Phosphorylation of SMAD5 at S463 and S465 is induced by bone morphogenetic protein (BMP) receptors, enabling SMAD5 to form a heteromeric complex with SMAD4. This complex translocates to the nucleus, where it binds Smad Binding Elements (SBEs) to regulate gene expression . Non-phosphorylated SMAD5 also influences cytoplasmic energy metabolism by interacting with hexokinase 1 (HK1) to modulate glycolysis and mitochondrial respiration . Dysregulation of SMAD5 phosphorylation is implicated in cancers, developmental disorders, and metabolic diseases .
Recombinant Production: Antibodies like CSB-RA859108A463phHU and SAB6010073 are produced via HEK293F cell systems, ensuring consistent lot-to-lot performance .
Epitope Specificity: Targets phosphorylated S463/S465 residues, validated via dot blot and competitive assays .
Cross-Reactivity: Most antibodies react with human, mouse, and rat SMAD5, though some (e.g., CSB-RA859108A463phHU) are human-specific .
Tissue | Antigen Retrieval | Dilution | Antibody | Source |
---|---|---|---|---|
Human cervical carcinoma | Tris/EDTA pH 9 | 1:800 | ab92698 | |
Human breast carcinoma | Tris/EDTA pH 9 | 1:100 | AP1518 | |
Rat liver | Tris/EDTA pH 9 | 1:800 | ab92698 |
Phosphatase Sensitivity: Treatment with alkaline phosphatase abolishes signal, confirming target specificity .
BMP-Induced Activation: BMP-4 treatment in HeLa cells increases p-SMAD5 levels, detectable at 58 kDa .
Leukemia Suppression: SMAD5 haploinsufficiency suppresses hematopoietic progenitor cell proliferation, with p-SMAD5 levels serving as a biomarker .
Tumor Microenvironment: Phospho-SMAD5 activation in endothelial cells co-cultured with cancer cells (e.g., MDA-MB-231) correlates with mesenchymal transition and metastasis .
Energy Metabolism: Non-phosphorylated SMAD5 interacts with HK1 to enhance glycolysis, a process disrupted in acidic environments .
Embryonic Hematopoiesis: SMAD5 regulates blood cell development, with dysregulated phosphorylation linked to developmental defects .
Parameter | Abcam ab92698 | Cusabio CSB-RA859108A463phHU | AssayGenie CABP1023 |
---|---|---|---|
Host | Rabbit | Rabbit | Rabbit |
Reactivity | Human, Mouse, Rat | Human | Human |
Primary Applications | WB, IHC-P, Dot blot | IHC, ELISA | WB, ELISA |
Dilution (WB) | 1:5000–1:10000 | Not recommended | 1:500–1:1000 |
Observed MW | 58 kDa | N/A | 52 kDa |
Price | ~$300 | ~$210 | ~$200 |
Multi-Species Studies: Use ab92698 or AP1518 for cross-reactivity with mouse/rat models .
Human-Specific IHC: Opt for CSB-RA859108A463phHU (Cusabio) or SAB6010073 (Sigma) .
Cost-Effective WB: CABP1023 (AssayGenie) offers affordability for human-centric research .
Band Variability: Observed molecular weights differ (52–58 kDa), likely due to post-translational modifications or isoforms .
Species-Specific Validation: Confirm antibody performance in non-human models, as some clones (e.g., CSB-RA859108A463phHU) are human-exclusive .
Signal Optimization: Use blocking buffers (e.g., 5% NFDM/TBST) and adjust exposure times to minimize background .
This phosphorylated SMAD5 antibody, CSB-RA859108A463phHU, is a recombinant monoclonal antibody produced through the expression of plasmids integrated with the phospho-SMAD5 (S463+S465) monoclonal antibody DNA sequence in cell lines. The phospho-SMAD5 (S463+S465) monoclonal antibody was generated from splenocytes isolated from animals immunized with human phospho-SMAD5 (S463+S465). This recombinant antibody is a rabbit IgG antibody. It was purified using an affinity-chromatography method. This antibody can detect human phospho-SMAD5 (S463+S465) and is suitable for ELISA and IHC analyses.
SMAD5 is a receptor-activated Smad that functions as an intracellular signal transducer for the transforming growth factor (TGF) superfamily. SMAD5 regulates cytoplasmic metabolic machinery and serves as an intracellular pH messenger to maintain cellular bioenergetic balance. Recent research by Bing Liu et al. has shown that SMAD5, in a haploinsufficiency form, acts as a negative regulator of embryonic hematopoiesis, providing insights into the cytogenetic mechanism by which SMAD5 functions as a leukemia suppressor.
Phosphorylation of SMAD5 at serine residues 463 and 465 represents a critical activation event in BMP (Bone Morphogenetic Protein) signaling. When BMP ligands bind to type I and type II serine/threonine kinase receptors, the type I receptors (ALK1, 2, 3, and 6) phosphorylate SMAD5 at these specific residues. This phosphorylation enables SMAD5 to form complexes with SMAD4 and translocate to the nucleus, where it regulates gene expression involved in development, cell proliferation, and differentiation . Particularly in hematopoietic cells, SMAD5 activation by BMP4 significantly increases the proliferation of erythroid progenitors and formation of glycophorin-A+ cells . This phosphorylation event serves as a fundamental mechanism for regulating both BMP and TGF-β signaling pathways, controlling gene expression, and coordinating cellular processes during development and tissue homeostasis .
The phosphorylation state of SMAD5 dramatically alters its cellular function and localization:
Phosphorylated SMAD5 (pSMAD5): Forms complexes with SMAD4 and predominantly functions in the nucleus as a transcription regulator, activating target genes involved in development and differentiation .
Non-phosphorylated SMAD5: Recent research indicates significant cytoplasmic roles independent of its transcriptional activity. Non-phosphorylated SMAD5 interacts with hexokinase 1 (HK1), a rate-limiting enzyme in glycolysis, enhancing its activity and cellular energy production . It also responds to changes in intracellular pH (pHi), functioning as a pH messenger that maintains bioenergetic homeostasis .
This dual functionality makes SMAD5 unique among SMAD proteins, as it exerts biological effects through both canonical (phosphorylation-dependent, nuclear) and non-canonical (cytoplasmic) mechanisms .
Several stimuli can induce SMAD5 phosphorylation at S463/S465:
BMP ligands: BMP2 and BMP4 are potent inducers of SMAD5 phosphorylation, with dose-dependent effects. Studies show concentration-dependent phosphorylation occurring within 30 minutes of exposure .
TGF-β: While primarily associated with SMAD2/3 activation, TGF-β also induces SMAD1/5 phosphorylation in various epithelial cells, including normal mammary epithelial lines (NMuMG, EpH4, MCF-10A), transformed mammary cell lines (MCF-7, MDA-MB-231), and pancreatic adenocarcinomas .
Viral infection: Some viral infections, such as Rift Valley fever virus, can trigger SMAD protein phosphorylation, including SMAD1/5 at S463/S465 .
Inorganic compounds: Certain ions, such as zirconium, can up-regulate the BMP/SMAD signaling pathway, resulting in increased phosphorylation of SMAD1/5 at S463/S465 .
For optimal induction and detection of SMAD5 phosphorylation:
Induction protocol:
BMP stimulation: Treat cells with 1-30 ng/mL of recombinant BMP4 or BMP2 for 30 minutes at 37°C .
Cell density: Seed cells at a density of 0.2-1.0 × 10^6 cells/mL for suspension cells .
Serum starvation: For cleaner results, serum-starve cells for 4-6 hours before stimulation.
Detection conditions:
Fixation: Fix cells with 4% paraformaldehyde for 20 minutes .
Antibody dilution: For Western blotting, use 1:500-1:5000 dilution; for immunohistochemistry, use 1:50-1:200; for immunofluorescence, use 1:100-1:400 .
Incubation: For maximum sensitivity, incubate with primary antibody overnight at 2-8°C .
Controls to include:
Unstimulated cells (negative control)
BMP receptor inhibitor (e.g., DMH-1) pre-treatment (inhibition control)
The Cell-Based ELISA protocol has been validated to provide reliable results when following these parameters, with fluorogenic detection at 600 nm for phosphorylated SMAD1/5 and 450 nm for a housekeeping protein such as GAPDH for normalization .
Each detection method has unique advantages and limitations when measuring phospho-SMAD5:
Technique | Advantages | Limitations | Optimal Sample Types | Recommended Controls |
---|---|---|---|---|
Western Blot | Quantitative, allows molecular weight verification | Requires larger cell numbers, semi-quantitative | Cell/tissue lysates | Total SMAD5, GAPDH/β-actin loading control |
ELISA | High-throughput, highly quantitative | Cannot visualize cellular localization | Cell/tissue lysates | Standard curve, secondary antibody-only wells |
Immunohistochemistry | Visualizes tissue distribution and localization | Semi-quantitative, fixation-sensitive | Fixed tissue sections | Isotype control, blocking peptide |
Immunofluorescence | High resolution of subcellular localization | Photobleaching, more qualitative | Fixed cells on coverslips | Secondary antibody-only control |
Cell-Based ELISA | Quantifies phosphorylation in intact cells | Limited to cell monolayers | Adherent cells | Quenching buffer control, unstimulated cells |
For cross-method validation:
Always normalize phospho-SMAD5 to total SMAD5 or a housekeeping protein
Use the same stimulation conditions across techniques
Apply matched antibody clones when possible
Include appropriate negative and positive controls for each method
When selecting a phospho-SMAD5 antibody, consider these critical factors:
Specificity considerations:
Cross-reactivity: Many antibodies recognize both phospho-SMAD1 (S463/S465) and phospho-SMAD5 (S463/S465) due to sequence homology. Some also detect phospho-SMAD9 (S465/S467). Check if your research requires SMAD5-specific detection .
Non-specific binding: Validate using knockout/knockdown controls or blocking peptides.
Application-specific parameters:
Western blotting: Select antibodies validated for this application with demonstrated specificity at ~60 kDa (observed molecular weight) .
Immunohistochemistry: Choose antibodies specifically validated in fixed tissues with optimized antigen retrieval protocols.
ELISA: Consider using matched antibody pairs (capture and detection) designed for ELISA applications .
Technical specifications:
Clone type: Monoclonal antibodies offer higher specificity but may have limited epitope recognition. Recombinant antibodies provide better lot-to-lot consistency .
Host species: Consider secondary antibody compatibility and potential cross-reactivity with your sample species.
Storage and handling: Most antibodies require storage at -20°C, with avoidance of repeated freeze/thaw cycles .
Validation documentation: Request data showing antibody validation in your specific application and cell/tissue type .
When encountering weak or absent phospho-SMAD5 signals, systematically address these potential issues:
Sample preparation issues:
Insufficient phosphorylation induction: Verify BMP ligand activity and increase concentration (up to 30-100 ng/mL). Consider time-course experiments (5-60 minutes) .
Rapid dephosphorylation: Add phosphatase inhibitors (25 mM NaF, 25 mM Na β-glycerophosphate) to all buffers .
Protein degradation: Use protease inhibitor cocktail and maintain cold temperature during sample handling.
Technical issues:
Antibody dilution: Optimize antibody concentration; try using more concentrated primary antibody (1:500-1:1000) .
Incubation conditions: Extend primary antibody incubation to overnight at 4°C rather than 2-3 hours .
Detection sensitivity: Switch to more sensitive detection systems (e.g., enhanced chemiluminescence for Western blot).
Biological variables:
Receptor expression: Confirm expression of BMP receptors (ALK2/3/6) in your cell model.
Pathway inhibition: Test for expression of SMAD pathway inhibitors (e.g., Smurfs, NOG).
Cell-specific differences: Some cell types may require different stimulation conditions or exhibit different baseline phosphorylation.
Validation experiment suggestions:
Include a positive control (e.g., C2C12 cells treated with BMP4)
Use phosphatase treatment as a negative control
Test multiple antibody clones if possible
Verify phosphorylation using a parallel technique (e.g., both Western blot and immunofluorescence)
Distinguishing true phospho-SMAD5 signals from cross-reactivity requires careful experimental design:
Understanding antibody specificity:
Many commercial phospho-SMAD5 (S463/S465) antibodies cross-react with phospho-SMAD1 (S463/S465) and sometimes phospho-SMAD9 (S465/S467) due to high sequence homology in the phosphorylation regions . This is evidenced by data sheets listing cross-reactivity with multiple SMAD proteins.
Validation strategies:
Knockout/knockdown controls: Use SMAD5-specific siRNA/shRNA while maintaining SMAD1/SMAD9 expression to determine the SMAD5-specific component of your signal.
Recombinant protein standards: Run purified phosphorylated forms of SMAD1, SMAD5, and SMAD9 alongside your samples to create a reference pattern.
Isoform-specific regions: Target antibodies to regions outside the phosphorylation site that differ between SMAD1, SMAD5, and SMAD9 for follow-up confirmation.
Phosphorylation-specific inhibitors: Use ALK receptor-specific inhibitors like DMH-1 (shown to inhibit SMAD1/5 phosphorylation in a dose-dependent manner) .
Data interpretation approach:
Report signals as "phospho-SMAD1/5" when using antibodies known to detect both
Perform parallel experiments with total SMAD5-specific antibodies to confirm expression
Use multiple antibody clones from different vendors and compare results
Consider mass spectrometry-based approaches for definitive identification
A comprehensive set of controls is essential for accurate interpretation of phospho-SMAD5 experiments:
Essential experimental controls:
Baseline/unstimulated control: Cells maintained in the same conditions without BMP/TGF-β stimulation to establish baseline phosphorylation .
Total SMAD5 measurement: Parallel detection of total (non-phosphorylated + phosphorylated) SMAD5 to normalize phosphorylation signals and account for expression variations .
Loading/housekeeping control: GAPDH, β-actin, or another stable reference protein to ensure equal protein loading across samples .
Pathway inhibition control: Pre-treatment with specific inhibitors like DMH-1 (BMP type I receptor inhibitor) to confirm signal specificity. Figure 3 from source demonstrates dose-dependent inhibition of BMP4-induced SMAD1/5 phosphorylation by DMH-1 .
Phosphatase treatment control: Sample aliquot treated with λ-phosphatase to confirm phosphorylation-specific detection.
Additional validation controls:
Time course: Multiple time points (5, 15, 30, 60, 120 minutes) following stimulation to capture phosphorylation dynamics.
Dose response: Range of BMP/TGF-β concentrations (1-100 ng/mL) to establish dose-dependent effects, as shown in Figures 1 and 2 from source .
Secondary antibody-only control: Omission of primary antibody to assess non-specific binding of secondary antibodies.
Known positive sample: Cell line with well-characterized SMAD5 phosphorylation (e.g., C2C12 cells treated with BMP4) .
Environmental variable control: For studies involving pH, temperature, or osmolarity changes, include appropriate vehicle controls to isolate effects on SMAD5 phosphorylation .
Recent research has revealed a novel, non-canonical role for cytoplasmic SMAD5 independent of its transcriptional activity . To investigate these functions:
Experimental approaches:
Subcellular fractionation coupled with immunoblotting:
Separate nuclear and cytoplasmic fractions
Immunoblot for SMAD5 in each fraction
Compare phosphorylated vs. total SMAD5 distribution
Mutant SMAD5 constructs:
Create phospho-mimetic mutants (S463E/S465E) that simulate constitutive phosphorylation
Create phospho-resistant mutants (S463A/S465A) that cannot be phosphorylated
Express these constructs and assess cytoplasmic functions
Co-immunoprecipitation to identify interacting partners:
Metabolic function assessment:
pH sensitivity experiments:
This research has shown that cytoplasmic SMAD5 interacts directly with HK1, enhancing glycolysis particularly under alkaline intracellular pH conditions, an interaction that becomes stronger as pHi increases .
The discovery that SMAD5 functions as an intracellular pH messenger through nucleocytoplasmic shuttling provides an exciting research area . To study this phenomenon:
Live-cell imaging approaches:
GFP-tagged SMAD5 constructs: Generate stable cell lines expressing GFP-SMAD5 for real-time visualization of localization changes.
Environmental manipulations:
Acidic conditions: Apply 10 mM hydrochloric acid treatments (induces nuclear localization)
Alkaline conditions: Apply 2 mM sodium hydroxide (induces cytoplasmic translocation)
Temperature changes: Cold (cytoplasmic) vs. heat (nuclear) treatments
Osmolarity: Hypotonic (nuclear) vs. hypertonic (cytoplasmic) conditions
Quantitative assessment methods:
Nuclear/cytoplasmic fluorescence ratio: Measure GFP intensity in defined nuclear and cytoplasmic regions over time.
High-content imaging: Automated measurement of subcellular distribution across many cells simultaneously.
pHi measurement: Use pHi-sensitive dyes (BCECF, SNARF) concurrently with localization studies.
Mechanistic investigations:
Mutation of charged amino acid clusters within the MH1 domain that serve as pH sensors.
Inhibition of nuclear export machinery using compounds like leptomycin B.
Biophysical measurements of proton dissociation from SMAD5 under varying pH conditions.
Research has demonstrated that SMAD5 responds to pHi changes within physiologically relevant ranges, with nuclear accumulation at pHi below 6.62 and cytoplasmic distribution at pHi above 7.62 .
Investigating SMAD5 phosphorylation in developmental processes versus pathological conditions requires sophisticated approaches:
Developmental context investigation:
Stem cell differentiation models:
Conditional knockout models:
Generate tissue-specific and temporally-controlled SMAD5 knockout systems
Introduce rescue constructs with wild-type or phospho-mutant SMAD5
Assess developmental outcomes and correlate with phosphorylation status
Developmental timing analysis:
Create phosphorylation state-specific reporter systems
Map phospho-SMAD5 patterns throughout embryonic development
Correlate with morphological and functional changes
Disease-specific approaches:
Patient-derived samples:
Compare phospho-SMAD5/total SMAD5 ratios in normal versus diseased tissues
Correlate phosphorylation status with disease progression
Develop tissue microarrays for high-throughput analysis
Disease models:
Therapeutic intervention assessment:
Research has shown that dysregulation of SMAD5 phosphorylation can have significant implications in developmental disorders and cancer progression, making it a promising therapeutic target .
Modern research increasingly examines SMAD5 phosphorylation within broader signaling networks:
Advanced multiplexing techniques:
Phosphoproteomics approaches:
Multi-parameter flow cytometry:
Single-cell analysis of phospho-SMAD5 alongside other phosphorylated proteins
Correlation with cell cycle status, differentiation markers, or viability parameters
Cell sorting based on phosphorylation patterns for downstream analysis
Spatial proteomics:
Multiplexed immunofluorescence using spectral unmixing
Imaging mass cytometry for high-parameter tissue analysis
Digital spatial profiling technologies
Integrated pathway analysis:
Combinatorial perturbation experiments:
Simultaneous modulation of BMP/TGF-β with intersecting pathways (MAPK, PI3K/AKT)
Compound effect assessment using phosphorylation readouts
Mathematical modeling of signaling cross-talk
Temporal dynamics investigation:
Kinetic measurements of phosphorylation events using time-course experiments
Pulse-chase approaches to determine phosphorylation stability
Live-cell imaging with phosphorylation-specific biosensors
Research using these approaches has revealed that phospho-SMAD5 signaling integrates with other pathways in complex ways, such as during viral infections when multiple signaling cascades activate simultaneously , providing a more comprehensive understanding of cellular response mechanisms.