CELMoDs (cereblon E3 ligase modulatory drugs) are molecular glues that bind to cereblon (CRBN), inducing conformational changes to activate ubiquitin ligase activity. This promotes selective degradation of target proteins.
Mechanism: CELMoDs shift CRBN from an inactive open state to a closed, active conformation, enabling substrate recruitment for proteasomal degradation .
Efficacy: Next-gen CELMoDs enhance CRBN activation efficiency, improving therapeutic outcomes in preclinical models .
CEL-2000, a peptide developed using L.E.A.P.S.™ technology, showed promising results in collagen-induced arthritis models:
Parameter | CEL-2000 | Enbrel® |
---|---|---|
Dosing Frequency | 2 doses (days 0, 7/14) | Every other day |
Disease Progression | Significant reduction | Moderate reduction |
Symptoms | Less severe | Comparable |
Serological Changes | Anti-inflammatory markers observed | Similar markers observed |
CEL-2000 demonstrated equivalent or superior efficacy to Enbrel® with fewer doses, suggesting potential for reduced toxicity and improved patient compliance .
University of Michigan-developed BET degraders, leveraging PROTAC technology, suppressed tumor growth in castration-resistant prostate cancer xenografts:
Efficacy: Treated mice exhibited 60% smaller tumors vs. controls .
Mechanism: Degraders tag BET proteins (BRD2/3/4) for proteasomal recycling, overcoming resistance seen with traditional inhibitors .
A U-M-developed menin-MLL1 inhibitor induced complete remission in mice with MLL1-rearranged leukemia:
Sample Size: Syngeneic models require more mice than PDXs/CDXs to achieve comparable accuracy .
Analysis: Linear mixed models (LMMs) account for heterogeneity in tumor growth and drug response across cohorts .
Carboxyl ester lipase (CEL), previously known as cholesterol esterase or bile salt-stimulated lipase, is an enzyme that breaks down fats (lipolytic enzyme). It can hydrolyze various lipids, including cholesteryl esters, phospholipids, triglycerides, diglycerides, monoglycerides, ceramides, and lysophospholipids. The enzyme's activity is regulated by its carboxyl terminus, which interacts with a surface loop to partially block the active site. The active site, consisting of serine, histidine, and aspartate, is located in the center of the enzyme and is partially covered by this loop. When bile salts bind to the loop domain, the active site becomes accessible to water-insoluble substrates. CEL is primarily produced in the pancreas and lactating mammary gland but is also found in the liver, macrophages, and blood vessel walls.
Recombinant Mouse CEL was expressed in Sf9 insect cells using a Baculovirus expression system. It is a single, glycosylated polypeptide chain containing 585 amino acids (21-599 aa), resulting in a molecular mass of 64.5 kDa. A 6-amino acid His-tag is fused to the C-terminus for purification purposes. The protein is purified using proprietary chromatographic techniques.
The CEL solution is provided at a concentration of 0.5 mg/ml in a buffer containing 10% glycerol and Phosphate-Buffered Saline (pH 7.4).
For short-term storage (2-4 weeks), store the CEL solution at 4°C. For extended storage, freeze the solution at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is recommended for long-term storage. Avoid repeated freeze-thaw cycles to maintain protein stability.
The specific activity of CEL is greater than 100,000 pmol/min/µg. This is determined by measuring the amount of enzyme required to hydrolyze 1.0 µmole of p-nitrophenyl butyrate to p-nitrophenol per minute at pH 7.5 and 25°C.
Bile salt-activated lipase, BAL, EC 3.1.1.13, EC 3.1.1.3, Bile salt-stimulated lipase, BSSL, Bucelipase, Carboxyl ester lipase, Cholesterol esterase, Pancreatic lysophospholipase, Sterol esterase, CEL, FAP, BSDL, CELL, FAPP, LIPA, Cease, MODY8.
AKLGAVYTEG GFVEGVNKKL SLLGGDSVDI FKGIPFATAK TLENPQRHPG WQGTLKATNF
KKRCLQATIT QDNTYGQEDC LYLNIWVPQG RKQVSHNLPV MVWIYGGAFL MGSGQGANFL
KNYLYDGEEI ATRGNVIVVT FNYRVGPLGF LSTGDANLPG NFGLRDQHMA IAWVKRNIAA
FGGDPDNITI FGESAGAASV SLQTLSPYNK GLIRRAISQS GMALSPWAIQ KNPLFWAKTI
AKKVGCPTED TGKMAACLKI TDPRALTLAY KLPVKKQEYP VVHYLAFIPV IDGDFIPDDP
INLYNNTADI DYIAGINNMD GHLFATIDVP AVDKTKQTVT EEDFYRLVSG HTVAKGLKGA
QATFDIYTES WAQDPSQENM KKTVVAFETD VLFLIPTEIA LAQHKAHAKS AKTYSYLFSH
PSRMPIYPKW MGADHADDLQ YVFGKPFATP LGYRPQDRAV SKAMIAYWTN FARSGDPNMG
NSPVPTHWYP YTLENGNYLD ITKTITSASM KEHLREKFLK FWAVTFEVLP TVTGDQDTLT
PPEDDSEVAP DPPSDDSQVV PVPPTDDSVE AQMPATIGFH HHHHH
Bile salt-activated lipase (CEL) is a 71,834 Da protein that plays a critical role in lipid metabolism in mice. The protein is encoded by the CEL gene (OMIM 609812) and functions primarily as a digestive enzyme that hydrolyzes cholesterol esters and other lipids . In mouse models, CEL serves as an important biomarker for various physiological and pathological processes related to lipid metabolism. Detection of CEL in mouse samples typically requires sensitive assays such as ELISA, which can detect the protein in serum, plasma, tissue homogenates, and cell culture supernatants with detection ranges as low as 18.75-1200 pg/ml . Understanding the baseline expression and function of CEL in healthy mice provides a foundation for interpreting alterations in disease models or experimental interventions.
Mouse models express CEL in patterns that share important similarities with humans but also exhibit notable differences that researchers must account for in experimental design. While both species express the protein, there are differences in tissue distribution, regulation, and functional characteristics.
Tissue-specific expression patterns
Regulatory mechanisms controlling gene expression
Post-translational modifications affecting enzyme activity
Interaction with other metabolic pathways
When utilizing mouse models for CEL research, it is essential to understand that findings may not directly translate to human biology due to these differences. Similar to how mouse models of human lung disease require careful consideration of anatomical and physiological differences , CEL researchers must account for species variations when designing experiments and interpreting results.
The quantification of CEL in mouse samples requires sensitive and specific detection methods. Based on current research practices, the following approaches provide reliable measurements:
Sandwich ELISA: The most widely used method employs antibodies specific for mouse CEL pre-coated onto microplates. This technique offers high sensitivity (as low as 4.68 pg/ml) and excellent specificity with minimal cross-reactivity . The assay typically involves:
Sample binding to immobilized antibody
Addition of biotin-conjugated antibody specific for CEL
Application of avidin-conjugated HRP
Substrate solution addition for colorimetric detection
Western Blotting: While less quantitative than ELISA, Western blotting provides information on protein size and potential modifications.
PCR-based methods: Quantitative PCR can measure CEL mRNA levels, though protein levels may not directly correlate with transcript abundance.
Mass Spectrometry: For more detailed characterization of CEL protein variants and modifications.
When selecting a method, researchers should consider sample type, expected concentration range, and required sensitivity. For most applications, sandwich ELISA represents the gold standard with intra-assay precision (CV%) less than 8% and inter-assay precision less than 10% .
Cell density represents a critical yet often overlooked variable in mouse cell culture systems studying CEL expression. Recent research has demonstrated that cell density serves as an effective tool for controlling how mouse cells pattern themselves into complex structures, which can significantly impact protein expression patterns, including CEL .
In studies conducted by Morsut and Thomson, mouse connective tissue cells and stem cells engineered with synthetic cellular communication systems showed dramatically different patterning behaviors based solely on cell density variations . This phenomenon has important implications for CEL research:
Threshold Effects: Above certain cell density thresholds, cellular signaling pathways can be attenuated, potentially affecting CEL expression and activity.
Dynamic Regulation: As cells proliferate at different rates, the constantly shifting density creates a dynamic regulatory environment that interacts with genetic circuits controlling CEL expression.
Reproducibility Challenges: Experiments involving genetically identical cells may produce different outcomes based on initial seeding density, explaining why some researchers observe inconsistent CEL expression patterns in seemingly identical experimental setups.
When designing in vitro experiments to study CEL, researchers should standardize initial seeding densities and monitor proliferation rates to account for this variable. Additionally, gradient experiments across different cell densities may reveal threshold effects that influence CEL expression and function .
Translating findings from CEL mouse models to human disease contexts requires careful consideration of several factors that influence interpretability and relevance:
Genetic Differences: Despite remarkable genetic homology between mice and humans, there are significant differences in gene expression and regulation that affect CEL function. The Mouse ENCODE Consortium has revealed divergence in transcription factor networks that may impact immune system function and metabolism-related pathways relevant to CEL activity .
Physiological Differences: Mouse metabolism operates at a different rate than humans, which affects how CEL functions in the digestive and metabolic systems. Additionally, differences in organ size, blood volume, and body temperature influence how CEL functions systemically.
Cell Biological Considerations: Mouse cells have longer telomeres (5-10 fold) than human cells, resulting in greater replicative capacity, which can affect long-term studies of CEL function . Additionally, mouse fibroblasts can spontaneously immortalize in culture, unlike human fibroblasts, potentially confounding in vitro studies.
Model Validation: Researchers should validate their mouse model by confirming whether it accurately recapitulates both the etiology and presentation of the human disease being studied. For CEL-related diseases, this means ensuring the model correctly mimics the pathophysiological mechanisms and responds to interventions in ways that parallel human responses .
A multilayered validation approach is recommended, comparing mouse and human CEL at the genomic, transcriptomic, proteomic, and functional levels before making translational claims about disease mechanisms or treatment efficacy.
Synthetic biology offers powerful approaches to manipulate and study CEL function in mouse models with unprecedented precision. Recent advances demonstrate how engineered genetic circuits can provide new insights into protein function and regulation:
SynNotch Systems: Researchers have developed synthetic cellular communication systems using engineered proteins like synNotch, which serves as a programmable sensor on cell surfaces. This technology can be adapted to study CEL by engineering mouse cells with synthetic circuits that respond to CEL activity or regulate CEL expression .
Fluorescent Reporters: By coupling CEL expression to fluorescent proteins, researchers can visualize CEL activity in real-time within living cells and tissues. This approach enables observation of dynamic patterns and spatial distribution of CEL activity .
Inducible Expression Systems: Synthetic biology tools allow for temporal control of CEL expression, enabling researchers to study the effects of CEL upregulation or downregulation at specific developmental stages or disease progression points.
Engineered Interaction Networks: By creating synthetic protein-protein interaction networks that include CEL, researchers can systematically map the functional relationships between CEL and other proteins in metabolic and signaling pathways.
These approaches can help address complex questions about CEL function that traditional genetic knockout or overexpression studies cannot answer. For example, synNotch systems could be designed to trigger CEL expression in response to specific metabolic states, allowing researchers to study the downstream effects with spatial and temporal precision .
Investigating protein-protein interactions (PPIs) involving CEL in mouse models requires specialized techniques that can detect both stable and transient interactions. The following methodological approaches have proven effective:
Co-Immunoprecipitation (Co-IP): This classical approach remains valuable for studying stable interactions between CEL and other proteins in mouse tissue lysates or cell extracts. The technique involves:
Preparation of mouse tissue/cell lysates under non-denaturing conditions
Immunoprecipitation using anti-CEL antibodies
Western blot analysis to identify co-precipitated proteins
Proximity Ligation Assays (PLA): This technique can detect protein interactions with high sensitivity in fixed mouse tissues or cells, providing spatial information about where interactions occur.
Menin-MLL1 Inhibition Paradigm: Drawing from recent advances in protein-protein interaction studies, researchers can apply approaches similar to those used in studying menin-MLL1 interactions . The MI-3454 compound development provides a methodological framework that can be adapted to study CEL interactions, particularly for identifying small molecules that might modulate CEL binding to partner proteins.
Transgenic Reporter Systems: Mouse models can be engineered with split-reporter systems where protein fragments reconstitute active reporter proteins only when the proteins of interest (including CEL) interact.
When investigating potential therapeutic compounds targeting CEL interactions, researchers should follow rigorous validation protocols similar to those used for the menin-MLL1 inhibitor, including in vitro binding assays, cell-based functional assays, and ultimately in vivo validation in appropriate mouse disease models .
Mouse strain selection represents a critical variable in CEL research that can significantly impact experimental outcomes and interpretability. Different inbred mouse strains exhibit substantial variation in CEL expression levels, activity, and regulation due to their distinct genetic backgrounds:
Baseline Expression Variations: Commonly used laboratory strains such as C57BL/6, BALB/c, and 129Sv may have different baseline CEL expression levels in relevant tissues, requiring strain-specific normalization in comparative studies.
Strain-Specific Responses: Genetic variation between strains can lead to different responses to the same experimental intervention affecting CEL. This phenomenon mirrors the genetic diversity in human populations and can provide valuable insights into potential variable responses in humans.
Immunological Differences: Mouse strains exhibit significant variation in immune system function and inflammatory responses, which can indirectly affect CEL expression and activity through cytokine signaling and other immune mediators .
To address these variations in experimental design:
Use consistent strains across all experimental groups
Include proper strain-matched controls
Consider using multiple strains to assess the robustness of findings
Report strain information in detail in publications
Consider the potential impact of the strain's genetic background on the specific CEL-related pathway being studied
Understanding the "genetic evolution of laboratory strains of mice, the effect of these genetic and immunological changes on mouse biology, and the impact on the translation of these results to human medicine" is crucial for proper experimental design and interpretation in CEL research .
Optimizing CEL ELISA assays for mouse samples requires attention to several technical parameters to ensure reliable and reproducible results:
Sample Preparation:
Serum/Plasma: Collect samples using consistent methods (e.g., cardiac puncture vs. tail vein) and anticoagulants
Tissue Homogenates: Use standardized buffer compositions and homogenization protocols
Cell Culture Supernatants: Consider collection timing relative to feeding schedules
Assay Conditions:
Temperature: Maintain consistent incubation temperature (typically room temperature or 37°C)
Incubation Times: Standardize all incubation periods
Washing Steps: Ensure thorough washing between steps to minimize background
Standard Curve Optimization:
Quality Control:
Data Analysis:
For specialized applications, such as measuring CEL in specific tissues or under certain experimental conditions, further optimization may be necessary to account for matrix effects and potential interferents.
Contradictory results in CEL mouse studies often stem from methodological variations and unrecognized confounding factors. Researchers can reconcile such discrepancies through systematic improvements in experimental design:
By implementing these strategies, researchers can transform seemingly contradictory results into valuable insights about the contextual factors that modulate CEL expression and function in mouse models.
Translating findings from CEL mouse models to human applications requires systematic approaches that address species differences while leveraging biological conservation:
Comparative Biology Framework:
Establish detailed molecular and functional comparison between mouse and human CEL
Map conservation at sequence, structure, expression pattern, and regulatory network levels
Identify species-specific differences that might limit translation
Validation Hierarchy:
Begin with in vitro validation using both mouse and human cells
Progress to ex vivo tissue preparations from both species
Confirm findings in multiple mouse models before moving to human studies
Verify results in human biospecimens when available
Cross-Species Experimental Design:
Computational Approaches:
Employ bioinformatic analyses to predict human responses based on mouse data
Use systems biology models to account for species-specific network differences
Apply machine learning algorithms trained on successful and failed translations
Translational Biomarkers:
Identify conserved biomarkers that predict therapeutic responses across species
Develop assays that function equivalently in mouse and human samples
Focus on pathway-level conservation rather than individual molecule identities
The successful translation of findings related to the menin-MLL1 protein-protein interaction inhibitor (MI-3454) from mouse models to human clinical trials provides an instructive paradigm for CEL research . This compound showed "profound" activity in mouse models before advancing to human studies, demonstrating effective translational progression.
CEL plays pivotal roles in mouse models of metabolic and digestive disorders, serving as both a biomarker and potential therapeutic target:
Lipid Metabolism Disorders:
CEL participates in cholesterol ester hydrolysis and fat absorption
Alterations in CEL activity correlate with lipid accumulation phenotypes
Mouse models with modified CEL expression show varying susceptibilities to high-fat diet-induced disorders
Pancreatic Function:
CEL is expressed in pancreatic tissue and impacts digestive enzyme secretion
Mouse models of pancreatitis show altered CEL expression profiles
CEL mutations in mice can lead to pancreatic dysfunction phenotypes
Diabetes Research:
CEL influences lipid metabolism, which intersects with insulin signaling pathways
Mouse models suggest CEL may play roles in β-cell function and lipotoxicity
Diabetes-prone mouse strains show characteristic patterns of CEL expression
Digestive Disorders:
CEL participates in intestinal fat digestion and absorption
Mouse models of malabsorption syndromes often show CEL abnormalities
CEL function impacts gut microbiome composition through effects on lipid availability
Age-Related Changes:
CEL expression and activity change throughout the mouse lifespan
Age-related metabolic disorders in mice correlate with altered CEL function
Interventions affecting CEL may impact age-related metabolic dysfunction
When designing studies to investigate these relationships, researchers should carefully consider the specific mouse strain being used, as genetic background significantly influences metabolic phenotypes and may interact with CEL function in complex ways .
Emerging technologies present unprecedented opportunities to deepen our understanding of CEL function in mouse models:
CRISPR-Cas9 Gene Editing:
Create precise modifications in CEL gene structure and regulatory elements
Generate knock-in reporters to visualize CEL expression in real-time
Develop inducible CEL expression systems for temporal control
Single-Cell Technologies:
Apply single-cell RNA sequencing to map CEL expression across diverse cell populations
Use single-cell proteomics to correlate CEL protein levels with other cellular markers
Implement spatial transcriptomics to visualize CEL expression patterns within intact tissues
Synthetic Biology Approaches:
Advanced Imaging:
Apply intravital microscopy to observe CEL activity in living mice
Use label-free imaging techniques to study CEL without introducing artifacts
Implement correlative light and electron microscopy to link CEL function to ultrastructural features
Artificial Intelligence Integration:
Develop machine learning algorithms to predict CEL interactions and functions
Use AI to identify patterns in multi-omic datasets related to CEL
Apply computational modeling to simulate CEL activity under various conditions
The integration of these technologies with established approaches will likely transform our understanding of CEL biology, particularly when combined with controlled manipulation of cell density, which has been shown to significantly influence cellular behavior and protein expression in mouse models .
Ethical considerations in CEL mouse model research require balancing scientific advancement with animal welfare:
These ethical considerations align with the concluding discussion in search result , which emphasizes the importance of balancing technological advances with ethical considerations in mouse model research, particularly as new methods become available that may reduce reliance on animal models while improving translational relevance.
Carboxyl Ester Lipase (CEL), also known as bile salt-stimulated lipase, is an enzyme with significant lipolytic capabilities. It is capable of hydrolyzing a variety of substrates, including cholesteryl esters, acylglycerols, and ceramide . This enzyme is highly expressed by pancreatic acinar cells and is secreted into the gastrointestinal tract, where it plays a crucial role in the digestion of dietary lipids .
CEL belongs to the Type-B carboxyl esterase/lipase family of enzymes. It possesses a catalytic triad consisting of serine, histidine, and aspartate, which enables it to hydrolyze both ester and amide bonds . The enzyme’s active site is partly shielded by a surface loop, which is controlled by the carboxyl terminus of the enzyme . Bile salt binding to the loop domain frees the active site, allowing accessibility by water-insoluble substrates .
Recombinant Mouse Carboxyl Ester Lipase is produced in various expression systems, including mouse myeloma cell lines and Sf9 Baculovirus cells . The recombinant protein is often tagged with a 6-His tag at the C-terminus to facilitate purification . The recombinant form is typically supplied as a sterile, filtered solution and is stored under specific conditions to maintain stability and activity .
Recombinant Mouse Carboxyl Ester Lipase is used in various research applications, including studies on lipid metabolism, enzyme kinetics, and digestive processes. Its ability to hydrolyze a wide range of substrates makes it a valuable tool for understanding the biochemical pathways involved in lipid digestion and absorption .