STAC antibodies are designed to detect and analyze STAC proteins, including STAC1 and STAC3, which regulate excitation-contraction coupling in skeletal muscle and calcium channel activity . These proteins interact with voltage-gated calcium channels (e.g., Ca<sub>v</sub>1.1, Ca<sub>v</sub>1.2) and ryanodine receptors (RyR1), facilitating calcium release during muscle contraction . Dysregulation of STAC proteins is linked to neuromuscular pathologies, making these antibodies vital for mechanistic and therapeutic studies .
Host: Rabbit
Reactivity: Human, Mouse, Rat
STAC3 is essential for skeletal muscle contraction, as demonstrated by knockout models where its absence caused paralysis and neonatal lethality . Structural studies reveal:
Domains: C1 domain (residues 93–150) binds Ca<sub>v</sub>1.2, while the SH3 domain mediates interactions with RyR1 .
Key Interaction: STAC3’s tandem SH3 domains bind the Ca<sub>v</sub>1.1 II-III loop (residues 745–765), enabling excitation-contraction coupling .
Muscular Dystrophy: STAC3 dysfunction disrupts calcium release, contributing to muscle degeneration .
Prion Disorders: STAC-BBB, a zinc finger-based delivery platform, shows potential for repressing tau mRNA in neurodegenerative diseases .
Antibody | Reactivity | Applications | Dilution |
---|---|---|---|
CAB15319 | Human, Mouse, Rat | WB, ELISA | 1:200–1:2000 |
Clone 2C5 | Human | WB, ELISA, IF | Not specified |
Neurological Delivery: STAC-BBB leverages AAV vectors for blood-brain barrier penetration, enabling neuron-specific gene repression (e.g., tau in Alzheimer’s) .
Muscle Disorders: Antibodies like CAB15319 aid in detecting STAC3 expression levels in myopathy models, guiding therapeutic development .
Ongoing research focuses on:
STAC (SH3 and cysteine-rich domain-containing protein) is a signaling adapter protein that has gained significant attention in research due to its involvement in multiple cellular pathways. The protein contains characteristic SH3 domains that mediate protein-protein interactions and is expressed in various tissues. In research settings, STAC antibodies are utilized to investigate the protein's role in signaling cascades, tissue-specific expression patterns, and potential implications in disease mechanisms. Studies have particularly focused on its expression in human tissues and its potential relevance to cellular signaling pathways .
STAC antibodies are available in multiple formats with varying specifications to accommodate different experimental needs. These include:
Antibody Type | Host Options | Applications | Features |
---|---|---|---|
Polyclonal | Rabbit | WB, ELISA, IF | Broader epitope recognition, unconjugated format available |
Monoclonal | Mouse | WB, ELISA | Clone-specific (e.g., 2C5), higher specificity to single epitope |
Researchers can select from STAC antibodies with different species reactivity profiles, including human-reactive variants, depending on their experimental model systems. These antibodies have been validated for various applications such as Western blotting (WB), enzyme-linked immunosorbent assay (ELISA), and immunofluorescence (IF) .
Selecting the appropriate STAC antibody requires methodical consideration of multiple factors:
Determine your target species (human, mouse, etc.) and ensure antibody reactivity matches this species
Identify your experimental application (WB, ELISA, IF, ICC) and select an antibody validated for that technique
Consider your detection system requirements and whether you need a conjugated or unconjugated antibody
Evaluate whether your research question requires the broader epitope recognition of polyclonal antibodies or the specificity of monoclonal antibodies
For cross-species studies, verify the conservation of the epitope sequence across species. Where possible, review published literature citing the use of specific STAC antibody clones in your application of interest to gauge expected performance .
For optimal Western blotting with STAC antibodies, follow these methodological guidelines:
Sample preparation: Extract proteins using a buffer containing appropriate protease inhibitors to prevent degradation of STAC proteins, which can affect epitope recognition
Gel separation: Use 10-12% SDS-PAGE gels for optimal resolution of STAC (predicted molecular weight ~40-45 kDa)
Transfer conditions: Cold transfer at 100V for 1 hour or 30V overnight to nitrocellulose or PVDF membranes
Blocking: 5% non-fat dry milk or BSA in TBST for 1 hour at room temperature
Primary antibody incubation: Dilute STAC antibody (typically 1:500-1:2000 for polyclonal and 1:1000-1:5000 for monoclonal) in blocking buffer and incubate overnight at 4°C
Detection: Use appropriate HRP-conjugated secondary antibodies and enhanced chemiluminescence detection systems
Include positive and negative controls in each experiment, such as lysates from cells known to express or not express STAC, respectively. This approach ensures reliable and reproducible detection of STAC proteins in your samples .
For immunofluorescence applications with STAC antibodies, implement the following protocol adaptations:
Cell/tissue preparation: Fix samples with 4% paraformaldehyde for 15 minutes, followed by permeabilization with 0.1% Triton X-100 for 5-10 minutes
Blocking: Use 1-3% BSA or 5-10% normal serum from the species of the secondary antibody in PBS for 30-60 minutes
Primary antibody: Incubate with diluted STAC antibody (typically 1:100-1:500 for polyclonal antibodies validated for IF) at 4°C overnight or 1-2 hours at room temperature
Secondary antibody: Use fluorophore-conjugated secondary antibodies at manufacturer-recommended dilutions (typically 1:200-1:1000)
Counterstaining: Include DAPI or other nuclear counterstains for proper localization assessment
Mounting: Use anti-fade mounting medium to preserve fluorescence signal
Include appropriate controls, such as secondary-only controls to assess background fluorescence and positive controls using tissues known to express STAC. Consider performing co-localization studies with markers of subcellular compartments to accurately determine STAC protein localization within cells .
Rigorous experimental design with STAC antibodies requires comprehensive controls:
Positive controls: Include samples known to express STAC protein (based on literature or verified expression)
Negative controls: Use samples known not to express STAC protein or samples where STAC has been knocked down/out
Technical controls:
Primary antibody omission: To assess non-specific binding of secondary antibody
Isotype controls: Use non-specific IgG of the same isotype and concentration as the STAC antibody
Peptide competition: Pre-incubate STAC antibody with excess immunizing peptide to demonstrate binding specificity
Validation controls:
Multiple antibody verification: Use multiple antibodies targeting different epitopes of STAC
Orthogonal methods: Confirm protein expression using complementary techniques (e.g., mass spectrometry, RNA-seq)
These controls help differentiate specific signal from background noise and validate the specificity of observed signals. For advanced applications, consider including genetic knockdown/knockout models or using cell lines transfected with STAC expression constructs as additional controls .
Validating STAC antibody specificity requires a multi-faceted approach:
Genetic validation: Use STAC knockout or knockdown models to confirm absence of signal when the target is removed
Overexpression validation: Transfect cells with STAC expression vectors and confirm increased signal intensity
Peptide competition assays: Pre-incubate antibody with the immunizing peptide to block specific binding
Cross-reactivity testing: Test antibody against samples containing related proteins to assess potential cross-reactivity
Molecular weight verification: Confirm that the detected band corresponds to the predicted molecular weight of STAC
Multiple antibody comparison: Compare results using antibodies recognizing different epitopes of STAC
Literature comparison: Verify that your observed expression patterns match published data on STAC distribution
Document all validation steps meticulously, including experimental conditions, controls, and any limitations identified. This comprehensive validation approach ensures scientific rigor and reproducibility in STAC antibody-based research .
Determining the optimal working dilution for STAC antibodies requires systematic titration:
Initial range finding: Test a broad range of dilutions based on manufacturer recommendations (typically 1:100 to 1:5000)
Narrow range titration: Once an approximate range is identified, test 3-5 dilutions within that range
Signal-to-noise assessment: For each dilution, calculate the ratio of specific signal to background
Sensitivity assessment: Determine the minimum amount of protein that can be reliably detected at each dilution
Dynamic range evaluation: Assess the linear range of detection at different antibody concentrations
For Western blotting, prepare a dilution series of your sample and test each antibody dilution. For immunostaining, prepare multiple identical sections/wells and test different antibody concentrations. Document the conditions that provide optimal signal-to-noise ratio while conserving antibody. Remember that optimal dilutions may vary between different lots of the same antibody and different experimental conditions .
Interpreting validation data for commercial STAC antibodies requires critical evaluation:
Validation method assessment: Examine what validation methods were used (Western blot, immunoprecipitation, immunofluorescence, etc.)
Control evaluation: Assess what positive and negative controls were employed in the validation
Specificity demonstration: Look for evidence of specificity testing such as knockout/knockdown validation or peptide competition assays
Cross-reactivity testing: Check if the antibody was tested against related proteins to rule out cross-reactivity
Application-specific validation: Ensure the antibody was validated for your specific application
Full blot/image disclosure: Look for complete blots or images rather than cropped versions
Reproducibility evidence: Check if validation was performed multiple times and under different conditions
Be cautious of validation data showing only the band/signal of interest without appropriate controls or full blots/images. Peer-reviewed publications citing the specific antibody for your application provide additional validation evidence. Remember that even well-validated antibodies may perform differently in your specific experimental system .
Common Western blotting issues with STAC antibodies and their resolutions include:
Issue | Possible Causes | Solutions |
---|---|---|
No signal | Insufficient protein, degraded antibody, incorrect dilution | Increase protein loading, verify antibody activity, optimize dilution |
Multiple bands | Cross-reactivity, protein degradation, non-specific binding | Use alternative STAC antibody, add protease inhibitors, increase blocking |
High background | Insufficient blocking, excessive antibody, suboptimal wash | Increase blocking time/concentration, dilute antibody further, extend washes |
Incorrect molecular weight | Post-translational modifications, isoforms, non-specific binding | Verify with alternative antibody, use denaturing conditions, confirm with IP |
Inconsistent results | Antibody batch variation, protocol inconsistencies | Standardize protocols, aliquot antibodies, use internal controls |
For persistent issues, consider alternative lysis buffers that may better preserve the STAC protein structure and epitopes. If multiple bands are observed, perform mass spectrometry analysis to identify the proteins in each band. Always include positive control samples known to express STAC protein to benchmark antibody performance across experiments .
Distinguishing true STAC signal from non-specific binding requires methodical verification:
Molecular weight verification: Confirm that the primary band appears at the expected molecular weight for STAC (~40-45 kDa)
Control samples: Compare signals between samples known to express or not express STAC
Signal patterns: True signals should show consistent patterns across biological replicates
Knockdown/knockout validation: Reduction/elimination of signal in STAC-depleted samples confirms specificity
Competition assays: Pre-incubation with immunizing peptide should eliminate specific signals but not non-specific binding
Multiple antibodies: Use antibodies targeting different STAC epitopes to confirm signal consistency
Alternative techniques: Confirm STAC expression using orthogonal methods (qPCR, mass spectrometry)
For immunostaining applications, include absorption controls where the antibody is pre-incubated with the immunizing peptide. The true STAC signal should disappear in absorption controls while non-specific binding remains. Always interpret results in the context of known STAC expression patterns from literature .
When faced with contradictory results between different STAC antibodies, follow this systematic approach:
Epitope comparison: Determine if the antibodies recognize different epitopes that might be differentially accessible
Validation assessment: Evaluate the validation evidence for each antibody (knockout validation, specificity testing)
Application optimization: Verify that each antibody has been optimized for your specific application
Isoform consideration: Assess whether different antibodies might recognize different STAC isoforms
Post-translational modifications: Consider whether modifications might affect epitope recognition in different contexts
Contextual factors: Evaluate cell/tissue type, fixation methods, and sample preparation differences
Technical replication: Perform side-by-side experiments controlling all variables except the antibody
To resolve contradictions, employ orthogonal methods such as mass spectrometry or RNA-seq to independently verify STAC expression patterns. Consider using genetic approaches (CRISPR/Cas9, RNAi) to manipulate STAC expression and determine which antibody most accurately reflects these changes. Document all contradictions carefully as they may reveal novel insights about STAC protein structure, processing, or function .
Integrating STAC antibodies in SIRT1-Activating Compounds research creates unique opportunities for mechanistic investigation:
Pathway analysis: Use STAC antibodies to monitor protein expression changes in response to SIRT1-activating compounds (STACs)
Target validation: Confirm whether STACs directly interact with STAC proteins using immunoprecipitation followed by mass spectrometry
Localization studies: Track changes in STAC protein subcellular localization upon SIRT1 activation using immunofluorescence
Pharmacodynamic markers: Employ STAC antibodies to develop biomarkers for STAC compound activity
Combination therapy research: Investigate how modulation of STAC protein expression affects cellular response to STACs
Research has shown that SIRT1-activating compounds inhibit pancreatic cancer cell growth and survival both in vitro and in vivo. STAC antibodies can be used to investigate whether these effects involve changes in STAC protein expression or function. This approach could reveal potential synergies between STAC-targeted therapies and SIRT1 activation strategies .
Advanced multiplexing with STAC antibodies enables complex protein interaction studies:
Multi-color immunofluorescence: Combine STAC antibodies with antibodies against interaction partners or pathway components using spectrally distinct fluorophores
CODEX/Cyclic immunofluorescence: Perform iterative staining and imaging cycles to analyze dozens of proteins including STAC in the same sample
Mass cytometry (CyTOF): Label STAC antibodies with rare earth metals for high-dimensional analysis without spectral overlap
Proximity ligation assays (PLA): Detect and visualize protein-protein interactions involving STAC proteins with single-molecule sensitivity
Simultaneous multiplexed immunoblotting: Use antibodies with distinct species origins to simultaneously probe for STAC and other proteins
Spectral unmixing techniques: Employ computational approaches to separate overlapping fluorescent signals in multicolor imaging
These techniques allow researchers to simultaneously investigate STAC protein expression alongside binding partners, signaling mediators, and cellular markers. For quantitative multiplexing, include appropriate controls for each antibody and standardize signal intensity using reference standards or housekeeping proteins .
Integrating STAC antibody data with Next-Generation Sequencing enables multi-omics insights:
RNA-protein correlation: Compare STAC protein levels (measured via antibodies) with STAC mRNA expression (from RNA-seq) to identify post-transcriptional regulation
Epitope validation: Use NGS data to verify conservation of antibody epitopes across species or between related protein family members
Isoform-specific detection: Design experiments combining isoform-specific antibodies with RNA-seq splice variant analysis
Regulatory network analysis: Correlate STAC protein expression with transcription factor binding (from ChIP-seq) to elucidate regulatory mechanisms
Single-cell multi-omics: Combine single-cell antibody-based detection with scRNA-seq for simultaneous protein and transcript analysis
Spatial transcriptomics integration: Align antibody-based spatial protein maps with spatially resolved transcriptomics data
This integrated approach requires careful experimental design, including:
Collecting matched samples for both antibody-based detection and sequencing
Normalizing data appropriately across platforms
Applying computational methods that can integrate protein and nucleic acid data
NGS data can help cluster and annotate sequences, showing diversity and region length plots that inform antibody selection and validation. This combined approach provides deeper understanding of STAC protein biology than either method alone .
Recent antibody engineering advances with implications for STAC research include:
Controllable antibody technologies: New methods enable antibodies to be turned "off" and "on" with temporal and spatial control using covalently tethered blocking constructs with protease-cleavable or photocleavable moieties
Nanobodies and single-domain antibodies: Smaller antibody fragments with superior tissue penetration for detecting STAC in complex tissues
Recombinant antibody production: Consistent, defined antibodies with reduced batch-to-batch variation
Site-specific conjugation: Precisely controlled attachment of labels or functional groups without compromising binding activity
Bispecific antibodies: Single molecules that can simultaneously bind STAC and another target for colocalization studies
Intrabodies: Antibodies engineered to function within cells for live-cell STAC protein tracking
Antibody fragments with enhanced tissue penetration: Fab, scFv, and other formats for improved tissue access
These technologies can address traditional limitations of STAC antibodies, such as batch variation, background signal, and limited accessibility to certain cellular compartments. For example, controllable antibody technologies could allow precise temporal activation of STAC antibodies during complex experimental procedures .
Optimizing STAC antibody protocols for difficult samples requires systematic adaptation:
Fixation optimization:
For formalin-fixed tissues: Test different antigen retrieval methods (heat-induced vs. enzymatic)
For frozen sections: Compare acetone, methanol, and paraformaldehyde fixation
For highly autofluorescent tissues: Consider Sudan Black B treatment or spectral unmixing
Permeabilization refinement:
Adjust Triton X-100 concentration (0.1-0.5%) or substitute with saponin (0.01-0.1%)
Test differential permeabilization with digitonin for selective membrane permeabilization
Consider dual detergent approaches (SDS followed by Triton X-100) for heavily cross-linked samples
Signal enhancement strategies:
Implement tyramide signal amplification for low abundance STAC detection
Use polymer detection systems instead of traditional secondary antibodies
Consider biotin-streptavidin amplification systems for enhanced sensitivity
Background reduction techniques:
Pre-absorb antibodies with tissue powder from the species being analyzed
Include species-specific blocking reagents matching the host of tissue samples
Employ longer/additional washing steps with increased detergent concentration
Document optimization steps methodically, changing only one variable at a time. For particularly challenging samples, consider consulting published protocols specific to your tissue type or fixation method that have successfully used antibodies against targets of similar abundance and cellular localization as STAC .
For high-throughput screening with STAC antibodies, consider these methodological requirements:
Assay miniaturization and automation:
Optimize antibody concentrations for microplate formats (96, 384, or 1536-well)
Establish automated liquid handling protocols with attention to consistent antibody dispensing
Validate performance across plate positions to identify edge effects
Quality control measures:
Include positive and negative controls on every plate
Calculate Z-factor scores to assess assay quality (aim for Z' > 0.5)
Implement regular antibody performance checks using reference standards
Data normalization strategies:
Determine appropriate normalization methods (percent of control, Z-score, etc.)
Account for plate-to-plate variation using standard curves
Consider data correction for systematic biases (row/column effects)
Reagent stability planning:
Evaluate antibody stability under screening conditions (time, temperature)
Prepare sufficient single-batch antibody aliquots for entire screening campaign
Determine maximum bench time before sensitivity loss
Validation cascades:
Design orthogonal secondary assays to confirm primary hits
Establish dose-response testing for confirmed hits
Develop counter-screens to identify false positives
Implement appropriate statistical methods for hit identification, including multiple testing correction. Consider using machine learning approaches for complex phenotypic data analysis when screening with imaging-based STAC antibody assays .
STAC antibodies enable mechanistic investigation of disease processes through multiple approaches:
Expression profiling in disease states:
Compare STAC protein levels between healthy and diseased tissues using immunohistochemistry
Perform quantitative analysis of STAC expression across disease progression stages
Correlate STAC protein levels with clinical outcomes or treatment responses
Protein interaction studies:
Use co-immunoprecipitation with STAC antibodies to identify altered protein interactions in disease
Employ proximity ligation assays to visualize disrupted or enhanced interactions in situ
Combine with mass spectrometry to identify novel interaction partners in disease contexts
Post-translational modification analysis:
Utilize modification-specific antibodies alongside total STAC antibodies to assess regulatory changes
Map phosphorylation, ubiquitination, or other modifications altered in disease states
Correlate modifications with altered STAC function or localization
Functional studies in disease models:
Track STAC protein dynamics during disease development using antibody-based imaging
Assess effects of disease-relevant stimuli on STAC expression and localization
Evaluate therapeutic interventions' impact on STAC expression or function
Research has shown that SIRT1-activating compounds inhibit pancreatic cancer cell growth and tumor development in vivo through mechanisms involving SIRT1 and lysosomes. STAC antibodies can help elucidate whether these effects involve direct interactions with STAC proteins or indirect pathway modulation, potentially revealing new therapeutic targets or biomarkers for disease progression and treatment response .
Next-generation antibody sequencing technologies are transforming STAC antibody research:
Repertoire sequencing advantages:
High-throughput analysis of millions of antibody sequences enables identification of optimal STAC-binding clones
Deep sequence analysis can reveal antibody maturation pathways and guide rational design
Paired heavy/light chain sequencing improves recombinant antibody production quality
Methodological applications:
NGS data analysis of antibody sequences allows clustering based on complementarity-determining regions (CDRs)
Sequence fingerprinting helps identify optimal antibody candidates with desired specificity profiles
Computational filtering based on framework and CDR sequences predicts cross-reactivity potential
Technical implementations:
Filtering and grouping sequences according to specific requirements optimizes antibody selection
Automated validation of sequences based on predefined rules improves quality control
Cluster diversity and region length plots inform epitope coverage strategies
Practical research benefits:
Accelerated discovery of high-affinity, specific STAC antibodies through computational screening
Enhanced reproducibility through sequence-defined recombinant antibodies
Improved antibody engineering through comprehensive understanding of sequence-function relationships
These technologies enable researchers to analyze antibody sequences at unprecedented scale and depth, facilitating more precise selection of antibodies for specific research applications and reducing reliance on traditional hybridoma approaches that may yield less consistent results .
Artificial intelligence is revolutionizing STAC antibody research through multiple applications:
Experimental design optimization:
Predictive models for optimal antibody dilutions and incubation conditions
Automated protocol optimization based on experimental parameters and target characteristics
Design of efficient validation experiments with minimal resource requirements
Image analysis enhancements:
Automated quantification of STAC protein expression in complex tissues
Deep learning algorithms for subcellular localization pattern recognition
Unbiased identification of co-localization patterns with other markers
Data interpretation frameworks:
Pattern recognition across large datasets to identify subtle expression changes
Integration of antibody-based data with other -omics datasets
Prediction of functional implications based on expression patterns
Quality control implementations:
Automated detection of non-specific binding or background issues
Consistency checking across experimental replicates
Objective assessment of staining quality and specificity
Literature mining capabilities:
Synthesis of published STAC protein data to inform experimental design
Identification of contradictions or inconsistencies across publications
Extraction of relevant methodological details from published protocols
Machine learning approaches can identify trends in large-scale antibody datasets that might not be apparent through conventional analysis, potentially revealing novel insights about STAC protein biology and facilitating more efficient experimental design. These methods are particularly valuable for complex multiplexed experiments where traditional analysis methods become unwieldy .
Researchers can advance STAC antibody validation standards through these practical approaches:
Implementation of rigorous validation protocols:
Employ genetic models (knockout/knockdown) to definitively establish antibody specificity
Use orthogonal detection methods to confirm STAC expression patterns
Document validation data comprehensively and share through publications or repositories
Transparent reporting practices:
Include complete details of antibody sources, catalog numbers, and lot numbers
Provide full unedited blot images and controls in publications
Describe all validation steps performed, including negative results
Community resource development:
Contribute validation data to antibody validation databases
Share optimized protocols through protocol repositories
Report antibody performance issues to manufacturers and the scientific community
Methodological standardization efforts:
Adopt consensus guidelines for antibody validation
Participate in multi-laboratory validation initiatives
Implement standardized reporting formats for antibody-based experiments
Educational initiatives:
Train junior researchers in proper antibody validation techniques
Advocate for inclusion of validation standards in journal submission requirements
Develop resources explaining the importance of antibody validation