Phospho-SMAD2 (S250) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Research Applications and Performance

Validated across multiple experimental modalities with characterized sensitivity:

Key Applications

  • Western Blot: Detects endogenous phospho-SMAD2 at 52-65 kDa depending on phosphorylation state

    • Optimal dilutions: 1:500–1:5000 (varies by clone)

  • Immunoprecipitation: Successfully IPs SMAD2 from PMA-treated HeLa lysates

  • Immunofluorescence: Localizes to nucleus/cytoplasm in TGF-β-activated cells

Validation Data

Cell ModelTreatmentObserved Band SizeSignal Specificity Confirmed BySource
HeLaPMA (0.2 μM)65 kDaAlkaline phosphatase sensitivity test
NIH/3T3PMA (200 nM)58 kDaKnockout cell line comparison
Human colon tissueNoneN/A (IHC)Phosphatase pretreatment controls

Biological Context and Functional Insights

Phosphorylation at Ser250 occurs in the linker region of SMAD2, modulating:

  1. Transcriptional activity: Enhances complex formation with SMAD4

  2. Subcellular trafficking: Promotes nuclear translocation upon TGF-β stimulation

  3. 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 .

Technical Considerations

  • Buffer compatibility: PBS with 0.05% BSA and 50% glycerol recommended for long-term storage

  • Cross-reactivity: Some clones (e.g., CABP1338) detect multiple phospho-sites (Ser245/250/255)

  • Critical controls:

    • Alkaline phosphatase treatment for phosphorylation specificity

    • SMAD2 knockout cell lines (e.g., ab255430)

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide, and 50% glycerol.
Description

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.

Form
Liquid
Lead Time
Typically, we can dispatch the products within 1-3 working days after receiving your orders. The delivery time may vary depending on the chosen purchasing method or location. Please consult your local distributors for specific delivery time information.
Synonyms
Drosophila, homolog of, MADR2 antibody; hMAD-2 antibody; HsMAD2 antibody; JV18 antibody; JV18-1 antibody; JV181 antibody; MAD antibody; MAD homolog 2 antibody; MAD Related Protein 2 antibody; Mad-related protein 2 antibody; MADH2 antibody; MADR2 antibody; MGC22139 antibody; MGC34440 antibody; Mother against DPP homolog 2 antibody; Mothers against decapentaplegic homolog 2 antibody; Mothers against decapentaplegic, Drosophila, homolog of, 2 antibody; Mothers against DPP homolog 2 antibody; OTTHUMP00000163489 antibody; Sma and Mad related protein 2 antibody; Sma- and Mad-related protein 2 MAD antibody; SMAD 2 antibody; SMAD family member 2 antibody; SMAD, mothers against DPP homolog 2 antibody; SMAD2 antibody; SMAD2_HUMAN antibody
Target Names
Uniprot No.

Target Background

Function

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.

Gene References Into Functions
  1. Overall, these findings suggest a more dominant role for SMAD3 and SMAD4 than SMAD2 in TGFbeta-induced chondrogenesis of human bone marrow-derived mesenchymal stem cells. PMID: 28240243
  2. the results of the present study indicated that miR4865p was upregulated in Osteoarthritis and may inhibit chondrocyte proliferation and migration by suppressing SMAD2. PMID: 29749497
  3. relevance of the discovered Sirt1-Smad2 interaction for the regulation of TGFbeta-dependent gene transcription PMID: 29187201
  4. Our present study indicated that S100A11 promotes EMT through accumulation of TGF-beta1 expression, and TGF-beta1-induced upregulation of p-SMAD2 and 3. PMID: 29569474
  5. the results of the present study indicated that miR2145p may promote the adipogenic differentiation of BMSCs through regulation of the TGFbeta/Smad2/COL4A1 signaling pathway, and potentially may be used to develop a novel drug for postmenopausal osteoporosis. PMID: 29532880
  6. High SMAD2 expression is associated with fibrosis in chronic pancreatitis and pancreatic cancer. PMID: 29328490
  7. The results suggest that co-expression of active SMAD2/3 could enhance multiple types of transcription factors (TF)-based cell identity conversion and therefore be a powerful tool for cellular engineering. PMID: 29174331
  8. We found that ITZ treatment was efficient in suppressing EMT and that the effect of ITZ was partially mediated by impaired TGF-b/SMAD2/3 signaling. The role of TGF-b/SMAD2/3 signaling in mediating the effect of ITZ was confirmed based on the results that recombinant TGF-b induced, but the TGF-b neutralizing antibody inhibited EMT as well as the invasion and migration of pancreatic cancer cells PMID: 29484419
  9. SMAD2/3 interactome reveals that TGFbeta controls m(6)A mRNA methylation in pluripotency PMID: 29489750
  10. This study's findings provide new insights into the mechanisms of how oscillatory shear stress regulates Smad2 signaling and pro-inflammatory genes through the complex signaling networks of integrins, transforming growth factor-beta receptors, and extracellular matrices, thus illustrating the molecular basis of regional pro-inflammatory activation within disturbed flow regions in the arterial tree. PMID: 29295709
  11. our findings demonstrated that thymoquinone suppressed the metastatic phenotype and reversed EMT of prostate cancer cells by negatively regulating the TGF-beta/Smad2/3 signaling pathway. These findings suggest that thymoquinone is a potential therapeutic agent against prostate cancer which functions by targeting TGF-beta PMID: 29039572
  12. MicroRNA-486-5p suppresses TGFB2-induced proliferation, invasion and epithelial-mesenchymal transition of lens epithelial cells by targeting Smad2. PMID: 29229876
  13. was found that treatment with iPSC-CM markedly reduced the proliferation of TGF-beta1-exposed cells, and the activities of TGF-beta1, Smad-2 and Smad-3. Accompanied by alterations in the expression of the indicated molecules, the lung structure of mice with PF was also markedly ameliorated. PMID: 29115383
  14. We found expression of pSmad2/3 and Smad4 in different liver tissues, with up-regulated expression of both antibodies in chronic hepatitis C with higher stage of fibrosis and higher grade of activity. PMID: 29924446
  15. TGFbeta and IL1beta signaling interact at the SMAD2/3 level in human primary MSC. Down-stream TGFbeta target genes were repressed by IL1beta independent of C-terminal SMAD2 phosphorylation. We demonstrate that SMAD2/3 linker modifications are required for this interplay and identified TAK1 as a crucial mediator of IL1beta-induced TGFbeta signal modulation. PMID: 28943409
  16. Our studies provide a molecular mechanism by which UCHL5 mitigates TGFbeta-1 signaling by stabilizing Smad2/Smad3. These data indicate that UCHL5 may contribute to the pathogenesis of idiopathic pulmonary fibrosis and may be a potential therapeutic target. PMID: 27604640
  17. we demonstrated that the downregulation of CLDN6 is regulated through promoter methylation by DNMT1, which depends on the SMAD2 pathway, and that CLDN6 is a key regulator in the SMAD2/DNMT1/CLDN6 pathway to inhibit EMT, migration and invasion of breast cancer cells PMID: 28867761
  18. High Expression of Smad2 is associated with liver cancer. PMID: 28415588
  19. Whereas autocrine signalling activates Smad2/3 in differentiating extravillous trophoblasts, paracrine factors contribute to Smad phosphorylation in these cells. PMID: 28864007
  20. Kidney samples from patients with advanced stages of diabetic nephropathy showed elevated pSmad2 staining. PMID: 28805484
  21. Smad2 (and myostatin) were significantly up-regulated in the failing heart of female patients, but not male patients. PMID: 28465115
  22. Nodal signaling through the Smad2/3 pathway up-regulated Slug, Snail and c-Myc to induce EMT, thereby promotingVasculogenic mimicry (VM) formation. PMID: 27659524
  23. this study shows that EGF induces epithelial-mesenchymal transition through phospho-Smad2/3-Snail signaling pathway in breast cancer cells PMID: 27829223
  24. Multiple myeloma cells adapted to long-term exposure to hypoxia exhibit stem cell characteristics with TGF-beta/Smad pathway activation. PMID: 29309790
  25. a novel heterozygous missense mutation (c.833C>T, p.A278V) in the SMAD2 gene in a family with early onset aortic aneurysms PMID: 28283438
  26. Data suggest that oncogenic Y-box binding protein 1 (YB-1) indirectly enhances transforming growth factor beta (TGFbeta) signaling cascades via Sma/Mad related protein 2 (Smad2)phospho-activation and may represent a promising factor for future diagnosis and therapy of breast cancer. PMID: 29187452
  27. Asiaticoside hindered the invasive growth of KFs by inhibiting the GDF-9/MAPK/Smad pathway. PMID: 28346732
  28. High Smad2 expression is associated with invasion and metastasis in pancreatic ductal adenocarcinoma. PMID: 26908446
  29. Data indicate that miR-206 inhibits neuropilin-1 (NRP1) and SMAD2 gene expression by directly binding to their 3'-UTRs. PMID: 27014911
  30. Results show that members of the Activin branch of the TGFbeta signaling pathway, namely Put and Smad2, are autonomously required for cell and tissue growth in the Drosophila larval salivary gland. PMID: 28123053
  31. CytoD modified MKL1, a coactivator of serum response factor (SRF) regulating CTGF induction, and promoted its nuclear localization. PMID: 27721022
  32. cells expressing mutant huntingtin have a dysregulated transcriptional response to epidermal growth factor stimulation PMID: 27988204
  33. CRT regulates TGF-beta1-induced-EMT through modulating Smad signaling PMID: 28778674
  34. P311 is a novel TGFbeta1/Smad signaling-mediated regulator of transdifferentiation in epidermal stem cells during cutaneous wound healing. PMID: 27906099
  35. human epidermal growth factor receptor 2 (HER-2) levels, were correlated well with TSP50/p-Samd2/3 and TSP50/p27 expression status. Thus, our studies revealed a novel regulatory mechanism underlying TSP50-induced cell proliferation and provided a new favorable intervention target for the treatment of breast cancer PMID: 28650473
  36. IL-17 can induce A549 alveolar epithelial cells to undergo epithelial-mesenchymal transition via the TGF-beta1 mediated Smad2/3 and ERK1/2 activation PMID: 28873461
  37. a critical role for miR-503-3p in induction of breast cancer EMT PMID: 28161325
  38. Nuclear localization of Smad2 was reduced in TGFbeta-1-stimulated primary tubular epithelial cells. Changes in nuclear Smad2 correlated with a reduced expression of the pro-fibrotic factor CTGF. Transient downregulation of Smad2 interfered with TGFbeta-1-induced CTGF synthesis. PMID: 27155083
  39. Low SMAD2 expression is associated with progression of hepatic fibrosis. PMID: 28423499
  40. In order to study the translation between mouse model and patients, we evaluated the signature of phosphorylated Sma- and Mad-related protein 2 (pSmad2), as molecular marker of TGF-beta/activin activity, in the kidneys of streptozotocin (STZ)-treated mice compared to that of type 1 diabetes (T1D) patients. PMID: 28064277
  41. SMAD2/SMAD3 signaling by bone morphogenetic proteins causes disproportionate induction of HAS2 expression and hyaluronan production in immortalized human granulosa cells. PMID: 26992562
  42. miR-27a contributed to cell proliferation and invasion by inhibiting TGF-beta-induced cell cycle arrest. These results suggest that miR-27a may function as an oncogene by regulating SMAD2 and SMAD4 in lung cancer. PMID: 28370334
  43. cPLA2alpha activates PI3K/AKT and inhibits Smad2/3 during epithelial-mesenchymal transition of hepatocellular carcinoma cells. PMID: 28649002
  44. selective inhibition of SMAD3 or CCT6A efficiently suppresses TGF-beta-mediated metastasis. Findings provide a mechanism that directs TGF-beta signaling toward its prometastatic arm and may contribute to the development of therapeutic strategies targeting TGF-beta for non-small-cell lung carcinoma. PMID: 28375158
  45. In response to TGF-beta, RASSF1A is recruited to TGF-beta receptor I and targeted for degradation by the co-recruited E3 ubiquitin ligase ITCH. RASSF1A degradation is necessary to permit Hippo pathway effector YAP1 association with SMADs and subsequent nuclear translocation of receptor-activated SMAD2. PMID: 27292796
  46. Smad2 is a key scaffold, allowing RIN1 to act as a GTP exchange factor for MFN2-GTPase activation to promote mitochondrial ATP synthesis and suppress superoxide production during mitochondrial fusion. PMID: 27184078
  47. Ang down-regulate the expression of Col-I, alpha-SMA and TGF-beta1/Smad2/3 and subsequently inhibits fibroblast-myofibroblast transition. PMID: 27543459
  48. Our findings suggest a stronger chondrogenic potential of CD105(+) SMSCs in comparison to that of CD105(-) SMSCs and that CD105 enhances chondrogenesis of SMSCs by regulating TGF-beta/Smad2 signaling pathway, but not Smad1/5. Our study provides a better understanding of CD105 with respect to chondrogenic differentiation. PMID: 27107692
  49. the findings show that TIEG1 is highly expressed in human keloids and that it directly binds and represses Smad7 promoter-mediated activation of TGF-beta/Smad2 signaling PMID: 28108300
  50. High expression of SMAD2 is associated with colorectal carcinoma. PMID: 27959430

Show More

Hide All

Database Links

HGNC: 6768

OMIM: 601366

KEGG: hsa:4087

STRING: 9606.ENSP00000262160

UniGene: Hs.12253

Protein Families
Dwarfin/SMAD family
Subcellular Location
Cytoplasm. Nucleus.
Tissue Specificity
Expressed at high levels in skeletal muscle, endothelial cells, heart and placenta.

Q&A

What is the biological significance of SMAD2 phosphorylation at serine 250?

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 .

How does the structure and function of SMAD2 relate to its phosphorylation status?

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.

What are the major cellular processes and pathways influenced by SMAD2 S250 phosphorylation?

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

What are the optimal conditions for detecting phospho-SMAD2 (S250) via Western blotting?

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

How can researchers validate the specificity of phospho-SMAD2 (S250) antibody detection?

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:

    • Smad2 (phospho S245+S250+S255) peptide

    • Smad2 (phospho S245+S250) peptide

    • Smad2 (phospho S245+S255) peptide

    • Smad2 (phospho S250+S255) peptide

    • Individual phosphorylation sites (S245, S250, S255)

    • Non-phosphorylated peptide as negative control

  • 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

What immunoprecipitation protocols yield the best results with phospho-SMAD2 (S250) antibodies?

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

How should researchers interpret discrepancies between calculated (52 kDa) and observed (60-65 kDa) molecular weights for phospho-SMAD2?

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 .

What factors might contribute to inconsistent phospho-SMAD2 (S250) detection in experimental samples?

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:

    • Antibody dilution optimization requirements

    • Blocking buffer compatibility issues (5% NFDM/TBST recommended)

    • Signal-to-noise ratio challenges with different detection systems

    • Batch-to-batch antibody variation

  • Biological stimulus timing:

    • Transient nature of many phosphorylation events

    • Optimal time points after stimulus application may vary (0.5h PMA treatment used in validation studies)

  • Troubleshooting approaches:

    • Include known positive controls (e.g., 293T+PMA, NIH/3T3+PMA)

    • Test multiple lysis buffer formulations with varying phosphatase inhibitor compositions

    • Optimize fixation methods for ICC/IF applications

    • Perform time-course experiments to identify peak phosphorylation

How can phospho-SMAD2 (S250) antibodies be utilized to investigate crosstalk between TGF-β and other signaling pathways?

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:

    • Given SMAD2's nucleocytoplasmic shuttling , compare subcellular distribution of phospho-S250 SMAD2

    • Correlate localization patterns with transcriptional outcomes

    • Apply techniques like proximity ligation assay to detect interactions between phospho-SMAD2 and other signaling components

  • 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

What are the implications of studying S250 phosphorylation in cancer and developmental biology research?

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

    • SMAD2 may act as a tumor suppressor in colorectal carcinoma

    • 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

How can researchers effectively study the dynamics of SMAD2 S250 phosphorylation in relation to other phosphorylation sites?

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

How does the detection of phospho-SMAD2 (S250) compare across different experimental techniques (WB, ELISA, IP, ICC/IF)?

Each experimental technique offers distinct advantages and limitations for phospho-SMAD2 (S250) detection:

TechniqueSensitivitySpecificityQuantificationSpatial ResolutionKey Methodological Considerations
Western BlotHighHighSemi-quantitativeNone- Band observed at 60-65 kDa (not predicted 52 kDa)
- Recommended dilutions: 1:500-1:5000
- 5% NFDM/TBST blocking buffer optimal
ELISAVery HighHighHighly quantitativeNone- Useful for high-throughput screening
- Can detect lower abundance modifications
- Requires careful validation of antibody specificity
ImmunoprecipitationModerateVery HighLowNone- Use 1/30 dilution (2μg per 0.35mg lysate)
- Requires verification with isotype control
- Useful for protein interaction studies
ICC/IFModerateModerate-HighLowSubcellular- Enables visualization of subcellular localization
- Can detect nuclear translocation
- Higher risk of non-specific binding
Dot BlotHighHighSemi-quantitativeNone- Excellent for antibody validation
- Tests reactivity with various phosphopeptides
- Cannot determine protein size

What considerations should be made when selecting cell lines or tissue samples for phospho-SMAD2 (S250) studies?

When selecting experimental systems for phospho-SMAD2 (S250) studies, researchers should consider:

  • Baseline expression and phosphorylation levels:

    • Human cell lines: HeLa and HepG2 cells are validated as suitable models

    • Rodent models: NIH/3T3 (mouse) and C6 (rat) cells show detectable levels after stimulation

    • Consider natural variation in SMAD2 expression across tissue types

  • Response to stimulation:

    • PMA treatment (0.2μM for 0.5h) has been validated to induce S250 phosphorylation

    • Different cell types may require optimization of stimulus concentration and duration

    • Consider multiple stimuli (PMA, TGF-β, serum, stress inducers) for comprehensive analysis

  • 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:

    • Antibody cross-reactivity with species-specific variants (human, mouse, rat)

    • Extraction efficiency of phosphorylated proteins from different sample types

    • Sample availability and ethical considerations for tissue samples

  • 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

What are the key technical differences between various commercially available phospho-SMAD2 (S250) antibodies?

When selecting a phospho-SMAD2 (S250) antibody, researchers should consider these key technical parameters:

CharacteristicConsiderationsTechnical Notes
Specificity- Cross-reactivity with other phosphorylation sites
- Recognition of multiple vs. single phosphorylation sites
Some antibodies recognize only S250, while others detect combinations (S245/S250/S255)
Species Reactivity- Human-specific vs. cross-reactive
- Validation in species of interest
Different antibodies show varying reactivity patterns with human, mouse, and rat samples
Applications- Validated applications (WB, ELISA, IP, ICC/IF, IHC)
- Performance metrics for each application
Application-specific dilutions vary (WB: 1:500-1:5000)
Clone Type- Monoclonal vs. polyclonal
- Recombinant vs. traditional hybridoma
Recombinant monoclonal antibodies offer greater batch-to-batch consistency
Host Species- Rabbit vs. mouse
- Compatibility with other antibodies for multiplexing
Rabbit monoclonal antibodies predominate for phospho-SMAD2 (S250)
Validation Depth- Peptide competition
- Specificity testing
- Knockout validation
Comprehensive validation data enhances reliability and reproducibility
Detection Format- Unconjugated vs. conjugated
- Compatible detection systems
Most are unconjugated, requiring appropriate secondary antibodies

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 2024 Thebiotek. All Rights Reserved.