ESM1 Human, SF9 is synthesized using baculovirus-infected Sf9 cells, which enable eukaryotic post-translational modifications critical for functional studies. The protein includes a C-terminal His tag for purification via affinity chromatography . Key steps include:
Transfection: Baculovirus vectors carrying the ESM1 gene infect Sf9 cells.
Harvesting: Secreted ESM1 is collected from cell culture media.
Purification: Proprietary chromatographic techniques ensure high purity (>95%) .
ESM1 promotes tumor angiogenesis by enhancing endothelial cell sprouting and vascular permeability . Overexpression correlates with advanced lung cancer and renal fibrosis, making it a biomarker for tumor progression . Antibodies targeting ESM1’s protein core are under investigation for cancer therapy .
Cytokines such as TNF-α and IL-1β upregulate ESM1 expression, while IFN-γ suppresses it . This regulatory mechanism links ESM1 to endothelial inflammation in conditions like hypertension and transplant rejection .
In murine models of unilateral ureteral obstruction, ESM1 overexpression induces endothelial-to-mesenchymal transition (EndoMT), marked by increased α-SMA and collagen deposition .
Parameter | ESM1 (SF9) | ESM1 (E. coli) | ESM1 (HEK293) |
---|---|---|---|
Glycosylation | Yes | No | Yes |
Molecular Mass | 19.2 kDa | 20.5 kDa | 19.5 kDa |
Expression Tag | C-terminal His tag | N-terminal His tag | C-terminal His tag |
Key Use Cases | Cell adhesion studies | Structural biology | Signal pathway analysis |
While ESM1 Human, SF9 replicates native glycosylation patterns, its in vivo stability requires optimization for therapeutic applications . Ongoing studies focus on:
ESM1, also known as Endothelial cell-specific molecule 1, is a protein encoded by the ESM1 gene in humans. This secreted protein is highly expressed in the endothelial cells of human kidney and lung tissues. Due to its regulation by cytokines, ESM1 is believed to be involved in endothelium-dependent pathological diseases.
Produced in Sf9 insect cells using a baculovirus expression system, ESM1 is a single, glycosylated polypeptide chain. This recombinant protein consists of 174 amino acids (20-184 a.a.), with a molecular weight of 19.2 kDa. However, it appears between 18-28 kDa on SDS-PAGE. The protein is expressed with a 9 amino acid His tag at the C-terminus and purified through proprietary chromatographic techniques.
The ESM1 protein solution is provided at a concentration of 0.25 mg/ml in Phosphate Buffered Saline (pH 7.4) with 10% glycerol.
The purity of the ESM1 protein is greater than 95.0% as determined by SDS-PAGE analysis.
Endothelial Cell Specific Molecule 1, Endothelial Cell-Specific Molecule 1, ESM-1, Endocan.
Sf9, Baculovirus cells.
ADLWSNNYAV DCPQHCDSSE CKSSPRCKRT VLDDCGCCRV CAAGRGETCY RTVSGMDGMK CGPGLRCQPS NGEDPFGEEF GICKDCPYGT FGMDCRETCN CQSGICDRGT GKCLKFPFFQ YSVTKSSNRF VSLTEHDMAS GDGNIVREEV VKENAAGSPV MRKWLNPRHH HHHH.
ESM1 (Endothelial-cell-specific molecule 1) is a secreted proteoglycan originally identified in vascular endothelial cells. It functions primarily in regulating endothelial cell function, angiogenesis, and inflammatory processes. ESM1 is expressed in various cell types, including human vascular endothelial cells, liver cells, and bronchial and pulmonary submucosal gland cells . Beyond endothelial cells, immunohistochemical studies have demonstrated ESM1 expression in the bronchial and renal epithelia, suggesting broader physiological roles than initially recognized . ESM1 plays a significant role in vascular contribution to organ-specific inflammation, with its expression regulated by inflammatory cytokines such as TNFα and IFNγ .
Intracellular ESM1 and its secreted counterpart show notable structural differences. Immunoblot analysis reveals that ESM1 present in cell lysates of human endothelial cells (HUVECs) has an apparent molecular weight of approximately 20 kD. In contrast, the secreted form exhibits a significant shift to an apparent molecular weight of 50 kD . This molecular weight difference indicates substantial post-translational modifications occur during the secretion process. Researchers investigating ESM1 should account for these structural differences when designing detection methods and interpreting experimental results, as antibodies may have different affinities for the two forms .
For comprehensive ESM1 detection, multiple antibody-based approaches targeting different epitopes are recommended. Research has established that developing mouse anti-ESM1 monoclonal antibodies against three distinct epitopes enables effective ELISA assay development, immunohistological staining, and immunoblot analysis . For intracellular ESM1 detection, standard western blotting of cell lysates is effective, while secreted ESM1 requires analysis of conditioned media or serum samples. When developing detection protocols, researchers should note that serum from healthy subjects contains ESM1 at an average concentration of 1.08 ng/ml, providing a baseline for pathological studies .
ESM1 demonstrates significant potential as a diagnostic biomarker for various cancers, particularly digestive tract malignancies. In stomach adenocarcinoma (STAD) and esophageal carcinoma (ESCA), receiver operating characteristic (ROC) curve analysis shows high diagnostic accuracy of ESM1. The area under the curve (AUC) values for ESM1 in different sample types are presented in the table below :
Cancer Type | Sample Type | Stage I AUC (95% CI) | Stage IV AUC (95% CI) |
---|---|---|---|
STAD | Plasma | 0.7978 (0.6574-0.9382) | 0.9222 (0.8367-1.000) |
STAD | Serum | 0.8179 (0.6847-0.9511) | 0.9056 (0.8079-1.000) |
STAD | Tissue | 0.8827 (0.7590-1.000) | 0.9778 (0.9357-1.000) |
ESCA | Plasma | 0.8611 (0.7582-0.9640) | 0.9444 (0.8599-1.000) |
ESCA | Serum | 0.7906 (0.6648-0.9164) | 0.8958 (0.7779-1.000) |
ESCA | Tissue | 0.9573 (0.9041-1.000) | 0.9931 (0.9717-1.000) |
These high AUC values (>0.79) across both early (Stage I) and advanced (Stage IV) disease stages indicate ESM1's utility as a sensitive biomarker across the disease spectrum .
When evaluating ESM1 as a biomarker, researchers should employ multi-sample type analysis for comprehensive assessment. Based on clinical research protocols, quantification should include:
RT-qPCR for mRNA expression analysis in tissue samples
Western blot for protein quantification
ELISA assays for measuring secreted ESM1 in plasma and serum samples
For clinical applications, establishing proper cutoff values is critical for distinguishing between normal and pathological levels. Research indicates significant upregulation of ESM1 at stages I, II, III, and IV in plasma, serum, and tissues of STAD and ESCA patients compared to healthy controls . When designing validation studies, researchers should include sufficient sample sizes across different disease stages to ensure robust statistical power for determining diagnostic accuracy.
ESM1 promotes cancer progression through multiple signaling networks, with the VEGFα pathway being particularly significant. In cervical squamous cell carcinoma (CSCC), ESM1 augments endothelial cell proliferation via the VEGFα/ERK signaling pathway . Gene Set Enrichment Analysis (GSEA) has demonstrated that high ESM1 expression is significantly enriched in carcinoma angiogenesis and VEGFα signaling pathways . The mechanistic relationship is bidirectional, as ESM1 knockdown decreases VEGFα and HIF-1α expression while reducing phosphorylation of VEGFR2 and ERK-1/2. Similarly, VEGFα inhibition dramatically decreases ESM1 and HIF-1α expression, suggesting a positive feedback loop between these factors in cancer cells .
ESM1 inhibition produces significant anti-cancer effects across multiple cancer cell lines. In vitro studies with both digestive tract cancers (AGS and TE1 cells) and cervical cancer lines (SiHa and ME-180) show that ESM1 knockdown consistently:
These effects have been quantitatively demonstrated in multiple experimental models. For example, flow cytometry analysis revealed that ESM1 downregulation dramatically increased cell apoptosis in SiHa cells (from 8.74% to 27.2%) and ME-180 cells (from 19.3% to 31.8%) . The consistency of these findings across different cancer types suggests ESM1 inhibition may represent a broadly applicable therapeutic strategy.
ESM1 plays a critical role in modulating the tumor microenvironment, particularly through promoting angiogenesis. Originally identified as an endothelial cell molecule, ESM1 facilitates vascular network formation essential for tumor growth . Mechanistically, ESM1 enhances HIF-1α expression, a master regulator of cellular response to hypoxia, which drives angiogenesis in tumors . The VEGFα signaling pathway is a key mediator of ESM1's pro-angiogenic effects, as demonstrated by reduced VEGFR2 phosphorylation following ESM1 knockdown . This angiogenic function represents a significant mechanism through which ESM1 promotes cancer progression beyond its direct effects on tumor cells themselves.
For comprehensive genomic and transcriptomic analysis of ESM1, researchers should employ integrated multi-platform approaches. Effective methodologies include:
Microarray dataset analysis for identifying differentially expressed genes (DEGs) associated with ESM1, as demonstrated in studies that screened 1265 abnormally expressed genes in ESCA samples and 93 in STAD samples
KEGG enrichment analysis for identifying functional pathways. Research has shown that ESM1-associated genes are involved in rheumatoid arthritis, protein digestion and absorption, and cytokine-cytokine receptor interaction pathways
TNMplot and TCGA (The Cancer Genome Atlas) data repositories for validating ESM1 expression patterns across different cancer types. These platforms have confirmed ESM1 overexpression in multiple cancer types compared to normal tissues
Gene Set Enrichment Analysis (GSEA) for identifying biological processes associated with ESM1 expression. This approach has revealed enrichment in carcinoma angiogenesis and VEGFα signaling pathways in high ESM1-expressing tumors
When designing experiments to evaluate ESM1's effects on cellular behavior, several methodological considerations are critical:
Selection of appropriate cell models: Both cancer cell lines (such as AGS, TE1, SiHa, and ME-180) and endothelial cells should be considered depending on the research question, as ESM1 functions in both cell types
Effective knockdown strategies: siRNA approaches have been successfully employed to achieve ESM1 silencing. Verification of knockdown efficiency should include both mRNA (RT-qPCR) and protein (Western blot) level assessments
Comprehensive functional assays: A battery of assays should be employed, including:
Cytokine regulation control experiments: As ESM1 expression is regulated by inflammatory cytokines (enhanced by TNFα, inhibited by IFNγ), experimental designs should account for these effects when interpreting results
Based on current research findings, several therapeutic approaches targeting ESM1 show promise:
Direct ESM1 inhibition: RNA interference approaches have demonstrated efficacy in preclinical models, with ESM1 knockdown suppressing cancer cell proliferation, migration, and invasion while increasing apoptosis
Disruption of the ESM1-VEGFα signaling axis: Targeting this pathway has shown potential in cervical squamous cell carcinoma, where the ESM1/VEGFα signaling pathway promotes cancer progression
Combination approaches: Since ESM1 expression is associated with resistance to certain cancer treatments, combining ESM1 inhibition with conventional therapies may overcome treatment resistance
Monitoring applications: Even without direct targeting, ESM1 levels could serve as biomarkers for treatment response and disease monitoring, given their correlation with disease stage and prognosis
Each approach requires careful validation in appropriate preclinical models before advancing to clinical investigation, with particular attention to potential effects on normal endothelial function given ESM1's physiological roles.
Despite significant advances, several important knowledge gaps remain in ESM1 research:
Mechanism of post-translational modification: While we know secreted ESM1 shifts from 20kD to 50kD, the exact nature and functional significance of these modifications remain incompletely characterized
Tissue-specific effects: ESM1 is expressed in various tissues, including vascular endothelium, bronchial and renal epithelia, but its tissue-specific functions are not fully elucidated
Regulatory network: The complete set of factors regulating ESM1 expression beyond TNFα and IFNγ requires further characterization
In vivo validation: Current studies acknowledge limitations in substantiation from in vivo data, suggesting a critical need for animal model studies to complement in vitro findings
Patient sample limitations: Clinical studies have been constrained by small sample sizes due to difficulties in sample collection, indicating a need for larger cohort studies
Developing ESM1-targeted therapeutics presents several methodological challenges:
Targeting specificity: Given ESM1's expression in normal tissues, particularly endothelial cells, developing cancer-specific targeting approaches is essential to minimize off-target effects
Delivery systems: For RNA interference approaches, effective delivery systems must be developed to ensure sufficient target engagement in tumor tissues
Biomarker standardization: Establishing standardized cutoff values and detection methodologies across different biological samples (plasma, serum, tissue) will be critical for clinical implementation
Resistance mechanisms: Understanding potential resistance mechanisms to ESM1-targeted therapies will be necessary for developing effective long-term treatment strategies
Combination strategies: Determining optimal combinations with existing therapies requires systematic evaluation in appropriate preclinical models and carefully designed clinical trials
ESM1 research has significant potential to integrate with several cutting-edge areas in cancer biology:
Liquid biopsy development: ESM1's detectable presence in plasma and serum makes it an excellent candidate for incorporation into liquid biopsy panels for cancer detection and monitoring
Tumor microenvironment modulation: As ESM1 influences angiogenesis and potentially other aspects of the tumor microenvironment, it may synergize with immunotherapy approaches that depend on favorable microenvironment conditions
Multi-omics integration: Combining ESM1 expression data with other molecular profiling approaches (genomics, proteomics, metabolomics) could enhance precision medicine approaches to cancer treatment
Artificial intelligence applications: Machine learning approaches could identify novel patterns in ESM1 expression and regulation across cancer types, potentially uncovering new therapeutic opportunities and biomarker applications
Biomarker panels: Including ESM1 in multi-marker panels may improve diagnostic and prognostic accuracy beyond single-marker approaches, particularly when combined with other angiogenesis and inflammation markers
ESM-1 is secreted into the bloodstream and can be detected in various tissues, including the lungs and kidneys . It plays a significant role in angiogenesis, promoting the sprouting of new blood vessels . The expression of ESM-1 is regulated by cytokines, suggesting its involvement in endothelium-dependent pathological disorders .
ESM-1 is involved in several critical biological processes:
Due to its aberrant expression under pathological conditions, ESM-1 serves as a diagnostic and prognostic indicator for various diseases, including cancer, sepsis, kidney diseases, and cardiovascular diseases . Its role in promoting angiogenesis and its presence in the bloodstream make it a potential target for therapeutic interventions, particularly in cancer treatment .
The recombinant form of ESM-1, produced using the Sf9 insect cell line, is used in research to study its functions and potential therapeutic applications. This recombinant protein retains the biological activity of the native molecule, making it a valuable tool for scientific investigations.