ENTPD6 Mouse Recombinant produced in HEK293 cells is a single, glycosylated polypeptide chain (33-455 a.a) containing 433 amino acids and having a molecular mass of 47.3 kDa.
ENTPD6 is fused to a 6 amino acid His-tag at C-terminus & purified by proprietary chromatographic techniques.
ectonucleoside triphosphate diphosphohydrolase 6 isoform1, 2700026H11Rik, Cd39l, Cd39l2, dJ738P15.3, NTPDa, NTPDase-6, Entpd6.
HEK293 cells.
DGSMKWHRAS AAQAFFTIAG AASGARWTQQ AFSSPGSAAR GHEVFYGIMF DAGSTGTRIH VFQFARPPGE TPTLTHETFK ALKPGLSAYA DDVEKSAQGI QELLNVAKQH IPYDFWKATP LVLKATAGLR LLPGEKAQKL LQKVKEVFKA SPFLVGDDCV SIMNGTDEGV SAWITVNFLT GSLKTPGSSS VGMLDLGGGS TQITFLPRVE GTLQASPPGH LTALQMFNRT
YKLYSYSYLG LGLMSARLAI LGGVEGKPAE NDKELVSPCL SPRFRGEWEH AEVTYRISGQ KAVGLYELCA SRVSEVLRNK VHRTEEAQHV DFYAFSYYYD LAASFGLIDA EKGGSLVVGD FEIAAKYVCR TLETQPPSSP FACMDLTYIS LLLHEFGFPG DKVLKLARKI DNVETSWALG AIFHYIDSLK RQKVPALHHH HHH
ENTPD6, also known as CD39L2, is an ectonucleoside triphosphate diphosphohydrolase that is encoded in the mouse genome. It is specifically located on chromosome 2 of mice . The gene encodes a protein that functions as an enzyme involved in nucleotide metabolism. In mouse models, ENTPD6 is studied for its potential roles in various physiological and pathological processes, often through conditional knockout approaches that enable tissue-specific investigation of its function .
Several ENTPD6 mouse models have been developed, with conditional knockout mice being particularly valuable for research. These models are typically generated using CRISPR/Cas technology, where sgRNA and ssDNA are designed to target the Entpd6 gene . One specific example is the conditional knockout model with the allele designation Entpd6tm1a(KOMP)Wtsi, which is a "Knockout First, Reporter-tagged insertion with conditional potential" type . This model was created through the International Knockout Mouse Consortium (IKMC) and is available through repositories such as the Wellcome Sanger Institute . The generation process typically involves high-throughput electroporation of fertilized eggs followed by sperm collection for cryopreservation after the mice reach sexual maturity .
Genotyping ENTPD6 mouse models requires a combination of PCR reactions to determine the presence of wild-type and mutant alleles. According to established protocols, three standard PCR reactions are recommended:
Assay Type | Primer Pair | Expected Band Size (bp) |
---|---|---|
Wild-type | Entpd6_35296_F + Entpd6_35296_R | 398 |
Mutant | Entpd6_35296_F + CAS_R1_Term | 309 |
Cassette | LacZ_2_small_F + LacZ_2_small_R | 108 |
The specific primer sequences are:
Entpd6_35296_F: CCACTCCTCCGAACATCAAAC
Entpd6_35296_R: AGAGCAGACAACTCACACCCTAAC
CAS_R1_Term: TCGTGGTATCGTTATGCGCC
LacZ_2_small_F: ATCACGACGCGCTGTATC
LacZ_2_small_R: ACATCGGGCAAATAATATCG
The PCR reaction should use standard amplification conditions with an annealing temperature of 58°C . The genotype is determined by interpreting the consolidated results from these reactions. For example, a heterozygous mouse would be positive for the cassette, mutant, and wild-type bands .
Mouse ENTPD6 (UniProt ID: Q3U0P5) is characterized as follows:
Characteristic | Information |
---|---|
Protein Name | Ectonucleoside triphosphate diphosphohydrolase 6 |
Gene Name | Entpd6 (Synonyms: Cd39l2) |
Organism | Mus musculus (Mouse) |
PRO ID | PR:Q3U0P5 |
PRO Name | ectonucleoside triphosphate diphosphohydrolase 6 (mouse) |
Definition | An ectonucleoside triphosphate diphosphohydrolase 6 encoded in the mouse genome |
Short Label | mENTPD6 |
Category | organism-gene |
Post-translational modifications identified for ENTPD6 include ubiquitination at K3 and phosphorylation at Y262, as reported in PhosphoSitePlus .
When designing experiments to evaluate tissue-specific effects of ENTPD6 deletion, researchers should employ a systematic approach that combines molecular validation with functional assessments. Begin by confirming the conditional knockout efficiency using tissue-specific PCR genotyping and protein expression analysis (western blotting or immunohistochemistry). For tissue-specific deletion, the Entpd6tm1a(KOMP)Wtsi conditional allele can be converted to functional knockout using tissue-specific Cre recombinase expression .
The experimental design should include:
Molecular validation: Confirm deletion efficiency at DNA (PCR), RNA (qRT-PCR), and protein levels (western blot/immunohistochemistry).
Physiological assessments: Based on the tissue targeted, include relevant functional tests. For neurological studies, consider behavioral assays; for metabolic tissues, include glucose tolerance tests; for immunological studies, incorporate cell population analysis and functional immune assays.
Control groups: Include both Cre-negative floxed mice and wild-type controls to distinguish between Cre-mediated effects and ENTPD6 deletion effects.
Temporal considerations: For inducible systems, establish appropriate timepoints post-induction to capture both immediate and long-term consequences of ENTPD6 deletion.
Remember that rapid conversion of the conditional alleles can be achieved using cell-permeable Cre recombinase as described by Ryder et al. (2013) , which may be advantageous for certain experimental designs.
Currently, comprehensive phenotypic characterization of ENTPD6-deficient mice appears limited in the available literature . The datasheet from the Wellcome Sanger Institute indicates that phenotypic data should be available through the International Mouse Phenotyping Consortium (IMPC), but specific phenotypes are not directly listed in the search results .
To address contradictory findings that may emerge in ENTPD6 research:
Genetic background considerations: Carefully document the exact strain background (e.g., C57BL/6JCya or C57BL/6N) as genetic background can significantly influence phenotypes . The EUCOMM/KOMP-CSD ES cell resources typically use C57BL/6N embryonic stem cells as described by Pettitt et al. (2009) .
Age and sex considerations: Stratify analyses by age and sex, as ENTPD6-related phenotypes may exhibit age or sex-specific manifestations.
Environmental factors: Standardize housing conditions, diet, and microbiome status across experimental cohorts.
Molecular mechanism analysis: When contradictory phenotypes are observed, conduct in-depth molecular analyses to identify potential compensatory mechanisms or strain-specific modifiers.
Cross-validation approach: Employ multiple experimental approaches and models (e.g., comparing conditional knockout with siRNA knockdown or pharmacological inhibition) to validate observed phenotypes.
When contradictory findings emerge, a systematic meta-analysis approach comparing experimental conditions, genetic backgrounds, and methodological differences can help resolve discrepancies.
Optimizing CRISPR/Cas9-mediated generation of ENTPD6 mouse models requires several advanced considerations beyond standard protocols:
Enhanced sgRNA design:
Utilize multiple prediction algorithms to select sgRNAs with minimal off-target effects
Design sgRNAs targeting conserved functional domains of ENTPD6
Include sgRNA efficiency validation in mouse cell lines before in vivo application
Delivery optimization:
Homology-directed repair (HDR) enhancement:
For precise mutations or insertions, include small molecule HDR enhancers (e.g., RS-1, SCR7)
Optimize homology arm length for specific modifications
Consider asymmetric donor design for improved HDR efficiency
Screening strategies:
Implement high-throughput genotyping workflows
Utilize droplet digital PCR for more sensitive detection of editing events
Consider next-generation sequencing for comprehensive assessment of on-target and potential off-target modifications
Mosaic reduction strategies:
Deliver editing components earlier in development
Consider maternal expression of Cas9 followed by zygotic sgRNA delivery
Implement strategies that select for non-mosaic founder animals
These optimizations can significantly improve the efficiency and precision of generating ENTPD6 mouse models, particularly when complex modifications are required.
Studying ENTPD6 enzymatic activity in mouse tissues requires specialized approaches due to its role as an ectonucleoside triphosphate diphosphohydrolase. The following methodological framework is recommended:
Tissue preparation optimization:
Fresh tissue collection in appropriate buffers that preserve enzymatic activity
Subcellular fractionation to isolate relevant membrane compartments where ENTPD6 is active
Careful temperature control throughout processing
Activity assays:
Colorimetric phosphate release assays using nucleotide substrates (ATP, ADP)
HPLC-based methods to detect substrate consumption and product formation
Real-time monitoring of enzymatic activity using fluorescent or bioluminescent reporters
Substrate specificity analysis:
Comparative analysis of different nucleotides as substrates
Kinetic parameter determination (Km, Vmax) for different tissues
Competition assays to determine substrate preferences
Inhibitor studies:
Use of specific inhibitors to validate assay specificity
Dose-response relationships in different tissues
Inhibitor competition studies to characterize binding sites
In situ activity visualization:
Enzyme histochemistry using lead phosphate precipitation methods
Fluorescent substrate analogues for live tissue imaging
Correlation of activity with protein localization using immunohistochemistry
It's essential to include appropriate controls, particularly tissues from ENTPD6 knockout mice, to differentiate ENTPD6-specific activity from other ectonucleotidases that may have overlapping substrate preferences.
Converting between different ENTPD6 conditional allele variants requires specific recombinase-mediated strategies. The Entpd6tm1a(KOMP)Wtsi allele, described in the search results, is a "Knockout First, Reporter-tagged insertion with conditional potential" that can be converted to other functional variants . For effective conversion:
tm1a to tm1c (conditional ready) conversion:
Use Flp recombinase to remove the FRT-flanked cassette
This maintains the loxP-flanked critical exon while removing the reporter and selection elements
Can be accomplished by crossing with Flp-expressing mice (e.g., FLPeR mice described by Farley et al., 2000)
Alternatively, cell-permeable Flp protein can be used in early embryos
tm1a to tm1b (lacZ-tagged null) conversion:
Use Cre recombinase to remove the loxP-flanked critical exon
Maintains the lacZ reporter for expression mapping
Can be accomplished by crossing with ubiquitous Cre-expressing mice
Results in a constitutive knockout with reporter expression
tm1c to tm1d (conditional knockout) conversion:
For validation of conversion, specific PCR strategies are required for each allele type. The EUCOMM/KOMP resource provides detailed genotyping guides for distinguishing between these allele variants, with specific primers and protocols available through the International Mouse Phenotyping Consortium (IMPC) website .
Maintaining ENTPD6 mouse colonies while minimizing genetic drift requires structured breeding strategies and regular quality control. Based on established practices for genetically modified mouse lines:
Breeding strategy implementation:
Establish a hierarchical breeding structure with dedicated breeders separated from experimental cohorts
Implement rotational breeding schemes to maintain genetic diversity
Regularly backcross to the original background strain (e.g., C57BL/6N for EUCOMM/KOMP-derived lines)
Maintain heterozygotes as breeding stock when homozygotes have reduced fertility
Cryopreservation program:
Establish cryopreserved stocks of sperm and/or embryos from early generations
As mentioned in the search results, sperm cryopreservation is performed after sexual maturity for ENTPD6 mouse models
Periodically refresh cryopreserved stocks to capture current colony status
Maintain detailed records of cryopreserved material, including generation number and genetic background
Quality control monitoring:
Genetic background considerations:
Be aware of subtle differences between C57BL/6 substrains (e.g., C57BL/6J vs. C57BL/6N)
The ENTPD6 mouse model described is on C57BL/6JCya background or C57BL/6N (EUCOMM/KOMP)
Note that genomic copy number variations have been documented in embryonic stem cells as reported by Liang et al. (2008)
Consider the potential impact of a spontaneous albinism observed in some C57BL/6N strains (Ryder et al., 2013)
These practices will help maintain colony integrity and ensure experimental reproducibility across generations.
Integrating ENTPD6 mouse models with other genetic models requires careful planning and strategic breeding approaches:
Breeding strategy design:
Map out the required crossings to achieve desired genotypes
Calculate the expected genotype frequencies at each generation
Plan breeding schemes that minimize the number of generations to achieve experimental cohorts
Consider whether sequential or parallel crossing strategies are more efficient
Compound mutant validation:
Develop multiplexed genotyping protocols that can simultaneously detect all relevant alleles
Confirm that multiple genetic modifications don't interfere with each other's expression or function
Validate that phenotypes in compound mutants represent true genetic interactions rather than additive effects
Check for unexpected recombination events between loci
Background strain harmonization:
Ensure all models are on the same genetic background or backcross to achieve uniformity
Be particularly careful with mixing C57BL/6 substrains, as the ENTPD6 models may be on C57BL/6JCya or C57BL/6N
Document the generation number and background strain percentage of all models
Consider using marker-assisted selection to accelerate background harmonization
Inducible system integration:
For temporal control, incorporate inducible systems (e.g., tetracycline-responsive or tamoxifen-inducible)
Validate that the ENTPD6 conditional allele is compatible with chosen inducible systems
Develop protocols for sequential activation of different genetic modifications
Optimize inducer dosing to achieve desired recombination efficiency
Experimental controls:
Generate all possible genetic combinations as controls
Include single-transgenic/single-mutant controls alongside compound mutants
Consider the impact of Cre or other recombinases alone on phenotypes
Maintain wild-type controls from the same colony for comparative analyses
This structured approach will facilitate the development of complex genetic models incorporating ENTPD6 modifications while maintaining experimental rigor.
ENTPD6 mouse models offer powerful tools for investigating nucleotide metabolism and signaling pathways. While the search results don't detail specific applications, the following represent key research directions based on the function of ENTPD6 as an ectonucleoside triphosphate diphosphohydrolase:
Purinergic signaling investigation:
Tissue-specific deletion of ENTPD6 can reveal compartmentalized roles in regulating extracellular ATP/ADP levels
Analysis of downstream purinergic receptor activation patterns in conditional knockout models
Integration with other ectonucleotidase knockouts to map nucleotide degradation pathways
Metabolic regulation studies:
Investigation of ENTPD6's role in cellular energy homeostasis
Analysis of interactions between nucleotide metabolism and other metabolic pathways
Potential roles in metabolic diseases through altered nucleotide availability
Immune regulation applications:
Given that CD39 family members regulate immune functions through ATP/adenosine modulation
Investigation of ENTPD6 in immune cell development and function
Potential applications in understanding inflammatory diseases
Developmental biology:
Analysis of ENTPD6's role during embryonic development
Tissue-specific requirements for nucleotide metabolism during organogenesis
Potential developmental timing mechanisms regulated by extracellular nucleotides
Pathophysiological models:
Integration of ENTPD6 conditional knockout with disease models to assess therapeutic potential
Investigation of ENTPD6 as a target in cancer, inflammation, and metabolic disorders
Comparative analysis with human ENTPD6-associated conditions
These applications represent promising research directions that leverage the flexibility of conditional ENTPD6 mouse models to address fundamental questions in nucleotide biology and disease mechanisms.
Integrating single-cell approaches with ENTPD6 mouse models can provide unprecedented insights into cell-specific functions and heterogeneity:
Single-cell transcriptomics applications:
Compare transcriptional profiles between wild-type and ENTPD6-deficient cells within tissues
Identify cell populations most affected by ENTPD6 deletion
Map compensatory transcriptional networks activated in response to ENTPD6 deficiency
Track developmental trajectories altered by ENTPD6 deletion in lineage-tracing studies
Single-cell proteomics integration:
Analyze proteome changes at single-cell resolution in ENTPD6-deficient tissues
Identify cell-specific post-translational modifications affected by altered nucleotide metabolism
Correlate protein expression patterns with enzymatic activity measurements
Spatial transcriptomics/proteomics approaches:
Map ENTPD6 expression patterns with spatial resolution in tissues
Correlate spatial expression with functional outcomes in conditional knockout models
Identify neighborhood effects where ENTPD6-expressing cells influence adjacent cell populations
Functional single-cell assays:
Develop microfluidic approaches to measure ENTPD6 activity in individual cells
Implement single-cell calcium imaging to assess purinergic signaling responses
Correlate metabolic profiles with ENTPD6 activity at single-cell resolution
Computational integration frameworks:
Develop multi-omics integration methods specific to nucleotide metabolism
Create cell-cell communication maps based on purinergic signaling networks
Model the impact of ENTPD6 deletion on tissue microenvironments
These approaches can be particularly powerful when applied to heterogeneous tissues where ENTPD6 function may vary between cell types, providing insights that would be masked in bulk tissue analyses.
Phenotyping ENTPD6 mouse models presents several challenges that require methodological considerations:
Enzymatic redundancy issues:
Challenge: Other ectonucleotidases may compensate for ENTPD6 deficiency
Solution: Implement comprehensive profiling of related enzymes (ENTPD1-8)
Approach: Combine targeted activity assays with gene expression analysis
Validation: Consider generating compound knockout models to address redundancy
Tissue-specific expression variability:
Challenge: ENTPD6 expression varies across tissues, complicating phenotype interpretation
Solution: Implement systematic tissue screening approaches
Approach: Use the lacZ reporter in tm1a or tm1b alleles to map expression patterns
Validation: Confirm expression patterns through multiple methods (qPCR, western blot, immunohistochemistry)
Nucleotide measurement challenges:
Challenge: Rapid turnover of extracellular nucleotides complicates accurate measurement
Solution: Develop optimized nucleotide sampling and preservation protocols
Approach: Consider using enzyme inhibitors during sample collection
Validation: Implement internal standards and rapid quenching methods
Phenotypic subtlety:
Challenge: ENTPD6 deficiency may produce subtle phenotypes easily missed
Solution: Implement high-sensitivity phenotyping protocols
Approach: Consider stress conditions that may unmask latent phenotypes
Validation: Increase cohort sizes for sufficient statistical power
Developmental compensation:
Challenge: Developmental adaptations may mask the importance of ENTPD6
Solution: Utilize inducible knockout strategies in adult mice
Approach: Compare constitutive versus acute ENTPD6 deletion
Validation: Time-course studies following inducible deletion
Addressing these challenges requires a combination of optimized methodologies, careful experimental design, and appropriate controls to detect and validate ENTPD6-specific phenotypes.
Inconsistent PCR genotyping results for ENTPD6 mouse models can stem from various technical factors. Here's a systematic troubleshooting approach:
Sample quality optimization:
Issue: Poor DNA quality from tail biopsies or other tissues
Solution: Implement standardized tissue collection and DNA extraction protocols
Approach: Compare commercial extraction kits versus in-house methods
Validation: Include DNA quality/quantity assessment before PCR
PCR reaction optimization:
Primer-specific troubleshooting:
Multiplex versus individual reactions:
Issue: Competition between primers in multiplex reactions
Solution: Compare multiplex versus individual PCR reactions
Approach: Adjust primer concentrations to balance amplification efficiency
Validation: Run side-by-side comparisons with known samples
Alternative genotyping strategies:
A standardized troubleshooting protocol incorporating these elements can systematically address inconsistent genotyping results and establish reliable workflows for ENTPD6 mouse colony management.
Ectonucleoside Triphosphate Diphosphohydrolase 6 (ENTPD6), also known as NTPDase 6 or CD39L2, is a member of the ectonucleoside triphosphate diphosphohydrolase family. These enzymes are crucial in the regulation of extracellular nucleotide levels, which play significant roles in various physiological processes, including purinergic signaling, inflammation, and tissue repair .
ENTPD6 is a protein-coding gene that encodes an enzyme responsible for the hydrolysis of nucleoside triphosphates and diphosphates. This enzyme operates in a calcium- or magnesium-dependent manner and exhibits a strong preference for nucleoside diphosphates. It preferentially hydrolyzes GDP, IDP, and UDP, with slower hydrolysis of CDP, ITP, GTP, CTP, ADP, and UTP, and virtually no hydrolysis of ATP .
The enzyme contains four apyrase-conserved regions, which are characteristic of NTPases. These regions are essential for its catalytic activity. The membrane-bound form of ENTPD6 might support glycosylation reactions in the Golgi apparatus and, when released from cells, might catalyze the hydrolysis of extracellular nucleotides .
ENTPD6 plays a pivotal role in purinergic signaling by regulating the availability of extracellular nucleotides at purinergic P2 receptors. This regulation is crucial for various cellular functions, including signal transmission, inflammation control, and tissue repair .
The enzyme also generates extracellular nucleosides for cellular reuptake and salvage via nucleoside transporters of the plasma membrane. The extracellular adenosine formed acts as an agonist of purinergic P1 receptors. Additionally, ENTPD6 can produce and hydrolyze extracellular inorganic pyrophosphate, which is significant in controlling bone mineralization .
Recombinant mouse ENTPD6 is widely used in research to study its biochemical properties, physiological roles, and potential therapeutic applications. The recombinant form allows for detailed investigations into the enzyme’s structure, function, and interactions with other molecules.