STK32A Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Phosphate Buffered Saline (PBS) containing 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the purchasing method and location. For specific delivery estimates, please contact your local distributor.
Synonyms
A930015B13Rik antibody; MGC22688 antibody; Serine/threonine kinase 32A antibody; Serine/threonine protein kinase 32A antibody; Serine/threonine-protein kinase 32A antibody; ST32A_HUMAN antibody; STK 32A antibody; Stk32a antibody; YANK 1 antibody; YANK1 antibody; Yet another novel kinase 1 antibody
Target Names
STK32A
Uniprot No.

Target Background

Database Links

HGNC: 28317

KEGG: hsa:202374

STRING: 9606.ENSP00000381030

UniGene: Hs.585069

Protein Families
Protein kinase superfamily, Ser/Thr protein kinase family
Subcellular Location
Cell membrane; Lipid-anchor.

Q&A

What is STK32A and what cellular functions does it perform?

STK32A (Serine/threonine kinase 32A, also known as YANK1) belongs to the Ser/Thr protein kinase family. Recent research has revealed its critical role in regulating hair cell planar polarity in the vestibular maculae of the inner ear. STK32A functions downstream of the transcription factor EMX2, which negatively regulates STK32A expression. The protein affects cellular metabolism through protein serine/threonine kinase phosphorylation .

STK32A is particularly important in the development of sensory receptor hair cells that detect linear acceleration and contribute to equilibrioception. These hair cells are organized with stereociliary bundles that have specific planar-polarized orientations. STK32A helps establish this planar-polarized organization by influencing the distribution of the transmembrane receptor GPR156 at hair cell boundaries .

What are the available types of STK32A antibodies and their respective applications?

Based on current research resources, STK32A antibodies are available in multiple formats with different applications:

Antibody TypeHostApplicationsReactivityExamples
MonoclonalMouseWB, IHC, ELISAHumanProteintech 67528-1-Ig
PolyclonalRabbitWB, ELISAHuman, Mouse, RatProteintech 16223-1-AP
PolyclonalRabbitIHCHumanHPA040236

The most commonly validated applications include Western Blotting (WB), Immunohistochemistry (IHC), and ELISA. Different antibodies show varying specificities and are recommended for different dilutions depending on the application .

How should I optimize sample preparation for STK32A detection in Western blotting?

For optimal STK32A detection in Western blotting:

  • Cell lysis: Use a complete lysis buffer containing protease inhibitors to prevent degradation of STK32A protein.

  • Protein concentration: Determine protein concentration using a standard method (Bradford or BCA) and load 20-40 µg of total protein per lane.

  • Electrophoresis conditions: Use 10-12% SDS-PAGE gels as STK32A has an observed molecular weight of approximately 46-50 kDa.

  • Transfer conditions: Transfer proteins to PVDF membranes (rather than nitrocellulose) for better protein retention.

  • Blocking: Block with 5% non-fat dry milk in TBST for 1-2 hours at room temperature.

  • Primary antibody incubation: Dilute STK32A antibody according to manufacturer recommendations (typically 1:500-1:10000 depending on the specific antibody).

  • Secondary antibody: Use appropriate HRP-conjugated secondary antibody at 1:5000-1:10000 dilution .

STK32A has been successfully detected in various cell lines including A549, HeLa, MCF-7, T-47D, and JAR cells, making these suitable positive controls .

What are the recommended antigen retrieval methods for STK32A immunohistochemistry?

For optimal STK32A detection in immunohistochemistry applications:

  • Primary fixation: 10% neutral buffered formalin fixation of tissues is recommended.

  • Section thickness: 4-5 μm sections provide optimal results.

  • Antigen retrieval: Two methods have been validated:

    • TE buffer pH 9.0 (primary recommendation)

    • Citrate buffer pH 6.0 (alternative method)

  • Antibody dilution: For most commercially available antibodies like Proteintech 67528-1-Ig, use at 1:500-1:2000 dilution.

  • Incubation conditions: Overnight incubation at 4°C yields optimal signal-to-noise ratio.

  • Detection system: A polymer-based detection system is recommended for enhanced sensitivity .

STK32A has been positively detected in human colon cancer tissue and human breast cancer tissue using these methods .

How can I validate STK32A antibody specificity for my particular experimental system?

Validating antibody specificity is critical for reliable experimental outcomes. For STK32A antibodies, implement the following comprehensive validation strategy:

  • Knockout/knockdown controls:

    • Generate STK32A knockdown in your experimental cell line using siRNA or shRNA

    • Use CRISPR-Cas9 to create STK32A knockout cells

    • Compare signal between wild-type and knockout/knockdown samples across all applications

  • Recombinant protein verification:

    • Test the antibody against recombinant STK32A protein (full-length and fragments)

    • Include closely related family members (STK32B/STK32C) to assess cross-reactivity

  • Epitope mapping:

    • Determine which region of STK32A the antibody recognizes

    • For monoclonal antibodies, consider testing multiple clones recognizing different epitopes

    • Available antibodies target different regions (AA 1-166, AA 1-30, AA 347-374, AA 90-170)

  • Orthogonal detection methods:

    • Confirm results using mass spectrometry

    • Compare results from antibodies from different vendors or different clones

    • Correlate protein detection with mRNA expression data

  • Peptide competition:

    • Pre-incubate the antibody with immunizing peptide before application

    • Signal should be significantly reduced if antibody is specific .

Despite the availability of several commercially available antibodies, researchers have reported challenges in identifying ones with sufficient specificity for certain applications, particularly for detecting endogenous STK32A in mouse tissues .

What strategies can be employed to detect subcellular localization of STK32A in hair cells?

Determining the subcellular localization of STK32A in hair cells presents unique challenges. Based on recent research approaches:

  • Fluorescent fusion proteins:

    • STK32A:EGFP fusion constructs have been successfully used to track localization

    • Both N- and C-terminal EGFP tags have been evaluated, with C-terminal tagging showing better functional capacity

    • These constructs can be delivered using adeno-associated virus (AAV) vectors for transduction into developing hair cells

  • Immunofluorescence optimization:

    • Use paraformaldehyde fixation (4%) followed by permeabilization with 0.2% Triton X-100

    • Counter-stain with phalloidin to visualize actin-rich stereociliary bundles

    • Use confocal microscopy with Z-stack acquisition for 3D visualization

  • Sub-compartment markers:

    • Co-stain with markers for specific cellular compartments

    • Phalloidin has been used to identify stereociliary bundles in relation to STK32A localization

  • Live-cell imaging:

    • For dynamic studies, time-lapse imaging from 12-24 hours post-transduction can capture the temporal aspects of STK32A localization

Using these approaches, researchers have determined that STK32A localizes to the apical compartment of hair cells and the stereociliary bundle, with detection possible between 12-24 hours post-transduction .

How does STK32A function in hair cell planar polarity development and what experimental approaches can investigate this mechanism?

STK32A plays a critical role in hair cell planar polarity through several mechanisms:

  • Relationship with EMX2:

    • STK32A is negatively regulated by the transcription factor EMX2

    • STK32A expression is repressed in lateral hair cells of the utricle where EMX2 is expressed

    • Experimental approach: Conditional knockout or overexpression of EMX2 can be used to study its regulatory effect on STK32A

  • GPR156 regulation:

    • STK32A influences the subcellular distribution of GPR156

    • In STK32A knockout mice, GPR156 appears aberrantly at the surface of EMX2-negative vestibular hair cells

    • STK32A overexpression in cochlear hair cells reduces GPR156 detection at the cell surface

    • Experimental approach: Co-immunoprecipitation studies to determine if STK32A directly interacts with GPR156 or regulates its trafficking partners

  • Basal body positioning:

    • STK32A appears to coordinate intracellular polarity with the planar cell polarity (PCP) axis

    • In STK32A knockout mice, hair cells in medial regions show random bundle orientations

    • Experimental approach: Live imaging of basal body movements in STK32A knockout versus wild-type cells

  • Kinase activity requirement:

    • STK32A's kinase function appears essential for its role in planar polarity

    • Experimental approach: Express kinase-dead mutants (e.g., K52R) and compare their effects with wild-type STK32A

To investigate these mechanisms, researchers have employed:

  • CRISPR-mediated mutagenesis to generate STK32A knockout mice

  • AAV-mediated gene delivery to express STK32A in specific cell populations

  • Fluorescently tagged STK32A to track subcellular localization

  • N-myristoylation mutants (STK32A Δ2G) to study membrane association requirements .

What are the optimal protocols for detecting endogenous STK32A in tissues where it may be expressed at low levels?

Detecting low-abundance endogenous STK32A requires specialized approaches:

  • Signal amplification techniques:

    • Tyramide signal amplification (TSA) for immunohistochemistry and immunofluorescence

    • RNAscope for in situ hybridization detection of mRNA with single-molecule sensitivity

    • Proximity ligation assay (PLA) for detecting protein interactions with enhanced sensitivity

  • Tissue-specific considerations:

    • For inner ear tissues: Careful micro-dissection of vestibular maculae followed by gentle fixation

    • Extended primary antibody incubation (48-72 hours) at 4°C may improve detection

    • Sample enrichment through laser capture microdissection prior to protein analysis

  • Antibody selection and validation:

    • Test multiple antibodies targeting different epitopes

    • Use tissues from STK32A knockout mice as negative controls

    • Consider using a cocktail of validated antibodies against different epitopes

  • Western blot enhancement:

    • Increase protein loading (50-100 μg)

    • Use high-sensitivity chemiluminescent substrates

    • Consider immunoprecipitation before Western blotting to concentrate the protein

  • Challenges in antibody validation:

    • Researchers have reported difficulties in identifying antibodies with sufficient specificity for detecting endogenous STK32A in mouse tissues

    • Multiple commercial antibodies (Σ, ProteinTech, RayBiotech) and custom antibodies have been tested with limited success

    • Alternative approaches such as tagged knockin mice may be necessary for reliable detection .

How can I design experiments to investigate the role of STK32A kinase activity in its biological functions?

To investigate STK32A kinase activity and its relevance to biological function:

  • Kinase-dead mutants:

    • Generate the K52R mutation which disrupts ATP binding

    • Compare the effects of wild-type STK32A versus K52R mutant on hair cell orientation

    • This approach has been used in research showing that kinase activity is required for STK32A's role in planar polarity

  • Identification of substrates:

    • Perform kinase assays with recombinant STK32A and candidate substrates

    • Use phosphoproteomic approaches to identify differentially phosphorylated proteins in STK32A-overexpressing versus control cells

    • BioID or proximity labeling methods can identify proteins in close proximity to STK32A

  • Pharmacological inhibition:

    • Test the effects of broad-spectrum serine/threonine kinase inhibitors

    • Development of more specific inhibitors may require structural information about STK32A

  • Structure-function analysis:

    • Generate deletion constructs to identify domains critical for STK32A function

    • The N-myristoylation of STK32A (disrupted in the Δ2G mutant) affects its localization to the apical compartment and stereociliary bundle

    • Research has shown that STK32A lacking N-myristoylation does not colocalize with phalloidin in the apical compartment or bundle

  • Temporal control of STK32A activity:

    • Develop conditionally active STK32A variants (e.g., using chemical-induced dimerization)

    • This would allow temporal dissection of when kinase activity is required during development

By combining these approaches, researchers can determine both the biochemical activity of STK32A and correlate it with its biological functions in hair cell development and other contexts .

What are the common challenges when using STK32A antibodies and how can they be addressed?

Researchers frequently encounter several challenges when working with STK32A antibodies:

  • Non-specific bands in Western blotting:

    • Problem: Multiple bands appearing at unexpected molecular weights

    • Solution: Increase antibody specificity by:

      • Using longer blocking times (2-3 hours) with 5% BSA instead of milk

      • Perform more stringent washes (5 x 5 minutes with 0.1% Tween-20)

      • Test multiple antibodies targeting different epitopes

      • Include peptide competition controls

  • Poor signal-to-noise ratio in immunohistochemistry:

    • Problem: High background staining obscuring specific signals

    • Solution:

      • Optimize antigen retrieval (compare citrate buffer pH 6.0 vs. TE buffer pH 9.0)

      • Increase blocking time and concentration (3% BSA + 10% normal serum)

      • Use lower primary antibody concentration with longer incubation times

      • Consider biotin-streptavidin amplification systems for weak signals

  • Inability to detect endogenous STK32A:

    • Problem: No detectable signal in tissues known to express STK32A

    • Solution:

      • Enrich samples through immunoprecipitation before Western blotting

      • Use highly sensitive detection methods (enhanced chemiluminescence)

      • Consider detecting STK32A mRNA through qPCR or in situ hybridization as an alternative approach

      • Employ tissue-specific expression systems or tagged knock-in mice

  • Inter-experimental variability:

    • Problem: Inconsistent results between experiments

    • Solution:

      • Standardize protein extraction methods

      • Include positive controls in each experiment

      • Document lot numbers of antibodies

      • Consider preparing larger batches of antibody dilutions to use across experiments

These solutions are based on reported challenges in STK32A research, where multiple commercial antibodies (Σ, ProteinTech, RayBiotech) and custom antibodies have been tested with variable success in detecting endogenous STK32A in mouse tissues .

How can I differentiate between STK32A and other closely related kinase family members?

Distinguishing STK32A from related family members (particularly STK32B and STK32C) requires careful experimental design:

  • Antibody selection and validation:

    • Choose antibodies targeting unique regions that differ between family members

    • Validate antibody specificity against recombinant STK32A, STK32B, and STK32C proteins

    • Perform peptide competition assays with specific peptides from each family member

  • Molecular approaches:

    • Design PCR primers or probes targeting unique regions for specific mRNA detection

    • Use siRNA or CRISPR targeting unique sequences to confirm antibody specificity

    • Express tagged versions of each family member and compare their migration patterns and subcellular localization

  • Expression pattern analysis:

    • Compare tissue distribution patterns of STK32A versus STK32B/STK32C

    • STK32A has been detected in specific regions of the inner ear, particularly in vestibular hair cells

    • Understanding the tissue-specific expression can help distinguish family members

  • Functional characterization:

    • Develop assays that distinguish the specific activities of each family member

    • For STK32A, its role in hair cell planar polarity and GPR156 regulation provides specific functional readouts

    • Overexpression studies in lateral hair cells can reveal STK32A-specific effects on bundle orientation

  • Mass spectrometry:

    • Use targeted mass spectrometry to identify specific peptides unique to STK32A

    • This approach can provide the highest specificity for distinguishing between closely related proteins .

What experimental strategies can resolve contradictory findings about STK32A subcellular localization and function?

When facing contradictory results regarding STK32A localization or function, consider these methodological approaches:

  • Standardize expression systems:

    • Problem: Different expression levels may lead to aberrant localization

    • Solution: Use inducible expression systems to achieve physiological levels

    • Compare endogenous protein localization with overexpressed protein

  • Tag position effects:

    • Problem: Different tag positions can alter protein localization

    • Solution: Compare N-terminal, C-terminal, and internal tags

    • Research has shown that C-terminal EGFP tags on STK32A have greater capacity to promote reversed bundle orientations in lateral hair cells compared to N-terminal tags

  • Temporal considerations:

    • Problem: Protein localization may change during development

    • Solution: Conduct time-course studies

    • STK32A:EGFP has been detected in the apical compartment between 12-24 hours post-transduction

  • Cell type specificity:

    • Problem: Function may vary by cell type

    • Solution: Study STK32A in multiple relevant cell types

    • Compare findings between in vitro cultures and in vivo models

  • Genetic background effects:

    • Problem: Phenotypes may vary by genetic background

    • Solution: Test mutations on multiple genetic backgrounds

    • Back-cross mutant lines to standardize genetic background

  • Methodological variations:

    • Problem: Different fixation or extraction methods may affect localization

    • Solution: Compare multiple preservation techniques

    • Use complementary approaches (live imaging vs. fixed cells)

These approaches have helped researchers resolve initial contradictions in STK32A studies, such as determining that STK32A localizes to the apical compartment of hair cells and stereociliary bundle, whereas EGFP alone remains cytosolic and strictly in the cell soma .

What is the current understanding of how STK32A regulates GPR156 distribution and affects hair cell polarization?

Recent research has revealed a complex mechanism by which STK32A influences hair cell polarization through GPR156 regulation:

  • GPR156 distribution model:

    • STK32A negatively regulates GPR156 distribution at the apical cell surface

    • In EMX2-negative hair cells (medial region), STK32A expression prevents GPR156 from being delivered or retained at the apical surface

    • This regulation creates a boundary of GPR156 expression that helps establish the line of polarity reversal (LPR) in vestibular maculae

  • Evidence from knockout studies:

    • In STK32A knockout mice, GPR156 appears aberrantly at the surface of EMX2-negative vestibular hair cells

    • This mislocalization leads to random hair cell bundle orientations in the medial region of the utricle and outer region of the saccule

  • Evidence from overexpression studies:

    • Ectopic overexpression of STK32A in cochlear hair cells dramatically reduces the amount of GPR156 detected at the cell surface

    • When STK32A is expressed in lateral hair cells of the utricle (where it's normally repressed), it can generate hair cells with medial bundle orientations

  • Potential mechanisms of GPR156 regulation:

    • STK32A may regulate GPR156 trafficking to the cell surface

    • Alternatively, it may affect GPR156 retention at the membrane

    • The kinase activity of STK32A appears necessary for this function, suggesting phosphorylation of GPR156 or its trafficking partners

  • Proposed model:

    • GPR156 functions at the apical cell surface to align stereociliary bundles with the underlying planar cell polarity (PCP) axis

    • STK32A serves a similar function in EMX2-negative regions by positioning the basal body opposite of PK2

    • The LPR is formed along the boundary of EMX2 expression through a two-step negative regulation process:

      1. EMX2 represses STK32A expression

      2. STK32A inhibits GPR156 delivery or retention at the apical cell surface .

What are the emerging techniques and methodologies for studying STK32A in developmental contexts?

Research on STK32A has benefited from several innovative approaches that could be applied to other developmental studies:

  • Advanced genetic manipulation:

    • CRISPR-mediated mutagenesis targeting specific exons

    • This approach was used to generate STK32A knockout mice by creating DNA breaks in introns flanking exon 2

    • AAV-mediated gene delivery for cell-specific expression in developing tissues

  • Ex vivo culture systems:

    • Utricle micro-dissection and explant culture techniques

    • These systems allow for experimental manipulation of developing sensory organs

    • Transduction with viral vectors to introduce wild-type or mutant proteins

  • Fluorescent fusion constructs:

    • STK32A:EGFP fusion proteins to track subcellular localization

    • Both N- and C-terminal EGFP tags have been evaluated

    • Bicistronic constructs (STK32A-P2A-EGFP) to identify transduced cells while expressing untagged protein

  • Structure-function analysis:

    • Creation of specific mutants:

      • Kinase-dead mutants (K52R)

      • N-myristoylation mutants (Δ2G)

    • These mutants help dissect the requirements for STK32A function and localization

  • Complementary techniques for protein localization:

    • Immunofluorescence with phalloidin co-staining to visualize actin-rich structures

    • Extraction of cellular profiles from image stacks to analyze protein distribution

  • Transcriptional regulation studies:

    • Analysis of STK32A as a target of the transcription factor EMX2

    • This has revealed a regulatory network controlling cell polarization in the inner ear

These methodologies provide a framework for investigating the roles of poorly characterized kinases in development and can be applied to study related proteins in different developmental contexts .

What are the most promising research avenues for understanding STK32A's broader roles beyond inner ear development?

While current research has focused on STK32A's role in inner ear development, several promising avenues exist for exploring its broader functions:

  • Potential roles in other polarized tissues:

    • Investigate STK32A expression and function in other epithelial tissues with planar polarity

    • Kidney tubules, respiratory epithelium, and other sensory organs would be logical targets

    • Research question: Does STK32A regulate planar polarity in these contexts similar to its function in the inner ear?

  • Association with human diseases:

    • STK32A maps to human chromosome 5, which contains genes associated with Cockayne syndrome (ERCC8) and familial adenomatous polyposis (APC)

    • Research question: Does STK32A contribute to the pathophysiology of these or other diseases?

  • Cancer biology connections:

    • STK32A antibodies have been validated in human colon and breast cancer tissues

    • Research question: Is STK32A expression altered in cancer, and does it affect cancer cell polarity or migration?

  • Signaling pathway integration:

    • Investigate how STK32A interacts with established planar cell polarity pathways

    • Research question: Does STK32A function independently or as part of known polarity pathways?

  • Identification of substrates:

    • Beyond potentially regulating GPR156, what are the direct substrates of STK32A kinase activity?

    • Research question: What proteins are phosphorylated by STK32A and how does this modify their function?

  • Evolutionary conservation:

    • Examine the conservation of STK32A function across species

    • Research question: Is the role of STK32A in polarity evolutionarily conserved, and can simpler model organisms be used to study it?

These research directions would build upon the current understanding of STK32A while expanding its known functions beyond the specialized context of inner ear development .

What technological advancements would most benefit STK32A research and similar studies of poorly characterized kinases?

Several technological advancements would significantly accelerate research on STK32A and other understudied or "dark" kinases:

  • Improved antibody development techniques:

    • Current challenge: Researchers have struggled to identify antibodies with sufficient specificity for detecting endogenous STK32A

    • Needed advancement: High-throughput antibody validation methods including parallel testing against knockout tissues

    • Benefit: Reliable detection of endogenous protein across multiple applications

  • Structural biology approaches:

    • Current challenge: Limited structural information about STK32A

    • Needed advancement: Cryo-EM or X-ray crystallography studies of STK32A alone and in complex with substrates

    • Benefit: Rational design of specific inhibitors and better understanding of activation mechanisms

  • Single-cell analysis technologies:

    • Current challenge: Understanding cell-type specific expression and function

    • Needed advancement: Integration of single-cell transcriptomics, proteomics, and spatial analysis

    • Benefit: Comprehensive maps of STK32A expression and activity at cellular resolution

  • Chemical biology tools:

    • Current challenge: Lack of specific activators or inhibitors

    • Needed advancement: Development of selective small molecule modulators or degraders (PROTACs)

    • Benefit: Temporal control of STK32A activity in experimental systems

  • Advanced imaging techniques:

    • Current challenge: Visualizing low-abundance proteins in native contexts

    • Needed advancement: Super-resolution microscopy combined with signal amplification methods

    • Benefit: Better visualization of protein localization and dynamics

  • Genome engineering refinements:

    • Current challenge: Generating subtle mutations or tagged endogenous proteins

    • Needed advancement: More efficient knock-in technologies and conditional alleles

    • Benefit: Study of STK32A variants in physiologically relevant contexts

  • Kinome-wide profiling methods:

    • Current challenge: Understanding STK32A in the context of the broader kinome

    • Needed advancement: Comprehensive activity-based profiling across tissues and developmental stages

    • Benefit: Positioning STK32A within signaling networks

These technological advancements would address the current limitations in STK32A research, particularly the challenges in detecting endogenous protein and understanding its precise biochemical functions .

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.