The ST13 PAT5C6AT Antibody is a monoclonal antibody (mouse IgG2a subclass) generated using a recombinant human ST13 protein (amino acids 1–369) as immunogen . It is purified via protein-A affinity chromatography and formulated in PBS with sodium azide and glycerol . Key features include:
The antibody has been validated for ELISA and Western blot, with recommended starting dilutions of 1:1000 for WB . Specificity is assured through rigorous testing, though titration is advised for optimal results .
The ST13 PAT5C6AT Antibody is critical for studying ST13’s role as a tumor suppressor and its interactions with molecular chaperones:
Co-chaperone Role: Mediates interactions between HSP70 and HSP90, facilitating glucocorticoid receptor maturation .
Tumor Suppression: Downregulated in colorectal carcinoma (CRC), with reduced expression correlating with primary tumors vs. adjacent mucosa .
Expression Patterns: Predominantly expressed in colorectal epithelia and adenocarcinoma cells .
The antibody enables detection of ST13 in CRC tissues, aiding studies on:
The ST13 PAT5C6AT Antibody differs from others in clone specificity and host origin. Below is a comparison:
ST13 (Suppression of Tumorigenicity 13) is an adaptor protein or co-chaperone that mediates the association between HSP70 and HSP90 and appears in early receptor complexes. Through common binding to both Hsp70 and Hsp90, ST13 functions as an adaptor that can integrate Hsp70 and Hsp90 interactions. ST13 plays a significant role in the assembly process of the glucocorticoid receptor, requiring the assistance of multiple molecular chaperones to achieve proper functioning. The protein has gained research importance because its expression is downregulated in colorectal carcinoma tissue compared to adjacent normal tissue, suggesting its role as a candidate tumor suppressor gene. The expression levels of the ST13 gene are significantly decreased in primary tumors compared with adjacent mucosa, making it an important research target in cancer biology .
The ST13 PAT5C6AT antibody is a mouse anti-human monoclonal antibody derived from the hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human ST13 protein (amino acids 1-369) purified from E. coli. It belongs to the IgG2a isotype with kappa light chain. The antibody has been purified from mouse ascitic fluids using protein-A affinity chromatography, ensuring high purity and specificity. It primarily reacts with human samples, though some versions may also show reactivity with mouse and rat samples .
The ST13 PAT5C6AT antibody has been validated for multiple research applications including:
Western Blot (WB): Typically used at dilutions of 1:1000-1:6000
Enzyme-Linked Immunosorbent Assay (ELISA)
Immunofluorescence (IF)
Immunocytochemistry (ICC)
Immunohistochemistry (IHC): Recommended dilutions of 1:200-1:1000
Flow Cytometry
Immunoprecipitation (IP): Using 0.5-4.0 μg for 1.0-3.0 mg of total protein lysate
The antibody has been tested in various cell lines and tissue samples, including HEK-293 cells, human colon cancer tissue, and mouse testis tissue, demonstrating its versatility for different experimental contexts .
For optimal Western blot results with ST13 PAT5C6AT antibody, consider the following methodological approach:
Sample preparation: Use 30 μg of protein under reducing conditions. The antibody has been validated with various sample types including human cell lines (HeLa, A431, 293T, SK-OV-3), rat tissues (testis, C6 cells), and mouse samples (testis, NIH/3T3 cells).
Gel electrophoresis: Use a 5-20% SDS-PAGE gradient gel for optimal separation. Run at 70V for the stacking gel and 90V for the resolving gel, with a total run time of 2-3 hours.
Protein transfer: Transfer proteins to a nitrocellulose membrane at 150 mA for 50-90 minutes.
Blocking: Block the membrane with 5% non-fat milk in TBS for 1.5 hours at room temperature.
Primary antibody incubation: Use ST13 antibody at 0.25 μg/mL (approximately 1:1000-1:6000 dilution) and incubate overnight at 4°C.
Washing: Wash with TBS-0.1% Tween three times, 5 minutes each.
Secondary antibody: Incubate with goat anti-rabbit or anti-mouse IgG-HRP secondary antibody at a dilution of 1:5000 for 1.5 hours at room temperature.
Detection: Develop signal using an Enhanced Chemiluminescent detection kit.
The expected band size for ST13 is approximately 41 kDa, though it may appear between 45-54 kDa on some gels due to post-translational modifications .
ST13 expression shows significant tissue-specific patterns in cancer, particularly in colorectal cancer, where its downregulation has been well-documented. The ST13 protein is mostly expressed in colorectal epithelia and adenocarcinoma cells, with expression levels significantly decreased in primary tumors compared to adjacent normal mucosa. This downregulation pattern suggests ST13 functions as a tumor suppressor.
When designing cancer research studies utilizing the ST13 PAT5C6AT antibody, researchers should consider:
Tissue-specific expression: Include appropriate positive controls from tissues known to express ST13, such as normal colorectal epithelia.
Comparative analysis: Design experiments that compare expression levels between tumor and adjacent normal tissue to identify differential expression patterns.
Functional studies: Consider complementing expression studies with functional assays to investigate how ST13 downregulation affects HSP70/HSP90 chaperone function and the downstream cellular processes they regulate.
Correlation with clinical parameters: Analyze ST13 expression in relation to tumor grade, stage, and patient outcomes to determine its prognostic value.
This antibody has been validated for immunohistochemistry in human colon cancer tissue, making it suitable for translational cancer research applications .
When performing immunofluorescence with ST13 PAT5C6AT antibody, appropriate controls are essential for result validation. Three main control types should be included:
Positive controls:
Include a sample known to express ST13 (such as HeLa cells or colorectal epithelia)
Use an antibody against a housekeeping protein (like β-actin or GAPDH) in a parallel experiment to confirm sample quality
Endogenous controls to assess cell/tissue health:
Include markers to check for cell stress (phospho-histone H2AX for DNA damage)
Examine mitochondrial morphology for stress indicators
For tissue samples, check for signs of cell death using antibodies against cleaved caspase or PARP
Look for morphological changes like condensed nuclei or cell blebbing
Holes in tissue architecture may indicate improper storage or embedding of samples
Negative controls:
Secondary antibody-only control to assess non-specific background staining
PBS-only treatment to evaluate autofluorescence
Isotype control (using the same IgG subclass as the ST13 antibody but with no specific target)
These controls will help set appropriate maximum exposure and laser power settings to avoid non-specific signal and ensure confidence that the antibody is binding specifically to ST133 .
For optimal IHC detection of ST13 in tissue samples using the PAT5C6AT antibody, follow these methodological steps:
Sample preparation:
Use properly fixed tissue sections (typically formalin-fixed, paraffin-embedded)
For frozen sections, ensure proper tissue embedding and sectioning to maintain morphology
Antigen retrieval:
Primary recommendation: Use TE buffer at pH 9.0
Alternative method: Citrate buffer at pH 6.0
Heat-induced epitope retrieval methods are typically more effective than proteolytic methods for ST13
Blocking:
Block with 10% normal goat serum to reduce non-specific binding
Include additional blocking steps if tissue contains high levels of endogenous peroxidase
Antibody dilution and incubation:
Use ST13 PAT5C6AT antibody at 1:200-1:1000 dilution
Incubate overnight at 4°C for optimal results
For sensitive detection, consider using amplification systems such as biotin-streptavidin
Detection system:
Use appropriate secondary antibodies compatible with the host species (anti-mouse)
For colorimetric detection, DAB (3,3'-diaminobenzidine) provides good contrast
For fluorescent detection, consider using Cy3-conjugated secondary antibodies
Counterstaining and mounting:
Hematoxylin counterstain for brightfield microscopy
DAPI for fluorescence applications
Use appropriate mounting media based on detection method (aqueous for fluorescence)
Parallel controls:
Run known positive tissue (colon cancer tissue has been validated)
Include negative controls (no primary antibody, isotype control)
Remember that optimal dilutions may vary based on tissue type and fixation method, so preliminary titration experiments are recommended to determine the optimal antibody concentration for specific experimental conditions .
When working with ST13 PAT5C6AT antibody in Western blot applications, researchers may encounter several challenges:
Multiple bands or unexpected molecular weight:
Expected molecular weight of ST13 is 41 kDa, but it may appear between 45-54 kDa
Solution: Use positive control lysates (HEK-293, HeLa cells) with known ST13 expression
Consider running gradient gels (5-20% SDS-PAGE) for better resolution
Post-translational modifications may cause shifts in apparent molecular weight
Weak or no signal:
Increase antibody concentration (try 1:1000 instead of 1:6000)
Extend primary antibody incubation time (overnight at 4°C)
Ensure protein transfer efficiency with Ponceau S staining
Verify sample preparation methods preserve protein integrity
Check if sample buffer contains appropriate reducing agents
High background:
Increase blocking time or concentration (try 5% BSA instead of milk)
Add 0.1-0.3% Tween-20 to washing buffer
Decrease secondary antibody concentration
Ensure membrane is completely covered during incubation steps
Use freshly prepared buffers
Non-specific bands:
Increase wash duration and frequency after primary and secondary antibody incubations
Try alternative blocking reagents (BSA, casein, commercial blocking buffers)
Validate with knockout or knockdown samples if available
Consider pre-adsorption of antibody with recombinant protein
For optimal results, ST13 antibody has been successfully used with multiple human cell lines (HeLa, A431, 293T, SK-OV-3) and mouse/rat samples (testis tissues, NIH/3T3, C6 cells) at concentrations of 0.25 μg/mL, with overnight incubation at 4°C .
Distinguishing genuine ST13 signal from artifacts in immunofluorescence experiments requires careful experimental design and systematic controls:
Subcellular localization assessment:
ST13 as a co-chaperone typically shows cytoplasmic localization
Unusual localization patterns (such as exclusively nuclear) may indicate non-specific binding
Co-stain with markers of cellular compartments to confirm expected localization
Signal intensity evaluation:
Genuine ST13 staining should show dose-dependent response with antibody concentration
Compare staining patterns between different cell types with known varying ST13 expression levels
Excessive brightness throughout all cells regardless of expression level suggests non-specific binding
Pattern consistency:
Compare with published literature on ST13 localization
Artifact staining often appears as:
Even staining across all cell types regardless of expected expression differences
Staining restricted to cell boundaries or nuclei only
Punctate pattern that doesn't correlate with expected distribution
Control experiments:
Secondary antibody-only controls help identify background staining
Competitive blocking with recombinant ST13 protein should reduce specific signal
Knockdown validation using siRNA against ST13 should diminish signal proportionally
Peptide competition assays can confirm antibody specificity
Technical considerations:
Use appropriate fixation methods (4% paraformaldehyde works well for ST13)
Permeabilize cells adequately (0.1% Triton X-100 is typically effective)
Block with 10% normal goat serum to reduce non-specific binding
Use optimized antibody concentration (5 μg/mL has been validated)
Counter-stain nuclei with DAPI to aid in cellular orientation
For validated IF protocol, successful staining has been demonstrated in HeLa cells using enzyme antigen retrieval, followed by blocking with 10% goat serum, incubation with 5 μg/mL ST13 PAT5C6AT antibody overnight at 4°C, and detection with Cy3-conjugated secondary antibody3 .
The ST13 PAT5C6AT antibody can be effectively utilized to investigate protein-protein interactions within the HSP70/HSP90 chaperone complex through several methodological approaches:
Co-immunoprecipitation (Co-IP):
Use 0.5-4.0 μg of ST13 antibody per 1.0-3.0 mg of protein lysate
Perform pull-down experiments to identify binding partners of ST13
Follow with Western blot analysis to detect HSP70, HSP90, or other suspected interacting proteins
This approach can reveal how ST13 functions as an adaptor between HSP70 and HSP90
Proximity Ligation Assay (PLA):
Combine ST13 PAT5C6AT antibody with antibodies against potential interacting partners
This technique allows visualization of protein-protein interactions in situ with high sensitivity
Quantify interaction signals under different cellular conditions or treatments
Immunofluorescence co-localization:
Use dual immunofluorescence with ST13 PAT5C6AT antibody and antibodies against HSP70 or HSP90
Analyze co-localization coefficients using appropriate software
This approach works particularly well in HeLa cells where ST13 antibody has been validated
FRET analysis:
Combine immunofluorescence using ST13 PAT5C6AT antibody with fluorescently tagged HSP70/HSP90
Measure energy transfer to assess proximity of proteins in living cells
This technique provides spatial information about chaperone complex assembly
Chromatin immunoprecipitation (ChIP):
For studying ST13's role in glucocorticoid receptor complex formation and DNA binding
Use ST13 antibody to pull down protein-DNA complexes
Analyze whether ST13 is present at specific genomic loci during transcriptional regulation
These approaches can be particularly valuable for understanding how ST13 downregulation in cancer affects chaperone complex formation and function, potentially providing insights into novel therapeutic approaches targeting the HSP70/HSP90 chaperone system .
To accurately measure differential ST13 expression between normal and cancer tissues, researchers can employ several quantitative approaches using the ST13 PAT5C6AT antibody:
Western blot densitometry:
Perform Western blot using standardized amounts of protein from matched normal/tumor samples
Use 1:1000-1:6000 dilution of ST13 PAT5C6AT antibody
Quantify band intensity using densitometry software
Normalize to loading controls such as β-actin or GAPDH
This method provides semi-quantitative comparison of protein levels
Quantitative immunohistochemistry (IHC):
Perform IHC with ST13 PAT5C6AT antibody at 1:200-1:1000 dilution on tissue microarrays
Use digital pathology software to quantify:
Staining intensity (0-3+ scale)
Percentage of positive cells
H-score (intensity × percentage)
Allred score or other standardized scoring systems
Compare scores between normal epithelia and matched tumor samples
This approach preserves tissue architecture and allows spatial analysis
Flow cytometry:
Prepare single-cell suspensions from fresh tissue or cell lines
Fix with 4% paraformaldehyde and permeabilize cells
Stain with ST13 PAT5C6AT antibody (1 μg per 1×10^6 cells)
Use fluorophore-conjugated secondary antibody
Measure median fluorescence intensity (MFI) as a quantitative measure of expression
This method allows analysis of ST13 expression in specific cell populations
Multiplexed protein assays:
Develop custom protein arrays with ST13 capture antibody
Quantify using standard curves
This approach allows simultaneous measurement of multiple proteins
Useful for correlating ST13 levels with other chaperone proteins
Mass spectrometry-based proteomics:
Use ST13 antibody for immunoprecipitation followed by mass spectrometry
Allows absolute quantification of ST13 and interacting partners
Can detect post-translational modifications
When analyzing colorectal cancer samples, studies have consistently shown decreased ST13 expression in tumor tissue compared to adjacent normal mucosa. Quantitative analysis can help establish whether this downregulation correlates with clinical parameters such as tumor stage, grade, or patient outcomes .
Ensuring reproducible results when working with ST13 PAT5C6AT antibody requires attention to several critical factors:
Antibody validation and characterization:
Verify antibody specificity using positive and negative controls
Document the exact clone (PAT5C6AT), host species (mouse), and isotype (IgG2a)
Note the specific epitope recognized (human ST13 protein, amino acids 1-369)
Maintain detailed records of antibody lot numbers as performance may vary between batches
Standardized protocols:
Develop detailed standard operating procedures (SOPs) for each application
Document critical parameters like antibody dilutions (1:1000-1:6000 for WB, 1:200-1:1000 for IHC)
Maintain consistent incubation times and temperatures (overnight at 4°C for primary antibody)
Use the same detection systems across experiments
For WB, standardize protein loading amounts (30 μg recommended)
Sample handling and preparation:
Use consistent sample collection and processing methods
Standardize lysis buffers and protein extraction protocols
For tissue samples, maintain consistent fixation times and conditions
Document storage conditions and freeze-thaw cycles of antibody and samples
Quantification methods:
Use consistent image acquisition settings
Apply standardized quantification methods (densitometry for WB, scoring systems for IHC)
Include calibration standards where possible
Document software and analysis parameters used
Experimental controls:
Include positive controls (HEK-293 cells, HeLa cells, colon tissue)
Use negative controls (secondary antibody only, isotype controls)
Include loading controls for normalization in quantitative experiments
Consider using siRNA knockdown or CRISPR knockout samples as gold-standard controls
Data reporting:
Report complete experimental conditions in publications
Include representative images of controls
Share raw data when possible
Document any deviations from standard protocols
By addressing these factors systematically, researchers can significantly improve the reproducibility of experiments using the ST13 PAT5C6AT antibody across different laboratory settings and experimental conditions .
The ST13 PAT5C6AT antibody has potential applications in several emerging research areas that extend beyond its established role in colorectal cancer:
Cancer immunotherapy and chaperone biology:
ST13's role in the HSP70/HSP90 chaperone system may influence antigen presentation
Research could explore how ST13 expression affects response to checkpoint inhibitors
ST13 antibodies can help characterize chaperone-dependent processes in immune cells
Understanding ST13's role in tumor immune microenvironment may identify new therapeutic targets
Stress response pathways in neurodegenerative diseases:
The HSP70/HSP90 system plays a crucial role in protein folding and degradation
ST13 antibodies could help investigate chaperone dysfunction in Alzheimer's and Parkinson's diseases
Potential links between ST13 expression and protein aggregation could be explored
ST13's role in neuronal stress response might offer insights into disease mechanisms
Drug resistance mechanisms:
HSP90 inhibitors are being developed as cancer therapeutics
ST13 antibodies can help study how co-chaperone expression affects drug sensitivity
Expression patterns might predict response to HSP90-targeted therapies
Understanding the dynamic interactions in the chaperone complex may improve drug design
Hormone receptor signaling beyond glucocorticoid receptors:
ST13's established role in glucocorticoid receptor assembly suggests potential involvement with other nuclear receptors
ST13 antibodies can help investigate co-chaperone requirements for estrogen, androgen, or progesterone receptor function
This research may identify new therapeutic approaches for hormone-dependent cancers
Tissue-specific roles in development and differentiation:
ST13 expression varies across tissues, suggesting specialized functions
Developmental studies using ST13 antibodies could reveal stage-specific roles
Understanding tissue-specific co-chaperone requirements may explain differential sensitivity to proteotoxic stress
Systems biology approaches to chaperone networks:
ST13 antibodies enable protein-protein interaction studies
Mapping the dynamic chaperone interactome under different conditions
Integration with other -omics data to build comprehensive models of cellular stress response
The HSP70 Interacting Protein Clone PAT5C6AT, Mouse Anti Human, is a monoclonal antibody used in various research applications. This antibody targets the Heat Shock Protein 70 (HSP70), a molecular chaperone that plays a crucial role in protein homeostasis by assisting in the folding, trafficking, and degradation of proteins.
HSP70 is a highly conserved protein found in almost all living organisms, from bacteria to humans. It is a part of the heat shock protein family, which is upregulated in response to stress conditions such as heat, toxins, and infection. HSP70 helps in the proper folding of nascent proteins, refolding of misfolded proteins, and prevention of protein aggregation. It also assists in the degradation of damaged proteins by delivering them to the proteasome .
Monoclonal antibodies are laboratory-produced molecules engineered to serve as substitute antibodies that can restore, enhance, or mimic the immune system’s attack on cells. They are designed to bind to specific targets, in this case, the HSP70 protein. The Clone PAT5C6AT is a mouse-derived monoclonal antibody that specifically targets human HSP70.
The HSP70 Interacting Protein Clone PAT5C6AT is used in various research applications, including:
Understanding the interactions and functions of HSP70 is crucial for studying various diseases, including cancer, neurodegenerative disorders, and infections. HSP70’s role in protein homeostasis makes it a potential target for therapeutic interventions. The Clone PAT5C6AT antibody aids researchers in exploring these interactions and developing new treatments.