AKR4C9 Antibody

Shipped with Ice Packs
In Stock

Description

AKR Enzyme Family Context

The aldo-keto reductase (AKR) superfamily comprises over 190 enzymes across species, categorized into 16 families (AKR1–AKR16) with distinct substrate specificities . Human AKRs include:

AKR FamilyKey MembersFunctions
AKR1AKR1C1–AKR1C4, AKR1D1Steroid metabolism, prostaglandin synthesis
AKR7AKR7A2/AKR7A3Detoxification of aflatoxins

No AKR4 family members (AKR4A–AKR4C) are documented in humans. AKR4 enzymes are primarily plant-specific (e.g., AKR4C8 in Arabidopsis), involved in stress response pathways .

Potential Misidentification or Typographical Errors

The term "AKR4C9" may derive from:

  • AKR1C9: A rat-specific 3α-hydroxysteroid dehydrogenase , but no human ortholog exists.

  • AKR1C1–AKR1C4: Human enzymes with validated antibodies (e.g., ab192785 for AKR1C1 , ab209899 for AKR1C3/4 ).

Research Implications

  • Gene Naming Discrepancies: AKR nomenclature follows strict guidelines (e.g., AKR1C3 = aldo-keto reductase family 1 member C3) . "AKR4C9" lacks annotation in UniProt, NCBI Gene, or BRENDA databases.

  • Experimental Validation Gaps: Antibody development requires epitope specificity confirmed by knockout/knockdown models , which are unavailable for AKR4C9.

Recommendations for Further Inquiry

  1. Verify gene/protein nomenclature using databases like:

  2. Explore orthologs in non-human species if studying AKR4C9 in model organisms.

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (Made-to-order)
Synonyms
AKR4C9 antibody; At2g37770 antibody; F13M22 antibody; T8P21.32NADPH-dependent aldo-keto reductase antibody; chloroplastic antibody; AtChlAKR antibody; EC 1.1.1.- antibody; Aldo-keto reductase family 4 member C9 antibody
Target Names
AKR4C9
Uniprot No.

Target Background

Function
This antibody targets an oxidoreductase enzyme with broad substrate specificity. It reduces a wide range of compounds, including ketosteroids, aromatic aldehydes, ketones, sugars, and other aliphatic aldehydes. Conversely, it oxidizes hydroxysteroids. This enzyme functions as an aldehyde reductase, catalyzing the reduction of aldehyde carbonyl groups in both saturated and α,β-unsaturated aldehydes. However, it exhibits no activity on α,β-unsaturated ketones. Confirmed substrates include propionaldehyde, butyraldehyde, methylglyoxal, (E)-2-pentenal, (E)-2-hexenal, (Z)-3-hexenal, and (E)-2-nonenal. In contrast, it does not utilize propenal (acrolein), crotonaldehyde, 2-butanone, 3-buten-2-one, or 1-penten-3-one. The enzyme's proposed function is detoxification; it reduces various toxic aldehydes and ketones generated during cellular stress.
Gene References Into Functions
Studies have implicated aldo-keto reductase (At2g37770) and aldehyde reductases (At1g54870 and At3g04000) in the reduction of aldehyde groups in saturated aldehydes and methylglyoxal, as well as α,β-unsaturated aldehydes within chloroplasts. [PMID: 21169366](https://www.ncbi.nlm.nih.gov/pubmed/21169366)
Database Links

KEGG: ath:AT2G37770

STRING: 3702.AT2G37770.2

UniGene: At.12803

Protein Families
Aldo/keto reductase family
Subcellular Location
Plastid, chloroplast.

Q&A

What is AKR4C9 and how does it relate to the broader aldo-keto reductase family?

AKR4C9 (Aldo-Keto Reductase Family 4 Member C9) is an enzyme that belongs to the aldo-keto reductase superfamily. This enzyme plays a significant role in the detoxification of lipid peroxidation products that result from oxidative stress in cells . The aldo-keto reductase family encompasses several related proteins including AKR1C1, AKR1C2, AKR1C3, and AKR1C4, which share similar structural and functional characteristics but differ in their substrate specificity and tissue distribution . AKR4C9 functions as part of cellular defense mechanisms against reactive oxygen species (ROS) and the resulting lipid peroxidation, working alongside other detoxification enzymes like ADR1, ADR2, and MO1 .

What are the primary applications of AKR4C9 antibodies in research?

AKR4C9 antibodies are primarily utilized in research for the detection, quantification, and localization of AKR4C9 protein in biological samples. While specific information about AKR4C9 antibodies is limited in the provided search results, we can infer applications based on related aldo-keto reductase antibodies. These applications typically include Western blotting for protein detection and quantification, immunohistochemistry (IHC) for tissue localization, immunocytochemistry/immunofluorescence (ICC/IF) for cellular localization, and enzyme-linked immunosorbent assay (ELISA) for protein quantification in solution . Researchers use these antibodies to study the expression patterns of AKR4C9 in different tissues, its regulation under various physiological and pathological conditions, and its role in detoxification pathways .

How do I determine the most appropriate antibody format for detecting AKR4C9 in my experimental system?

Selecting the most appropriate antibody format depends on your specific experimental requirements and system. Consider these methodological steps:

  • Determine your application needs: For protein quantification in cell/tissue lysates, consider Western blot-validated antibodies. For protein localization in fixed tissues, choose IHC-validated antibodies. For live-cell imaging, consider non-fixative-requiring antibodies .

  • Evaluate antibody specificity: Review the antibody's validation data to ensure it specifically recognizes AKR4C9 without cross-reactivity to other AKR family members. This is particularly important given the high homology among AKR family proteins, as demonstrated by the challenges in developing specific antibodies for AKR1C3 .

  • Consider antibody type: Monoclonal antibodies typically offer higher specificity but recognize a single epitope (potentially limiting detection if that epitope is altered), while polyclonal antibodies recognize multiple epitopes (offering greater detection probability but potentially more cross-reactivity) .

  • Review conjugation requirements: Based on your detection method, select appropriate conjugation (unconjugated for traditional two-step detection, or directly conjugated with fluorophores or enzymes for one-step detection) .

An example approach is the development of the highly specific 10B10 monoclonal antibody for AKR1C3, which demonstrates excellent performance across multiple assay formats and clear differentiation from other highly homologous family members .

What are the optimal conditions for using AKR4C9 antibodies in Western blotting protocols?

For optimal Western blotting with AKR4C9 antibodies, consider these methodological guidelines based on successful protocols with related AKR family antibodies:

  • Sample preparation:

    • Lyse cells or tissues in a buffer containing protease inhibitors

    • Use reducing conditions (include β-mercaptoethanol or DTT in sample buffer)

    • Heat samples at 95°C for 5 minutes before loading

  • Electrophoresis parameters:

    • Use 10-12% polyacrylamide gels for optimal resolution of AKR proteins (~37 kDa)

    • Load appropriate protein amounts (typically 20-50 μg total protein per lane)

  • Transfer and blocking:

    • Transfer to PVDF membrane (preferred over nitrocellulose for AKR proteins)

    • Block with 5% non-fat dry milk or BSA in TBST for 1 hour at room temperature

  • Antibody incubation:

    • Use optimized antibody concentration (typically 0.5-5 μg/mL, based on titration)

    • Incubate with primary antibody overnight at 4°C

    • For detection, use HRP-conjugated secondary antibodies

  • Controls and validation:

    • Include positive control lysates from cells known to express AKR4C9

    • Use recombinant AKR4C9 protein as a standard

    • Consider knockout or knockdown controls to verify specificity

Evidence from related AKR antibodies shows that these protocols yield specific bands at approximately 36-37 kDa under reducing conditions, as demonstrated with the AKR1C3 antibody in A549 and HepG2 cell lines .

How can I optimize immunohistochemistry protocols for AKR4C9 detection in tissue sections?

For optimal immunohistochemical detection of AKR4C9 in tissue sections, follow these methodologically sound steps:

  • Tissue preparation:

    • Fix tissues in 10% neutral buffered formalin (24-48 hours)

    • Process and embed in paraffin following standard protocols

    • Section at 4-5 μm thickness

  • Antigen retrieval:

    • Perform heat-induced epitope retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)

    • Heat in a pressure cooker or microwave until boiling, then maintain for 10-20 minutes

    • Cool sections to room temperature gradually

  • Blocking and antibody incubation:

    • Block endogenous peroxidase activity with 3% hydrogen peroxide (10 minutes)

    • Block non-specific binding with 5-10% normal serum from the same species as the secondary antibody

    • Apply optimized concentration of primary antibody (typically 1-5 μg/mL)

    • Incubate at 4°C overnight or at room temperature for 1-2 hours

  • Detection system selection:

    • Use biotin-free detection systems to avoid background from endogenous biotin

    • Consider polymer-based detection systems for enhanced sensitivity

    • For fluorescence detection, select fluorophores with spectral properties compatible with your microscopy setup

  • Controls and validation:

    • Include tissue sections known to express AKR4C9 as positive controls

    • Include negative controls (omitting primary antibody)

    • Validate staining patterns against known expression profiles

This approach has proven effective for related AKR family members, as demonstrated by the successful IHC application of the AKR1C4 antibody (2C11) on formalin-fixed paraffin-embedded human liver tissue at 3 μg/mL concentration .

What considerations should be made when designing immunofluorescence experiments to study AKR4C9 cellular localization?

When designing immunofluorescence experiments for AKR4C9 cellular localization, implement these methodological considerations:

  • Cell preparation and fixation:

    • Choose appropriate fixative based on epitope sensitivity (4% paraformaldehyde preserves most epitopes while maintaining cellular architecture)

    • Optimize fixation time (typically 10-20 minutes at room temperature)

    • Consider membrane permeabilization requirements (0.1-0.5% Triton X-100 for cytoplasmic proteins)

  • Antibody selection and validation:

    • Confirm antibody validation for immunofluorescence applications

    • Titrate antibody to determine optimal concentration (typically starting at 1-10 μg/mL)

    • Consider fluorophore-conjugated primary antibodies to reduce background and simplify protocol

  • Co-localization studies:

    • Select appropriate subcellular markers (e.g., DAPI for nucleus, phalloidin for actin cytoskeleton)

    • Choose fluorophores with minimal spectral overlap

    • Include markers for expected cellular compartments based on known AKR4C9 localization (primarily cytoplasmic)

  • Imaging parameters:

    • Optimize exposure settings to prevent photobleaching and signal saturation

    • Use appropriate filters to minimize bleed-through

    • Consider confocal microscopy for precise subcellular localization

  • Quantification approaches:

    • Develop clear criteria for positive vs. negative staining

    • Use automated image analysis software for unbiased quantification

    • Analyze multiple fields and biological replicates for statistical validity

Based on related AKR family members, expect primarily cytoplasmic localization, as demonstrated with AKR1C3 antibody in LNCaP cells, which showed specific staining localized to cell surfaces and cytoplasm , and AKR1C4 antibody (2C11) immunofluorescence analysis on HepG2 cells .

How can I validate the specificity of an AKR4C9 antibody against other AKR family members?

Validating antibody specificity for AKR4C9 against other highly homologous AKR family members requires a comprehensive approach:

  • Recombinant protein testing:

    • Express and purify recombinant AKR4C9 and related family proteins (AKR1C1, AKR1C2, AKR1C3, AKR1C4)

    • Perform Western blot analysis with equivalent amounts of each protein

    • Quantify cross-reactivity percentages across family members

  • Overexpression systems:

    • Transfect cells with individual AKR family member expression constructs

    • Analyze antibody binding using Western blot, flow cytometry, or immunofluorescence

    • Compare signal intensity across different AKR-expressing cell lines

  • Knockout/knockdown validation:

    • Generate CRISPR/Cas9 knockout or siRNA knockdown of AKR4C9

    • Confirm loss of antibody signal in knockout/knockdown samples

    • Verify that signals for other AKR family members remain unchanged

  • Epitope mapping:

    • Identify the specific epitope recognized by the antibody

    • Compare sequence homology of this region across AKR family members

    • Design peptide competition assays with specific and non-specific peptides

  • Immunoprecipitation followed by mass spectrometry:

    • Perform IP using the antibody of interest

    • Analyze precipitated proteins by mass spectrometry

    • Identify all proteins captured and quantify specificity

This rigorous approach was demonstrated in the development of the 10B10 monoclonal antibody for AKR1C3, which was extensively tested against highly homologous family members AKR1C1, AKR1C2, and AKR1C4 to ensure specificity .

What techniques can be used to minimize non-specific binding when using AKR4C9 antibodies?

To minimize non-specific binding when using AKR4C9 antibodies, implement these methodological strategies:

  • Optimize blocking conditions:

    • Test different blocking agents (BSA, normal serum, commercial blockers)

    • Extend blocking time to 1-2 hours at room temperature

    • Consider adding 0.1-0.5% detergent (Tween-20, Triton X-100) to reduce hydrophobic interactions

  • Antibody dilution optimization:

    • Perform titration experiments to determine optimal antibody concentration

    • Use the minimum effective concentration that produces specific signal

    • Dilute antibodies in blocking buffer containing 0.1% detergent

  • Sample preparation refinements:

    • For tissue sections, implement additional blocking steps for endogenous biotin, peroxidase, and phosphatase

    • For cells, optimize fixation and permeabilization conditions to maintain epitope integrity

    • Consider antigen retrieval methods that maximize specific epitope exposure

  • Incubation condition modifications:

    • Extend primary antibody incubation time (overnight at 4°C rather than 1 hour at room temperature)

    • Increase washing duration and number of washes between steps

    • Consider adding carrier proteins or reducing agents to antibody diluent

  • Advanced specificity controls:

    • Perform peptide competition assays with immunizing peptide

    • Use isotype control antibodies at the same concentration

    • Include knockout/knockdown samples as negative controls

These approaches have proven effective with related antibodies, such as the AKR1C3 monoclonal antibody 10B10, which demonstrated high specificity and sensitivity across multiple assay formats after optimization of these parameters .

How does epitope selection affect the performance of AKR4C9 antibodies in different applications?

Epitope selection significantly impacts AKR4C9 antibody performance across different applications through these key mechanisms:

  • Epitope accessibility variations across applications:

    ApplicationProtein StateOptimal Epitope Characteristics
    Western BlotDenaturedLinear epitopes, internally located sequences
    ELISANative or denaturedSurface-exposed epitopes, distinctive sequences
    IHC/ICCPartially denaturedSemi-conformational epitopes, fixative-resistant
    IPNativeSurface-exposed conformational epitopes
  • Structural considerations:

    • Antibodies targeting highly conserved regions may cross-react with other AKR family members

    • Epitopes in catalytic domains may affect enzyme function in certain applications

    • N-terminal or C-terminal epitopes might be more accessible in native proteins but could be proteolytically cleaved in some samples

  • Post-translational modification effects:

    • Epitopes containing phosphorylation, glycosylation, or other modification sites may show variable antibody binding

    • Some applications may require modification-specific antibodies

    • Consider application-specific requirements for detecting different protein states

  • Application-specific performance:

    • For immunoprecipitation, conformational epitopes on protein surfaces perform best

    • For Western blotting, linear epitopes resistant to SDS denaturation are preferable

    • For immunohistochemistry, epitopes resistant to fixation and embedded tissue processing are essential

  • Validation requirements:

    • Each application requires specific validation approaches

    • Performance in one application doesn't guarantee performance in others

This understanding has been applied in developing specific antibodies for related proteins, as seen with the AKR1C3 antibody 10B10, which was specifically designed and validated to perform well across multiple assay formats, including Western blot, immunohistochemistry, and ELISA .

What approaches can be used to study the regulation of AKR4C9 expression under oxidative stress conditions?

To study AKR4C9 regulation under oxidative stress conditions, implement these methodological approaches:

  • Stress induction and time course analysis:

    • Expose cells/tissues to controlled oxidative stressors (H₂O₂, paraquat, tBHP)

    • Perform time-course experiments (0-48 hours) to capture dynamic expression changes

    • Monitor concurrent cellular ROS levels using fluorescent indicators (DCF-DA, MitoSOX)

  • Transcriptional regulation assessment:

    • Quantify AKR4C9 mRNA levels using RT-qPCR after stress induction

    • Analyze promoter activity using reporter assays (luciferase constructs with AKR4C9 promoter)

    • Identify transcription factor binding using ChIP assays focusing on stress-responsive factors like Nrf2, ANAC102

  • Protein expression quantification:

    • Measure protein levels via Western blot using validated AKR4C9 antibodies

    • Implement pulse-chase experiments to determine protein stability changes

    • Assess subcellular localization changes using immunofluorescence

  • Signaling pathway investigation:

    • Use specific pathway inhibitors to delineate regulatory mechanisms

    • Perform phosphoproteomic analysis to identify post-translational modifications

    • Implement genetic approaches (overexpression, knockdown) of pathway components

  • In vivo models:

    • Analyze AKR4C9 expression in oxidative stress-related disease models

    • Assess tissue-specific expression patterns using IHC

    • Correlate expression with pathological findings and oxidative damage markers

This approach is supported by research on related genes like ANAC102, which regulates detoxification-related genes including AKR4C9 in response to oxidative stress . Similar methods have been applied to study AKR1C3, revealing its protective role against oxidative stress in various cellular contexts .

How can AKR4C9 antibodies be utilized in studying protein-protein interactions and complex formation?

For investigating AKR4C9 protein-protein interactions and complex formation, implement these advanced methodological approaches:

  • Co-immunoprecipitation (Co-IP) strategies:

    • Use AKR4C9 antibodies to precipitate the protein complex from cell lysates

    • Perform reciprocal Co-IP with antibodies against suspected interaction partners

    • Analyze precipitated complexes via Western blot or mass spectrometry

    • Consider crosslinking approaches to stabilize transient interactions

  • Proximity labeling techniques:

    • Generate AKR4C9 fusion constructs with BioID or APEX2 proximity labeling enzymes

    • Express constructs in relevant cell types and induce labeling

    • Purify biotinylated proteins using streptavidin beads

    • Identify proximal proteins via mass spectrometry

  • FRET/BRET applications:

    • Create fluorescent protein fusions with AKR4C9 and potential interactors

    • Perform live-cell FRET measurements to detect direct interactions

    • Utilize BRET assays for detecting interactions with minimal perturbation to cellular physiology

    • Quantify interaction dynamics under different cellular conditions

  • Pull-down assays with recombinant proteins:

    • Express and purify recombinant AKR4C9 with affinity tags

    • Perform pull-down experiments with cell lysates

    • Identify interacting proteins via mass spectrometry

    • Validate direct interactions using purified recombinant proteins

  • Antibody-based proximity assays:

    • Use in situ proximity ligation assay (PLA) to visualize protein interactions in fixed cells

    • Implement co-localization studies using immunofluorescence with appropriate controls

    • Quantify interaction dynamics under different experimental conditions

These approaches can reveal interactions similar to those observed with related proteins like AKR1C3, which has been shown to interact with specific cellular components in prostate cancer cells using antibody-based detection methods .

What are the considerations for using AKR4C9 antibodies in high-throughput screening of potential inhibitors or activators?

When using AKR4C9 antibodies for high-throughput screening of modulators, implement these methodological considerations:

  • Assay format selection and optimization:

    Assay FormatAdvantagesConsiderations for AKR4C9 Screening
    ELISA-basedHigh throughput, quantitativeRequires highly specific antibodies with minimal cross-reactivity
    Cell-based reporterPhysiologically relevantRequires careful validation of reporter construct specificity
    Antibody-based imagingAllows subcellular analysisNeeds optimization for automated image acquisition and analysis
    AlphaLISA/HTRFNo-wash format, sensitiveRequires pairs of antibodies recognizing different epitopes
  • Antibody validation requirements:

    • Verify specificity against other AKR family members

    • Determine optimal antibody concentration for signal-to-noise optimization

    • Assess antibody performance in the presence of DMSO and other vehicle controls

    • Validate reproducibility across multiple lots and extended time periods

  • Assay development considerations:

    • Establish Z-factor >0.5 for statistical robustness

    • Develop appropriate positive and negative controls

    • Optimize enzyme concentration, substrate concentration, and reaction time

    • Implement counter-screens to eliminate false positives

  • Data analysis strategies:

    • Develop algorithms for identifying true hits versus artifacts

    • Implement dose-response confirmation of primary hits

    • Establish criteria for hit selection based on both potency and efficacy

    • Consider computational approaches to predict off-target effects

  • Secondary validation approaches:

    • Confirm hits with orthogonal assays using different detection methods

    • Verify target engagement using cellular thermal shift assays

    • Assess compound effects on AKR4C9 expression and stability

    • Evaluate selectivity against other AKR family members

This approach is supported by successful development of highly specific antibodies for related proteins like AKR1C3 (the 10B10 monoclonal antibody), which enabled sensitive detection across multiple assay formats and facilitated the development of AKR1C3-targeting therapeutics .

How should I address inconsistent results when using AKR4C9 antibodies across different experimental systems?

When encountering inconsistent results with AKR4C9 antibodies across experimental systems, implement this systematic troubleshooting approach:

  • Antibody-related variables assessment:

    • Verify antibody specificity using recombinant AKR4C9 and related family proteins

    • Test multiple antibody lots and storage conditions (avoid freeze-thaw cycles)

    • Consider epitope availability differences across applications

    • Validate antibody performance in each experimental system independently

  • Sample preparation evaluation:

    • Standardize lysis buffers and protein extraction protocols

    • Verify protein integrity through total protein staining methods

    • Consider native versus denaturing conditions for epitope accessibility

    • Standardize sample handling to minimize degradation

  • Experimental system comparison:

    • Document differences in expression levels across cell types/tissues

    • Consider species-specific variations in protein sequence and epitope conservation

    • Evaluate post-translational modifications in different systems

    • Assess protein interactions that might mask epitopes

  • Protocol optimization for each system:

    SystemOptimization ParametersValidation Approach
    Cell linesCell density, passage numberUse consistent positive control cell line
    Primary cellsIsolation method, culture conditionsInclude matched cell line controls
    Tissue sectionsFixation time, antigen retrievalUse consistent positive control tissue
    Animal modelsSpecies, tissue processingValidate antibody for cross-reactivity
  • Control implementation strategies:

    • Include recombinant protein standards across experiments

    • Implement knockdown/knockout controls when possible

    • Use competitive peptide blocking to confirm specificity

    • Consider alternative antibodies targeting different epitopes

This systematic approach has proven effective for troubleshooting other members of the AKR family, as demonstrated in the development and characterization of the highly specific AKR1C3 antibody (10B10), which was validated across multiple experimental systems to ensure reliable and reproducible results .

What statistical approaches are recommended for quantifying AKR4C9 expression levels in immunohistochemistry studies?

For robust quantification of AKR4C9 expression in immunohistochemistry studies, implement these statistical and methodological approaches:

  • Semi-quantitative scoring systems:

    • Develop a clear scoring system (e.g., 0-3+ intensity scale)

    • Implement H-score methodology (intensity × percentage of positive cells)

    • Use Allred scoring (intensity + proportion) for comprehensive assessment

    • Ensure multiple independent pathologists score samples blindly

  • Digital image analysis optimization:

    • Standardize image acquisition parameters (magnification, exposure, white balance)

    • Segment tissue compartments using machine learning algorithms

    • Quantify positive pixel area, intensity, and distribution

    • Validate algorithm against expert pathologist scoring

  • Statistical analysis selection:

    • Determine appropriate statistical tests based on data distribution

    • Use non-parametric tests for ordinal scoring data (Mann-Whitney, Kruskal-Wallis)

    • Apply parametric tests for continuous measurements after confirming normal distribution

    • Implement ANOVA with post-hoc tests for multiple group comparisons

  • Correlation and multivariate analysis:

    • Correlate AKR4C9 expression with clinical parameters

    • Perform multivariate analysis to identify independent associations

    • Use machine learning approaches for pattern recognition

    • Consider survival analysis (Kaplan-Meier, Cox regression) for prognostic value

  • Quality control and reproducibility measures:

    • Calculate inter-observer and intra-observer kappa statistics

    • Implement tissue microarrays for standardization across samples

    • Use internal reference standards in each batch

    • Report detailed methods for reproducibility

This approach aligns with methodologies used for quantifying expression of related proteins like AKR1C3 and AKR1C4 in immunohistochemical studies, where specific antibodies have been validated for tissue expression analysis in different pathological contexts .

How can conflicting results between transcriptomic data and antibody-based detection of AKR4C9 be reconciled?

To reconcile conflicting results between transcriptomic data and antibody-based AKR4C9 detection, implement this systematic investigative approach:

  • Technical verification of both methods:

    • Validate RNA integrity and quality metrics for transcriptomic data

    • Confirm antibody specificity using recombinant proteins and knockout controls

    • Repeat experiments with alternative primers/probes and different antibody clones

    • Verify that both methods are targeting the same gene/protein isoform

  • Post-transcriptional regulation assessment:

    • Analyze microRNA expression that might target AKR4C9 mRNA

    • Assess mRNA stability through actinomycin D chase experiments

    • Investigate RNA binding proteins that might influence translation efficiency

    • Consider alternative splicing that might affect antibody epitope presence

  • Post-translational regulation investigation:

    • Examine protein stability through cycloheximide chase experiments

    • Assess post-translational modifications that might affect antibody binding

    • Investigate proteasomal or lysosomal degradation pathways

    • Consider protein localization changes that might affect detection

  • Time-course resolution studies:

    • Perform detailed time-course experiments to capture temporal dynamics

    • Analyze both mRNA and protein levels at multiple timepoints

    • Consider delay between transcription and translation (typically 4-6 hours)

    • Examine protein half-life in relation to mRNA half-life

  • Experimental system considerations:

    • Evaluate cell type-specific differences in post-transcriptional regulation

    • Consider microenvironmental factors that might affect protein but not mRNA

    • Assess developmental or stress-dependent regulatory mechanisms

    • Examine epigenetic modifications that might influence protein expression

This approach is supported by research on stress-responsive genes like AKR4C9, which can exhibit complex regulation patterns where transcription factors like ANAC102 influence expression in response to cellular stress conditions, potentially leading to discrepancies between mRNA and protein levels under different conditions .

How can single-cell analysis techniques be applied to study AKR4C9 expression heterogeneity in complex tissues?

For investigating AKR4C9 expression heterogeneity at the single-cell level, implement these advanced methodological approaches:

  • Single-cell RNA sequencing (scRNA-seq) applications:

    • Apply droplet-based platforms (10x Genomics) for high-throughput analysis

    • Implement Smart-seq2 for full-length transcript coverage when isoform detection is crucial

    • Use computational tools (Seurat, Monocle) to identify cell clusters with differential AKR4C9 expression

    • Perform trajectory analysis to link AKR4C9 expression with cellular differentiation states

  • Protein-level single-cell analysis:

    • Apply mass cytometry (CyTOF) with metal-conjugated AKR4C9 antibodies

    • Implement imaging mass cytometry for spatial context preservation

    • Use cyclic immunofluorescence (CycIF) for multiplexed protein detection

    • Consider single-cell Western blotting for protein isoform discrimination

  • Integrated multi-omics approaches:

    • Apply CITE-seq for simultaneous mRNA and protein detection

    • Implement spatial transcriptomics to correlate AKR4C9 expression with tissue architecture

    • Use paired single-cell RNA-seq and ATAC-seq to link expression with chromatin accessibility

    • Consider single-cell proteogenomics approaches for comprehensive profiling

  • In situ analysis methodologies:

    • Apply multiplexed RNA fluorescence in situ hybridization (FISH) techniques

    • Implement proximity ligation assay (PLA) for protein interaction studies

    • Use highly multiplexed immunofluorescence platforms (CODEX, MIBI)

    • Consider spatial metabolomics to link AKR4C9 expression with metabolic activity

  • Analysis and visualization strategies:

    • Implement dimensionality reduction techniques (t-SNE, UMAP) for visualization

    • Use spatial statistical methods to quantify expression patterns

    • Apply machine learning approaches for pattern recognition

    • Develop integrative computational frameworks to synthesize multi-modal data

This approach leverages cutting-edge technologies that have been applied to study related proteins, allowing researchers to understand the heterogeneous expression patterns of enzymes like AKR4C9 in complex tissues and their correlation with cellular states and tissue microenvironments .

What are the latest advances in antibody engineering that might improve AKR4C9 detection specificity and sensitivity?

Recent advances in antibody engineering offer significant improvements for AKR4C9 detection through these innovative approaches:

  • Phage display technology optimization:

    • Implement negative selection strategies against homologous AKR family members

    • Use structural information to target unique epitopes on AKR4C9

    • Apply deep sequencing of selection rounds to identify rare high-specificity binders

    • Develop computational models to predict antibody specificity profiles based on sequence

  • Recombinant antibody fragment development:

    • Engineer smaller antibody formats (scFv, Fab, nanobodies) for improved tissue penetration

    • Design bispecific antibodies targeting two distinct AKR4C9 epitopes for increased specificity

    • Implement affinity maturation through directed evolution

    • Create site-specific conjugation strategies for consistent labeling

  • Computational design approaches:

    • Apply machine learning algorithms to predict optimal antibody-antigen interfaces

    • Implement structure-based design using homology models of AKR4C9

    • Use molecular dynamics simulations to optimize binding interactions

    • Develop epitope-specific antibodies based on in silico epitope mapping

  • Novel detection technologies:

    • Implement proximity-based detection systems (SplitTEV, SUPRA) for enhanced sensitivity

    • Develop aptamer-antibody hybrid systems for dual recognition

    • Apply DNA-barcoded antibodies for ultrasensitive detection

    • Implement amplification-free digital detection methods

  • Validation and quality control advances:

    Engineering ApproachSpecificity EnhancementSensitivity ImprovementValidation Method
    CDR optimizationSequence-guided mutationsAffinity maturationSPR/BLI binding kinetics
    Negative selectionCross-reactivity eliminationSequential panningCross-adsorption testing
    Structural designEpitope-focused engineeringParatope optimizationX-ray crystallography
    ML-guided selectionSpecificity predictionSignal-to-noise modelingHigh-content screening

These approaches align with the latest developments in antibody engineering for highly homologous proteins, as exemplified by the design of highly specific antibodies through phage display experiments and computational models that can predict and design novel antibody sequences with customized specificity profiles .

How might multiplexed imaging approaches using AKR4C9 antibodies enhance our understanding of metabolic pathway interactions?

Multiplexed imaging approaches with AKR4C9 antibodies can reveal complex metabolic pathway interactions through these advanced methodological implementations:

  • Highly multiplexed immunofluorescence platforms:

    • Implement cyclic immunofluorescence (CycIF) for 30+ marker detection

    • Apply CODEX or MIBI for highly multiplexed tissue imaging

    • Use DNA-barcoded antibody systems (Immuno-SABER) for signal amplification

    • Implement clearing-enhanced 3D imaging for volumetric analysis

  • Multi-parameter correlation analysis:

    • Simultaneously detect AKR4C9 with related metabolic enzymes

    • Co-visualize detoxification pathway components (ADR1, ADR2, ALDH7B4)

    • Map key transcription factors (ANAC102) regulating metabolic responses

    • Correlate with oxidative stress markers and lipid peroxidation products

  • Spatial metabolomics integration:

    • Combine antibody-based imaging with mass spectrometry imaging

    • Correlate AKR4C9 distribution with metabolite profiles

    • Implement MALDI-imaging mass spectrometry for metabolite localization

    • Use computational approaches to integrate proteomic and metabolomic data

  • Dynamic process visualization:

    • Develop live-cell compatible antibody-based sensors

    • Implement optogenetic perturbation with simultaneous imaging

    • Use biosensors to correlate enzyme activity with localization

    • Apply fluorescence lifetime imaging to detect protein-protein interactions

  • Data analysis and systems biology integration:

    • Implement advanced image analysis algorithms for cellular segmentation

    • Apply machine learning for pattern recognition across multiple parameters

    • Develop computational models to predict metabolic pathway interactions

    • Use network analysis to identify key regulatory nodes

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.