EMAP II Human

Endothelial-Monocyte Activating Polypeptide II Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Biosynthesis

EMAP II is a 166-amino acid polypeptide (18.3 kDa) produced via proteolytic cleavage of the 34 kDa precursor proEMAP II/p43. Key structural features include:

  • OB-fold domain: A bacterial tRNA-binding homolog forming a pseudo-dimer with degenerate symmetry .

  • Monomeric state: Unlike bacterial Trbp proteins (dimers), EMAP II remains monomeric due to interdomain interactions .

  • Crystallographic resolution: Solved at 1.14 Å, revealing a compact architecture with an N-terminal β-barrel and C-terminal helical domain .

Table 2: EMAP II in Non-Hodgkin’s Lymphoma (NHL)

ParameterNHL Patients (n=80)Healthy Controls (n=20)
EMAP II+ lymphocytes32.5% ± 8.73.1% ± 1.2
CD36 co-expression24.8% ± 6.30%
Serum LDH correlationr = 0.71N/A

Pulmonary Disease

  • BPD pathogenesis: Sustained EMAP II expression in bronchial club cells drives macrophage infiltration, alveolar arrest, and pulmonary hypertension .

  • Therapeutic neutralization: Anti-EMAP II antibodies reduce IL-1β, CCL2, and CCL9 levels, restoring lung compliance in murine models .

Infectious Disease

  • COVID-19 severity: EMAP II Ct ≤34 predicts mortality (AUC = 0.97) and correlates with lymphopenia (r = -0.82) and hypoxia (r = -0.75) .

Mechanistic Insights

  • Hypoxia vs. apoptosis: Hypoxia triggers non-caspase-dependent EMAP II release, while apoptosis relies on caspase-7 cleavage .

  • ER stress and mitophagy: EMAP II disrupts mitochondrial dynamics, activating PERK/ATF4 pathways in glioblastoma .

Therapeutic Applications

  • Recombinant EMAP II: Produced in E. coli (18.3 kDa, 1 mg/ml solubility) for preclinical studies .

  • Dual-targeting strategies: Combining EMAP II with VEGF inhibitors or autophagy enhancers shows promise in oncology .

Challenges and Future Directions

  • Biomarker validation: Large-scale studies needed to confirm EMAP II’s prognostic utility in NHL and COVID-19 .

  • Delivery optimization: Improving tissue-specific targeting to mitigate systemic inflammation risks .

Product Specs

Introduction
EMAP-II, also known as SCYE1, is a cytokine produced by tumors that exhibits a wide range of effects on endothelial cells, monocytes, and neutrophils. It acts as an inhibitor of endothelial cell proliferation, vasculogenesis, and neovessel formation, and it can also trigger apoptosis. Additionally, EMAP-II exhibits chemotactic properties towards neutrophils and monocytes, and it stimulates myeloperoxidase activity in neutrophils. Clinically, EMAP-II holds promise in inhibiting angiogenesis in vascular beds and suppressing the growth of both primary and secondary tumors without affecting healthy tissues. SCYE1 is specifically produced during apoptosis and plays a role in regulating angiogenesis, inflammation, and wound healing. The release of SCYE1 increases the sensitivity of tumor-associated vasculature to tumor necrosis factor. Interestingly, the precursor protein of EMAP-II is identical to the p43 subunit, a component of the multi-tRNA synthetase complex, and it regulates the aminoacylation activity of tRNA synthetase in healthy cells. EMAP-2 contributes to the stimulation of inflammatory responses following proteolytic cleavage within tumor cells.
Description
Recombinant Human EMAP-II, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 166 amino acids. It has a molecular weight of 18.3 kDa. The purification of EMAP-II is achieved through proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
Lyophilized from a concentrated (1mg/ml) solution in 20mM sodium Phosphate buffer pH 7.5 and 130mM sodium chloride.
Solubility
To reconstitute lyophilized EMAP-II, it is recommended to dissolve it in sterile 18 MΩ-cm H2O at a concentration of at least 100 µg/ml. The resulting solution can be further diluted in other aqueous solutions as needed.
Stability
Lyophilized EMAP-II remains stable at room temperature for up to 3 weeks. However, for long-term storage, it is recommended to store it desiccated below -18°C. After reconstitution, EMAP-II should be stored at 4°C for 2-7 days. For extended storage, it should be kept at -18°C. Avoid repeated freeze-thaw cycles.
Purity
Greater than 98.0% as determined by (a) RP-HPLC analysis and (b) SDS-PAGE analysis.
Biological Activity
The biological activity of EMAP-II is determined by its apoptotic effect on MCF-7 cells. The effective concentration range for inducing apoptosis is 20-30 ng/ml.
Synonyms
AIMP1, EMAP2, EMAP-2, EMAPII, SCYE1, Multisynthetase complex auxiliary component p43, Endothelial monocyte-activating polypeptide 2, EMAP-II, p43.
Source
Escherichia Coli.
Amino Acid Sequence
SKPIDVSRLD LRIGCIITAR KHPDADSLYV EEVDVGEIAP RTVVSGLVNH VPLEQM QNRM VILLCNLKPA KMRGVLSQAM VMCASSPEKI EILAPPNGSV PGDRITFDAF PGEPDKELNP KKKIWEQIQP DLHTNDECVA TYKGVPFEVK GKGVCRAQTM SNSGIK.

Q&A

What is the molecular structure and synthesis of EMAP II in humans?

EMAP II is synthesized as a 34-kDa intracellular protein (proEMAP II/p43), which is cleaved to produce the extracellular mature form of 20-22 kDa with cytokine activity . The protein is closely related or identical to the p43 auxiliary protein of the multisynthase complex involved in protein synthesis . EMAP II is found in the cellular cytoplasm, but various stress factors including viral infection, lipopolysaccharides, hypoxia, or apoptosis can promote extracellular EMAP II secretion and increase its synthesis levels .

Methodologically, researchers studying EMAP II structure should consider:

  • Using immunoblotting techniques for protein quantification

  • Applying confocal microscopy to detect cellular localization

  • Implementing qRT-PCR to measure mRNA expression levels

What are the primary physiological functions of EMAP II in normal human tissues?

In the adult organism, strong EMAP II mRNA expression is predominantly restricted to thymus, testis, and brain . EMAP II exhibits pleiotropic effects on neutrophils, macrophages/monocytes, and endothelial cells. It functions as a chemoattractant for monocytes and granulocytes and is involved in the recruitment of phagocytic cells to sites of programmed cell death .

The physiological roles of EMAP II include:

  • Triggering von Willebrand factor release and expression of P- and E-selectin in endothelial cells

  • Activating the neutrophil respiratory burst

  • Stimulating chemotaxis of macrophages and neutrophils

  • Activating dendritic cells and macrophages to enhance T-helper 1 responses and interleukin-12 release

  • Contributing to immunosurveillance in the brain, particularly in the recruitment of microglial cells

How does hypoxia regulate EMAP II expression and processing compared to apoptotic mechanisms?

Hypoxia and apoptosis represent two distinct mechanisms for EMAP II generation in human cells. This distinction is critical for understanding EMAP II's role in pathological conditions like cancer.

In hypoxic conditions:

In apoptotic conditions:

  • Caspase-7 has been identified as a key enzyme in the cleavage of the 34-kDa proEMAP II

  • Caspase inhibitors can effectively block the processing of proEMAP II to mature EMAP II in apoptotic cells

This differential processing suggests that researchers should carefully distinguish between hypoxia-induced and apoptosis-induced EMAP II when designing experiments, as they may represent functionally distinct pathways with different downstream effects.

What methodological approaches are most effective for studying EMAP II's role in human tumor microenvironments?

When investigating EMAP II in tumor contexts, researchers should implement a multi-modal approach:

  • Spatial distribution analysis:

    • Use confocal microscopy to correlate proEMAP II/p43 expression with hypoxic and apoptotic regions in tumor samples

    • Employ immunohistochemistry to map EMAP II expression in relation to areas of tissue necrosis

  • Functional assessments:

    • Conduct in vitro co-culture experiments with cancer cells and immune cells (e.g., Jurkat T cells) to assess EMAP II's immunomodulatory effects

    • Use antibody neutralization studies to determine the contribution of EMAP II to cancer cell-induced lymphocyte apoptosis

  • Molecular mechanism investigation:

    • Compare mRNA and protein levels to distinguish between transcriptional regulation and post-translational processing

    • Apply caspase inhibitors to differentiate between apoptosis-dependent and hypoxia-dependent EMAP II generation

The data from these approaches should be integrated to understand EMAP II's complex role in creating immunosuppressive tumor microenvironments.

How can EMAP II be effectively utilized as a biomarker in respiratory diseases including COVID-19?

EMAP II shows significant potential as a biomarker for respiratory disease severity, particularly in COVID-19. Research methodologies should include:

  • Sample collection and processing:

    • Collect nasopharyngeal samples for EMAP II mRNA detection using qRT-PCR

    • Standardize collection timing relative to disease onset

  • Quantification approaches:

    • Utilize real-time PCR with SYBR green for EMAP II mRNA detection

    • Assess EMAP II expression in terms of cycle threshold (Ct) values, with lower values indicating higher expression

  • Statistical analysis:

    • Correlate EMAP II Ct values with clinical parameters (oxygen saturation, lymphocyte percentages, inflammatory markers)

    • Establish appropriate cutoff values for disease severity prediction

Research has demonstrated that EMAP II RNA is not detected in nasopharyngeal swabs of normal controls and mild to asymptomatic COVID-19 patients but is detectable in severe COVID-19 patients . A Ct value cutoff of ≤34 predicts worse outcomes with 100% sensitivity and specificity . This suggests that EMAP II detection protocols could be standardized for clinical implementation in severe respiratory disease assessment.

What are the key considerations when designing in vivo models to study EMAP II's role in pulmonary pathologies?

When designing in vivo models for studying EMAP II in pulmonary diseases, researchers should consider:

  • Model selection and establishment:

    • For bronchopulmonary dysplasia (BPD) studies, utilize neonatal hyperoxia exposure models that mimic the clinical conditions of premature infants

    • Ensure appropriate control groups (e.g., normoxia vs. hyperoxia)

  • EMAP II administration protocols:

    • Define appropriate dosing regimens that produce pathophysiological effects without excessive toxicity

    • Consider recombinant EMAP II administration (intravenous or intratracheal) to study direct effects

  • Comprehensive outcome measurements:

    • Assess structural changes through histological analysis of alveolar formation (radial alveolar count, mean linear intercept)

    • Measure functional parameters including pulmonary biophysical properties (pressure-volume relationships)

    • Evaluate secondary effects like right ventricular hypertrophy and pulmonary hypertension

    • Quantify inflammatory cell infiltration, particularly macrophages

  • Molecular assessments:

    • Monitor EMAP II protein levels using immunoblotting at various timepoints

    • Assess potential compensatory mechanisms (e.g., surfactant protein-C expression)

Studies using this approach have demonstrated that exogenous EMAP II administration leads to significantly impaired lung structure and function, with larger distal airspaces, decreased radial alveolar count, increased mean linear intercepts, and compromised pulmonary biophysical properties .

How should researchers design experiments to distinguish between EMAP II's direct effects and downstream inflammatory cascades?

This methodological challenge requires careful experimental design:

  • Temporal sequence studies:

    • Implement time-course experiments to determine whether EMAP II elevation precedes or follows other inflammatory markers

    • Use conditional knockout or inducible expression systems to control the timing of EMAP II expression

  • Pathway inhibition approaches:

    • Employ selective inhibitors at various points in the inflammatory cascade

    • Utilize EMAP II neutralizing antibodies to block its activity while monitoring downstream effects

  • Receptor identification and blockade:

    • Identify and characterize specific EMAP II receptors on target cells

    • Use receptor antagonists or receptor-deficient cell lines/animals to distinguish direct from indirect effects

  • Cell-type specific responses:

    • Conduct in vitro studies with purified cell populations (endothelial cells, monocytes, lymphocytes)

    • Compare responses between different cell types to map the sequence of inflammatory activation

These approaches can help establish whether EMAP II functions as an initiator or amplifier of inflammatory responses in various pathological conditions.

What are the most promising therapeutic strategies targeting EMAP II in human diseases?

Based on current research, several therapeutic approaches targeting EMAP II show promise:

  • Neutralizing antibodies:

    • Anti-EMAP II monoclonal antibodies have shown efficacy in preclinical models of lung injury, including virus-induced damage

    • This approach may be particularly valuable for acute inflammatory conditions

  • Inhibitors of EMAP II processing:

    • Given the distinct processing mechanisms in hypoxia versus apoptosis, targeted inhibitors of the relevant proteases could provide selective therapeutic effects

    • This approach may allow context-specific intervention without disrupting physiological EMAP II functions

  • Receptor antagonists:

    • Development of specific antagonists for EMAP II receptors on target cell populations

    • This strategy could offer more selective modulation of EMAP II effects

  • RNA interference approaches:

    • siRNA or antisense oligonucleotides targeting EMAP II mRNA

    • This could be particularly useful in chronic inflammatory conditions where sustained EMAP II suppression is desired

Each approach has distinct advantages depending on the disease context, with neutralizing antibodies currently showing the most advanced development for conditions like BPD and viral-induced lung injury .

How can EMAP II measurement be standardized for clinical applications in disease prognosis?

Standardization for clinical implementation requires addressing several methodological considerations:

  • Sample type optimization:

    • Determine optimal biological samples (serum, plasma, bronchoalveolar lavage fluid, or tissue biopsies)

    • For respiratory diseases, nasopharyngeal swabs have shown utility for EMAP II mRNA detection

  • Detection method standardization:

    • Establish standardized qRT-PCR protocols with appropriate controls

    • Consider development of ELISA or other protein-based detection systems for mature EMAP II

  • Reference ranges and cutoffs:

    • Determine normal reference ranges in healthy populations

    • Establish disease-specific cutoff values, such as the Ct value of ≤34 identified for severe COVID-19

  • Integration with clinical data:

    • Develop algorithms that integrate EMAP II measurements with other clinical parameters

    • Create prognostic models that incorporate EMAP II with established biomarkers

For COVID-19 specifically, EMAP II Ct values have shown strong correlations with clinical parameters including positive correlations with lymphocyte percentages and oxygen saturation, and negative correlations with age, serum CRP, ferritin, and D-dimer levels .

How should researchers address contradictory findings regarding EMAP II's pro-inflammatory versus immunosuppressive effects?

EMAP II demonstrates seemingly contradictory effects - pro-inflammatory in some contexts and immunosuppressive in others. Resolving these apparent contradictions requires:

  • Contextual analysis:

    • Carefully document experimental conditions including cell types, tissue context, and disease state

    • Consider that EMAP II may have biphasic effects depending on concentration and timing

  • Distinguishing between direct and indirect effects:

    • Determine whether observed immunosuppression (e.g., lymphocyte apoptosis) is a direct effect of EMAP II or a secondary response to initial pro-inflammatory activity

    • Identify cell-type specific responses that may explain differential effects

  • Molecular form consideration:

    • Distinguish between effects of proEMAP II/p43 and mature EMAP II

    • Investigate whether different cleavage products have distinct biological activities

  • Receptor expression analysis:

    • Examine whether target cell receptor profiles determine pro-inflammatory versus immunosuppressive responses

    • Identify whether receptor expression changes during disease progression

These approaches can help reconcile the observation that EMAP II acts as a pro-inflammatory mediator on endothelial cells and monocytes while inducing apoptosis in lymphocytes , potentially contributing to an immunosuppressive tumor microenvironment.

What methodological approaches can distinguish between correlation and causation in EMAP II studies?

Establishing causality in EMAP II research requires rigorous experimental approaches:

  • Intervention studies:

    • Implement gain-of-function studies using recombinant EMAP II administration

    • Conduct loss-of-function studies using neutralizing antibodies or genetic approaches

  • Dose-response relationships:

    • Establish clear dose-response curves for EMAP II effects

    • Determine whether threshold effects exist that may explain contradictory findings

  • Temporal sequence analysis:

    • Document the precise timing of EMAP II elevation relative to disease progression

    • Use inducible systems to control EMAP II expression at specific time points

  • Genetic evidence:

    • Utilize genetic variants affecting EMAP II expression or function

    • Apply mendelian randomization approaches in human studies when possible

  • Mechanism verification:

    • Confirm proposed mechanisms through multiple independent experimental approaches

    • Validate findings across different model systems and species

These approaches can help determine whether EMAP II elevation is merely a biomarker of disease severity or an active participant in pathogenesis, particularly in complex conditions like COVID-19 and BPD.

What are the most critical knowledge gaps in understanding EMAP II's role in human disease pathogenesis?

Several significant knowledge gaps require further investigation:

  • Receptor identification and signaling:

    • The specific receptors mediating EMAP II's diverse effects remain incompletely characterized

    • The downstream signaling pathways triggered by EMAP II binding need further elucidation

  • Processing regulation:

    • While caspase-7 has been identified in apoptotic processing, the proteases responsible for EMAP II cleavage under hypoxic conditions remain unknown

    • The regulatory mechanisms controlling EMAP II release in different pathological states require further study

  • Tissue-specific effects:

    • Comprehensive mapping of EMAP II effects across different human tissues and cell types

    • Understanding the basis for tissue-specific vulnerability to EMAP II-mediated damage

  • Long-term consequences:

    • The long-term effects of transient EMAP II elevation in acute conditions like COVID-19

    • Potential chronic inflammatory sequelae of EMAP II dysregulation

  • Genetic variability:

    • The impact of genetic polymorphisms on EMAP II expression, processing, and function

    • How genetic variation might explain differential susceptibility to EMAP II-mediated pathology

Addressing these gaps will require integrated approaches combining molecular, cellular, and clinical research methodologies.

What novel technologies might advance EMAP II research in the next decade?

Emerging technologies with significant potential to advance EMAP II research include:

  • Single-cell technologies:

    • Single-cell RNA sequencing to identify cell-specific responses to EMAP II

    • Single-cell proteomics to characterize cell-type specific signaling pathways

  • Advanced imaging approaches:

    • Intravital microscopy to visualize EMAP II effects in real-time in vivo

    • Multiplexed imaging technologies to simultaneously map EMAP II and multiple inflammatory markers

  • Organ-on-chip platforms:

    • Microfluidic devices modeling complex tissue environments to study EMAP II effects

    • Integration of multiple organ systems to understand systemic consequences of EMAP II elevation

  • CRISPR-based approaches:

    • Precise genome editing to study specific domains of EMAP II

    • CRISPRi/CRISPRa systems for temporal control of EMAP II expression

  • Computational modeling:

    • Systems biology approaches to integrate EMAP II into inflammatory network models

    • AI-driven analysis of large-scale datasets to identify novel EMAP II-associated biomarkers

These technologies promise to provide more nuanced understanding of EMAP II's contextual effects and facilitating the development of precision therapeutic approaches targeting EMAP II in specific disease states.

Product Science Overview

Structure and Synthesis

EMAP-II is synthesized as the C-terminal portion of a biologically inactive precursor protein that contains a propeptide of 146 amino acid residues . The mature form of EMAP-II is an 18.3 kDa protein . Recombinant Human EMAP-II (rHuEMAP-II) is produced in Escherichia coli and is a single non-glycosylated polypeptide chain containing 166 amino acids .

Biological Functions

EMAP-II exerts several critical functions:

  • Inhibition of Endothelial Cell Proliferation: It prevents the proliferation of endothelial cells, which is crucial for the formation of new blood vessels .
  • Induction of Apoptosis: EMAP-II can induce programmed cell death in various cell types .
  • Chemotactic Activity: It attracts neutrophils and monocytes to the site of action .
  • Myeloperoxidase Activity: EMAP-II induces myeloperoxidase activity from neutrophils .
Clinical Significance

One of the most significant aspects of EMAP-II is its role in inhibiting angiogenesis in vascular beds, which suppresses the growth of primary and secondary tumors without affecting normal tissues . This makes it a potential candidate for anti-cancer therapies.

Research and Applications

Recombinant Human EMAP-II is used extensively in research to study its effects on cell proliferation, apoptosis, and angiogenesis . It is also used to explore its potential therapeutic applications in cancer treatment .

Storage and Handling

Recombinant Human EMAP-II is typically supplied as a sterile filtered, white lyophilized powder. It is recommended to reconstitute it in sterile distilled water or an aqueous buffer containing 0.1% BSA to a concentration of 0.1-1.0 mg/mL . The product should be stored at -20°C to -70°C and should avoid repeated freeze-thaw cycles to maintain its stability .

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.