GSDMA is a pore-forming protein expressed predominantly in epithelial tissues, such as the skin and gastrointestinal tract . It plays dual roles in:
Pyroptosis induction: Cleaved by proteases (e.g., SpeB from Streptococcus pyogenes) to release its N-terminal domain, which forms membrane pores and triggers inflammatory cell death .
Tissue homeostasis: Regulates epithelial maintenance and mucin secretion in the colon .
GSDMA antibodies are validated for multiple experimental applications:
Overexpression in Psoriatic Skin: GSDMA expression is significantly elevated in psoriatic plaques compared to healthy skin, correlating with keratinocyte hyperproliferation and inflammation .
Biomarker Potential: Serum GSDMA levels are higher in psoriasis patients, though urinary GSDMA/creatinine ratios show no diagnostic utility .
Prognostic Marker: Overexpression of GSDMA is linked to tumor progression in ovarian, lung adenocarcinoma, and clear-cell renal cell carcinoma (ccRCC) .
Immune Microenvironment: GSDMA expression positively correlates with immune cell infiltration (e.g., macrophages, dendritic cells) in tumors .
Antimicrobial Defense: GSDMA-NT pores induced by S. pyogenes SpeB protease trigger pyroptosis, limiting bacterial invasion .
Sample Preparation: Use RIPA buffer for tissue lysates; heat-mediated antigen retrieval (pH 9.0 Tris-EDTA) for IHC .
Blocking: 5% non-fat dry milk/TBST reduces background noise in WB .
Controls: Include secondary antibody-only and non-transfected cell lysates to validate specificity .
CSB-PA850415LA01HU is an IgG rabbit polyclonal GSDMA antibody designed for the detection of human GSDMA through ELISA, Western Blotting, Immunohistochemistry, and Immunofluorescence techniques. The antibody is generated through immunization of rabbits with recombinant human GSDMA protein (amino acids 64-172). This GSDMA antibody is provided in the non-conjugated IgG form and exhibits a purity exceeding 95%, achieved through protein G purification.
GSDMA protein acts as a regulator of cell death and inflammation. Predominantly expressed in epithelial tissues, GSDMA plays a crucial role in regulating immune responses and maintaining tissue homeostasis. It is also involved in various physiological processes, including skin development and the preservation of skin barrier function. Dysregulation of GSDMA can lead to the development of skin disorders, inflammatory bowel disease, cancer, and other diseases.
GSDMA (gasdermin A) is a protein encoded by the GSDMA gene in humans. It may also be known by alternative names including FKSG9, GSDM, GSDM1, and gasdermin-1. The protein has a molecular weight of approximately 49.4 kilodaltons . GSDMA belongs to the gasdermin family, which plays crucial roles in regulated cell death pathways and inflammatory responses. Research into GSDMA is particularly important for understanding epithelial maintenance and homeostasis, as demonstrated by studies showing its expression in specific epithelial tissues . The protein's functional significance makes it a target of interest in research related to cell death mechanisms, inflammation, and epithelial biology.
When selecting a GSDMA antibody, consider these methodological factors:
Epitope recognition: Determine whether you need an antibody targeting N-terminal, C-terminal, or internal epitopes based on your experimental questions.
Validated applications: Verify the antibody has been validated for your specific application (WB, ELISA, IF, IHC, etc.) .
Species reactivity: Confirm cross-reactivity with your species of interest. Many antibodies react with human GSDMA, while others may cross-react with mouse, rat, or other species .
Clonality: Monoclonal antibodies offer high specificity for a single epitope, while polyclonal antibodies recognize multiple epitopes and may provide stronger signals.
Conjugation: Determine if you need unconjugated antibodies or those conjugated with fluorescent dyes (FITC), enzymes, or other tags based on your detection method .
Validation data: Review published literature or supplier validation data demonstrating specificity and performance in applications similar to yours.
Distinguishing between gasdermin family members requires careful antibody selection and experimental controls:
Sequence alignment analysis: Before selecting antibodies, perform sequence alignment of GSDMA with other family members (GSDMB, GSDMC, GSDMD, GSDME) to identify unique regions.
Epitope specificity: Select antibodies raised against unique regions with minimal homology to other gasdermin family members.
Knockout/knockdown controls: Include GSDMA knockout or knockdown samples to confirm specificity.
Cross-reactivity testing: Test the antibody against recombinant proteins of multiple gasdermin family members.
Multiple antibody approach: Use antibodies recognizing different epitopes of GSDMA to confirm observations.
Western blotting discrimination: Utilize the molecular weight differences between gasdermin family members (GSDMA is 49.4 kDa) as an additional specificity check .
For optimal detection of GSDMA in tissues, consider these methodological approaches:
Immunohistochemistry (IHC):
Use paraffin-embedded sections (IHC-p) with heat-induced epitope retrieval (HIER) in citrate buffer (pH 6.0)
Apply primary GSDMA antibody at 1:100-1:500 dilution (optimize for your specific antibody)
Detect using appropriate secondary antibody systems (HRP/DAB or fluorescent)
Include positive controls (epithelial tissues) and negative controls (antibody omission)
Immunofluorescence (IF):
RNAscope® in situ hybridization: For GSDMA mRNA detection when protein expression is low or antibody specificity is questionable
Western blotting: For quantitative comparison of expression levels across samples
To effectively investigate GSDMA's role in cell death pathways:
Overexpression studies:
Knockout/knockdown approaches:
Domain-specific investigations:
Create constructs expressing only N-terminal or C-terminal domains
Evaluate pore-forming activity of N-terminal fragments
Study regulatory mechanisms involving C-terminal domains
Cell death assays:
LDH release assay for membrane integrity
Caspase activation assays
TUNEL assay for DNA fragmentation
Propidium iodide/Annexin V staining for flow cytometry
Live-cell imaging with membrane-impermeable dyes
Activation mechanisms:
Test potential activating stimuli (inflammatory triggers, specific proteases)
Use protease inhibitors to block cleavage
Perform co-immunoprecipitation to identify interaction partners
For quantitative Western blotting of GSDMA:
Sample preparation:
Lyse cells or tissues in RIPA buffer supplemented with protease inhibitors
Homogenize tissues thoroughly (using mechanical or ultrasonic methods)
Clarify lysates by centrifugation (12,000-15,000 × g, 15 min, 4°C)
Determine protein concentration (BCA or Bradford assay)
Gel electrophoresis and transfer:
Immunoblotting protocol:
Block membranes in 5% non-fat milk or BSA for 1 hour at room temperature
Incubate with primary GSDMA antibody (1:500-1:2000 dilution) overnight at 4°C
Wash 3× with TBST (10 minutes each)
Incubate with appropriate HRP-conjugated secondary antibody (1:5000-1:10000) for 1 hour
Wash 3× with TBST (10 minutes each)
Develop using enhanced chemiluminescence (ECL) reagents
Quantification approach:
Include housekeeping protein controls (β-actin, GAPDH)
Use a range of sample amounts to ensure linearity of signal
Capture images with dynamic range-appropriate systems
Perform densitometric analysis using ImageJ or similar software
Express GSDMA signal relative to loading control
Understanding the differences between mouse and human GSDMA is critical for translational research:
Detecting GSDMA activation and cleavage presents several methodological challenges:
Antibody epitope considerations:
Choose antibodies recognizing either full-length GSDMA, N-terminal fragments, or C-terminal fragments
Use antibody pairs recognizing different regions to confirm cleavage events
Consider raising custom antibodies against cleavage sites or neo-epitopes
Temporal dynamics:
Cleavage may be rapid and products transient
Design time-course experiments with appropriate sampling intervals
Use protease inhibitors or stabilizing agents to capture transient intermediates
Subcellular localization:
N-terminal fragments may localize to membranes while C-terminal fragments remain cytosolic
Use subcellular fractionation techniques to isolate different cellular compartments
Complement biochemical approaches with imaging techniques
Technical approaches:
Use gradient gels (4-20%) to resolve both high and low molecular weight species
Consider native PAGE for intact complexes
Implement immunoprecipitation followed by mass spectrometry to identify precise cleavage sites
Use proximity ligation assays to detect protein-protein interactions related to activation
Controls and standards:
Generate recombinant GSDMA cleavage products as positive controls
Include known activating conditions/stimuli as positive controls
Use mutants resistant to cleavage as negative controls
To investigate GSDMA's role in inflammatory pathways:
Cell culture models:
Select appropriate cell types (epithelial cells, keratinocytes)
Apply inflammatory stimuli (cytokines, TLR ligands, inflammasome activators)
Measure GSDMA expression, localization, and cleavage
Assess inflammatory outcomes (cytokine release, inflammasome activation)
Gain/loss-of-function approaches:
Overexpress wild-type or mutant GSDMA
Create knockout or knockdown systems
Compare inflammatory response magnitudes between modified and control cells
Measure downstream inflammatory markers (IL-1β, IL-18, NF-κB activation)
Signaling pathway dissection:
Use specific pathway inhibitors to identify regulatory connections
Perform co-immunoprecipitation to identify interaction partners
Conduct phospho-proteomic analysis to identify post-translational modifications
Use reporter assays (NF-κB, ISRE) to measure transcriptional responses
In vivo inflammation models:
Human patient samples:
Analyze GSDMA expression in inflammatory disease tissues
Correlate expression with disease severity and inflammatory markers
Use single-cell approaches to identify cell-type specific effects
To improve GSDMA antibody specificity and sensitivity:
Validation approaches:
Optimizing signal-to-noise ratio:
Titrate antibody concentration (test serial dilutions)
Optimize blocking conditions (5% BSA may reduce background compared to milk for phospho-epitopes)
Try longer but more dilute antibody incubations (overnight at 4°C)
Increase washing stringency (add 0.1-0.5% SDS or increase salt concentration)
Antigen retrieval optimization for IHC/IF:
Compare different buffers (citrate pH 6.0 vs. EDTA pH 9.0)
Test different retrieval times and temperatures
Consider alternative fixation methods if using freshly prepared samples
Cross-reactivity management:
Pre-absorb antibody with recombinant proteins of related gasdermin family members
Use alternative antibodies targeting different epitopes for confirmation
Consider monoclonal antibodies for highest specificity
Signal amplification strategies:
Implement tyramide signal amplification for low-abundance detection
Use polymer-based detection systems rather than traditional ABC methods
Consider more sensitive substrates (SuperSignal West Femto vs. standard ECL)
When facing contradictory GSDMA expression data:
Methodological reconciliation:
Compare detection methods (qPCR vs. Western blot vs. IHC)
Assess antibody target regions (different epitopes may be accessible in different contexts)
Consider post-translational modifications affecting epitope recognition
Evaluate protein extraction methods (some may yield better recovery)
Biological context considerations:
Technical validation strategies:
Use multiple antibodies targeting different epitopes
Implement orthogonal detection methods (RNA-seq, proteomics)
Perform subcellular fractionation to identify compartmentalization
Consider absolute quantification approaches with recombinant protein standards
Experimental design improvements:
Include positive and negative controls in all experiments
Test multiple antibody lots
Standardize sample collection and processing
Blind analysis to prevent bias
Reporting recommendations:
Document detailed methods (including antibody catalog numbers, dilutions, exposure times)
Report all experimental conditions that might affect results
Consider sharing raw data in repositories
When encountering unexpected bands with GSDMA antibodies:
Common explanations for higher molecular weight bands:
Post-translational modifications (phosphorylation, ubiquitination, SUMOylation)
Protein complexes resistant to SDS denaturation
Dimerization or oligomerization
Cross-reactivity with related proteins
Incomplete reduction of disulfide bonds
Common explanations for lower molecular weight bands:
Proteolytic cleavage (physiological or artifactual during sample preparation)
Alternative splice variants
Degradation products
Non-specific antibody binding
Internal translation initiation sites
Investigative approaches:
Vary sample preparation conditions (different buffers, protease inhibitors)
Test different reducing agents or concentrations
Perform peptide competition assays to determine specificity
Use mass spectrometry to identify unexpected bands
Compare patterns across different tissues/cell types
Validation experiments:
Use GSDMA knockout/knockdown samples as negative controls
Overexpress tagged GSDMA and detect with both anti-tag and anti-GSDMA antibodies
Test multiple antibodies targeting different GSDMA epitopes
Perform immunoprecipitation followed by Western blotting or mass spectrometry
To investigate GSDMA in tissue-specific pathologies:
Tissue expression profiling:
Perform IHC on tissue microarrays covering multiple normal and diseased tissues
Quantify GSDMA expression levels and correlate with pathological features
Combine with markers of cell death, inflammation, or tissue-specific differentiation
Compare expression patterns of human GSDMA with mouse orthologs
Disease-specific methodology:
Skin disorders: Analyze GSDMA expression in different layers of skin, correlate with differentiation markers
Gastrointestinal pathologies: Examine expression in different cell types of the GI tract epithelium
Respiratory conditions: Assess airway epithelial cells for GSDMA expression changes
Cancer studies: Compare GSDMA expression in tumor vs. adjacent normal tissue
Functional studies in tissue contexts:
Use organoid cultures to study GSDMA in 3D tissue architecture
Implement ex vivo tissue explant cultures
Consider tissue-specific conditional knockout animals
Use tissue-specific promoters for transgenic expression
Clinical correlation approaches:
Correlate GSDMA expression with clinical parameters and outcomes
Perform longitudinal studies using sequential biopsies
Integrate with multi-omics data from the same tissues
Consider genetic variation in GSDMA in relation to disease phenotypes
Advanced techniques for studying GSDMA pore formation include:
Membrane integrity assays:
Propidium iodide uptake in live cells
LDH release assays
Calcein release from liposomes
SYTOX dye uptake kinetics
Live cell imaging with membrane-impermeant fluorescent dyes
Structural biology approaches:
Cryo-electron microscopy of GSDMA pores in membranes
Atomic force microscopy to visualize pore architecture
X-ray crystallography of GSDMA fragments
NMR spectroscopy for dynamic structural analysis
Biophysical characterization:
Patch-clamp electrophysiology to measure pore conductance
Black lipid membrane conductance measurements
Surface plasmon resonance for membrane binding kinetics
Fluorescence recovery after photobleaching (FRAP) for membrane dynamics
Reconstitution systems:
Liposome-based pore formation assays
Cell-free expression systems with artificial membranes
Nanodiscs containing purified GSDMA
Giant unilamellar vesicles (GUVs) with fluorescent markers
Advanced microscopy techniques:
Super-resolution microscopy (STORM, PALM, SIM)
Correlative light and electron microscopy (CLEM)
Single-molecule tracking of labeled GSDMA
FRET-based assays to detect conformational changes
To investigate interactions between GSDMA and other gasdermin family members:
Co-expression analysis:
Perform multiplexed immunofluorescence for multiple gasdermin family members
Analyze single-cell RNA-seq data for co-expression patterns
Compare expression in normal vs. pathological conditions
Study developmental regulation of different family members
Functional redundancy assessment:
Interaction studies:
Conduct co-immunoprecipitation experiments between family members
Perform proximity ligation assays to detect in situ interactions
Use FRET/BRET approaches to detect direct interactions
Implement mammalian two-hybrid systems for interaction mapping
Domain swap experiments:
Create chimeric constructs swapping domains between family members
Test functional consequences of domain exchanges
Identify critical regions for unique vs. shared functions
Assess evolutionary conservation of functional domains
Systems biology approaches:
Perform network analysis of gasdermin signaling pathways
Use mathematical modeling to predict combined effects
Implement CRISPR screens targeting multiple family members
Conduct proteome-wide interaction screens