Purification: Affinity chromatography to ensure high specificity .
Quality Control: Tested via ELISA, Western blot (WB), immunohistochemistry (IHC), and flow cytometry (FC) .
The antibody demonstrates stringent specificity for pS199:
Peptide Competition Assays: Blocks binding with phosphopeptide but not non-phosphopeptide .
Phosphatase Treatment: Loss of signal in dephosphorylated lysates confirms phosphorylation dependency .
Cross-Reactivity: No binding to MAP2 or MAP4 in transfected cells .
Phosphorylation at S199 is an early event in Alzheimer’s disease, impairing tau’s microtubule-stabilizing function .
Mechanistic Insights:
Diagnostic Biomarker: Detects early phosphorylation events in tauopathies .
Immunotherapy Target: Monoclonal antibodies targeting pS199 may reduce pathological tau spread .
Clone | Applications | Key Features |
---|---|---|
23E8 | IHC, FC, ELISA | High sensitivity for human tau in native tissues |
EPR2401Y | WB, Dot Blot | Cross-reactivity with mouse and human samples |
The phospho-MAPT (S199) recombinant monoclonal antibody is produced through a well-established process. The antibody-encoding gene for MAPT is initially inserted into expression vectors. These vectors are subsequently introduced into host cells via polyethyleneimine-mediated transfection. Upon culturing, the host cells produce and secrete the antibodies. Following affinity chromatography purification, the antibodies undergo rigorous functionality assessment through ELISA, IHC, and FC tests. These tests validate their ability to specifically recognize the human MAPT protein phosphorylated at S199.
MAPT, when phosphorylated at S199, plays a crucial role in regulating microtubule assembly, neuronal morphology, and is closely linked to neurodegenerative diseases such as Alzheimer's disease and tauopathies.
MAPT plays a vital role in promoting microtubule assembly and stability, potentially contributing to the establishment and maintenance of neuronal polarity. Its C-terminus binds axonal microtubules, while the N-terminus interacts with neural plasma membrane components, suggesting a role as a linker protein between these structures. Neuronal polarity is determined by the localization of MAPT within the cell body, specifically within the domain defined by the centrosome. Short MAPT isoforms enable cytoskeletal plasticity, whereas the longer isoforms may preferentially contribute to cytoskeletal stabilization.
Phospho-MAPT (S199) refers to the microtubule-associated protein tau (MAPT) that is phosphorylated at the serine 199 residue. This specific phosphorylation event is significant in neurodegenerative research because:
It represents one of the early phosphorylation events in the development of tau pathology
S199 phosphorylation, particularly when combined with phosphorylation at S202 and T205 (collectively recognized by the AT8 antibody), disrupts the "paperclip" conformation of tau, potentially exposing the phosphatase-activating domain (PAD)
The S199 site shows dramatic increases in phosphorylation at later stages of Alzheimer's disease compared to other tau residues
It serves as a biomarker for disease progression and can be detected in various biological samples including brain tissue, cerebrospinal fluid, and plasma
It is implicated in the formation of neurofibrillary tangles (NFTs), a hallmark pathological feature of Alzheimer's disease and other tauopathies
Phospho-MAPT (S199) antibodies are versatile research tools with multiple applications:
Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative detection of phosphorylated tau in solution (dilution range typically 1:2000-1:10000)
Immunohistochemistry (IHC): For visualization of phosphorylated tau in tissue sections (typical dilution 1:50-1:200)
Flow Cytometry (FC): For measuring phosphorylated tau in cell populations (typical dilution 1:50-1:200)
Immunofluorescence (IF): For subcellular localization studies of phosphorylated tau (typical dilution 1:50-1:200)
Western Blotting: For detecting phosphorylated tau protein and fragments in tissue homogenates and cellular lysates
Recombinant production of Phospho-MAPT (S199) antibodies in expression systems like HEK293F cells offers several advantages over traditional hybridoma methods:
Consistency: Recombinant antibodies show reduced batch-to-batch variation
Defined Sequence: The antibody sequence is precisely known and can be engineered
No Animal Requirements: Production doesn't require ongoing use of animals after initial sequence determination
Scalability: Can be readily scaled up in cell culture systems
Reduced Contaminants: Lower risk of contamination with animal pathogens or other antibodies
Reproducibility: Enhanced experimental reproducibility due to consistent performance characteristics
The recombinant monoclonal antibodies against Phospho-MAPT (S199) are typically produced in HEK293F cells, purified by affinity chromatography, and validated for specific detection of tau phosphorylated exclusively at the S199 site .
Validating antibody specificity is crucial for obtaining reliable results:
Phosphatase Treatment Control: Treat duplicate samples with lambda phosphatase to remove phosphorylation and confirm loss of signal
Peptide Competition Assay: Pre-incubate the antibody with phosphorylated and non-phosphorylated peptides spanning the S199 region to demonstrate specificity for the phosphorylated epitope
Knockout/Knockdown Controls: Use MAPT knockout or knockdown samples as negative controls
Multiple Antibody Validation: Compare results with other phospho-tau antibodies targeting the same site but from different sources or clones
Immunoprecipitation-Mass Spectrometry: Confirm the identity of the immunoprecipitated protein by mass spectrometry
Dot Blot Analysis: Test antibody against arrays of phosphorylated and non-phosphorylated tau peptides to confirm epitope specificity
Sample preparation varies by experimental application and tissue/cell type:
For Brain Tissue:
Rapid post-mortem processing is essential to preserve phosphorylation status
Use phosphatase inhibitors in all buffers (sodium fluoride, sodium orthovanadate, β-glycerophosphate, and sodium pyrophosphate)
For frozen tissue: homogenize in RIPA buffer with protease and phosphatase inhibitors
For fixed tissue: optimal fixation is 4% paraformaldehyde for 24-48 hours, followed by careful temperature-controlled paraffin embedding to preserve epitopes
For Cell Culture:
Lyse cells directly in wells using buffer containing 20 mM Tris (pH 7.5), 0.5 mM DTT, 0.15 M NaCl, and 0.5% Triton X-100 supplemented with protease and phosphatase inhibitors
Sonicate lysates with short pulses (4-5 seconds each) followed by centrifugation at 12,000×g for 10 minutes at 4°C
For Sarkosyl Fractionation (to separate soluble from insoluble tau):
This method separates normal tau from aggregated pathological tau
The ratio of Sarkosyl insoluble to Sarkosyl soluble phospho-tau can indicate the degree of pathological aggregation
When incorporating Phospho-MAPT (S199) antibodies in multiplex assays:
Antibody Cross-reactivity: Ensure no cross-reactivity with other phospho-epitopes or proteins in the multiplex panel
Species Compatibility: Confirm compatibility with other antibodies in terms of species origin to avoid cross-reactivity of secondary antibodies
Signal Optimization: Balance signal intensities across all analytes in the panel, as phospho-tau signals may be significantly weaker than other proteins
Epitope Masking: Consider the possibility that binding of one antibody might mask nearby epitopes due to steric hindrance
Dephosphorylation Risk: Minimize time between sample collection and assay to prevent loss of phosphorylation
Signal Normalization: Include total tau measurement to normalize phospho-tau signals for more accurate quantification
The relationship between S199 phosphorylation and other sites involves complex temporal and functional dynamics:
Sequential Phosphorylation: Evidence suggests that S199 phosphorylation may precede phosphorylation at other sites in the disease process, potentially acting as a priming event
Combined Effects: When S199 is phosphorylated alongside S202 and T205 (the AT8 epitope), it causes disruption of tau's normal "paperclip" conformation, leading to exposure of the phosphatase-activating domain (PAD)
Differential Distribution: Phosphorylation at different sites shows distinct spatial distribution patterns in the brain. While pTau-T231 is preferentially located in cytoplasm surrounding nuclei, pTau-S396 is predominantly found in nerve fibers and strongly associated with amyloid plaques
Functional Consequences: Phosphomimetic mutations at Ser199/Ser202/Thr205 (psTau) have been shown to impair axonal transport in rat hippocampal neurons through a PAD-dependent mechanism involving protein phosphatase 1 (PP1γ)
Biomarker Utility: In comparative studies, plasma p-tau217 typically shows stronger associations with brain amyloid-β deposition than p-tau181 and p-tau231, though phosphorylation at multiple sites including S199 increases with disease progression
Table 1: Comparison of Key Tau Phosphorylation Sites in Alzheimer's Disease
Phosphorylation Site | Disease Stage Association | Primary Cellular Location | Key Antibodies | Functional Consequences |
---|---|---|---|---|
Ser199 | Early-Late stages | Cytoplasmic and neuritic | Anti-pS199 | Paperclip conformation disruption, PAD exposure |
Ser202/Thr205 | Early-Mid stages | Cytoplasmic | AT8 | Microtubule binding inhibition |
Thr231 | Early stages | Primarily cytoplasmic | AT180 | Priming site for other kinases |
Ser396/Ser404 | Mid-Late stages | Primarily in nerve fibers | PHF1 | Associated with tangle formation |
Blood-based detection of phosphorylated tau faces several methodological challenges:
Low Abundance: Phosphorylated tau exists at significantly lower concentrations in blood compared to cerebrospinal fluid (CSF), requiring highly sensitive detection methods
Matrix Effects: Plasma and serum contain various proteins and factors that can interfere with antibody binding and assay performance
Epitope Stability: Phosphorylation at S199 may be unstable in blood samples due to phosphatase activity, necessitating rapid processing and effective phosphatase inhibitors
Standardization Issues: Different assay platforms and antibody clones yield varying absolute values, making cross-study comparisons difficult
Threshold Generation: Establishing clinically meaningful cutoff values requires large datasets from diverse populations
Fragment Variability: Tau exists in multiple fragments in blood, and the phosphorylation profile may differ between fragments (e.g., 50 kDa, 38 kDa, and 25 kDa fragments show different phosphorylation patterns)
Pre-analytical Variables: Collection tubes, processing time, freeze-thaw cycles, and storage conditions can all affect phosphorylation detection
Emerging immunoassay platforms combining immunoprecipitation with mass spectrometry (IP-MS) have shown promise for more accurate quantification of site-specific phosphorylated tau in blood samples .
Several experimental approaches can be employed to study functional consequences of S199 phosphorylation:
Phosphomimetic Mutations: Using S199E or S199D mutations to mimic constitutive phosphorylation
Cell-Based Functional Assays:
Axonal transport assays in primary neurons to assess effects on cargo movement
Microtubule binding assays to determine effects on tau-microtubule interactions
Protein-protein interaction studies (co-immunoprecipitation) to identify altered binding partners
PAD exposure assays to determine conformational changes
Kinase/Phosphatase Modulation:
Animal Models:
Advanced Imaging:
FRET-based sensors to detect conformational changes associated with S199 phosphorylation
Live-cell imaging to track trafficking and aggregation dynamics
Super-resolution microscopy to visualize subcellular localization
Researchers commonly encounter these technical challenges:
High Background Signal
Cause: Non-specific binding, excessive antibody concentration, inadequate blocking
Solution: Optimize antibody dilution (start with 1:200 for IHC/IF), extend blocking time, use alternative blockers (5% BSA or 5% normal serum from secondary antibody species)
Weak or Absent Signal
Cause: Epitope masking, dephosphorylation during processing, insufficient antigen retrieval
Solution: Ensure phosphatase inhibitors are fresh and used at appropriate concentrations; for IHC, try heat-induced epitope retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)
Non-specific Bands in Western Blot
Cause: Cross-reactivity, protein degradation, non-specific binding
Solution: Include appropriate controls (phosphatase-treated samples), optimize washing steps, try alternative blocking agents
Variable Results Between Experiments
Cause: Batch-to-batch antibody variation, inconsistent sample handling
Solution: Use the same antibody lot when possible, standardize all protocols, include positive control samples in every experiment
Poor Reproducibility in Quantitative Assays
Cause: Phosphorylation instability, assay variability
Solution: Establish tight time controls for sample processing, include internal calibrators, perform technical replicates
When faced with conflicting data from different detection methods:
Understand Methodological Differences:
Different antibodies may have varying specificities and affinities even when targeting the same epitope
Detection platforms have different sensitivity thresholds and dynamic ranges
Sample preparation can affect epitope availability
Consider Technical Validation:
Compare results using multiple antibody clones against the same phospho-epitope
Assess antibody specificity using phosphatase-treated controls and peptide competition assays
Validate findings using orthogonal methods (e.g., mass spectrometry)
Evaluate Sample-Specific Factors:
Tau fragments may vary between sample types, affecting epitope presentation
Matrix effects can influence antibody binding in complex biological samples
Post-translational modifications beyond phosphorylation may mask epitopes
Establish Context-Specific Benchmarks:
Report Comprehensive Methodological Details:
Include complete information about antibody clones, dilutions, incubation conditions
Specify exact sample processing steps, including timing and temperature
Report all controls and validation steps performed
For reliable quantification of relative changes:
Western Blotting with Dual Detection:
Probe for phospho-S199 and total tau simultaneously (using differently labeled secondary antibodies)
Calculate the ratio of phospho-S199 to total tau for each sample
Include a concentration gradient of a standard sample to ensure linearity of detection
ELISA-Based Approaches:
Sandwich ELISA with capture antibody against total tau and detection antibody against phospho-S199
Include a standard curve using recombinant phospho-tau
Normalize to total tau measured in parallel assays
Quantitative Immunofluorescence:
Co-stain with antibodies against phospho-S199 and total tau
Use automated image analysis to calculate mean intensity ratios
Include internal control regions to normalize between sections/samples
Mass Spectrometry-Based Methods:
Targeted MS approaches (multiple reaction monitoring) for absolute quantification
Immunoprecipitation followed by MS for enhanced sensitivity
Isotope-labeled internal standards for accurate quantification
Flow Cytometry:
Dual staining for phospho-S199 and total tau
Calculate mean fluorescence intensity ratios
Include calibration beads to standardize between experiments
Phospho-MAPT (S199) antibodies play crucial roles in therapeutic development:
Target Validation:
Confirming the presence and abundance of phospho-S199 tau in patient samples
Correlating phospho-S199 levels with disease severity and progression
Therapeutic Antibody Development:
Serving as templates for therapeutic antibody engineering
Competing with therapeutic candidates in binding assays to confirm epitope targeting
Vaccine Development Assessment:
Drug Screening:
Quantifying changes in phospho-S199 levels in response to kinase or phosphatase modulators
High-throughput screening assays to identify compounds that reduce S199 phosphorylation
Biomarker for Clinical Trials:
Monitoring treatment effects on phospho-tau levels in biological fluids
Participant stratification based on baseline phospho-tau profiles
Target Engagement Studies:
Confirming that therapeutic interventions effectively engage with phosphorylated tau
Determining dose-response relationships for investigational treatments
S199 phosphorylation has specific implications for axonal transport:
PAD Exposure Mechanism:
PP1γ Activation and Consequences:
Experimental Evidence:
Expression of psTau causes significant impairment of axonal transport in primary rat hippocampal neurons
Deletion of PAD in psTau significantly reduces interaction with PP1γ and rescues axonal transport impairment
Similar effects have been observed in squid axoplasm models with phosphomimetic mutations
Disease Relevance:
Axonal transport disruption is an early event in Alzheimer's disease and other tauopathies
The PAD-dependent mechanism may represent a potential therapeutic target
Compounds that prevent PAD exposure or block PAD-PP1γ interaction might protect against transport deficits
Plasma phospho-tau biomarkers are rapidly evolving:
Diagnostic Applications:
Monitoring Disease Progression:
Technical Advancements:
Differential Diagnosis:
Special Populations:
Table 2: Comparison of Different Plasma Phospho-Tau Biomarkers in Alzheimer's Disease
Biomarker | Diagnostic Performance | Association with Amyloid | Correlation with Cognition | Stage of Development |
---|---|---|---|---|
p-tau181 | Good | Moderate | Moderate-High | Advanced clinical validation |
p-tau217 | Very Good | Strong | High | Emerging, promising results |
p-tau231 | Good | Moderate | Moderate | Early clinical validation |
p-tau199 | Under investigation | Under investigation | Preliminary evidence | Early research phase |
Interpretation of phospho-S199 changes requires contextual understanding:
Model-Specific Considerations:
Transgenic Mouse Models: Different tau transgenic lines show varying phosphorylation profiles and progression rates
Cell Culture Models: Neuronal versus non-neuronal cells may show different phosphorylation regulation
Acute vs. Chronic Models: Acute treatments (like okadaic acid) may induce different phosphorylation patterns than chronic disease models
Temporal Dynamics:
Early increases may represent physiological stress responses
Persistent elevation suggests pathological processes
The relationship between S199 phosphorylation and other sites may shift over disease course
Regional Variations:
Relationship to Other Pathologies:
Molecular Context:
Several knowledge gaps persist in our understanding:
The phospho-S199-PP1γ interaction reveals a specific mechanism of pathology:
Molecular Mechanism:
Functional Consequences:
Activated PP1γ triggers signaling cascades that impair anterograde fast axonal transport
This leads to disruption of cargo delivery to synapses and axon terminals
Expression of phosphomimetic tau at S199/S202/T205 (psTau) impairs axonal transport in primary rat hippocampal neurons
Deletion of PAD rescues this transport impairment, confirming the PAD-dependent mechanism
Amplification Cycle:
Transport deficits may further compromise neuronal health
Compromised neurons may exhibit altered kinase/phosphatase balance
This could lead to additional tau phosphorylation, creating a pathological cycle
Therapeutic Implications:
Targeting the tau-PP1γ interaction or downstream signaling represents a potential intervention strategy
Compounds that prevent PAD exposure or block its interaction with PP1γ might protect against transport deficits
Small molecule inhibitors of this protein-protein interaction could be therapeutic candidates
Figure 1: Model of S199 Phosphorylation-Induced PAD Exposure and PP1γ Activation
The figure would illustrate the proposed mechanism where tau phosphorylation at S199 (along with S202/T205) disrupts the paperclip conformation, exposing PAD, which then interacts with and activates PP1γ, leading to axonal transport impairment and subsequent neurodegeneration.