ELANE Mouse refers to the murine ortholog of the human neutrophil elastase (ELANE/ELA2), a serine protease encoded by the Elane gene located on chromosome 10 (10:79,722,146-79,724,050) in mice . This enzyme is primarily expressed in neutrophil azurophil granules and plays critical roles in immune defense by degrading bacterial outer membrane proteins (e.g., E. coli OmpA) and elastin in connective tissues . Recombinant ELANE Mouse protein is widely used in biochemical and cancer research due to its species-specific functional properties and therapeutic potential .
ELANE Mouse exhibits selective cytotoxicity against cancer cells through mechanisms distinct from human ELANE :
Mechanistic Feature | ELANE Mouse | Human ELANE |
---|---|---|
SLPI Inhibition | Yes (co-released with SLPI in neutrophils) | No |
CD95/Histone H1 Interaction | Preserved | Preserved |
In Vivo Efficacy | Attenuated due to serpin inhibition (A1AT, SLPI) | Enhanced with porcine elastase homologs |
Abscopal Effect | Not observed | Observed (immune-mediated distant metastasis suppression) |
Key findings:
ELANE Mouse induces cancer cell death by liberating the CD95 death domain, which interacts with histone H1 isoforms overexpressed in malignant cells .
Murine ELANE’s anticancer activity is neutralized by secretory leukocyte peptidase inhibitor (SLPI), limiting its therapeutic utility compared to human ELANE .
The Quantikine® Mouse ELA2 ELISA Kit (MELA20) enables precise measurement of ELANE Mouse in biological samples :
Parameter | Details |
---|---|
Assay Range | 31.2–2,000 pg/mL |
Sensitivity | 10 pg/mL |
Precision | Intra-assay CV: 1.2–2.1%; Inter-assay CV: 6.1–9.7% |
Recovery | 90–125% in plasma/serum; 91–102% in cell culture media |
Neutropenia Models: Mutations in Elane disrupt translational regulation via internal ribosome entry site (IRES) activity. A 18S rRNA-complementary region downstream of ATG2 in Elane mRNA is critical for IRES function .
Unfolded Protein Response: Certain Elane mutations (e.g., truncations) trigger endoplasmic reticulum stress, impairing granulopoiesis in transgenic mice .
Serpin Resistance: Porcine pancreatic elastase, an ELANE homolog less susceptible to serpin inhibition, shows enhanced in vivo efficacy in murine cancer models .
Timing Dependency: Prolonged ELANE exposure (6–24 hours) induces apoptosis, while brief exposure (1 hour) promotes cancer cell proliferation .
Elastase-2, also known as ELANE, is a serine protease of the chymotrypsin family. This enzyme plays a crucial role in breaking down elastin, a key component of connective tissue that provides elasticity. It works in conjunction with collagen to regulate the mechanical properties of these tissues. In neutrophils, a type of white blood cell, ELANE targets and degrades the Outer membrane protein A (OmpA) found in E. coli and other Gram-negative bacteria. The activity of ELANE is tightly regulated by alpha 1-antitrypsin (A1AT), an acute-phase protein. A1AT acts as a potent inhibitor by binding almost irreversibly to the active site of elastase, effectively preventing it from breaking down elastin. This inhibition is crucial because a deficiency in A1AT, often referred to as A1AD, can lead to the uncontrolled breakdown of elastic fiber by elastase, potentially resulting in various health complications.
This product consists of the recombinant mouse ELANE protein, produced in Sf9 insect cells. It is a single, glycosylated polypeptide chain that encompasses amino acids 27 to 265 from the original sequence. The protein also includes a 6 amino acid His Tag fused at the C-terminus for purification purposes. With a total of 245 amino acids, the recombinant ELANE protein has a molecular weight of 26.8 kDa. Under reducing conditions on SDS-PAGE, it appears as multiple bands between 28-40 kDa. The protein has been purified using proprietary chromatographic techniques to ensure high purity.
The ELANE protein is supplied in a solution at a concentration of 0.25 mg/ml. The solution is buffered with 20mM Tris-HCl at a pH of 8.0 and also contains 30% glycerol and 0.1M NaCl for stability.
To ensure product stability, store the vial at 4°C if it will be used within 2-4 weeks. For long-term storage exceeding this period, it is recommended to freeze the product at -20°C. To further enhance stability during prolonged storage, consider adding a carrier protein such as HSA or BSA to a final concentration of 0.1%. Repeated freezing and thawing of the product should be avoided to maintain optimal product integrity.
The purity of ELANE Mouse is greater than 90%, as determined by SDS-PAGE analysis.
Elane, Ela2, F430011M15Rik, NE, Neutrophil elastase, Elastase-2, Leukocyte elastase.
Sf9, Insect cells.
SEIVGGRPAR PHAWPFMASL QRRGGHFCGA TLIARNFVMS AAHCVNGLNF RSVQVVLGAH
DLRRQERTRQ TFSVQRIFEN GFDPSQLLND IVIIQLNGSA TINANVQVAQ LPAQGQGVGD
RTPCLAMGWG RLGTNRPSPS VLQELNVTVV TNMCRRRVNV CTLVPRRQAG ICFGDSGGPL
VCNNLVQGID SFIRGGCGSG LYPDAFAPVA EFADWINSII RSHNDHLLTH PKDREGRTNH HHHHH
ELANE (Elastase, Neutrophil Expressed) encodes neutrophil elastase, a serine protease expressed in neutrophil azurophil granules. This protein plays a crucial role in innate immunity and neutrophil function. In research contexts, ELANE has significant importance as mutations in this gene are associated with severe congenital neutropenia (SCN) in humans. Mouse models with ELANE mutations are developed to study the molecular mechanisms underlying neutropenia and to explore potential therapeutic approaches. These models allow researchers to investigate the effects of specific ELANE mutations found in human patients with neutropenia in a controlled experimental environment. ELANE has also recently been found to exhibit selective anticancer activity, making it a gene of interest in both immunology and oncology research .
ELANE mouse models are typically generated through targeted mutation approaches using homologous recombination in embryonic stem (ES) cells. The process involves several key steps:
Design of a targeting vector containing the desired mutation in the Elane gene
Introduction of this vector into embryonic stem cells where homologous recombination occurs
Selection of ES cell clones with the properly integrated mutation
Microinjection of these ES cells into blastocysts
Implantation into pseudopregnant foster females
Breeding of chimeric offspring to establish germline transmission
For example, the G193X Elane mouse model was created by introducing a mutation that produces a stop codon at amino acid G193, deleting the carboxyl-terminal 27 amino acids of the mature neutrophil elastase protein. This mutation reproduces the G192pter ELANE mutation found in some SCN patients . The successful ES cell clones were identified and used to generate chimeric mice, with subsequent breeding to establish stable transgenic lines. Genotyping of these mice typically involves PCR using specific primers targeting the Elane locus, followed by restriction enzyme digestion to distinguish between wild-type and mutant alleles .
Interestingly, despite expectations based on human SCN, many ELANE mutant mice do not exhibit overt neutropenia under basal conditions. For instance, the G193X Elane mice show normal growth, development, and fertility, and are grossly indistinguishable from wild-type littermates . This suggests complex species-specific differences in neutrophil development regulation.
Key characteristics observed in various ELANE mouse models include:
Normal basal granulopoiesis in many models, contrasting with human SCN phenotypes
Increased sensitivity of granulocytic precursors to chemical induction of ER stress
Rapid degradation of mutant NE protein in some models (e.g., G193X Elane mice)
Normal stress granulopoiesis following myeloablative therapy
Molecular evidence of unfolded protein response (UPR) activation in some models
The G193X Elane mice, while phenotypically normal, show complete absence of detectable NE protein despite normal mRNA expression, indicating post-translational degradation of the truncated protein . This rapid degradation may explain the lack of neutropenia in these mice, as the misfolded protein is efficiently removed before triggering substantial ER stress.
The unfolded protein response (UPR) has emerged as a key mechanism in the pathogenesis of ELANE-associated neutropenia. Research suggests a model where ELANE mutations result in NE protein misfolding, which induces endoplasmic reticulum (ER) stress, activates the UPR, and ultimately blocks granulocytic differentiation .
In G193X Elane mice, though basal UPR activation is not significant, granulocytic precursors show increased sensitivity to chemical induction of ER stress . This suggests that while the mutant protein alone may not trigger sufficient ER stress to impair granulopoiesis, it may predispose cells to ER stress when faced with additional challenges.
The UPR involves three main sensors: PERK, IRE1, and ATF6. Experiments with G193X Elane mice found that inactivation of PERK (protein kinase RNA-like ER kinase), one of the major sensors of ER stress, either alone or in combination with the G193X Elane mutation, did not significantly affect granulopoiesis . This indicates that either the PERK branch of the UPR is not critical for ELANE mutation-associated neutropenia, or that compensatory mechanisms exist in mice that are absent in humans.
The discrepancy between human ELANE mutations causing severe neutropenia and mouse models showing normal neutrophil counts represents an important research puzzle. Several hypotheses explain this species difference:
Protein sequence divergence: Human and mouse NE share only 76% amino acid identity, which may affect how mutations impact protein folding and function .
Differential protein quality control: Mice may have more efficient systems for degrading misfolded proteins, preventing accumulation of toxic intermediates.
Species-specific granulopoiesis regulation: The mechanisms controlling neutrophil development may have species-specific features that make mice more resistant to perturbations in NE function.
Compensatory pathways: Mice may activate compensatory mechanisms that humans lack or cannot sufficiently upregulate.
For example, the V72M Elane mouse model, which reproduces a mutation found in SCN patients, showed normal granulopoiesis despite valine at amino acid 72 being conserved between species. This suggests that this specific mutation may induce different structural changes in mouse NE compared to human NE . Similarly, the G193X Elane mice produce a truncated NE protein that is rapidly degraded, potentially preventing the protein from accumulating and causing ER stress .
These differences highlight the importance of considering species-specific factors when using mouse models to study human diseases and may explain why different ELANE mutations have varying phenotypic consequences in mice versus humans.
Recent research has revealed that ELANE possesses remarkable cancer-selective cytotoxic properties. Cui et al. demonstrated that ELANE can selectively kill a wide range of cancer cells while sparing proximal non-cancer cells and significantly attenuate tumorigenesis . This selective killing spans 35 different human or murine cancer cell lines across 11 tumor types, making ELANE a potential broad-spectrum anticancer agent .
The mechanism of ELANE's selective toxicity involves:
Liberation of the CD95 death domain (DD) that interacts with histone H1 isoforms
Triggering of an abscopal effect mediated by CD8+ T cells that prevents distant metastasis
Several factors contribute to this cancer-selective property:
Higher levels of CD95 in cancer cells compared to normal cells
Increased expression of histone H1 isoforms in malignant cells
The interaction between CD95-DD and histone H1 isoforms occurs predominantly in cancer cells
This discovery presents a promising direction for developing selective anticancer therapies based on the naturally evolved specificity of neutrophil elastase against genetically diverse targets. Mouse models with controlled ELANE expression could be valuable tools for further investigating these anticancer mechanisms and developing therapeutic approaches.
Researchers employ multiple complementary techniques to verify ELANE expression and function in mouse models:
1. RNA Expression Analysis:
RNA isolation from bone marrow using TRIzol reagent
Reverse transcription and PCR with gene-specific primers
Restriction enzyme digestion of PCR products to distinguish wild-type from mutant transcripts
2. Protein Expression Analysis:
Western blotting using specific anti-NE antibodies
Treatment with N-glycosidase F (PNGase F) to remove N-linked oligosaccharides for better detection
Generation of custom antibodies (e.g., rabbit anti-mouse NE antibody) targeting specific regions of the NE protein
3. Enzymatic Activity Assays:
Measurement of elastase activity using chromogenic, neutrophil elastase-specific substrates such as N-methoxysuccinyl Ala-Ala-Pro-Val p-nitroanilide
Normalization of elastolytic activity per microgram of protein
4. Functional Assays:
Analysis of granulopoiesis under basal and stress conditions
Evaluation of ER stress response in isolated granulocytic precursors
Assessment of neutrophil function and bacterial killing capacity
For example, in G193X Elane mice, researchers confirmed normal mRNA expression but found no detectable NE protein, suggesting post-translational degradation of the mutant protein . This combination of techniques provides a comprehensive assessment of ELANE expression and function at multiple levels, crucial for accurately characterizing mouse models.
Isolation and culture of neutrophil precursors from ELANE mouse models are essential for studying granulopoiesis and the effects of ELANE mutations. The following methodological approach is typically used:
Isolation of Hematopoietic Progenitors:
Harvest bone marrow cells from femurs and tibias of mice
Isolate c-Kit+lineage- hematopoietic progenitors using magnetic cell separation or fluorescence-activated cell sorting (FACS)
For fetal liver-derived progenitors, harvest fetal livers at 12-14 days post-conception
Culture Conditions for Granulocytic Differentiation:
Culture isolated progenitors in medium containing:
Kit ligand (stem cell factor)
Granulocyte-colony stimulating factor (G-CSF)
Appropriate base medium (e.g., RPMI 1640) supplemented with 10-20% fetal bovine serum
Maintain cultures for 3-7 days to obtain populations enriched for granulocytic precursors
Analysis of Differentiation:
Monitor cell morphology by Wright-Giemsa staining
Assess surface marker expression (e.g., Gr-1, Mac-1) by flow cytometry
Evaluate cell proliferation and viability using appropriate assays
Induction of ER Stress:
Treat cultured cells with ER stress-inducing agents such as tunicamycin or thapsigargin
Assess activation of UPR pathways through expression of markers like BiP, CHOP, and XBP1 splicing
Using this approach, researchers have demonstrated that granulocytic precursors from G193X Elane mice show increased sensitivity to chemical induction of ER stress, even though basal UPR activation is not significant . This methodology allows for detailed analysis of cell-autonomous effects of ELANE mutations on neutrophil development and response to stress.
The choice of genetic background can significantly influence the phenotypic expression of ELANE mutations in mouse models. Based on research practices, several genetic backgrounds have been used for ELANE mouse studies, each with advantages and considerations:
Common Genetic Backgrounds:
Inbred 129/SvJ background: Provides genetic homogeneity and reproducibility
Inbred C57Bl/6 background: Widely used standard background, obtained by backcrossing for 6+ generations
Outbred 129/SvJ × C57Bl/6 hybrid background: Offers genetic diversity that may better reflect human population variability
Considerations for Background Selection:
Genetic background can influence penetrance and expressivity of mutations
Strain-specific modifiers may compensate for or exacerbate ELANE mutation effects
Background strain characteristics (e.g., hematopoietic profile, immune system features) should align with research questions
Experimental Approach for Background Effect Assessment:
Generate and characterize the same ELANE mutation on multiple backgrounds
Compare phenotypes across backgrounds to identify strain-dependent effects
Consider using F2 intercrosses or recombinant inbred strains to map modifier loci
In studies with the G193X Elane mouse model, researchers analyzed mice on three different genetic backgrounds (inbred 129/SvJ, outbred 129/SvJ × C57Bl/6, and inbred C57Bl/6) to assess potential background effects on phenotype . This approach allows for detection of genetic modifiers that may influence the manifestation of ELANE mutations.
For transplantation studies, immunodeficient recipients such as NOD,B6.SCID Il2rγ−/−KitW41/W41 (NBSGW) mice may be preferred as they support multilineage engraftment of human hematopoietic cells without irradiation .
Interpreting discrepancies between different ELANE mouse models requires a systematic approach that considers multiple factors:
Methodological Considerations:
Mutation-specific effects: Different ELANE mutations may affect protein structure, stability, and function in distinct ways. For example, the G193X mutation causes truncation and rapid degradation of the NE protein, while other mutations might result in stable but misfolded proteins .
Model generation techniques: The method used to introduce mutations (e.g., knock-in, transgenic overexpression, CRISPR/Cas9) can influence phenotype severity and specificity.
Experimental conditions: Basal versus stress conditions may reveal phenotypes not apparent under standard housing conditions. G193X Elane mice show normal basal granulopoiesis but increased sensitivity to ER stress inducers .
Analytical Framework:
Compare multiple models side-by-side using standardized assays
Assess phenotypes at multiple levels:
Molecular (protein expression, activity)
Cellular (neutrophil development, function)
Organismal (susceptibility to infection, response to stress)
Consider species-specific differences in neutrophil biology
Integration with Human Data:
Correlate findings in mouse models with observations in patients carrying the corresponding ELANE mutations
Use patient-derived cells (e.g., induced pluripotent stem cells) to validate mechanisms observed in mouse models
The comparison of different models can provide valuable insights. For instance, while the V72M Elane mice showed normal granulopoiesis despite this mutation causing SCN in humans, the G193X Elane mice revealed a potential mechanism - rapid degradation of mutant protein - that might explain the lack of phenotype in some mouse models . These apparent discrepancies can thus drive deeper mechanistic understanding when systematically analyzed.
ELANE mouse models face several important limitations when used to study human neutropenia:
Phenotypic Divergence:
Most ELANE mutant mouse models fail to develop neutropenia despite carrying mutations homologous to those causing SCN in humans
Species-specific differences in neutrophil development regulation may account for this divergence
Molecular Differences:
Only 76% amino acid identity between human and mouse NE proteins
Structural differences may alter how mutations affect protein folding and function
Species-specific differences in protein quality control mechanisms
Compensatory Mechanisms:
Mice may have redundant pathways or compensatory mechanisms absent in humans
The UPR may function differently between species, with varying consequences for granulopoiesis
Experimental Challenges:
Limited access to primary human samples for direct comparison
Difficulties in studying cell-autonomous effects in complex in vivo systems
Limitations in accurately modeling human disease kinetics and progression
Future Directions to Address Limitations:
Development of humanized models (e.g., mice expressing human ELANE)
Combined approaches using mouse models and patient-derived iPSCs
Systems biology approaches to identify species-specific differences in neutrophil development networks
Study of ELANE knockout models compared to mutation models to distinguish loss-of-function from gain-of-toxic-function mechanisms
A promising approach has been the development of CRISPR/Cas9-mediated ELANE knockout models, which enable neutrophilic maturation of primary hematopoietic stem and progenitor cells and induced pluripotent stem cells from SCN patients . These findings suggest that complete absence of ELANE may be less detrimental than expression of mutant forms, highlighting the likely "gain-of-toxic-function" nature of ELANE mutations in SCN.
ELANE mouse models have emerging applications beyond neutropenia research, opening new avenues for investigation:
Cancer Research:
ELANE has demonstrated selective cytotoxicity against multiple cancer cell lines while sparing normal cells
ELANE mouse models could serve as platforms to study mechanisms of cancer-specific cell death
Potential for developing targeted cancer therapeutics based on ELANE's selectivity for cancer cells
Opportunity to investigate the abscopal effect mediated by CD8+ T cells that prevents distant metastasis
Inflammatory Disorders:
ELANE plays roles in several inflammatory conditions beyond neutropenia
Mouse models can help study neutrophil elastase's contribution to tissue damage in inflammatory diseases
Potential applications in studying auto-inflammatory syndromes as indicated by ClinVar annotations
Protein Folding and Quality Control:
ELANE mutations provide a model system for studying protein misfolding and ER stress responses
Insights may apply to other conditions involving protein misfolding (neurodegenerative diseases, etc.)
Opportunity to test therapeutic approaches targeting protein quality control mechanisms
Innate Immunity:
ELANE's role in neutrophil function makes these models valuable for studying innate immune responses
Models can be used to investigate neutrophil's role in eliminating genetically diverse pathogens
Potential applications in studying host-pathogen interactions
Therapeutic Development:
Testing of treatments that modulate UPR or enhance protein folding
Evaluation of gene editing approaches (like CRISPR/Cas9) for correcting ELANE mutations
Development of small molecules that might stabilize specific mutant forms of ELANE
Recent studies showing ELANE's selective anticancer activity represent a particularly promising direction. Cui et al. demonstrated that ELANE can kill 35 different human or murine cancer cells across 11 tumor types while sparing healthy cells . This broad-spectrum anticancer activity operates through mechanisms involving CD95 death domain liberation and interaction with histone H1 isoforms, suggesting therapeutic applications beyond neutropenia treatment.
Recombinant Mouse Neutrophil Elastase/ELA2 is derived from a mouse myeloma cell line, NS0. The protein sequence ranges from Ser27 to Arg260 and includes a C-terminal 10-His tag for purification purposes . The predicted molecular mass of the protein is approximately 27 kDa, but it appears as a 34 kDa band under reducing conditions in SDS-PAGE due to post-translational modifications .
Neutrophil Elastase/ELA2 is involved in various biological processes, including:
Recombinant Mouse Neutrophil Elastase/ELA2 is used in various research applications, including: