The DHRS2 antibody is a polyclonal rabbit antibody designed to target the DHRS2 (dehydrogenase/reductase member 2) protein, a member of the short-chain dehydrogenase/reductase (SDR) superfamily. DHRS2 plays critical roles in lipid metabolism, redox regulation, and tumor suppression, with its expression often downregulated in cancers such as ovarian and esophageal squamous cell carcinoma (ESCC) . The antibody is primarily used in research settings for protein detection via Western blotting (WB) and immunofluorescence (IF), enabling investigations into DHRS2’s cellular localization and functional mechanisms.
Tumor Suppression: Studies using DHRS2 antibodies demonstrated its tumor-suppressive role in ovarian cancer (OC) and ESCC. DHRS2 overexpression inhibits cell growth and metastasis by disrupting choline metabolism via downregulation of choline kinase α (CHKα) .
HDACi Resistance: In ovarian cancer cells, DHRS2 knockdown correlates with resistance to histone deacetylase inhibitors (HDACi), as shown by reduced apoptosis and G2/M arrest in treated cells .
Post-Transcriptional Regulation: The antibody was used to confirm DHRS2’s post-transcriptional degradation of CHKα mRNA, linking it to AKT signaling inhibition and reduced phosphorylcholine (PC)/glycerophosphorylcholine (GPC) ratios in OC .
Redox and Signaling Pathways: DHRS2 stabilizes p53, reduces reactive oxygen species (ROS), and inhibits p38 MAPK/MMP2 pathways, as validated by immunoblotting in ESCC models .
Western Blotting: Recommended dilutions range from 1:250–1:500, with observed bands at 27–32 kDa .
Immunofluorescence: Mitochondrial localization of DHRS2 is evident at 1:500 dilution (e.g., HepG2 cells) .
Cross-Reactivity: Proteintech’s antibody (15735-1-AP) exhibits cross-reactivity with mouse and rat samples, expanding its utility in preclinical models .
DHRS2 is an NADPH-dependent oxidoreductase that belongs to the short-chain dehydrogenase/reductase (SDR) family. It primarily functions as a dicarbonyl reductase, catalyzing the reduction of various dicarbonyl compounds including 3,4-hexanedione, 2,3-heptanedione, and 1-phenyl-1,2-propanedione . At the cellular level, DHRS2 has several important functions:
Acts as an enzymatic inactivator of reactive carbonyls involved in the covalent modification of cellular components
Displays minor hydroxysteroid dehydrogenase activity toward bile acids such as ursodeoxycholic acid (UDCA)
Attenuates MDM2-mediated p53/TP53 degradation, leading to p53 stabilization
Reduces proliferation, migration, and invasion of cancer cells
Decreases the production of reactive oxygen species (ROS) in cancer cells
DHRS2 is primarily localized in the mitochondrion matrix, though a minor fraction can be translocated to the nucleus after cleavage of the targeting signal .
DHRS2 antibodies are versatile research tools employed in multiple experimental methodologies:
Researchers should select antibodies based on their specific application requirements and validate them in their experimental system .
For maintaining optimal antibody activity, adhere to these storage and handling guidelines:
Store at -20°C in aliquots to minimize freeze-thaw cycles
Most commercial DHRS2 antibodies are supplied in buffer containing 50% glycerol and stabilizers
Avoid repeated freeze-thaw cycles as they can degrade antibody quality
When working with the antibody, keep it on ice or at 4°C
For long-term storage (>12 months), some manufacturers recommend -80°C
Before immunofluorescence applications, centrifuge antibody vials briefly to collect solution at the bottom
Many DHRS2 antibodies remain stable for 12 months when stored properly at -20°C . When shipping is required, antibodies should be transported with ice packs and immediately stored at the recommended temperature upon receipt .
DHRS2 antibodies vary significantly in their properties and applications:
When selecting an antibody, researchers should consider the specific epitope recognized, as this affects detection efficiency of specific DHRS2 isoforms or post-translationally modified forms .
Comprehensive validation of DHRS2 antibodies requires a multi-faceted approach:
Positive and negative control samples:
Molecular weight verification:
Cross-validation with multiple antibodies:
Use antibodies recognizing different epitopes of DHRS2 (e.g., N-terminal vs. C-terminal)
Compare results from polyclonal and monoclonal antibodies
Subcellular localization confirmation:
Immunoprecipitation followed by mass spectrometry:
For ultimate validation, perform IP with the DHRS2 antibody followed by MS identification
For immunofluorescence validation, researchers should observe DHRS2 protein specifically at mitochondria, as confirmed in HepG2 cells with GTX123431 antibody .
Cancer research involving DHRS2 antibodies requires specific methodological considerations:
Cell line selection:
Functional assays:
Mechanistic studies:
Clinical correlation:
When designing experiments, remember that DHRS2 has been implicated as both a tumor suppressor and oncogene depending on cancer type, underscoring the importance of context-specific validation .
When troubleshooting Western blotting with DHRS2 antibodies, consider these specific issues:
Unexpected band sizes:
Multiple bands:
May indicate splice variants or post-translational modifications
Can result from cross-reactivity with other SDR family members
Solution: Use antibodies targeting different epitopes to confirm specificity
Weak or no signal:
Optimization guidance table:
Background issues:
Increase washing frequency and duration
Optimize blocking conditions (try 5% BSA instead of milk)
Decrease antibody concentration if using too much
Cell-specific considerations:
When possible, include both positive controls (cells with known DHRS2 expression) and negative controls (DHRS2 knockdown cells) in troubleshooting experiments .
DHRS2 has emerged as a significant factor in chemoresistance, particularly in colorectal cancer:
DHRS2 upregulation in chemoresistant cells:
DHRS2-mediated resistance mechanisms:
Experimental approaches using antibodies:
Knockdown experiments: Transfect cells with DHRS2 siRNA and measure chemosensitivity
Protein interaction studies: Use co-immunoprecipitation with DHRS2 antibodies to detect interactions with MDM2
EMT marker analysis: Use Western blotting to detect changes in E-cadherin expression
Signaling pathway analysis: Examine p53 and ERCC1 expression levels
Research findings on DHRS2 and chemoresistance:
Researchers can use DHRS2 antibodies to identify potential therapeutic targets for simultaneously addressing cancer metastasis and chemoresistance .
DHRS2 antibodies have proven valuable in studying RNA-protein interactions through RNA immunoprecipitation (RIP) assays:
RIP assay protocol for DHRS2:
Applications in cancer biology:
RNA stability assays in conjunction with DHRS2 antibodies:
Technical considerations:
Antibody selection is crucial; use antibodies validated for immunoprecipitation
Include appropriate controls (IgG, input samples)
Consider crosslinking to stabilize transient RNA-protein interactions
Verify DHRS2 pull-down efficiency by Western blotting before RNA analysis
These approaches have been instrumental in uncovering DHRS2's role in regulating mRNA stability and gene expression, expanding our understanding beyond its canonical enzymatic functions .
DHRS2 demonstrates tumor suppressor activity in multiple cancer types through several mechanisms:
Inhibition of cell proliferation:
Inhibition of invasion and metastasis:
Regulation of p53 pathway:
Research approaches using antibodies:
In vivo evidence:
DHRS2 antibodies provide essential tools for investigating these tumor suppressor mechanisms across different cancer types, enabling both expression analysis and mechanistic studies .
Immunofluorescence with DHRS2 antibodies requires specific technical considerations:
Sample preparation protocol:
Antibody dilution optimization:
Mitochondrial localization visualization:
Signal detection and image acquisition:
Common pitfalls and solutions:
| Problem | Potential Cause | Solution |
|---|---|---|
| No signal | Insufficient permeabilization | Optimize detergent concentration and incubation time |
| High background | Excessive antibody concentration | Increase dilution factor of primary antibody |
| Non-specific staining | Cross-reactivity | Use antibodies validated for IF applications |
| Weak mitochondrial signal | Fixation affecting epitope | Try alternative fixation methods (methanol, acetone) |
When performing IF with DHRS2 antibodies, it's essential to verify subcellular localization patterns consistent with expected mitochondrial and occasional nuclear distribution .
Designing effective genetic manipulation experiments for DHRS2 requires careful planning and validation:
DHRS2 knockdown strategies:
DHRS2 overexpression strategies:
Validation using antibodies:
Functional assays following manipulation:
Experimental design considerations:
| Experiment Type | Control Selection | Antibody Application | Expected Outcome |
|---|---|---|---|
| siRNA knockdown | Non-targeting siRNA | WB confirmation of knockdown | Enhanced growth/invasion in cancer cells |
| shRNA stable knockdown | Non-targeting shRNA | WB and IF for expression/localization | Increased tumorigenicity in vivo |
| Overexpression | Empty vector | WB confirmation of expression | Reduced proliferation and invasion |
| In vivo xenografts | Vector control cells | IHC of tumor sections | Delayed tumor formation, reduced size |
For rigorous validation, researchers should confirm protein-level changes using antibodies targeting different epitopes of DHRS2 to ensure specificity of manipulation .
DHRS2 has a complex relationship with the p53 pathway that can be investigated using antibody-based techniques:
DHRS2-MDM2-p53 interaction mechanism:
Experimental approaches using antibodies:
DHRS2 in p53-dependent chemoresistance:
Context-dependent regulation:
Antibody panel for p53 pathway investigation:
| Target Protein | Purpose in DHRS2-p53 Studies | Recommended Application |
|---|---|---|
| DHRS2 | Confirm expression manipulation | WB, IF, IHC |
| p53 | Monitor stabilization levels | WB, IP, IF |
| MDM2 | Detect interaction with DHRS2 | Co-IP, WB |
| p21 | Assess downstream p53 activity | WB, IF |
| ERCC1 | Examine DNA repair pathway regulation | WB |
This relationship between DHRS2 and p53 explains how DHRS2 can function as both a tumor suppressor and a mediator of chemoresistance in different contexts .
Proper control design is critical for reliable DHRS2 antibody experiments:
Positive controls for DHRS2 detection:
Cell lines with confirmed DHRS2 expression:
Recombinant DHRS2 protein: For antibody validation and band size confirmation
Negative controls for specificity:
Application-specific controls:
| Application | Essential Controls | Purpose |
|---|---|---|
| Western Blotting | Loading control (β-actin, GAPDH) | Normalize protein loading |
| Molecular weight marker | Confirm band size (expected ~30 kDa) | |
| Immunofluorescence | Secondary antibody only | Detect non-specific binding |
| DHRS2 knockdown cells | Validate signal specificity | |
| Mitochondrial co-staining | Confirm subcellular localization | |
| Immunoprecipitation | IgG control | Identify non-specific binding |
| Input sample | Confirm presence in starting material | |
| RIP Assay | IgG antibody (1:20) | Control for non-specific RNA binding |
Validation across multiple methods:
Implementing these comprehensive controls ensures reliable interpretation of DHRS2 antibody experimental results and helps distinguish true signals from technical artifacts .
DHRS2 localizes to multiple cellular compartments, requiring specific detection approaches:
Mitochondrial DHRS2 detection:
Primary localization: DHRS2 is predominantly found in the mitochondrion matrix
Immunofluorescence protocol:
Nuclear DHRS2 detection:
Secondary localization: A minor fraction translocates to the nucleus after cleavage of targeting signal
Detection considerations:
Cell type-specific considerations:
Technical optimization for subcellular detection:
Fixation: Optimize to preserve both mitochondrial and nuclear structure
Antibody concentration: May require different dilutions for different compartments
Image acquisition: Use confocal microscopy for precise localization
Signal amplification: Consider tyramide signal amplification for low abundance detection
Understanding DHRS2's dual localization is crucial for experimental design and interpretation, particularly when studying its non-enzymatic functions in the nucleus versus its enzymatic roles in mitochondria .
Quantitative analysis of DHRS2 expression in cancer tissues requires standardized approaches:
Immunohistochemistry (IHC) protocol optimization:
Scoring systems for DHRS2 expression:
Semi-quantitative scoring:
Staining intensity: 0 (negative), 1 (weak), 2 (moderate), 3 (strong)
Percentage of positive cells: 0-100%
H-score calculation: Intensity × percentage (range: 0-300)
Digital image analysis:
Use software to quantify optical density or immunoreactive area
Enables more objective and reproducible quantification
Comparative analysis frameworks:
| Tissue Analysis Approach | Methodology | Quantification Method | Advantages |
|---|---|---|---|
| Tumor vs. normal tissue | Paired sample analysis | Direct comparison of H-scores | Controls for patient variability |
| TMA analysis | Multiple patient samples | Statistical comparison across groups | High throughput, reduced variability |
| Correlation with clinical outcomes | Survival analysis | Kaplan-Meier with optimal cutoffs | Links expression to prognosis |
| Subcellular distribution analysis | High-resolution imaging | Ratio of nuclear/mitochondrial staining | Insight into functional state |
Validation and quality control:
Clinical correlation considerations:
DHRS2 expression varies by cancer type with both tumor-suppressive and oncogenic roles reported
Consider chromosome 14q11.2 status, as this region shows high-frequency loss of heterozygosity
Correlate with markers of p53 pathway activation (MDM2, p21)
For chemoresistance studies, correlate with ERCC1 expression
These approaches enable robust quantitative analysis of DHRS2 expression in cancer tissues, facilitating comparisons across patients and correlation with clinical outcomes .
Several innovative applications of DHRS2 antibodies are advancing cancer research:
Biomarker development for chemoresistance prediction:
Therapeutic target validation:
RNA-protein interaction studies:
Single-cell analysis applications:
Adaptation of DHRS2 antibodies for mass cytometry (CyTOF)
Flow cytometry applications for heterogeneity assessment
Correlation with other cancer markers at single-cell resolution
Pathway mapping applications:
| Pathway | DHRS2 Antibody Application | Cancer Relevance |
|---|---|---|
| p53-MDM2 axis | Co-IP to detect interactions | Tumor suppression mechanisms |
| EMT pathway | Monitor E-cadherin changes after DHRS2 manipulation | Metastasis and invasion |
| DNA repair | Correlate with ERCC1 expression | Chemoresistance mechanisms |
| Cell cycle regulation | Assess cyclin D1 levels in response to DHRS2 | Growth inhibition mechanisms |
Combination therapy response prediction:
Using DHRS2 antibodies to stratify patients for clinical trials
Monitoring DHRS2 expression changes during treatment
Correlating with treatment outcomes in patient-derived xenograft models
These emerging applications highlight the versatility of DHRS2 antibodies beyond traditional detection methods, positioning them as valuable tools in translational cancer research .