryonic stem cell-derived endothelial cells (hESC-ECs)

    • CRISPR-Cas9 gene editing to study loss-of-function effects in hematopoietic stem cells

  • In Vivo Models:

    • Zebrafish models to assess angiogenesis defects resulting from MECOM depletion

    • Serial transplantation assays to evaluate long-term hematopoietic stem cell function

  • Computational Approaches:

    • RNA velocity analysis to predict cellular dynamics using scVelo

    • Trajectory inference using tools like Slingshot

    • Protein-protein interaction network analysis using STRING database

How can researchers accurately analyze MECOM's role in endothelial cell differentiation?

The analysis of MECOM in endothelial differentiation requires multi-modal approaches:

  • Single-Cell Resolution Analysis:

    • Implement single-cell RNA-Seq to verify that MECOM-positive cells are exclusively enriched in bona fide endothelial cells derived from induced pluripotent stem cells

    • Apply pseudobulk gene expression analysis to minimize the confounding influence of allelic dropout

    • Use RNA velocity analysis to predict direction and magnitude of cellular dynamics during differentiation

  • Functional Validation:

    • Perform siRNA-mediated knockdown of MECOM in Day 7 human embryonic stem cell-derived endothelial cells (hESC-EC) using transfection with predesigned siRNA at 5 nM concentration

    • Isolate CD144+ hESC-EC by magnetic activated cell sorting after transfection to assess pure endothelial populations

    • Quantify changes in endothelial marker expression and function following MECOM depletion

  • Molecular Pathway Analysis:

    • Integrate findings from Hi-C, DNase-Seq, ChIP-Seq, and RNA-Seq data to identify how MECOM binds enhancers that form chromatin loops to regulate endothelial cell identity genes

    • Use DESeq2 to identify differentially expressed genes following MECOM knockdown, focusing on those with absolute fold change values >1.5 and adjusted P-values <0.05

    • Apply clusterProfiler with Benjamini-Hochberg correction to identify over-represented KEGG pathways among MECOM-regulated genes

What approaches reveal MECOM's function in hematopoietic stem cell regulation?

Research into MECOM's role in hematopoiesis requires specialized methodologies:

  • Genetic Perturbation and Functional Assessment:

    • Implement CRISPR-Cas9 gene editing to create heterozygous MECOM edits in hematopoietic stem cells

    • Verify editing efficiency at both DNA and RNA levels using PCR and sequencing

    • Assess transcriptional consequences of MECOM editing, including potential nonsense-mediated decay

  • In Vivo Functional Analysis:

    • Perform primary and secondary transplantation assays to evaluate the long-term repopulating capacity of MECOM-edited hematopoietic stem cells

    • Use human-specific PCR primers to sensitively detect human cells in secondary recipient animals

    • Sequence recovered cells to determine the persistence of MECOM edits in serially-repopulating stem cells

  • Transcriptional Network Analysis:

    • Conduct differential gene expression analysis between MECOM-edited and control cells

    • Implement permutation testing to ensure robust identification of differentially expressed genes

    • Perform gene set enrichment analysis to identify biological pathways affected by MECOM depletion

How should researchers approach MECOM expression analysis across cancer types?

Cancer studies of MECOM require sophisticated analytical frameworks:

Significant survival differences based on MECOM expression:

Cancer TypeHigh MECOM ExpressionP-value
CHOL (Cholangiocarcinoma)Worse OS0.044
KIRP (Kidney renal papillary cell carcinoma)Worse OS and DFS0.015 (OS), 0.013 (DFS)
LGG (Brain lower grade glioma)Worse OS and DFS0.00044 (OS), 0.04 (DFS)
PAAD (Pancreatic adenocarcinoma)Worse OS0.042
LUAD (Lung adenocarcinoma)Better OS0.041
OV (Ovarian cancer)Better OS0.046
UCS (Uterine carcinosarcoma)Worse DFS0.046

What methodologies best identify genes and pathways regulated by MECOM?

Identifying MECOM's regulatory targets requires integrative approaches:

  • Multi-Omic Data Integration:

    • Combine Hi-C, DNase-Seq, ChIP-Seq, and RNA-Seq data to comprehensively map MECOM's regulatory landscape

    • Apply protein-protein interaction network analysis using STRING to identify proteins that potentially interact with MECOM

    • Use GEPIA2's 'Similar Gene Detection' module to determine the top MECOM-correlated target genes

  • Regulatory Network Inference:

    • Implement SCENIC workflow for gene regulatory analysis in single-cell data

    • Calculate pairwise gene-gene Pearson correlation analyses using GEPIA2's 'Correlation Analysis' module

    • Generate heatmaps of selected genes with their partial correlations and P-values using TIMER2's 'Gene_Corr' module

  • Functional Validation Protocol:

    • Perform siRNA-mediated knockdown of MECOM followed by bulk RNA sequencing

    • Map and quantify reads using RSEM and the GENCODE primary assembly transcriptome

    • Process isoform read counts with tximport and DESeq2 to obtain gene-level P-values and fold changes

    • Focus on genes with absolute fold change >1.5 and adjusted P-value <0.05

Research has identified the VEGF signaling pathway as a key target of MECOM regulation, which was verified through integrative analysis of genomic data .

How can MECOM research inform regenerative medicine approaches?

MECOM's critical role in endothelial cell development presents significant implications for regenerative medicine:

  • Therapeutic Target Identification:

    • MECOM functions as an endothelial lineage regulator, making it a prime therapeutic candidate for vascular regeneration

    • Understanding MECOM's role in maintaining the transcriptional identity of arterial endothelial cells can guide directed differentiation protocols

  • Optimization of Endothelial Cell Generation:

    • Protocols for human embryonic stem cell-derived endothelial cell (hESC-EC) differentiation can be refined based on MECOM expression patterns

    • Monitoring MECOM expression during differentiation can serve as a quality control measure for generating functional endothelial cells

  • Disease Modeling Approaches:

    • MECOM dysfunction is associated with vascular development disorders and embryonic lethality

    • Creating experimental models with controlled MECOM expression can help study these conditions and test potential interventions

How does MECOM haploinsufficiency contribute to human disease?

MECOM haploinsufficiency has significant pathological implications:

  • Hematopoietic Consequences:

    • In serial transplantation studies, MECOM-edited hematopoietic stem cells show profound loss of repopulating capacity

    • This finding is consistent with HSC loss observed in patients with MECOM haploinsufficiency

    • Analysis reveals near complete absence of MECOM edits in serially-repopulating long-term HSCs, indicating strong selective pressure against MECOM reduction

  • Clinical Manifestations:

    • MECOM testing is included in panels for bone marrow failure syndrome, thrombocytopenia, and bleeding disorders, suggesting its role in these conditions

    • MECOM germline variants may contribute to skeletal dysplasias and growth disorders

  • Research Models:

    • Zebrafish models with MECOM depletion show impaired angiogenesis, providing an in vivo system to study vascular defects

    • Human embryonic stem cell-derived endothelial cells with MECOM knockdown can model aspects of vascular disease

What are the current challenges and limitations in MECOM research?

Researchers face several methodological and conceptual challenges:

  • Technical Limitations:

    • Single-cell genotyping frequently encounters allelic dropout, potentially underestimating the true percentage of heterozygous edits

    • Current genetic testing approaches may not detect complex inversions, gene conversions, balanced translocations, or deeper non-coding variants in MECOM

    • Germline testing for MECOM is not designed for detection of somatic variants in tumor tissue

  • Biological Complexity:

    • MECOM exhibits context-dependent effects across different tissues and cancer types, complicating interpretation of its role

    • The gene can have opposing prognostic associations in different cancers (e.g., high expression predicting worse outcomes in some cancers but better outcomes in others)

  • Translational Barriers:

    • The complex regulatory networks involving MECOM make targeting it therapeutically challenging

    • Balancing MECOM function across multiple tissues requires careful consideration to avoid unintended consequences in clinical applications

What emerging technologies could enhance MECOM functional studies?

Several cutting-edge approaches show promise for advancing MECOM research:

  • Spatial Transcriptomics:

    • Combining single-cell RNA-seq with spatial information could reveal tissue-specific MECOM functions during development

    • In situ hybridization methods have already been used to validate MECOM localization in developing human heart

  • Multi-Modal Single-Cell Analysis:

    • Integrating single-cell transcriptomics with epigenomics and proteomics could provide deeper insights into MECOM's regulatory mechanisms

    • Methods like RNA velocity analysis and trajectory inference tools (Slingshot) are already showing value in understanding cellular dynamics

  • Advanced Genome Editing:

    • Base editing or prime editing could enable more precise manipulation of MECOM than traditional CRISPR-Cas9

    • Inducible systems would allow temporal control of MECOM expression to study stage-specific effects

How can researchers better integrate multi-omics data to understand MECOM function?

Integration of diverse data types requires sophisticated computational approaches:

  • Comprehensive Framework Development:

    • Combine results from Hi-C, DNase-Seq, ChIP-Seq, and RNA-Seq data to map MECOM's regulatory landscape

    • Apply machine learning algorithms to identify patterns across multiple data types

    • Develop specialized visualization tools to represent complex multi-omic relationships

  • Cross-Platform Data Integration:

    • Harmonize data from different sources (TCGA, GEO, HPA) to enable meta-analyses

    • Apply batch correction methods like Harmony to integrate single-cell datasets from different experiments

    • Normalize data using approaches like MultiBatchNormalisation before merging datasets

  • Functional Validation Pipeline:

    • Systematically test predictions from multi-omic analyses using targeted experiments

    • Implement pseudobulk analysis to minimize technical artifacts like allelic dropout

    • Apply robust statistical methods including permutation testing to ensure reliable results

Product Science Overview

Structure and Function

The MECOM locus is located on the long arm of chromosome 3 at position 3q26.2 . It includes several alternative transcripts, with EVI1 being a prominent oncogenic zinc-finger transcription factor. EVI1 is known for its role in myeloid malignancies, where its overexpression contributes to disease progression and poor clinical outcomes .

EVI1 exists in two main forms:

  1. EVI1: The shorter isoform, which is abundant and oncogenic.
  2. MDS1-EVI1: The longer isoform, created by splicing the MDS1 gene upstream to EVI1 .

EVI1 contains ten zinc fingers arranged in two sets: seven in the N-terminal and three in the C-terminal. These zinc fingers allow EVI1 to bind DNA and regulate gene expression .

Role in Hematopoiesis

The MDS1-EVI1 (ME) isoform is critical for the long-term function of hematopoietic stem cells (HSCs). Studies have shown that ME is exclusively expressed in the stem cell compartment and is essential for maintaining the quiescence and long-term repopulation capacity of HSCs . ME deficiency leads to a reduction in HSC numbers and a shift from quiescence to active cycling, which can result in hematopoietic defects .

Implications in Leukemogenesis

The dysregulation of EVI1 is strongly associated with leukemogenesis. Overexpression of EVI1 is linked to poor outcomes in myeloid malignancies, including acute myeloid leukemia (AML). The oncogenic properties of EVI1 are attributed to its ability to disrupt normal gene expression and promote uncontrolled cell proliferation .

Therapeutic Potential

Understanding the biology of MECOM and its associated transcripts has significant implications for developing targeted therapies. Current research is focused on identifying novel therapeutic interventions that can modulate EVI1 activity and improve clinical outcomes for patients with myeloid malignancies .

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.