Validated across multiple experimental modalities with characterized sensitivity:
Western Blot: Detects endogenous phospho-SMAD2 at 52-65 kDa depending on phosphorylation state
Immunoprecipitation: Successfully IPs SMAD2 from PMA-treated HeLa lysates
Immunofluorescence: Localizes to nucleus/cytoplasm in TGF-β-activated cells
Phosphorylation at Ser250 occurs in the linker region of SMAD2, modulating:
Transcriptional activity: Enhances complex formation with SMAD4
Subcellular trafficking: Promotes nuclear translocation upon TGF-β stimulation
Protein stability: Regulates ubiquitin-mediated degradation via SMURF2 interaction
Post-translational modifications at adjacent residues (Ser245/Ser255) create phosphorylation clusters detectable by multi-phospho-specific clones .
The phospho-SMAD2 (S250) recombinant monoclonal antibody is meticulously crafted using cutting-edge techniques in protein and DNA recombinant technology. The process commences with the immunization of animals using a synthetic peptide derived from human phospho-SMAD2 (S250). This immunization elicits the production of B cells, from which positive clones are carefully selected and identified. The genes encoding the phospho-SMAD2 (S250) antibody are subsequently amplified through PCR and inserted into a plasmid vector, resulting in a recombinant vector. This recombinant vector is then transfected into host cells to facilitate the expression of the phospho-SMAD2 (S250) antibody. Finally, the phospho-SMAD2 (S250) recombinant monoclonal antibody is purified from the cell culture supernatant using affinity chromatography. This antibody serves as a reliable tool for precise and accurate detection of human phospho-SMAD2 (S250) protein in ELISA and WB applications.
Phospho-SMAD2 (S250) is a receptor-regulated SMAD (R-SMAD) that acts as an intracellular signal transducer and transcriptional modulator, activated by TGF-beta (transforming growth factor) and activin type 1 receptor kinases. It binds to the TRE element within the promoter region of numerous genes regulated by TGF-beta. Upon formation of the SMAD2/SMAD4 complex, it activates transcription. This protein may function as a tumor suppressor in colorectal carcinoma. Notably, it positively regulates PDPK1 kinase activity by promoting its dissociation from the 14-3-3 protein YWHAQ, which acts as a negative regulator.
SMAD2 phosphorylation at serine 250 represents one of several key regulatory modifications within the TGF-β signaling pathway. SMAD2 functions as a receptor-regulated SMAD (R-SMAD) that acts as an intracellular signal transducer and transcriptional modulator activated by TGF-beta and activin type 1 receptor kinases . The phosphorylation at S250 occurs in the linker region of SMAD2, distinct from the canonical C-terminal phosphorylation at S465/467 that occurs directly through TGF-β receptor activation. Linker region phosphorylation, including at S250, represents an integration point for crosstalk between TGF-β and other signaling pathways, allowing for context-dependent modulation of SMAD2's transcriptional activities. This phosphorylation plays critical roles in regulating cell proliferation, differentiation, and apoptotic responses .
SMAD2 belongs to the SMAD family of proteins, evolutionarily conserved from the Drosophila gene 'mothers against decapentaplegic' (Mad) and C. elegans gene Sma . Structurally, SMAD2 contains conserved MH1 and MH2 domains connected by a less conserved linker region where S250 resides. Upon TGF-β receptor activation, SMAD2 undergoes C-terminal phosphorylation, which induces dissociation from the SMAD anchor for receptor activation (SARA) protein and subsequent association with SMAD4 . This SMAD2/SMAD4 complex translocates to the nucleus, binds the TRE element in promoter regions of TGF-β-regulated genes, and activates transcription . The linker region phosphorylation at sites including S250 provides additional regulatory control, often mediated by non-TGF-β pathways, creating a sophisticated mechanism for signal integration and contextual responses to TGF-β.
SMAD2 S250 phosphorylation influences multiple cellular processes within the broader context of TGF-β signaling, including:
Cell proliferation regulation: Phosphorylation at S250 can modulate the growth inhibitory effects of TGF-β signaling in various cell types
Cellular differentiation: SMAD2 promotes TGF-β1-mediated transcription of differentiation genes, with S250 phosphorylation potentially regulating this activity
Apoptotic responses: The phosphorylation status influences cell survival signals
Cancer progression: SMAD2 may act as a tumor suppressor in colorectal carcinoma, with its phosphorylation status potentially affecting this function
PDPK1 kinase activity: Phosphorylated SMAD2 positively regulates PDPK1 by stimulating its dissociation from the inhibitory 14-3-3 protein YWHAQ
For optimal Western blot detection of phospho-SMAD2 (S250), researchers should follow these methodological guidelines:
Sample preparation: Use whole cell lysates from appropriate cell lines (HeLa, HepG2, NIH/3T3, C6) with or without treatment with PMA (0.2μM for 0.5h) as a positive control
Protein loading: Load 20-30μg of protein per lane for standard detection
Blocking conditions: Use 5% non-fat dry milk in TBST as blocking and diluting buffer
Primary antibody dilution: Apply the phospho-SMAD2 (S250) antibody at dilutions between 1:500-1:5000 depending on the specific product and sensitivity required
Detection considerations: Be aware that phospho-SMAD2 (S250) typically appears as a band at approximately 60-65 kDa, though the calculated molecular weight is 52 kDa
Potential degradation: Note that bands around and below the 50-kDa marker could represent degradation fragments
Secondary antibody: Use an HRP-conjugated anti-rabbit secondary antibody at approximately 1:100,000 dilution
Validating antibody specificity for phospho-SMAD2 (S250) is crucial for experimental reliability. A comprehensive validation approach should include:
Peptide competition assays: Use phosphorylated and non-phosphorylated peptides to demonstrate binding specificity. Test against peptides containing different phosphorylation sites (S245, S250, S255) and combinations thereof to establish site-specificity
Dot blot analysis: Apply a systematic dot blot with various phospho-peptides:
Signal induction: Compare signals between untreated cells and cells treated with PMA (0.2μM for 0.5h), which induces phosphorylation
Immunoprecipitation: Perform IP followed by Western blot to confirm specificity
Knockout/knockdown controls: Include SMAD2 knockdown or knockout samples as negative controls
For optimal immunoprecipitation of phospho-SMAD2 (S250), follow these methodological guidelines:
Sample preparation: Start with 0.35-0.5 mg of whole cell lysate from cells treated with phosphorylation inducers (e.g., 0.2μM PMA for 0.5h)
Antibody amount: Use the phospho-SMAD2 (S250) antibody at approximately 1/30 dilution (about 2μg antibody per 0.35mg lysate)
Immunoprecipitation procedure:
Incubate lysate with antibody overnight at 4°C
Add protein A/G beads and incubate for 2-4 hours
Wash beads thoroughly to remove non-specific binding
Western blot detection: Perform Western blotting on immunoprecipitated material using the same phospho-SMAD2 (S250) antibody at 1/1000 dilution
Secondary antibody: Use a specialized IP detection reagent (like VeriBlot for IP Detection Reagent) at 1/5000 dilution to avoid heavy and light chain interference
Controls: Include a parallel IP with isotype control antibody (e.g., rabbit monoclonal IgG) to identify non-specific binding
The discrepancy between the calculated molecular weight (52 kDa) and observed molecular weight (60-65 kDa) of phospho-SMAD2 is a common observation that requires careful interpretation:
Post-translational modifications: Multiple phosphorylation sites beyond S250 (including canonical C-terminal sites S465/467 and other linker region sites) can contribute to reduced electrophoretic mobility, causing the protein to migrate more slowly than predicted
Glycosylation or other modifications: Additional post-translational modifications may further alter migration patterns
Isoform considerations: Alternative splicing variants of SMAD2 exist and may display different migration patterns
Technical factors affecting migration:
Buffer composition and pH
Acrylamide percentage in SDS-PAGE gels
Running conditions
Verification strategies:
Use total SMAD2 antibody alongside phospho-specific antibody to confirm identity
Include phosphatase-treated samples as controls to verify phosphorylation-dependent migration shift
Consider isoform-specific primers if investigating at mRNA level
The consistent observation of the 60-65 kDa band across multiple independent studies strongly suggests this represents the authentic phosphorylated form of SMAD2 .
Inconsistent detection of phospho-SMAD2 (S250) can result from several experimental and biological factors:
Cell type and context dependence:
Different cell lines may exhibit varying baseline and inducible S250 phosphorylation levels
Growth conditions and cell density can affect phosphorylation status
Cell cycle phase may influence phosphorylation patterns
Sample processing issues:
Rapid phosphatase activity during lysate preparation can dephosphorylate target residues
Inadequate phosphatase inhibitors in lysis buffers
Protein degradation during sample preparation or storage
Freeze-thaw cycles degrading phospho-epitopes
Technical considerations:
Biological stimulus timing:
Troubleshooting approaches:
Phospho-SMAD2 (S250) antibodies provide powerful tools for investigating signaling crosstalk through these methodological approaches:
Dual pathway stimulation experiments:
Treat cells with TGF-β in combination with activators of other pathways (MAPK, PI3K/AKT, Wnt)
Analyze S250 phosphorylation status alongside canonical C-terminal phosphorylation (S465/467)
Compare temporal dynamics of different phosphorylation events
Pathway inhibitor studies:
Apply specific inhibitors of non-TGF-β pathways and measure effects on S250 phosphorylation
Use pharmacological inhibitors in combination with genetic approaches (siRNA, CRISPR)
Create dose-response and time-course matrices to map pathway interactions
Nuclear-cytoplasmic fractionation:
Chromatin immunoprecipitation (ChIP):
Use phospho-S250 antibodies for ChIP to identify genomic targets specifically regulated by this modification
Compare with targets of canonically phosphorylated SMAD2
Perform sequential ChIP to examine co-occupancy with other transcription factors
Proteomics approaches:
Conduct immunoprecipitation with phospho-S250 antibodies followed by mass spectrometry
Identify differential interaction partners compared to non-phosphorylated or C-terminally phosphorylated SMAD2
Apply phosphoproteomics to map global signaling changes following pathway perturbations
Studying SMAD2 S250 phosphorylation has significant implications for both cancer and developmental biology research:
Cancer research applications:
TGF-β pathway exhibits context-dependent tumor suppressor or promoter functions
S250 phosphorylation potentially serves as a biomarker for cancer progression or treatment response
Linker phosphorylation may mediate resistance to TGF-β-induced growth inhibition in cancer cells
Targeting enzymes responsible for S250 phosphorylation could represent therapeutic strategies
Developmental biology implications:
SMAD2 promotes TGF-β1-mediated transcription of differentiation genes
S250 phosphorylation may regulate context-specific responses during embryonic development
The balance between different SMAD2 phosphorylation states may direct cell fate decisions
Temporal dynamics of phosphorylation could coordinate developmental timing
Methodological approaches for both fields:
Tissue-specific expression analysis using immunohistochemistry with phospho-S250 antibodies
In vivo models with phosphorylation site mutations (S250A/S250E)
Single-cell analyses to examine heterogeneity in phosphorylation patterns
Correlation of phosphorylation status with differentiation markers or tumor progression indicators
Studying the dynamic interplay between different SMAD2 phosphorylation sites requires sophisticated methodological approaches:
Multiplex phosphorylation analysis:
Use antibodies specific for individual phosphorylation sites (S245, S250, S255, S465/467)
Apply multiplexed Western blotting with different fluorescent secondary antibodies
Develop ELISA-based assays to quantitatively measure multiple phosphorylation sites
Consider phospho-flow cytometry for single-cell resolution of multiple phosphorylation events
Time-course experiments:
Apply stimulus (TGF-β, PMA, etc.) and collect samples at multiple time points
Compare temporal patterns of different phosphorylation events
Use mathematical modeling to infer causality between phosphorylation events
Mutagenesis approaches:
Generate phospho-mimetic (S→D/E) and phospho-null (S→A) mutations at different sites
Create combination mutants to study interdependence of phosphorylation events
Analyze functional consequences of mutations on SMAD2 localization, protein interactions, and transcriptional activity
Mass spectrometry-based approaches:
Apply phosphoproteomics to quantitatively assess multiple phosphorylation sites simultaneously
Use SILAC or TMT labeling for comparative analysis across conditions
Perform targeted mass spectrometry for enhanced sensitivity to specific phosphopeptides
Advanced imaging techniques:
Develop and apply phosphorylation-specific FRET biosensors
Use phospho-specific antibodies for super-resolution microscopy
Apply live cell imaging to track phosphorylation dynamics in real-time
Each experimental technique offers distinct advantages and limitations for phospho-SMAD2 (S250) detection:
When selecting experimental systems for phospho-SMAD2 (S250) studies, researchers should consider:
Baseline expression and phosphorylation levels:
Response to stimulation:
Experimental system characteristics:
TGF-β pathway status (receptor expression, pathway mutations)
Context of other relevant signaling pathways
Growth characteristics and culture requirements
Transfection/transduction efficiency if genetic manipulation is required
Technical considerations:
Disease relevance:
For cancer studies, consider cell lines that reflect the cancer type of interest
For developmental studies, consider models that recapitulate relevant differentiation processes
Patient-derived samples may provide greater clinical relevance but introduce heterogeneity
When selecting a phospho-SMAD2 (S250) antibody, researchers should consider these key technical parameters: