Phospho-MAPT (S202) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Production

The Phospho-MAPT (S202) Recombinant Monoclonal Antibody is engineered through genetic engineering to bind specifically to the phosphorylated S202 residue of MAPT. Key characteristics include:

ParameterDetail
ClonalityRecombinant monoclonal
ImmunogenSynthetic peptide derived from human MAPT phosphorylated at S202
Host CellsHEK293F cells (for expression)
PurificationAffinity chromatography
ConjugateUnconjugated (native IgG)
Species ReactivityHuman; cross-reactivity with mouse/rat reported in some variants

The production process involves:

  1. Gene Cloning: Incorporation of the MAPT antibody-encoding gene into expression vectors.

  2. Transfection: Introduction of vectors into host cells (e.g., HEK293F) using polyethyleneimine .

  3. Culturing: Secretion of antibodies into culture media.

  4. Purification: Affinity chromatography to isolate high-purity antibodies .

Clinical and Research Significance

Phosphorylation at S202 is a hallmark of pathological tau in AD and related dementias. This antibody enables:

Disease Mechanism Insights

  • Tau Hyperphosphorylation: S202 phosphorylation destabilizes microtubules and promotes tau aggregation into β-sheet-rich structures, a precursor to NFTs .

  • Diagnostic Potential: Detects early-stage tauopathy in brain tissue and biofluids .

Applications in Research

TechniqueDilution RecommendationsKey Observations
ELISA1:1000–1:10,000 High specificity for pS202 vs. non-phosphorylated tau
Flow Cytometry1:50–1:200 Stains neurons with aberrant tau phosphorylation
Immunohistochemistry1:50–1:200 Highlights neurofibrillary tangles in AD brain sections
Western Blotting1:1000 Detects bands at 32–72 kDa (varies by isoform phosphorylation)

Specificity and Cross-Reactivity

  • Target Validation:

    • Positive Control: Tau S202-phosphorylated peptide .

    • Negative Controls: Non-phosphorylated tau peptides (e.g., T181, S214) .

  • Cross-Reactivity: Minimal to non-phosphorylated tau or unrelated proteins (e.g., BSA) .

Molecular Weight and Isoform Detection

IsoformPredicted MWObserved MWSource
Full-length tau~79 kDa32–72 kDa
Truncated tau~40–50 kDaVaries

Discrepancies in observed MW stem from alternative splicing (e.g., 2N, 3R, 4R isoforms) and post-translational modifications .

Role in Tauopathy Pathogenesis

  • Phosphomimetic Mutations: Introduction of S202D (aspartate) mutations in recombinant tau promotes assembly into paired helical filaments (PHFs), mimicking AD-associated tau folding .

  • Kinase Involvement: GSK-3β, PKA, and cdk5 phosphorylate S202, linking metabolic stress to tau pathology .

Diagnostic and Therapeutic Potential

  • Biomarker Development: S202 phosphorylation is detectable in cerebrospinal fluid (CSF) and plasma, aiding early AD diagnosis .

  • Immunotherapy: Targeting pS202 tau may disrupt NFT formation, though clinical trials are pending .

Product Specs

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

The expression of phospho-MAPT (S202) recombinant monoclonal antibody typically involves the initial step of incorporating the MAPT antibody-encoding gene into expression vectors. These vectors are then introduced into host cells via polyethyleneimine-mediated transfection. Subsequent culturing of the host cells leads to the production and secretion of these antibodies. After purification through affinity chromatography, the antibodies' functionality is evaluated using ELISA and FC assays, demonstrating their specific binding to the human MAPT protein phosphorylated at S202.

MAPT, when phosphorylated at S202 and other sites, can modulate the stability and dynamics of microtubules in neurons. Phosphorylation of MAPT at S202, in particular, is associated with tau hyperphosphorylation and aggregation, contributing to neuronal damage and cognitive decline in Alzheimer's disease.

Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery times may vary depending on the shipping method or location. Please consult your local distributors for specific delivery times.
Synonyms
Microtubule-associated protein tau (Neurofibrillary tangle protein) (Paired helical filament-tau) (PHF-tau), MAPT, MAPTL MTBT1 TAU
Target Names
Uniprot No.

Target Background

Function

MAPT, also known as Tau, plays a crucial role in promoting microtubule assembly and stability within neurons. This function is essential for maintaining neuronal polarity. The C-terminus of MAPT binds to axonal microtubules, while the N-terminus interacts with neural plasma membrane components. This suggests that MAPT functions as a linker protein between these two structures. Axonal polarity is determined by the localization of MAPT within the neuronal cell body, specifically in the domain defined by the centrosome. The shorter isoforms of MAPT allow for cytoskeletal plasticity, while the longer isoforms may preferentially contribute to its stabilization.

Gene References Into Functions
  1. Genetic manipulation of Sirt3 revealed that amyloid-beta increased levels of total tau and acetylated tau through its modulation of Sirt3. PMID: 29574628
  2. Research suggests that both the small heat shock protein HspB1/Hsp27 and the constitutive chaperone Hsc70/HspA8 interact with tau to prevent tau-fibril/amyloid formation. Chaperones from different families play distinct but complementary roles in preventing tau-fibril/amyloid formation. (HspB1 = heat shock protein family B small member 1; Hsc70 = heat shock protein family A Hsp70) PMID: 29298892
  3. A 2.0-kDa peptide, biochemically and immunologically resembling the injected amino terminal tau 26-44, was endogenously detected in vivo and present in hippocampal synaptosomal preparations from Alzheimer's disease subjects. PMID: 29508283
  4. A study identified new bona fide human brain circular RNAs produced from the MAPT locus. PMID: 29729314
  5. TAU attaches to brain lipid membranes where it self-assembles in a cation-dependent manner. PMID: 29644863
  6. Microtubule hyperacetylation enhances KL1-dependent micronucleation under a Tau deficiency in mammary epithelial cells. PMID: 30142893
  7. This article presents key studies of tau in oligodendrocytes and select important studies of tau in neurons. The extensive work on tau in neurons has considerably advanced the understanding of how tau promotes either health or disease. [review] PMID: 30111714
  8. Zn2 + enhances Tau aggregation-induced apoptosis and toxicity in neuronal cells. PMID: 27890528
  9. Tau binds to synaptic vesicles via its N-terminal domain and interferes with presynaptic functions. PMID: 28492240
  10. A study identifies a potential "two-hit" mechanism in which tau acetylation disengages tau from microtubules (MT) and also promotes tau aggregation. Thus, therapeutic approaches to limit tau K280/K281 acetylation could simultaneously restore MT stability and ameliorate tau pathology in Alzheimer's disease and related tauopathies. PMID: 28287136
  11. In vitro neuroprotective effects of naringenin nanoemulsion against beta-amyloid toxicity through the regulation of amyloidogenesis and tau phosphorylation. PMID: 30001606
  12. To confirm the neuroprotective role of 24-OH, in vivo experiments were run on mice that express human tau without spontaneously developing tau pathology (hTau mice), by means of the intracerebroventricular injection of 24-OH. PMID: 29883958
  13. These findings suggest a relatively homogeneous clinicopathological phenotype in P301L MAPT mutation carriers in this series. This phenotype might help in the differential diagnosis from other tauopathies and be a morphological hint for genetic testing. The haplotype analysis results suggest a founder effect of the P301L mutation in this area. PMID: 28934750
  14. Report that the interaction of Tau with vesicles results in the formation of highly stable protein/phospholipid complexes. These complexes are toxic to primary hippocampal cultures and are detected by MC-1, an antibody recognizing pathological Tau conformations. The core of these complexes is comprised of the PHF6* and PHF6 hexapeptide motifs, the latter in a beta-strand conformation. PMID: 29162800
  15. A more selective group of neurons appears to be affected in frontotemporal lobar degeneration (FTLD)-TDP and FTLD-FUS than in FTLD-tau PMID: 28984110
  16. Our data show that the hyperacetylation of Tau by p300 histone acetyltransferase (HAT) disfavors liquid-liquid phase separation, inhibits heparin-induced aggregation, and impedes access to LLPS-initiated microtubule assembly PMID: 29734651
  17. Because neurofibrillary tangles are aberrant intracellular inclusions formed in AD patients by hyperphosphorylated tau, it was initially proposed that phosphorylated and/or aggregated intracellular tau protein was causative of neuronal death. However, recent studies suggest a toxic role for non-phosphorylated and non-aggregated tau when it is located in the brain extracellular space. [review] PMID: 29584657
  18. MAPT rs242557G/A genetic polymorphism is associated with susceptibility to sporadic AD, and individuals with a GG genotype of rs242557G/A might be at a lower risk. PMID: 29098924
  19. A study indicates that there are at least two common patterns of TDP-43 and tau protein misfolding in human brain aging. In patients lacking substantial Alzheimer's disease pathology, cerebral age-related TDP-43 with sclerosis (CARTS) cases tend to have tau neurofibrillary tangles in the hippocampal dentate granule neurons, providing a potential proxy indicator of CARTS. PMID: 28281308
  20. Patients with Kii amyotrophic lateral sclerosis and parkinsonism-dementia complex (Kii ALS/PDC) had dislocated, multinucleated Purkinje cells and various tau pathologies in the cerebellum. These cerebellar abnormalities may provide new insights into the pathomechanism of Kii ALS/PDC and may provide a neuropathological marker for the condition. PMID: 28236345
  21. The studies findings indicate that p.E372G is a pathogenic microtubule-associated protein tau mutation that causes microtubule-associated protein tau similar to p.G389R. PMID: 27529406
  22. Solven ionic strength, temperature and polarity altered tau conformation dynamics. PMID: 29630971
  23. MAPT alternative splicing is associated with Neurodegenerative Diseases. PMID: 29634760
  24. High tau expression is associated with blood vessel abnormalities and angiogenesis in Alzheimer's disease. PMID: 29358399
  25. We identified common splice factors hnRNP F and hnRNP Q regulating the haplotype-specific splicing of MAPT exon 3 through intronic variants rs1800547 and rs17651213 PMID: 29084565
  26. Cognitive impairment in progressive supranuclear palsy is associated with severity of progressive supranuclear palsy-related tau pathology. PMID: 29082658
  27. These observations indicate the ability of QUE to decrease tau protein hyperphosphorylation and thereby attenuate the associated neuropathology... these results support the potential of QUE as a therapeutic agent for AD and other neurodegenerative tauopathies. PMID: 29207020
  28. Increasing microtubule acetylation rescues human tau-induced microtubule defects and neuromuscular junction abnormalities in Drosophila. PMID: 28819043
  29. The findings reveal the ability of Bin1 to modify actin dynamics and provide a possible mechanistic connection between Bin1 and tau-induced pathobiological changes of the actin cytoskeleton. PMID: 28893863
  30. We find that both the generation of Abeta and the responsiveness of TAU to A-beta are affected by neuronal cell type, with rostral neurons being more sensitive than caudal neurons. PMID: 29153990
  31. The results of the current study indicate that variations in microtubule-associated protein tau influence cognition in progressive supranuclear palsy. PMID: 29076559
  32. The identification of mutations in MAPT, the gene that encodes tau, causing dementia and parkinsonism established the notion that tau aggregation is responsible for the development of disease. PMID: 28789904
  33. CSF tau proteins and their index differentiated between Alzheimer's disease or other dementia patients and cognitively normal subjects, while CSF levels of neurofilaments expressed as their index seem to contribute to the discrimination between patients with neuroinflammation and normal controls or AD patients PMID: 28947837
  34. Comparison of the distributions of tau pTyr18 and double-phosphorylated Syk in the transgenic mouse brain and human hippocampus showed that the phosphorylation of tyrosine 18 in tau already occurs at an early stage of tauopathy and increases with the progression of neurodegeneration. Syk appears unlikely to be a major kinase that phosphorylates tyrosine 18 of tau at the early stage of tauopathy. PMID: 28919467
  35. Study confirmed that Western diet did not exacerbate tau pathology in hTau mice, observed that voluntary treadmill exercise attenuates tau phosphorylation, and reported that caloric restriction seems to exacerbate tau aggregation compared to control and obese hTau mice. PMID: 28779908
  36. Study showed a gradual accumulation of nuclear tau in human cells during aging and its general co-localization with the DAPI-positive heterochromatin, which seems to be related to aging pathologies (neurodegenerative or cancerous diseases), where nuclear AT100 decreases drastically, a condition very evident in the more severe stages of the diseases. PMID: 28974363
  37. Methamphetamine can impair the endoplasmic reticulum-associated degradation pathway and induce neuronal apoptosis through endoplasmic reticulum stress, which is mainly mediated by abnormal CDK5-regulated Tau phosphorylation. PMID: 29705343
  38. Aha1 colocalized with tau pathology in brain tissue, and this association positively correlated with Alzheimer disease progression. PMID: 28827321
  39. Assessed the subcellular localization of tau45-230 fragment using tau45-230-GFP-transfected hippocampal neurons as well as neurons in which this fragment was endogenously generated under experimental conditions that induced neurodegeneration. Results suggested that tau45-230 could exert its toxic effects by partially blocking axonal transport along microtubules, contributing to the early pathology of Alzheimer's disease. PMID: 28844006
  40. frontotemporal dementia and parkinsonism linked to chromosome 17 tau with a mutation in the C-terminal region had different banding patterns, indicating a different phosphorylation pattern. PMID: 27641626
  41. Study demonstrated the presence of the smaller Tau isoform (352 amino acids), whose amount increases in differentiated SK-N-BE cells, with Tau-1/AT8 nuclear distribution related to the differentiation process. PMID: 29684490
  42. In primary-culture fetal astrocytes, streptozotocin increases phosphorylation of Tau at Ser396. alpha-boswellic acid reduced hyperphosphorylated tau (Ser404). Interruption in astroglial Reelin/Akt/Tau signaling pathways may have a role in Alzheimer disease. PMID: 27567921
  43. Screening of MAPT, GRN and CHCHD10 genes in Chinese patients with frontotemporal dementia (FTD) identified about 4.9% mutation carriers. Among the known FTD causative genes tested, MAPT and CHCHD10 play the most important roles in Chinese patients with sporadic FTD. PMID: 28462717
  44. Data show that aggregation of the Tau protein correlates with destabilization of the turn-like structure defined by phosphorylation of Ser202/Thr205. PMID: 28784767
  45. Deletion or inhibition of the cytoplasmic shuttling factor HDAC6 suppressed neuritic tau bead formation in neurons. PMID: 28854366
  46. We propose that the H2 haplotype, which expresses reduced 4R tau compared with the H1 haplotype, may exert a protective effect as it allows for more fluid mitochondrial movement along axons with high energy requirements, such as the dopaminergic neurons that degenerate in PD. PMID: 28689993
  47. Results find that overexpression of hTau increases intracellular calcium, which in turn activates calpain-2 and induces degradation of alpha4 nAChR. PMID: 27277673
  48. When misfolded tau assemblies enter the cell, they can be detected and neutralized via a danger response mediated by tau-associated antibodies and the cytosolic Fc receptor tripartite motif protein 21 (TRIM21) PMID: 28049840
  49. Stress granules and TIA-1 play a central role in the cell-to-cell transmission of Tau pathology. PMID: 27460788
  50. A clinicopathologic study shows inter- and intra-familial clinicopathologic heterogeneity of FTDP-17 due to MAPT p.P301L mutation, including globular glial tauopathy in one patient. PMID: 27859539
Gene References Into Functions Show More Less

Show More

Hide All

Database Links

HGNC: 6893

OMIM: 157140

KEGG: hsa:4137

STRING: 9606.ENSP00000340820

UniGene: Hs.101174

Involvement In Disease
Frontotemporal dementia (FTD); Pick disease of the brain (PIDB); Progressive supranuclear palsy 1 (PSNP1); Parkinson-dementia syndrome (PARDE)
Subcellular Location
Cytoplasm, cytosol. Cell membrane; Peripheral membrane protein; Cytoplasmic side. Cytoplasm, cytoskeleton. Cell projection, axon. Cell projection, dendrite. Secreted.
Tissue Specificity
Expressed in neurons. Isoform PNS-tau is expressed in the peripheral nervous system while the others are expressed in the central nervous system.

Q&A

What is the molecular structure and function of the MAPT protein?

MAPT (microtubule-associated protein tau) is a neuronal protein that promotes microtubule assembly and stability, potentially contributing to the establishment and maintenance of neuronal polarity. Structurally, MAPT functions as a linker protein where the C-terminus binds axonal microtubules while the N-terminus binds neural plasma membrane components. This arrangement allows MAPT to serve as a crucial bridge between cytoskeletal elements and membrane structures . The protein has a calculated molecular weight of approximately 79 kDa and contains multiple domains that regulate its binding affinity to various cellular components . Tau's functionality is heavily regulated through post-translational modifications, particularly phosphorylation at specific residues including serine 202, which can dramatically alter its binding properties and cellular function.

How does a phospho-specific antibody differ from general anti-tau antibodies?

Phospho-specific antibodies like the Phospho-MAPT (S202) antibody are engineered to recognize tau protein only when phosphorylated at specific residues (in this case, serine 202). This high specificity distinguishes them from general anti-tau antibodies that bind to tau regardless of its phosphorylation state . The epitope of phospho-specific antibodies contains the phosphorylated residue, making them valuable tools for distinguishing between normal tau and pathological hyperphosphorylated forms. For instance, the Phospho-MAPT (S202) antibody shows no cross-reactivity with normal (non-phosphorylated) tau, allowing researchers to selectively detect and quantify the phosphorylated species that are associated with disease states . This specificity is critical for research applications examining tau pathology progression and for diagnostic approaches that aim to detect early changes in tau phosphorylation before overt pathology develops.

How does phosphorylation at S202 affect tau's "paperclip" conformation and PAD exposure?

Recent structural studies using FRET analysis have revealed that phosphorylation at the S202 site (particularly in combination with S199 and T205) disrupts tau's native "paperclip" conformation. This conformational change causes the N-terminus to extend outward, exposing the Phosphatase Activating Domain (PAD) . The exposed PAD then becomes available for interaction with protein phosphatase 1 (PP1), particularly the γ isoform. This structural alteration is significant because it represents a mechanistic link between tau phosphorylation and downstream signaling effects. Experimentally, researchers have demonstrated this phenomenon using phosphomimetic mutations (S/T to E) at these sites, which reproduce the conformational changes seen with actual phosphorylation . The functional consequence of this PAD exposure includes aberrant activation of PP1γ and subsequent disruption of axonal transport, connecting molecular changes in tau structure directly to neuronal dysfunction observed in tauopathies.

What is the relationship between S202 phosphorylation and other tau phosphorylation sites in disease progression?

Tau phosphorylation in pathological conditions occurs at multiple sites, with complex temporal and spatial relationships. S202 phosphorylation often occurs in conjunction with other sites, particularly T205 and S199, collectively forming epitopes recognized by antibodies such as AT8 . Recent evidence suggests AT8 recognition depends on phosphorylation at combinations of sites including S198, S199, S202, T205, S208, and/or S210 . In disease progression, phosphorylation at S202 appears to be an early event that precedes extensive tangle formation. The sequential phosphorylation pattern suggests a hierarchical process where certain phosphorylation events (like S202) may prime tau for additional modifications. Experimental models indicate that phosphomimetic mutations at the combination of S199/S202/T205 (psTau) significantly impair axonal transport in primary rat hippocampal neurons, highlighting how these specific phosphorylation events contribute to neuronal dysfunction . Understanding this complex phosphorylation cascade is critical for developing targeted therapeutic approaches that could interrupt the pathological progression at specific stages.

What molecular mechanisms link S202 phosphorylation to axonal transport impairment?

S202 phosphorylation contributes to axonal transport impairment through several interconnected molecular mechanisms. Primary among these is the PAD-dependent activation of PP1γ. When S202 (along with S199 and T205) becomes phosphorylated, the resulting conformational change exposes the PAD region, enhancing tau's interaction with PP1γ and increasing levels of active PP1γ in cells . This aberrant phosphatase activation then leads to dephosphorylation of motor proteins and their regulatory components, disrupting kinesin-based transport. Experimental evidence demonstrates that expression of phosphomimetic tau at S199/S202/T205 (psTau) significantly increases pause frequency in axonal transport in primary rat hippocampal neurons . Critically, deletion of the PAD domain in psTau significantly reduces interaction with PP1γ, eliminates increases in active PP1γ levels, and rescues axonal transport impairment, confirming the PAD-dependent nature of this pathogenic pathway . This mechanistic insight connects molecular tau modifications directly to functional deficits in neurons, providing potential targets for therapeutic intervention.

What are the optimal protocols for expression and purification of Phospho-MAPT (S202) recombinant monoclonal antibodies?

Expression and purification of Phospho-MAPT (S202) recombinant monoclonal antibodies involves a systematic multi-step process. The recommended protocol begins with incorporating the MAPT antibody-encoding gene into appropriate expression vectors . These vectors are then introduced into host cells (commonly HEK293F cells) via polyethyleneimine-mediated transfection . After transfection, host cells are cultured under optimized conditions to maximize antibody production and secretion. The subsequent purification process typically employs affinity chromatography to isolate the antibody with high purity .

A standardized protocol includes:

  • Vector preparation with antibody-encoding gene (24-48 hours)

  • Transfection of host cells using polyethyleneimine (4-6 hours)

  • Cell culture and antibody expression (5-7 days)

  • Harvest of cell culture supernatant containing secreted antibodies

  • Purification via protein A/G affinity chromatography

  • Quality control testing using ELISA and flow cytometry to confirm specific binding to phosphorylated MAPT (S202)

For optimal results, maintain sterile conditions throughout the process and validate antibody functionality using positive controls containing known phosphorylated tau proteins.

What experimental methods are most effective for validating the specificity of Phospho-MAPT (S202) antibodies?

Validating the specificity of Phospho-MAPT (S202) antibodies requires multiple complementary approaches to ensure they recognize only phosphorylated S202 epitopes without cross-reactivity. The most reliable validation protocol combines:

  • Western blotting with phosphatase controls: Sample treatment with lambda phosphatase should eliminate antibody binding, confirming phospho-specificity .

  • Peptide competition assays: Pre-incubation of the antibody with phosphorylated and non-phosphorylated peptides containing the S202 site. Only phosphorylated peptides should block antibody binding .

  • Immunoreactivity comparison: Testing against samples containing:

    • Wild-type tau

    • Tau with S202A mutation (prevents phosphorylation)

    • Tau with S202E mutation (phosphomimetic)

    • Hyperphosphorylated tau from AD brain tissue

  • Cross-reactivity assessment: Testing against other phosphorylated sites in tau to confirm epitope specificity, particularly against phosphorylation at nearby residues S199 and T205 .

  • Flow cytometry validation: For cell-based applications, using established tau-expressing neuronal cell lines with and without phosphatase inhibitor treatment .

A thorough validation should demonstrate no cross-reactivity with normal tau while showing strong binding to PHF-tau from AD tissue samples .

What are the optimal sample preparation methods for detecting phosphorylated tau at S202 in different biological specimens?

Sample preparation methods must be tailored to the specific biological specimen while preserving phosphorylation status. The following approaches are recommended based on sample type:

For tissue samples:

  • Rapid post-mortem collection (<12 hours) is critical to prevent dephosphorylation

  • Immediate snap-freezing in liquid nitrogen

  • Homogenization in buffer containing phosphatase inhibitors (typically sodium fluoride, sodium pyrophosphate, sodium orthovanadate, and β-glycerophosphate)

  • Use of detergents appropriate for subcellular location (e.g., RIPA buffer for total tau, sucrose buffer for cytoskeletal fraction)

For CSF samples:

  • Collection in polypropylene tubes to prevent protein adsorption

  • Addition of phosphatase inhibitor cocktail immediately upon collection

  • Processing within 4 hours of collection or storage at -80°C

  • Recommended dilution of 5-fold to 50-fold for ELISA applications

For cell cultures:

  • Direct lysis in phosphatase-inhibitor containing buffer

  • Differential extraction to separate soluble and insoluble tau fractions

  • When using flow cytometry, fixation with 4% paraformaldehyde followed by permeabilization with 0.1% Triton X-100

The detection ranges for quantitative assays typically span from 2.7 pM to 1961 pM with sensitivity around 0.93 pM . To ensure optimal results, all samples should be processed with appropriate positive and negative controls, including non-phosphorylated tau and hyperphosphorylated tau standards.

How can researchers distinguish between physiological and pathological levels of S202 phosphorylation?

Distinguishing between physiological and pathological S202 phosphorylation requires multiple analytical approaches and careful data interpretation. Physiological tau phosphorylation occurs transiently and at much lower levels than pathological hyperphosphorylation. To differentiate between these states:

  • Quantitative comparison: Use quantitative ELISA with a sensitivity of 0.93 pM and detection range of 2.7-1961 pM to establish baseline phosphorylation levels in control samples versus pathological specimens . Typically, a 3-5 fold increase above baseline indicates potential pathology.

  • Co-occurrence analysis: Examine phosphorylation at multiple sites simultaneously. Pathological tau typically shows concurrent phosphorylation at S202 along with T205, S199, and other sites, while physiological phosphorylation may be more site-specific .

  • Solubility fractionation: Pathological phospho-tau increasingly shifts to detergent-insoluble fractions, while physiological phospho-tau remains largely soluble.

  • Cellular localization: Use immunocytochemistry to assess subcellular distribution. Pathological phospho-tau redistributes from axons to soma and dendrites, while physiological phospho-tau remains predominantly axonal .

  • Conformation-specific analysis: Use antibodies that detect specific pathological conformations that result from hyperphosphorylation, such as those recognizing the "paperclip" conformational change .

It's important to include age-matched controls in any analysis, as baseline phosphorylation levels may increase with normal aging but not to the extent seen in pathological conditions.

What are the most common methodological pitfalls when working with Phospho-MAPT (S202) antibodies and how can they be avoided?

Working with Phospho-MAPT (S202) antibodies presents several methodological challenges that can compromise research results. Common pitfalls and their solutions include:

  • Post-mortem dephosphorylation:

    • Pitfall: Rapid dephosphorylation after death or sample collection

    • Solution: Collect samples quickly (<12 hours post-mortem) and immediately add phosphatase inhibitors

  • Fixation artifacts:

    • Pitfall: Over-fixation can mask epitopes; under-fixation can allow dephosphorylation

    • Solution: Optimize fixation protocol (typically 10-15 minutes with 4% paraformaldehyde) and validate with positive controls

  • Cross-reactivity misinterpretation:

    • Pitfall: Some antibodies recognize multiple phosphorylation sites

    • Solution: Use multiple antibodies targeting different epitopes and perform blocking experiments with specific phosphopeptides

  • Inconsistent sample processing:

    • Pitfall: Variable phosphorylation levels due to processing delays

    • Solution: Standardize time from collection to processing and maintain consistent temperature conditions

  • Quantification errors:

    • Pitfall: Nonlinear relationship between signal and phosphorylation level

    • Solution: Use quantitative standards and ensure measurements fall within the antibody's validated detection range (2.7-1961 pM)

  • Storage-induced dephosphorylation:

    • Pitfall: Phosphorylation loss during sample storage

    • Solution: Store samples at -80°C with phosphatase inhibitors and minimize freeze-thaw cycles

  • Binding interference:

    • Pitfall: Buffer components affecting antibody-epitope interaction

    • Solution: Test antibody performance in your specific buffer system and optimize dilution ratios (typically FC:1:50-1:200)

Implementing rigorous controls, including phosphatase-treated samples, non-phosphorylated tau, and known phosphorylated standards, is essential for validating results and avoiding misinterpretation.

How can researchers reconcile conflicting data when different phospho-specific antibodies show varying results for the same S202 epitope?

Conflicting results between different phospho-specific antibodies targeting the S202 epitope represent a significant challenge in tau research. This discrepancy often stems from variations in epitope recognition and can be reconciled through systematic analysis:

  • Epitope mapping characterization:
    Different antibodies may recognize slightly different epitopes surrounding S202. Some antibodies require only S202 phosphorylation, while others (like AT8) recognize combinations including S202 plus T205 and/or S199 . Create an epitope map for each antibody by testing against synthetic peptides with defined phosphorylation patterns.

  • Antibody validation comparison:
    Assess each antibody's specificity using:

    • Phosphatase treatment sensitivity

    • Peptide competition assays

    • Reactivity against site-directed mutants (S202A, S202E)

    • Cross-reactivity with other phosphorylation sites

  • Conformational influences:
    S202 accessibility varies with tau's conformation. Some antibodies may preferentially detect specific conformational states that expose S202 differently . Test antibodies against both soluble and aggregated tau to determine conformational preferences.

  • Technical approach harmonization:
    Different detection methods (WB, ELISA, IHC) may affect epitope accessibility. When possible, compare antibodies using the same detection platform and sample preparation method.

  • Multiple antibody consensus approach:
    Use a panel of antibodies with overlapping epitopes and consider results reliable only when multiple antibodies show consistent findings.

  • Mass spectrometry validation:
    For definitive resolution, use phospho-proteomics to directly measure S202 phosphorylation independent of antibody-based detection.

A systematic reconciliation table comparing antibody characteristics can help interpret conflicting results:

CharacteristicAntibody AAntibody BReconciliation Approach
Epitope requirementsS202 onlyS202+T205Check for multi-site phosphorylation
Conformation dependenceLinear epitopeConformationalTest in native and denaturing conditions
Sensitivity (pM)0.935.2Consider detection threshold differences
Cross-reactivityMinimalModerateValidate with blocking peptides

By implementing these approaches, researchers can better understand the source of discrepancies and develop more nuanced interpretations of phosphorylation data.

How does the pattern of S202 phosphorylation differ across various tauopathies?

S202 phosphorylation patterns show distinct characteristics across different tauopathies, providing valuable diagnostic and mechanistic insights. The pattern variations include:

  • Alzheimer's Disease (AD):

    • Early and extensive S202 phosphorylation, typically co-occurring with T205 phosphorylation

    • Predominantly associated with 3R/4R tau isoforms in neurofibrillary tangles

    • Progressive spread following Braak staging pattern

  • Progressive Supranuclear Palsy (PSP):

    • S202 phosphorylation primarily on 4R tau isoforms

    • More pronounced in subcortical regions compared to AD

    • Often colocalized with phosphorylation at other sites but with distinctive regional distribution

  • Corticobasal Degeneration (CBD):

    • S202 phosphorylation on both neuronal and glial tau aggregates

    • Distinctive pattern in astrocytic plaques and threadlike inclusions

    • Predominantly affects 4R tau isoforms

  • Pick's Disease:

    • S202 phosphorylation primarily on 3R tau isoforms

    • Characteristic distribution in Pick bodies within the dentate gyrus and frontal cortex

    • Often lower intensity of S202 phosphorylation compared to AD

  • Frontotemporal Dementia (FTD):

    • Variable S202 phosphorylation depending on subtype

    • In FTDP-17, pattern depends on specific tau mutation

    • Some mutations alter the propensity for S202 phosphorylation

These disease-specific patterns suggest distinct pathogenic mechanisms and kinase/phosphatase imbalances underlying different tauopathies. The analysis of S202 phosphorylation patterns, particularly when combined with other phosphorylation sites, can aid in differential diagnosis and may guide targeted therapeutic approaches for specific tauopathies.

What experimental approaches can determine the temporal relationship between S202 phosphorylation and tau aggregation?

Understanding the temporal relationship between S202 phosphorylation and tau aggregation requires sophisticated experimental approaches that can capture the dynamic progression of these events. Researchers can employ the following methods:

  • Time-course studies in cellular models:

    • Express tau in neuronal cell lines and induce phosphorylation using specific kinase activators

    • Sample at defined intervals (0-72 hours) and simultaneously measure:

      • S202 phosphorylation levels via quantitative ELISA (sensitivity: 0.93 pM)

      • Tau oligomerization via non-denaturing PAGE

      • Insoluble tau formation via sarkosyl fractionation

    • This approach can establish whether S202 phosphorylation precedes or follows aggregation onset

  • Inducible transgenic animal models:

    • Use doxycycline-regulated expression of human tau in mice

    • After induction, track progressive changes in:

      • S202 phosphorylation using phospho-specific antibodies

      • Conformational changes using FRET-based sensors

      • Aggregation using thioflavin-S staining and biochemical fractionation

    • Allows temporal mapping in a physiologically relevant context

  • Seeding and propagation experiments:

    • Apply tau seeds with defined phosphorylation states to neuronal cultures

    • Monitor progression of S202 phosphorylation in relation to seed-induced aggregation

    • Use phosphomimetic mutations (S202E) to determine if phosphorylation at this site enhances seeding propensity

  • Super-resolution microscopy combined with phospho-specific labeling:

    • Apply techniques like STORM or STED with phospho-S202 antibodies

    • Co-label with aggregation-specific markers

    • Permits visualization of spatial and temporal relationships at nanometer resolution

  • In vitro aggregation kinetics with recombinant tau:

    • Compare aggregation rates between:

      • Non-phosphorylated tau

      • Enzymatically phosphorylated tau (using GSK-3β or other kinases)

      • Phosphomimetic tau (S202E)

    • Monitor aggregation using thioflavin T fluorescence, dynamic light scattering, and electron microscopy

These complementary approaches can resolve whether S202 phosphorylation represents an early initiating event in tau pathology or occurs as a consequence of initial aggregation, providing crucial insights for therapeutic targeting.

How can Phospho-MAPT (S202) antibodies be utilized in developing novel therapeutic approaches for tauopathies?

Phospho-MAPT (S202) antibodies offer multiple avenues for therapeutic development in tauopathies, serving as both tools for target validation and potential therapeutic agents themselves. Strategic applications include:

  • Antibody-based immunotherapies:

    • Passive immunization approaches using humanized versions of phospho-S202 antibodies

    • These can facilitate clearance of phosphorylated tau species before aggregation progresses

    • Target validation studies should assess antibody penetration across the blood-brain barrier and measure engagement with phospho-S202 tau using quantitative assays (detection range: 2.7-1961 pM)

  • Screening platforms for kinase inhibitors:

    • Develop high-throughput assays using phospho-S202 antibodies to screen compounds that reduce S202 phosphorylation

    • Focus on kinases known to phosphorylate S202 (GSK-3β, cdk5, PKA)

    • Measure changes in phosphorylation level using flow cytometry (recommended dilution: 1:50-1:200)

  • PAD-targeting therapeutic development:

    • Since S202 phosphorylation exposes the PAD domain and leads to axonal transport deficits

    • Design compounds that either:

      • Prevent PAD exposure despite phosphorylation

      • Block PAD interaction with downstream effectors like PP1γ

    • Validate using axonal transport rescue assays in primary neurons

  • Biomarker development for clinical trials:

    • Use quantitative phospho-S202 assays (sensitivity: 0.93 pM) to monitor treatment efficacy

    • Develop CSF and plasma assays for phospho-S202 tau as pharmacodynamic markers

    • Correlate changes in phospho-S202 levels with clinical outcomes

  • Conformation-stabilizing approaches:

    • Design peptides or small molecules that prevent the conformational change induced by S202 phosphorylation

    • Target the "paperclip" conformation to prevent PAD exposure even when phosphorylation occurs

    • Validate using FRET-based conformational assays

  • Gene therapy approaches:

    • Develop viral vectors expressing intrabodies derived from phospho-S202 antibodies

    • These intracellular antibodies can bind nascent phosphorylated tau before aggregation

    • Target validation requires demonstration of specific binding to S202-phosphorylated tau without affecting normal tau function

Each therapeutic approach requires careful validation using phospho-specific antibodies to confirm target engagement and mechanism of action, with particular attention to potential effects on physiological tau function.

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.