AKR1E2 antibodies are developed using recombinant proteins or synthetic peptides corresponding to specific regions of the AKR1E2 protein. Key features include:
AKR1E2 antibodies undergo rigorous validation to ensure specificity and reproducibility:
Western Blot: Detects endogenous AKR1E2 at ~37 kDa in human cell lysates .
Immunohistochemistry: Validated in human tissue samples, showing cytoplasmic localization in hepatic and breast cancer cells .
Epitope Mapping: Antibodies targeting AA 291–320 (C-terminal) show high specificity due to low homology with other AKR family members .
A rabbit polyclonal antibody (HPA037822) demonstrated strong reactivity in IHC (1:1,000 dilution) and WB (0.04–0.4 µg/mL) .
Mouse monoclonal antibodies (e.g., ABIN529454) exhibit no cross-reactivity with AKR1C1, AKR1C2, or AKR1C3 isoforms .
AKR1E2 is implicated in drug resistance and tumor progression. In tamoxifen-resistant breast cancer, elevated AKR1E2 expression correlates with poor therapeutic outcomes, potentially due to its role in steroid metabolism .
AKR1E2 participates in detoxification by reducing reactive aldehydes and ketones, leveraging NADPH as a cofactor . This activity is critical in liver and kidney tissues, where AKR1E2 mitigates oxidative stress .
AKR1E2 (Aldo-Keto Reductase Family 1, Member E2) is a cytoplasmic enzyme that functions primarily as a 1,5-anhydro-D-fructose reductase. The protein catalyzes the NADPH-dependent reduction of 1,5-anhydro-D-fructose (AF) to 1,5-anhydro-D-glucitol. Additionally, AKR1E2 demonstrates low NADPH-dependent reductase activity toward 9,10-phenanthrenequinone in vitro, suggesting potential roles in detoxification pathways . This enzyme belongs to the wider aldo/keto reductase superfamily, sharing structural and functional characteristics with other AKR proteins involved in various metabolic processes . Its highly tissue-specific expression pattern suggests specialized functions in testicular biology that warrant further investigation through antibody-based research techniques.
AKR1E2 demonstrates a highly tissue-specific expression pattern, being predominantly expressed in the testis according to multiple independent studies . More specifically, the protein is localized within testicular germ cells and testis interstitial cells . This restricted expression profile suggests AKR1E2 may play specialized roles in male reproductive physiology or spermatogenesis. When designing experiments to study AKR1E2, researchers should select appropriate positive controls (testicular tissue) and negative controls (non-testicular tissues) to validate antibody specificity. The cytoplasmic localization of AKR1E2 further informs appropriate cellular fractionation techniques when preparing samples for antibody-based detection methods.
AKR1E2 has a calculated molecular weight of 36,589 Da , which researchers should consider when validating antibody specificity by Western blot. The protein is also known by several alternative names including AKR1CL2, AKRDC1, HTSP1, LoopADR, TAKR, and hTSP . When selecting antibodies, researchers should verify which protein epitopes are targeted, as some commercially available antibodies are developed against the C-terminal region (amino acids 291-320) while others may target different regions. Understanding the protein's tertiary structure and post-translational modifications is essential for selecting antibodies that target accessible epitopes in native versus denatured conditions.
When selecting an AKR1E2 antibody, researchers should consider:
For Western blot applications, polyclonal antibodies targeting the C-terminal region (amino acids 291-320) of AKR1E2 have been validated . These antibodies are purified through protein A columns followed by peptide affinity purification to ensure specificity . For immunohistochemistry, researchers should consider antibodies validated specifically for fixed tissue samples and optimize protocols for testicular tissue, where AKR1E2 is predominantly expressed .
A comprehensive validation strategy for AKR1E2 antibodies should include:
Positive and negative tissue controls: Use testicular tissue (positive) versus non-testicular tissues (negative) based on the known expression profile .
Peptide competition assay: Pre-incubate the antibody with the immunizing peptide before application to verify signal extinction.
Molecular weight verification: Confirm detection of a band at approximately 36.6 kDa in Western blot applications .
Knockout/knockdown controls: If available, use AKR1E2 knockout models or siRNA-mediated knockdown samples as negative controls.
Multiple antibody comparison: Use antibodies targeting different epitopes of AKR1E2 to corroborate findings.
For IHC applications, include peptide blocking controls and compare staining patterns with literature reports describing testicular germ cells and interstitial cell localization . For ELISA applications, establish a standard curve using recombinant AKR1E2 protein and verify detection sensitivity within the expected range (5.0-100 ng/mL) .
To maintain optimal antibody performance, store AKR1E2 antibodies according to manufacturer recommendations. Generally, antibodies should be stored at -20°C for long-term storage, with small aliquots to prevent freeze-thaw cycles that can degrade antibody quality . For short-term use (up to one month), storage at 4°C is acceptable . Most commercial AKR1E2 antibodies are supplied in PBS containing preservatives such as 0.09% sodium azide or 50% glycerol with 0.5% BSA . When working with these antibodies, researchers should document lot numbers and maintain consistent storage conditions throughout a research project to minimize variability. Additionally, avoid exposing antibodies to direct light and minimize exposure to room temperature, as these conditions can accelerate protein degradation.
For optimal Western blot detection of AKR1E2:
Sample preparation:
Extract proteins from testicular tissue or cells using RIPA buffer supplemented with protease inhibitors
Determine protein concentration using BCA or Bradford assay
Load 20-50 μg total protein per lane
Electrophoresis conditions:
Use 10-12% SDS-PAGE gels
Include molecular weight markers spanning 25-50 kDa range
Transfer and blocking:
Transfer to PVDF membrane at 100V for 60-90 minutes
Block with 5% non-fat milk in TBST for 1 hour at room temperature
Antibody incubation:
Detection:
For validation purposes, include positive control (testis lysate) and negative control (non-testicular tissue) samples . If multiple bands appear, consider using peptide competition assays to verify specificity.
For immunohistochemical detection of AKR1E2 in testicular tissues:
Tissue preparation:
Fix tissues in 10% neutral-buffered formalin for 24 hours
Process and embed in paraffin
Section at 4-5 μm thickness
Antigen retrieval:
Perform heat-induced epitope retrieval using citrate buffer (pH 6.0)
Heat in pressure cooker or microwave for 15-20 minutes
Cool sections to room temperature (approximately 20 minutes)
Blocking and antibody incubation:
Detection and visualization:
Apply HRP-conjugated secondary antibody for 1 hour at room temperature
Develop with DAB substrate
Counterstain with hematoxylin
Dehydrate, clear, and mount
Always include positive control (normal testis) and negative controls (primary antibody omission and non-testicular tissue) . Optimize antibody concentration specifically for your tissue samples and fixation conditions. Focus analysis on cytoplasmic staining in testicular germ cells and interstitial cells, consistent with the known localization of AKR1E2 .
When implementing ELISA for AKR1E2 quantification:
ELISA format selection:
Sample preparation:
Standard curve preparation:
Antibody dilution:
Controls and validation:
Include blank wells (no sample)
Run samples in duplicate or triplicate
Include positive control (testis extract) and negative control (non-testicular tissue)
For quantitative analysis, ensure all equipment is properly calibrated, particularly the microplate reader measuring absorbance at 450 nm . Analytical software should incorporate a 4- or 5-parameter logistic curve fit for optimal quantification accuracy.
For investigating AKR1E2 protein interactions:
Co-immunoprecipitation (Co-IP):
Prepare testicular cell or tissue lysates in non-denaturing buffer
Pre-clear lysate with Protein A/G beads for 1 hour
Incubate cleared lysate with AKR1E2 antibody overnight at 4°C
Add Protein A/G beads and incubate for 2-4 hours
Wash beads 4-5 times with wash buffer
Elute bound proteins and analyze by Western blot using antibodies against suspected interacting partners
Proximity Ligation Assay (PLA):
Fix cells or tissue sections and permeabilize
Block non-specific binding sites
Incubate with AKR1E2 antibody and antibody against putative interacting protein
Follow manufacturer's protocol for PLA probes and detection
Analyze fluorescent signals indicating proteins in close proximity (<40 nm)
Pull-down assays:
Express and purify recombinant AKR1E2 with appropriate tag
Immobilize on matrix or beads
Incubate with cellular lysates
Wash, elute, and identify binding partners by mass spectrometry
Focus investigations on potential interactions with NADPH-dependent enzymes or proteins involved in testicular metabolism, considering AKR1E2's enzymatic function and tissue-specific expression . Use appropriate negative controls including IgG controls and samples from tissues that do not express AKR1E2.
To study AKR1E2 enzymatic activity alongside antibody detection:
Activity assays following immunoprecipitation:
Immunoprecipitate AKR1E2 from testicular lysates using validated antibodies
Measure NADPH-dependent reduction of 1,5-anhydro-D-fructose to 1,5-anhydro-D-glucitol
Monitor NADPH consumption by spectrophotometric methods (decrease in absorbance at 340 nm)
Correlate activity with protein levels determined by Western blot
In situ activity assays combined with immunohistochemistry:
Perform enzymatic activity assay on tissue sections
Follow with immunohistochemical detection of AKR1E2
Compare patterns of activity with protein localization
Structure-function analysis:
Generate AKR1E2 mutants affecting catalytic sites
Express in appropriate cell systems
Correlate antibody detection of protein levels with enzymatic activity
Use to identify critical residues for catalysis
Inhibitor studies:
Test effects of potential inhibitors on AKR1E2 activity
Verify target engagement using antibody-based methods
Correlate inhibition with structural interactions
When designing these experiments, researchers should consider the optimal buffer conditions for AKR1E2 activity, including pH, temperature, and cofactor concentrations. Control experiments should include enzyme kinetics characterization with recombinant AKR1E2 protein to establish baseline parameters.
For developmental studies of AKR1E2 in testicular biology:
Temporal expression analysis:
Spatial expression patterns:
Cell-type specific expression:
Isolate specific testicular cell populations using flow cytometry or magnetic separation
Verify AKR1E2 expression by Western blot and RT-PCR
Correlate protein levels with cell differentiation state
Functional studies during development:
Design loss-of-function or gain-of-function experiments
Monitor effects on spermatogenesis and testicular development
Use AKR1E2 antibodies to confirm experimental manipulation effectiveness
These approaches should incorporate appropriate controls, including tissues where AKR1E2 is not expressed . Experimental design should account for the potential influence of hormonal regulation on AKR1E2 expression during developmental transitions, particularly during puberty and sexual maturation.
When troubleshooting unexpected Western blot banding patterns:
| Issue | Possible Causes | Corrective Actions |
|---|---|---|
| Multiple bands | Protein isoforms or alternative splicing | Verify against transcript data; use different antibodies targeting distinct epitopes |
| Post-translational modifications | Treat samples with phosphatases or deglycosylation enzymes | |
| Protein degradation | Add fresh protease inhibitors; reduce sample handling time | |
| No signal | Low expression level | Increase protein loading; use more sensitive detection method |
| Epitope masking | Try different antibody targeting different epitope; optimize antigen retrieval | |
| Higher MW than expected | Post-translational modifications | Verify with deglycosylation treatment |
| Protein complexes resistant to denaturation | Increase SDS concentration; add reducing agents |
For AKR1E2 specifically, validate all bands by peptide competition assay using the synthetic peptide corresponding to amino acids 291-320 . Consider that AKR1E2 may interact with other proteins in the aldo-keto reductase family, potentially leading to cross-reactivity. Always compare your results with positive controls (testis lysate) where the expected 36.6 kDa band should be visible .
When facing inconsistencies between different detection methods:
Western blot vs. IHC discrepancies:
Western blot detects denatured proteins while IHC detects native conformations
Epitope accessibility may differ between methods
Verify antibody performance in each application separately
Consider using multiple antibodies targeting different epitopes
ELISA vs. Western blot inconsistencies:
General approach to resolving inconsistencies:
Systematically evaluate each method's controls
Verify antibody batch consistency and storage conditions
Consider third method validation (e.g., mass spectrometry)
Evaluate sample preparation effects on protein stability and detection
Document all methodological details, including antibody dilutions (1:500-1:2000 for WB; 1:100-1:300 for IHC; 1:10,000 for ELISA) and detection conditions to facilitate troubleshooting. Remember that different detection methods may reveal different aspects of AKR1E2 biology, potentially reflecting biologically relevant differences rather than methodological errors.
For rigorous analysis of AKR1E2 tissue-specific expression:
Essential tissue controls:
Antibody validation controls:
Peptide competition: Pre-incubate antibody with immunizing peptide
Isotype control: Use matched IgG at same concentration
Antibody omission: Perform protocol without primary antibody
Method-specific controls:
Biological validation:
When interpreting results, researchers should acknowledge that very low expression levels might be detected by sensitive methods like ELISA (detection limit 5.0 ng/mL) but not by less sensitive techniques. Additionally, post-translational modifications may affect epitope recognition differently across tissues, potentially leading to false negatives. Using antibodies targeting different regions of AKR1E2 can help address this limitation.
For investigating AKR1E2's role in reproductive physiology:
Functional genomics approaches:
Animal model studies:
Metabolomic analysis:
Clinical correlations:
Analyze AKR1E2 expression in testicular biopsies from patients with:
Male infertility
Testicular cancer
Developmental disorders
Correlate expression with clinical parameters and outcomes
These experimental approaches should incorporate appropriate controls and validation using antibodies with confirmed specificity for AKR1E2. Researchers should consider the cytoplasmic localization of AKR1E2 when interpreting results and designing functional studies.
To investigate post-translational modifications (PTMs) of AKR1E2:
Phosphorylation analysis:
Immunoprecipitate AKR1E2 using specific antibodies
Analyze by Western blot using phospho-specific antibodies
Alternatively, use mass spectrometry to identify phosphorylation sites
Validate functional significance with phosphatase treatments
Glycosylation studies:
Treat samples with deglycosylation enzymes (PNGase F, O-glycosidase)
Compare molecular weight shifts by Western blot
Use lectins to probe for specific glycan structures
Correlate glycosylation status with enzymatic activity
Ubiquitination and SUMOylation:
Immunoprecipitate AKR1E2 under denaturing conditions
Probe with anti-ubiquitin or anti-SUMO antibodies
Use proteasome inhibitors to enhance detection
Correlate modifications with protein stability and turnover
PTM site mapping:
Generate recombinant AKR1E2 mutants at predicted modification sites
Express in appropriate cell systems
Compare PTM patterns and functional consequences
Validate with site-specific antibodies if available
These approaches should include appropriate controls and consider the tissue-specific expression of AKR1E2 in testis . Mass spectrometry analyses should target the 36.6 kDa region and adjacent molecular weights to capture modified forms. Researchers should correlate PTMs with AKR1E2's NADPH-dependent reductase activity to establish functional significance.
For multiplexed detection of AKR1E2 in testicular samples:
Multiplex immunofluorescence:
Combine AKR1E2 antibodies with markers for:
Cell type-specific markers (VASA for germ cells, SOX9 for Sertoli cells)
Developmental stage markers (SYCP3 for meiotic cells)
Functional pathway components (NADPH-producing enzymes)
Use spectrally distinct fluorophores and multispectral imaging
Perform quantitative colocalization analysis
Mass cytometry (CyTOF):
Label AKR1E2 antibodies with rare earth metals
Combine with multiple markers for cellular phenotyping
Analyze testicular cell suspensions at single-cell resolution
Perform high-dimensional data analysis to identify cell populations
Single-cell analysis workflows:
Sort testicular cells based on surface markers
Analyze AKR1E2 expression in specific populations by Western blot
Correlate with single-cell transcriptomics data
Identify regulatory relationships
Spatial transcriptomics with protein detection:
Perform in situ hybridization for AKR1E2 mRNA
Follow with immunodetection of AKR1E2 protein
Analyze spatial relationships with other markers
Correlate mRNA and protein expression patterns
These multiplexed approaches should include appropriate controls for antibody specificity, including peptide competition controls and validation in tissues known to lack AKR1E2 expression. Consider the detection range (5.0-100 ng/mL) and sensitivity (1.0 ng/mL) when designing experiments to ensure signals remain within quantifiable ranges.
For applying AKR1E2 as a potential biomarker:
Tissue microarray analysis:
Liquid biopsy development:
Multimarker panel development:
Combine AKR1E2 detection with established testicular markers
Perform multivariate analysis to improve diagnostic accuracy
Validate in independent cohorts
Determine sensitivity and specificity for specific conditions
Longitudinal monitoring protocols:
Establish baseline AKR1E2 levels in healthy individuals
Monitor changes during disease progression or treatment
Correlate with clinical response
Develop standardized testing protocols
These approaches should incorporate appropriate quality control measures, including standard curves using recombinant AKR1E2 protein and spike-recovery experiments to validate detection in complex biological matrices. Given AKR1E2's testis-specific expression , researchers should evaluate its potential as a tissue-specific marker for testicular damage or dysfunction.
To investigate AKR1E2's role in testicular metabolic pathways:
Metabolic substrate profiling:
Purify AKR1E2 using immunoprecipitation with specific antibodies
Screen against panel of potential substrates beyond known targets
Monitor NADPH consumption spectrophotometrically
Identify novel substrates relevant to testicular metabolism
Metabolomic analysis in AKR1E2-manipulated systems:
Pathway intersection analysis:
Use AKR1E2 antibodies for co-immunoprecipitation
Identify interacting proteins by mass spectrometry
Map interactions to known metabolic pathways
Validate functional relationships with enzymatic assays
Flux analysis:
Use isotope-labeled substrates in testicular cells or tissues
Track metabolite conversions in presence/absence of AKR1E2
Correlate with protein expression levels
Define rate-limiting steps in relevant pathways
These approaches should include appropriate controls and consider AKR1E2's NADPH-dependent reductase activity . Researchers should validate findings across multiple experimental systems and correlate with the known cytoplasmic localization of AKR1E2 in testicular cells.
To address antibody performance variability in cross-study comparisons:
Standardization strategies:
Develop standard reference materials (recombinant AKR1E2)
Establish calibration curves across detection methods
Use consistent positive controls (testicular tissue lysates)
Report antibody catalog numbers, lot numbers, and validation data
Cross-validation protocols:
Test multiple antibodies on identical samples
Compare detection limits and dynamic ranges
Establish conversion factors between different antibodies
Document epitope differences between antibodies
Meta-analysis approach:
Standardize data reporting formats
Apply normalization methods to account for antibody differences
Use relative rather than absolute quantification when comparing studies
Weight studies by quality of antibody validation
Reporting guidelines:
Researchers should establish internal reference standards when comparing historical data and consider generating bridging datasets when transitioning between antibody reagents. When possible, validation with orthogonal methods such as mass spectrometry can provide antibody-independent confirmation of findings.
Emerging technologies for AKR1E2 research include:
Nanobody and recombinant antibody development:
Generate single-domain antibodies against AKR1E2
Engineer antibodies with enhanced specificity for distinct epitopes
Develop antibody fragments maintaining specificity but with improved tissue penetration
Create genetically encoded intrabodies for live-cell imaging
Advanced imaging technologies:
Super-resolution microscopy of AKR1E2 localization
Live-cell imaging using fluorescent AKR1E2 antibody fragments
Correlative light and electron microscopy for ultrastructural localization
Expansion microscopy for enhanced spatial resolution
Microfluidic and single-cell applications:
Develop microfluidic antibody-based capture systems
Combine with single-cell transcriptomics
Create droplet-based enzymatic activity assays
Enable high-throughput screening of AKR1E2 modulators
Synthetic biology approaches:
Design split-protein systems based on AKR1E2 interactions
Create biosensors for AKR1E2 substrates and products
Develop optogenetic tools to control AKR1E2 activity
Engineer cellular reporters for AKR1E2 expression and function
These technologies should build upon current knowledge of AKR1E2's structure, tissue distribution, and enzymatic function . Researchers should validate new approaches against established methods using well-characterized antibodies with documented specificity and performance characteristics.
For integrated multi-omics investigation of AKR1E2:
Proteogenomic integration:
Correlate AKR1E2 protein levels (antibody-based detection) with:
Genomic variants affecting expression or function
Transcriptomic data from various testicular cell types
Epigenetic modifications affecting gene regulation
Identify regulatory mechanisms controlling tissue-specific expression
Structural biology with functional validation:
Determine AKR1E2 crystal structure
Map epitopes recognized by various antibodies
Correlate structural features with enzymatic function
Design structure-based inhibitors and activators
Systems biology modeling:
Integrate AKR1E2 into testicular metabolic network models
Predict metabolic flux changes under various conditions
Validate predictions using antibody-based quantification
Identify emergent properties from network analysis
Spatially resolved multi-omics:
Combine immunodetection with spatial transcriptomics
Map AKR1E2 protein distribution relative to substrate availability
Correlate with local metabolite concentrations
Develop computational models of spatial regulation