The antibody is a rabbit polyclonal IgG raised against a synthetic phosphopeptide encompassing the phosphorylated Thr181 site (P-K-T(p)-P-P) derived from human Tau-F isoform (P10636-8) . Affinity purification ensures specificity, with non-phospho antibodies removed via chromatography . The antibody is unconjugated, formulated in PBS with 50% glycerol and 0.02% sodium azide .
Tau phosphorylation at Thr181 is a hallmark of neurodegenerative diseases, including:
Alzheimer’s Disease (AD): Hyperphosphorylation disrupts microtubule stability, promoting neurofibrillary tangle formation .
Parkinson’s Disease (PD): LRRK2 kinase phosphorylates Tau at Thr181, with mutant G2019S linked to elevated phosphorylation .
Frontotemporal Dementia (FTD): Pathogenic Tau isoforms exhibit Thr181 phosphorylation .
Phosphorylation at Thr181 is mediated by proline-directed kinases (e.g., CDK5, GSK3β) and reciprocally regulates O-GlcNAcylation at adjacent residues . This interplay modulates Tau’s ability to bind microtubules and stabilize axonal polarity .
Phospho-MAPT (Thr181) antibodies are available in both monoclonal and polyclonal formats. Monoclonal antibodies, such as clone AT270 from Thermo Fisher Scientific and clone M7004D06 from BioLegend, provide high specificity and reproducibility for consistent experimental results . Polyclonal antibodies, including those from St John's Labs and Boster Bio, often recognize multiple epitopes around the phosphorylation site and may provide enhanced signal in certain applications . The choice between monoclonal and polyclonal depends on your experimental needs - monoclonals offer better specificity and lot-to-lot consistency, while polyclonals may provide higher sensitivity due to recognition of multiple epitopes within the target region .
When selecting a Phospho-MAPT (Thr181) antibody, cross-species reactivity is an important consideration. Many commercially available antibodies demonstrate reactivity across human, mouse, and rat samples, making them versatile for comparative studies across disease models . For example, the rabbit polyclonal antibody from St John's Labs (STJ90425) has validated reactivity to human, rat, and mouse samples . The antibody from ARG51609 similarly shows broad cross-reactivity . Species reactivity is determined during antibody validation using species-specific positive controls and depends on the conservation of the epitope sequence around Thr181 across species. Verify the specific reactivity claims with validation data and consider testing small amounts if working with less common research models or when absolute cross-reactivity confirmation is required for your particular application .
Host species selection for Phospho-MAPT (Thr181) antibodies can significantly impact experimental outcomes. Mouse-derived monoclonal antibodies like AT270 typically show excellent specificity for the phospho-epitope with minimal cross-reactivity to non-phosphorylated tau . These antibodies are ideal for applications requiring high discrimination between phosphorylated and non-phosphorylated states. Rabbit-derived polyclonal antibodies often provide enhanced sensitivity due to recognition of multiple epitopes within the target region, which can be advantageous in detecting low abundance phosphorylated tau species . The choice between hosts should also consider your experimental system - if using mouse tissue with mouse-derived primary antibodies, additional blocking steps may be required to prevent non-specific binding to endogenous mouse immunoglobulins. Similarly, secondary antibody selection must align with the host species to ensure proper detection .
For optimal Western blot results with Phospho-MAPT (Thr181) antibodies, careful sample preparation and protocol optimization are essential. Begin with appropriate dilutions - most manufacturers recommend starting dilutions between 1:500-1:2000 for Western blotting . Sample preparation should include phosphatase inhibitors to preserve the phosphorylation state of tau proteins. When detecting the ~75 kDa phospho-tau species, ensure adequate protein loading (20-40 μg of total protein) and use 8-10% polyacrylamide gels for optimal separation . Transfer conditions should be optimized for high molecular weight proteins, with longer transfer times at lower voltages often yielding better results. Blocking with 5% BSA rather than milk is recommended as milk contains phospho-proteins that may increase background. For detection, the phospho-tau signal can sometimes appear as multiple bands due to different tau isoforms and varying degrees of phosphorylation . Always include appropriate positive controls (such as brain lysates from tauopathy models) and negative controls (dephosphorylated samples) to validate specificity .
Successful immunohistochemistry (IHC) with Phospho-MAPT (Thr181) antibodies requires attention to several critical factors. First, tissue fixation significantly impacts epitope accessibility - while 10% neutral buffered formalin is commonly used, overfixation can mask the phospho-epitope . Antigen retrieval is essential, with heat-induced epitope retrieval in citrate buffer (pH 6.0) being most effective for exposing the Thr181 phospho-epitope . For formalin-fixed paraffin-embedded sections, recommended antibody concentrations range from 1.0-10 μg/ml, but optimization for your specific tissue is advisable . Background staining can be minimized by using appropriate blocking solutions containing both serum matching the host of the secondary antibody and bovine serum albumin. When interpreting results, be aware that phospho-tau distribution patterns vary depending on disease stage and type - early stage tauopathies may show primarily neuronal staining while advanced cases display both neuronal and glial pathology . Always include positive control tissues (such as Alzheimer's disease brain sections) and negative controls (primary antibody omission and non-phosphorylated controls) to ensure specificity and validate staining patterns .
Validating the specificity of Phospho-MAPT (Thr181) antibody staining requires multiple complementary approaches. First, perform side-by-side comparisons with total tau antibodies on sequential sections to confirm co-localization of signals . Include phosphatase treatment controls - treating one set of samples with lambda phosphatase should abolish or significantly reduce Phospho-MAPT (Thr181) staining while leaving total tau staining intact . For immunodepletion validation, pre-incubate the antibody with excess phosphorylated and non-phosphorylated peptides representing the Thr181 region - specific signal should be blocked only by the phosphorylated peptide . Additionally, compare staining patterns across tissues with known levels of phosphorylated tau, such as Alzheimer's disease brain tissue (positive control) versus age-matched controls (lower levels) . In cell culture models, treatment with kinase activators (like okadaic acid) to increase tau phosphorylation versus kinase inhibitors to decrease phosphorylation can further validate specificity . Finally, confirming consistent results across multiple antibody lots and different phospho-tau antibodies targeting the same epitope provides additional validation of specificity .
Immunodepletion experiments using Phospho-MAPT (Thr181) antibodies require careful consideration of antibody-antigen kinetics. As demonstrated in the kinetics data for similar phospho-tau antibodies (PHF6 and PHF13), association rates (kon), dissociation rates (koff), and equilibrium dissociation constants (KD) significantly impact depletion efficiency . For optimal immunodepletion, antibodies with high affinity (low KD values) and slow dissociation rates are preferred. The kinetics data shown in Figure 1 reveals that effective phospho-tau antibodies typically have kon values in the 104-105 M-1s-1 range and koff values in the 10-4-10-5 s-1 range, resulting in KD values in the nanomolar range . The immunodepletion protocol should allow sufficient incubation time (typically 12-24 hours at 4°C) to reach equilibrium binding. Protein A/G bead capacity must exceed the expected antibody-antigen complex concentration to ensure complete capture. When performing immunodepletion with Phospho-MAPT (Thr181) antibodies, verify depletion efficiency by measuring both phospho-tau and total tau levels in the depleted sample using ELISA or Western blot . Successful immunodepletion should significantly reduce phospho-tau levels while having a more modest effect on total tau levels, as observed in the Triton-insoluble MC1 staining reductions demonstrated in Figure 1D .
Phosphorylation at Thr181 is part of a complex network of post-translational modifications (PTMs) that collectively regulate tau function. Research demonstrates significant crosstalk between phosphorylation and acetylation, as shown in Figure 2, where tau acetylation modulates phosphorylation at specific epitopes including Thr181 . Active CBP (CREB-binding protein, an acetyltransferase) expression leads to reduced phosphorylation at multiple sites, visualized as a reduction in the ~75 kDa phospho-tau species (marked by solid black arrows in Figure 2a) . Experiments with acetylation-mimicking mutants (4KQ) show reduced phosphorylation compared to acetylation-deficient mutants (4KR), suggesting acetylation at lysine residues affects the ability of kinases to phosphorylate nearby threonine residues including Thr181 . This interplay is particularly evident under conditions of enhanced phosphorylation, such as okadaic acid treatment (Figure 2c) . Mechanistically, acetylation may alter tau conformation, affecting kinase accessibility to phosphorylation sites or changing tau's interaction with phosphatases. For comprehensive tau PTM analysis, researchers should employ complementary approaches including mass spectrometry, site-specific antibodies for different modifications, and mutational studies to delineate the hierarchical relationship between phosphorylation at Thr181 and other modifications .
Accurate quantification of Phospho-MAPT (Thr181) across subcellular fractions requires specialized approaches due to tau's differential distribution and phosphorylation state in cellular compartments. When analyzing tau pathology, it's critical to examine both soluble and insoluble fractions. The Triton-insoluble fraction, as demonstrated in Figure 1B and 1C, contains aggregated phosphorylated tau species that are particularly relevant to pathological conditions . Sequential extraction protocols typically begin with low-stringency buffers (PBS or Tris-buffered saline) to extract soluble cytosolic tau, followed by detergent extraction (1% Triton X-100) for membrane-associated tau, and finally high-stringency conditions (RIPA buffer, sarkosyl, or formic acid) to solubilize aggregated tau . When quantifying Phospho-MAPT (Thr181) across these fractions, normalize phospho-tau signals to total tau within each fraction rather than to total protein, as tau distribution itself varies between fractions. Different detection methods (Western blot, ELISA) may have varying sensitivities across fractions - ELISA may be more sensitive for soluble fractions while Western blot may better detect aggregated species in insoluble fractions . For immunohistochemical quantification, distinguish between different cellular compartments (neuronal soma vs. neurites vs. neuropil) as phosphorylation patterns may vary across these regions . Finally, phosphatase inhibitors are particularly critical during subcellular fractionation procedures to prevent artificial dephosphorylation during the extended processing times required for fractionation .
Phospho-MAPT (Thr181) antibodies demonstrate variable performance across tauopathy models, reflecting different tau phosphorylation profiles in various pathologies. In Alzheimer's disease models, these antibodies robustly detect NFT pathology, as phosphorylation at Thr181 occurs early in disease progression . The experimental data in Figure 1B and 1C demonstrates successful detection of tau pathology using phospho-tau antibodies in both primary neuronal cultures treated with tau preformed fibrils (PFF) and in rTg4510 transgenic mouse brain extracts . These antibodies can distinguish between pathological conditions (PFF treatment or transgenic models) and controls (untreated cultures or control tTA brain extracts). In frontotemporal dementia models, the detection pattern may differ due to distinct phosphorylation profiles. When selecting appropriate models, consider that phosphorylation at Thr181 is regulated by proline-directed protein kinases including CDK5, GSK3, and MAPK . Therefore, cellular stress models that activate these kinases (such as okadaic acid treatment shown in Figure 2c) can artificially enhance Thr181 phosphorylation . For quantitative assessments across models, standardized protocols and identical antibody concentrations are essential, as the absolute signal intensity may vary between models. Researchers should validate findings across multiple models when possible, as no single model recapitulates all aspects of human tauopathies .
Phosphorylation at Thr181 serves as a critical biomarker in the progression of tauopathies like Alzheimer's disease. This site is phosphorylated by proline-directed protein kinases (PDPK1, CDK1, CDK5, GSK3, MAPK) and shows increased phosphorylation during disease development . Mechanistically, phosphorylation at Thr181, which is located in the proline-rich region of tau, reduces tau's binding affinity for microtubules, promoting detachment and potentially contributing to microtubule disassembly and aggregation of free tau . Research has demonstrated that phosphorylation within tau's repeat domain or flanking regions (including Thr181) diminishes tau's interaction with microtubules and plasma membrane components . In disease models, immunodepletion with phospho-tau antibodies targeting sites including Thr181 reduces tau pathology in neuronal cultures, as evidenced by significant declines in Triton-insoluble MC1 staining (Figure 1D) . This suggests that targeting phosphorylated tau epitopes could have therapeutic potential. Temporally, Thr181 phosphorylation increases with disease progression but can show fluctuations based on cellular stress and activity of regulatory phosphatases. The interplay with other post-translational modifications, particularly acetylation as shown in Figure 2, indicates that Thr181 phosphorylation is part of a complex regulatory network governing tau function and dysfunction .
Phospho-MAPT (Thr181) antibodies serve as valuable tools for evaluating therapeutic interventions targeting tau pathology. For kinase inhibitor studies, these antibodies can quantify reductions in Thr181 phosphorylation following treatment, serving as pharmacodynamic markers of target engagement . When evaluating immunotherapies, Phospho-MAPT (Thr181) antibodies can assess the efficacy of therapeutic antibodies through competition assays, epitope mapping, and measurement of residual phospho-tau levels after treatment . The immunodepletion data in Figure 1D demonstrates how phospho-tau antibodies (PHF6 and PHF13) can reduce Triton-insoluble MC1 staining in neuronal cultures, providing proof-of-concept for immunotherapy approaches . For small molecule modulators of tau acetylation, these antibodies can measure consequent changes in phosphorylation states, as illustrated in Figure 2 where acetylation modulates phosphorylation at multiple epitopes . In clinical trials, Phospho-MAPT (Thr181) levels in cerebrospinal fluid serve as biomarkers of disease progression and treatment response. Methodologically, establish baseline phosphorylation levels before intervention, include appropriate controls (vehicle, inactive compound), and use multiple readouts (Western blot, ELISA, immunohistochemistry) for comprehensive assessment . Time-course analyses are essential to distinguish transient from sustained effects and to identify optimal therapeutic windows for intervention .
Non-specific binding with Phospho-MAPT (Thr181) antibodies can compromise experimental results, but several strategies can mitigate these issues. For Western blotting, increasing blocking stringency by using 5% BSA rather than milk (which contains phosphoproteins) can reduce background . Titrating antibody concentration is essential - start with manufacturer-recommended dilutions (typically 1:500-1:2000 for Western blot) and adjust based on signal-to-noise ratio . Pre-absorbing the antibody with non-specific proteins from the species being studied can reduce cross-reactivity. For immunohistochemistry applications, additional blocking steps including avidin/biotin blocking (if using biotinylated secondary antibodies) and mouse-on-mouse blocking (when using mouse primary antibodies on mouse tissue) may be necessary . False positive signals can arise from endogenous phosphatases - always include phosphatase inhibitors in all buffers during sample preparation . To definitively distinguish specific from non-specific binding, include appropriate controls: phosphatase-treated samples (should show reduced or eliminated signal), peptide competition assays with phosphorylated and non-phosphorylated peptides, and immunodepleted samples as demonstrated in Figure 1D . If high background persists, consider alternative antibody clones or switching from polyclonal to monoclonal antibodies, which typically offer higher specificity .
Maintaining optimal Phospho-MAPT (Thr181) antibody performance requires precise storage and handling practices. Most manufacturers recommend storing undiluted antibodies at -20°C for long-term preservation, with aliquoting to avoid repeated freeze-thaw cycles that can degrade antibody quality . For frequent use, short-term storage at 4°C (up to one month) is acceptable for most formulations . The typical antibody formulation includes phosphate-buffered solution (pH 7.2-7.4) containing preservatives like sodium azide (0.02-0.09%) and stabilizers such as glycerol (50%) and BSA (0.5-1%) . These components maintain antibody stability and prevent microbial growth, though sodium azide can inhibit horseradish peroxidase and should be diluted sufficiently for immunohistochemical applications . Working dilutions should be prepared fresh and used within 24 hours for optimal results. When using diluted antibodies, store at 4°C and protect from light, particularly for fluorophore-conjugated variants. Monitor for signs of degradation, including precipitation, color changes, or diminished performance in positive controls. If performance decreases, verify storage conditions and consider obtaining a new lot. Maintain detailed records of antibody performance across different lots to track potential variability .
Detecting low abundance Phospho-MAPT (Thr181) in biological samples requires optimized protocols to enhance sensitivity while maintaining specificity. Begin with sample enrichment strategies - phosphorylated tau can be concentrated using immunoprecipitation with total tau antibodies followed by phospho-specific detection . For Western blotting, sensitivity can be enhanced by using high-sensitivity chemiluminescent substrates, increasing protein loading (up to 50-100 μg total protein), and employing signal amplification systems such as biotinylated secondary antibodies with streptavidin-HRP . Extended exposure times may reveal faint signals, but always monitor for increased background. In immunohistochemistry applications, signal amplification through tyramide signal amplification (TSA) or polymer-based detection systems can significantly enhance sensitivity compared to conventional ABC methods . For ELISA-based detection, sandwich ELISA formats using a capture antibody against total tau and detection with Phospho-MAPT (Thr181) antibodies can lower detection thresholds to pg/ml ranges . When working with cerebrospinal fluid or dilute samples, consider sample concentration through ultrafiltration prior to analysis. Regardless of the method, optimize blocking conditions carefully - insufficient blocking increases background while excessive blocking may mask low-abundance epitopes . Finally, compare multiple detection methods, as some phospho-epitopes may be better preserved in certain applications - native conditions in ELISA may maintain some phospho-epitopes better than the denaturing conditions of Western blot .
Multiplexed phospho-tau profiling using Phospho-MAPT (Thr181) antibodies alongside other site-specific phospho-tau antibodies provides comprehensive characterization of tau phosphorylation states. This approach is particularly valuable given the demonstrated interactions between different phosphorylation sites and other post-translational modifications shown in Figure 2 . For multiplex immunoassays, carefully select antibodies with minimal cross-reactivity and compatible host species to allow simultaneous detection. Multiplex Western blotting can be achieved through sequential probing with different phospho-tau antibodies after complete stripping between applications, or using spectrally distinct fluorescent secondary antibodies for simultaneous detection . For immunohistochemistry, multiplex approaches include multi-color immunofluorescence with primary antibodies from different host species or using tyramide signal amplification to allow multiple antibodies from the same host . Advanced techniques like Luminex xMAP technology enable simultaneous quantification of multiple phospho-tau epitopes in solution phase assays with high sensitivity. When designing multiplex experiments, consider the hierarchical relationship between phosphorylation sites - phosphorylation at one site may influence accessibility or phosphorylation status of other sites . Data analysis should include correlation analyses between different phospho-epitopes to identify co-regulated sites and potential priming relationships. Multiplexed approaches can reveal disease-specific phosphorylation signatures that may not be apparent when examining individual sites in isolation .
Single-cell analysis using Phospho-MAPT (Thr181) antibodies represents a frontier in tauopathy research, allowing examination of cell-to-cell variability in tau phosphorylation. This approach can reveal distinct neuronal subpopulations with varying vulnerability to tau pathology . For single-cell immunofluorescence, high-affinity monoclonal antibodies with minimal background are optimal, with titers carefully optimized to detect physiological phosphorylation levels without artifacts . Quantitative image analysis should include single-cell segmentation and intensity measurements normalized to total tau or neuronal markers. Mass cytometry (CyTOF) with metal-conjugated Phospho-MAPT (Thr181) antibodies allows simultaneous measurement of multiple tau epitopes and cellular markers in single cells without spectral overlap concerns. For spatial context preservation, multiplexed immunofluorescence or imaging mass cytometry can map phospho-tau distribution within tissue while maintaining information about cellular relationships and microenvironment . Single-nucleus RNA-seq combined with immunostaining for Phospho-MAPT (Thr181) can correlate tau phosphorylation status with transcriptomic signatures of individual cells. When designing single-cell experiments, consider that fixation and permeabilization protocols significantly affect epitope accessibility and must be optimized for single-cell applications . Data integration across multiple single-cell modalities provides the most comprehensive view of cellular heterogeneity in tauopathies and can identify specific cellular subpopulations that may be targeted for therapeutic intervention .
Standardization of Phospho-MAPT (Thr181) measurements across laboratories requires rigorous protocol alignment and reference materials. For Western blot applications, standardized protocols should specify protein extraction methods, loading amounts (typically 20-40 μg), gel percentage (8-10% for optimal separation of phospho-tau species), transfer conditions, blocking agents (preferably 5% BSA), and antibody dilutions (1:500-1:2000) . Quantification should employ densitometry with background subtraction and normalization to housekeeping proteins, with results reported as fold-change relative to control samples rather than absolute values . For ELISA applications, standard curves should utilize recombinant tau phosphorylated at Thr181, with concentrations verified by mass spectrometry . Reference materials should include well-characterized positive controls (brain extracts from tauopathy models like rTg4510 as shown in Figure 1B) and negative controls (dephosphorylated samples) . Inter-laboratory proficiency testing with distribution of identical samples analyzed by different laboratories can identify methodological variables affecting results. Digital image sharing of original blots and immunostaining allows direct comparison of band patterns and staining intensity across laboratories. When comparing results across antibodies, epitope mapping and cross-reactivity profiles should be thoroughly documented . Finally, method validation reports should include limit of detection, linear range, precision, and accuracy for each protocol to ensure comparability of measurements across studies .
Quantitative reference data for Phospho-MAPT (Thr181) levels shows considerable variation across experimental models, providing important benchmarks for researchers. In primary neuronal cultures, baseline Phospho-MAPT (Thr181) levels are typically low but increase significantly upon treatment with tau preformed fibrils (PFF), as demonstrated in Figure 1B and 1C where PFF treatment increased Triton-insoluble tau pathology markers . In transgenic mouse models like rTg4510, Phospho-MAPT (Thr181) levels are substantially elevated compared to control tTA mice, with immunodepletion studies showing significant reductions in both phospho-tau and total tau levels following antibody treatment (Figure 1D) . In cellular models, treatment with phosphatase inhibitors like okadaic acid significantly increases Phospho-MAPT (Thr181) levels, as shown in Figure 2C, where this treatment amplified differences between wild-type and acetylation-mimicking tau mutants . Quantitatively, the association and dissociation kinetics data in Figure 1A provide reference values for antibody-antigen interactions, with calculated association rates (kon), dissociation rates (koff), and equilibrium dissociation constants (KD) in the nanomolar range . These quantitative parameters are essential for comparing antibody performance and designing immunodepletion or therapeutic antibody experiments. When establishing quantitative baselines for new models, researchers should compare phospho-tau levels to total tau levels rather than using absolute values, as total tau expression varies widely across models .