MAPT Human Recombinant (Isoform 3) fused with a 20 amino acid His tag at N-terminus produced in E.Coli is a single, non-glycosylated, polypeptide chain containing 403 amino acids (1-383 a.a.) and having a molecular mass of 42.1kDa (Molecular size on SDS-PAGE will appear higher). The MAPT is purified by proprietary chromatographic techniques.
MGSSHHHHHH SSGLVPRGSH MAEPRQEFEV MEDHAGTYGL GDRKDQGGYT MHQDQEGDTD AGLKAEEAGI GDTPSLEDEA AGHVTQARMV SKSKDGTGSD DKKAKGADGK TKIATPRGAA PPGQKGQANA TRIPAKTPPA PKTPPSSGEP PKSGDRSGYS SPGSPGTPGS RSRTPSLPTP PTREPKKVAV VRTPPKSPSS AKSRLQTAPV PMPDLKNVKS KIGSTENLKH QPGGGKVQII NKKLDLSNVQ SKCGSKDNIK HVPGGGSVQI VYKPVDLSKV TSKCGSLGNI HHKPGGGQVE VKSEKLDFKD RVQSKIGSLD NITHVPGGGN KKIETHKLTF RENAKAKTDH GAEIVYKSPV VSGDTSPRHL SNVSSTGSID MVDSPQLATL ADEVSASLAK QGL.
Recombinant MAPT Human 383a.a. serves multiple experimental purposes in neurodegenerative disease research. Researchers typically employ this protein for:
Aggregation studies: Investigating the kinetics and mechanisms of tau aggregation, which is crucial for understanding pathological processes in tauopathies.
Fibrillization experiments: Examining the formation of tau fibrils under various conditions to model neurofibrillary tangle development.
Assay development: Creating standardized assays for tau protein detection, quantification, and characterization.
Standard reference material: Utilizing as internal controls or calibration standards in analytical procedures.
SDS-PAGE analysis: Studying protein migration patterns and structural properties .
When designing experiments using this protein, researchers should consider that commercially available preparations typically have >90% purity as determined by SDS-PAGE .
Recombinant MAPT Human 383a.a. is typically produced through recombinant DNA technology using bacterial expression systems. The methodological approach involves:
Expression system: The protein is commonly expressed in Escherichia coli using a DNA sequence encoding the human tau-383 (0N4R isoform) .
Purification process: Following expression, the protein undergoes purification procedures to achieve >90% purity as verified by SDS-PAGE analysis .
Final formulation: The purified protein is generally provided as a white lyophilized powder for stability and convenience in shipping and storage .
For optimal storage and handling:
Storage temperature: -20°C is the recommended storage temperature to maintain protein integrity .
Shipping conditions: The protein can be shipped at ambient temperature, though immediate transfer to -20°C upon receipt is advised .
Working concentration: Researchers should reconstitute the lyophilized powder according to experimental requirements, documenting concentration and buffer conditions.
These standardized production and storage protocols ensure batch-to-batch consistency in both purity and quality, which is critical for reproducible research results .
When investigating MAPT Human 383a.a. in the context of neurofibrillary tangle formation, a multi-modal research approach is recommended:
In vitro aggregation assays:
Thioflavin T fluorescence assays to monitor fibril formation kinetics
Electron microscopy to characterize fibril morphology
Dynamic light scattering to assess oligomer and aggregate sizes
Sedimentation assays to quantify insoluble tau fractions
Cellular models:
Primary neuronal cultures expressing MAPT Human 383a.a.
Differentiated human iPSC-derived neurons for studying tau pathology in human cellular context
Microfluidic chamber systems to investigate cell-to-cell propagation mechanisms
Tissue analysis techniques:
Immunohistochemistry with phospho-tau specific antibodies
FRET-based approaches to study tau conformational changes
Super-resolution microscopy to visualize early-stage aggregates
Physiological readouts:
Electrophysiology to correlate tau aggregation with neuronal dysfunction
Calcium imaging to assess impact on neuronal signaling
Mitochondrial function assays to evaluate metabolic consequences
Research has demonstrated that MAPT pathology, particularly related to mutations such as P301L, affects specific neuronal populations, including excitatory neurons in the medial entorhinal cortex and GABAergic interneurons in the hippocampus . Experimental designs should account for these regional and cell-type specificities.
Post-translational modifications (PTMs) significantly influence tau function and pathogenicity. When designing experiments to study PTMs of MAPT Human 383a.a., consider the following methodological framework:
PTM site identification:
Mass spectrometry (MS) approaches:
Liquid chromatography-tandem mass spectrometry (LC-MS/MS)
MALDI-TOF MS for peptide mapping
Top-down proteomics for intact protein analysis
Site-directed mutagenesis to create PTM-mimetic or PTM-deficient variants
Functional impact assessment:
Microtubule binding assays to evaluate effects on tau-microtubule interactions
Aggregation assays comparing modified and unmodified protein
Structural analyses using circular dichroism or NMR spectroscopy
Enzymatic studies:
Kinase/phosphatase assays to study modification dynamics
In vitro enzymatic modification systems
Inhibitor screening for modulating specific PTMs
Temporal dynamics:
Pulse-chase experiments to track PTM acquisition and turnover
Time-resolved structural studies
Sequential modification analysis
Spatial organization:
Proximity ligation assays in cellular systems
Subcellular fractionation combined with PTM-specific antibodies
Live-cell imaging with PTM-sensitive fluorescent reporters
Experimental controls should include:
Unmodified MAPT Human 383a.a. as negative control
Chemically modified standards as positive controls
Enzymatically modified standards to validate antibody specificity
When designing comparative studies between wild-type MAPT Human 383a.a. and mutant variants (such as P301L), researchers should implement the following methodological considerations:
Experimental system selection:
Cell-free systems for direct biochemical comparisons
Neuronal cell lines for cellular phenotypes
Primary neurons for physiological relevance
Animal models for in vivo complexity
Expression level considerations:
Matched expression levels between wild-type and mutant proteins
Inducible expression systems to control timing and magnitude
Single-cell analysis to account for expression heterogeneity
Structural and functional comparisons:
Detailed structural analysis using CD spectroscopy, NMR, or X-ray crystallography
Microtubule binding affinity assays
Aggregation propensity comparisons under identical conditions
Protein-protein interaction network mapping
Phenotypic readouts:
Cytoskeletal dynamics (microtubule stability, axonal transport)
Synaptic function measurements
Neuronal morphology analysis
Cell survival and stress response metrics
Advanced comparative approaches:
Differential interactome analysis
Unbiased multi-omics comparisons (proteomics, transcriptomics)
Computational modeling of structural differences
Research has shown that MAPT mutations can affect multiple biological processes, including neurovascular coupling and immune responses . For example, the P301L mutation has been associated with increased expression of adenosine A2A receptors in human frontal cortex, suggesting immune function alterations . Therefore, experimental designs should incorporate measurements of these broader physiological processes beyond direct tau protein analysis.
To thoroughly investigate MAPT Human 383a.a. interactions with microtubules, researchers should consider implementing the following experimental methodologies:
In vitro microtubule binding assays:
Co-sedimentation assays with pre-formed taxol-stabilized microtubules
Surface plasmon resonance to determine binding kinetics
Isothermal titration calorimetry for thermodynamic parameters
Microscale thermophoresis for binding affinity measurement
Structural analysis of the tau-microtubule complex:
Cryo-electron microscopy of tau-decorated microtubules
Hydrogen-deuterium exchange mass spectrometry to map binding interfaces
FRET-based approaches to measure binding dynamics
Cross-linking mass spectrometry to identify precise contact points
Functional microtubule dynamics assessments:
Turbidity assays to measure microtubule assembly rates
Total internal reflection fluorescence (TIRF) microscopy to visualize microtubule dynamics
Video-enhanced differential interference contrast microscopy
Optical tweezers to measure mechanical properties of tau-stabilized microtubules
Cellular analysis of tau-microtubule interactions:
Live-cell imaging with fluorescently tagged tau and tubulin
FRAP (Fluorescence Recovery After Photobleaching) to measure tau mobility
Proximity ligation assays to visualize tau-tubulin interactions in situ
Super-resolution microscopy to map tau distribution along microtubules
Experimental validation approaches:
Competition assays with microtubule-binding drugs
Domain deletion experiments to map critical binding regions
Phosphorylation-dependent binding studies
Research design should account for the presence of the four microtubule binding regions in MAPT Human 383a.a. (0N4R isoform) , which contribute to its specific binding characteristics and functional properties in stabilizing microtubule structures.
When investigating MAPT Human 383a.a. aggregation kinetics, researchers should implement the following methodological approaches to ensure robust and reproducible results:
Aggregation induction and monitoring techniques:
Thioflavin T/S fluorescence assays with continuous real-time monitoring
Dynamic light scattering for early oligomer formation detection
Turbidity measurements for macroscopic aggregation assessment
Atomic force microscopy for structural characterization at different time points
Sedimentation analysis for quantitative measurement of insoluble fractions
Experimental condition standardization:
Precisely defined buffer conditions (pH, ionic strength, temperature)
Removal of preformed aggregates through filtration or centrifugation
Consistent protein concentration determination methods
Controlled agitation conditions (stirring vs. shaking vs. static)
Seed-dependent vs. de novo aggregation protocols
Inducer/inhibitor testing methodology:
Concentration-response relationships for aggregation inducers (heparin, RNA, fatty acids)
IC50 determination for potential aggregation inhibitors
Time-of-addition experiments to determine stage-specific effects
Conformation-specific antibody probes to detect specific aggregate species
Kinetic analysis approaches:
Lag phase, growth phase, and plateau phase quantification
Nucleation-elongation kinetic modeling
Seeding efficiency calculations
Critical concentration determination
Data validation strategies:
Multiple complementary techniques to confirm findings
Independent preparation replicates to assess reproducibility
Internal standards for inter-experimental normalization
Statistical analysis appropriate for non-linear kinetic data
Researchers should be aware that MAPT Human 383a.a. is particularly suitable for aggregation studies and fibrillization experiments , making it an excellent model for investigating the molecular mechanisms underlying pathological tau aggregation in neurodegenerative diseases.
When designing crossover experiments involving MAPT Human 383a.a., researchers should implement rigorous methodological approaches based on established principles of experimental design:
Crossover design selection:
Period determination and washout considerations:
Include adequate washout periods between treatments
Assess potential carryover effects of MAPT Human 383a.a. interventions
Consider the biological half-life of administered treatments
Implement baseline measurements at each period start
Statistical analysis approach:
Practical implementation for MAPT experiments:
When comparing wild-type vs. mutant MAPT Human 383a.a. effects
When testing different post-translational modification states
When evaluating different aggregation inducers or inhibitors
Design validation:
Power analysis to determine appropriate sample size
Simulation studies to verify design efficiency
Pilot studies to estimate variability and effect sizes
Crossover designs are particularly valuable when investigating MAPT Human 383a.a. under multiple experimental conditions, as they allow for within-subject comparisons, reducing variability and increasing statistical power with smaller sample sizes .
Response surface methodology (RSM) provides a powerful approach for optimizing experimental conditions and understanding complex relationships between factors affecting MAPT Human 383a.a. behavior. Researchers should consider the following implementation framework:
Experimental design selection:
Factor selection and optimization targets:
For MAPT aggregation: temperature, pH, ionic strength, inducer concentration
For microtubule binding: tau concentration, tubulin concentration, GTP levels, buffer conditions
For phosphorylation studies: kinase concentration, ATP levels, incubation time, cofactors
Model fitting and analysis:
Optimization strategies:
Canonical analysis to identify stationary points
Ridge analysis for constrained optimization
Overlaid contour plots for multi-response optimization
Numerical optimization using desirability functions
Implementation in R:
RSM is particularly valuable for optimizing conditions for MAPT Human 383a.a. research applications such as:
Identifying optimal buffer conditions for tau protein stability
Determining ideal conditions for reproducible aggregation kinetics
Optimizing assay conditions for drug screening against tau aggregation
Maximizing yield and purity in recombinant tau production
Designing experiments to investigate region-specific effects of MAPT Human 383a.a. requires careful consideration of neuroanatomical specificity and appropriate model systems. Researchers should implement the following methodological framework:
Model system selection based on research question:
Brain region-specific primary neuronal cultures
Organotypic brain slice cultures maintaining regional architecture
Brain region-specific differentiated iPSCs
Spatially-resolved in vivo models with region-specific expression
Delivery and expression approaches:
Region-specific viral delivery systems
Cell type-specific promoters for targeted expression
Inducible expression systems with spatial control
Stereotactic injection techniques for precise anatomical targeting
Analysis of regional vulnerability:
Comparative susceptibility assays across neuronal populations
Regional protein interaction network mapping
Transcriptomic profiling of regional responses to MAPT
Proteomic analysis of region-specific post-translational modifications
Functional readouts with spatial resolution:
Region-specific electrophysiology
Spatial transcriptomics for molecular responses
Circuit-specific functional imaging
Spatially-resolved proteomics
Validation strategies:
Cross-validation across multiple model systems
Correlation with human neuropathological data
Comparison with in vivo imaging from patient studies
Research has demonstrated regional specificity in MAPT pathology, with studies showing that P301L mutations affect the medial entorhinal cortex, disrupting grid cells that control spatial navigation and impairing spatial memory . Additionally, entorhinal cortical neurons expressing wolframin-1 that project to hippocampal CA1 neurons have been implicated in propagating misfolded tau and disrupting hippocampal synapses . Experimental designs should account for these regional vulnerabilities and connectivity patterns.
For tau aggregation experiments specifically, consider kinetic curve fitting with non-linear mixed effects models to account for batch-to-batch variability while extracting meaningful parameters such as lag time, maximum rate, and final plateau .
When faced with contradictory findings in the MAPT Human 383a.a. research literature, researchers should implement a systematic approach to interpretation and resolution:
Methodological comparison framework:
Detailed examination of experimental conditions:
Protein preparation methods and purity
Buffer composition and pH
Temperature and other physical conditions
Presence of additives or co-factors
Analysis of detection methods and their sensitivity/specificity
Evaluation of model systems (cell-free, cellular, animal models)
Assessment of statistical approaches and power
Meta-analytical approaches:
Systematic review of contradictory findings
Quantitative meta-analysis where appropriate
Subgroup analyses to identify condition-dependent effects
Publication bias assessment
Mechanistic reconciliation strategies:
Hypothesis generation for condition-dependent effects
Design of critical experiments to test competing hypotheses
Development of integrated models that account for apparent contradictions
Identification of potentially unknown variables or confounders
Experimental validation approaches:
Direct replication attempts under original conditions
Systematic variation of key parameters to identify sensitivity factors
Independent verification using complementary methodologies
Collaborative cross-laboratory validation studies
Synthesis and communication:
Development of consensus statements where possible
Clear articulation of remaining uncertainties
Transparent reporting of methodological details
Open data sharing to enable community reanalysis
When interpreting contradictions, consider that recent research has demonstrated heterogeneity in P301L mutation effects across brain regions, with varying patterns of atrophy and pathology observed in different carriers . This biological heterogeneity may explain some apparently contradictory findings in the literature.
When incorporating MAPT Human 383a.a. into drug discovery campaigns, researchers should implement the following methodological framework to maximize efficiency and translational relevance:
Assay development and validation:
Primary screening assays:
Thioflavin T aggregation inhibition assays
Microtubule binding competition assays
Cellular tau aggregation reporter systems
Post-translational modification modulator screens
Validation criteria:
Z' factor >0.5 for high-throughput screening
Signal-to-background ratio optimization
Minimized coefficient of variation across replicates
Appropriate positive and negative controls
Screening cascade design:
Primary screens at single concentration (typically 10 μM)
Dose-response confirmation for hits (IC50 determination)
Orthogonal assay validation of confirmed hits
Counter-screens for selectivity and specificity
ADME/Tox profiling of validated hits
Target engagement demonstration:
Cellular thermal shift assays (CETSA)
Surface plasmon resonance for direct binding
NMR-based fragment screening
Hydrogen-deuterium exchange mass spectrometry
Advanced mechanistic characterization:
Time-resolved structural studies
Kinetic mechanism of action determination
Structure-activity relationship development
Computational modeling of binding interactions
Translational validation approaches:
Neuronal models with endogenous tau expression
Ex vivo validation in brain slice cultures
In vivo proof-of-concept studies in relevant models
Biomarker development for clinical translation
Researchers should leverage the high purity (>90% by SDS-PAGE) of recombinant MAPT Human 383a.a. preparations for consistent assay performance , while considering the specific pathophysiological relevance of the 0N4R isoform in tauopathies.
When investigating the cell-to-cell propagation of MAPT Human 383a.a., researchers should implement the following methodological framework to accurately capture this complex process:
Model system selection and validation:
Microfluidic chamber systems for directional propagation
Co-culture systems with donor and recipient populations
3D organoid models for complex cellular architectures
In vivo models with spatially restricted initial expression
Seed preparation and characterization:
Defined aggregation protocols for reproducible seed generation
Detailed characterization of seed size distribution and morphology
Fluorescent labeling approaches for tracking
Assessment of seed stability under experimental conditions
Uptake and propagation monitoring:
Live-cell imaging with fluorescently tagged tau
Time-lapse microscopy to capture dynamic processes
Super-resolution approaches to visualize small aggregates
Flow cytometry for quantitative analysis of population-level changes
Mechanistic dissection approaches:
Genetic manipulation of endocytosis pathways
Pharmacological inhibition of specific transfer mechanisms
Isolation and characterization of extracellular vesicles
Identification of cellular receptors mediating uptake
Functional consequence assessment:
Electrophysiological measurements of neuronal function
Synaptic connectivity and plasticity evaluation
Network activity analysis using multi-electrode arrays
Correlation of propagation with cellular dysfunction
Experimental designs should account for research showing that entorhinal cortical neurons expressing wolframin-1 that project to hippocampal CA1 neurons are implicated in the propagation of misfolded tau and subsequent disruption of hippocampal synapses and memory function . This highlights the importance of considering specific neuronal subpopulations and their connectivity in propagation studies.
The field of MAPT research is rapidly evolving, with several emerging methodological approaches that offer new insights into tau biology and pathology. Researchers should consider implementing the following cutting-edge techniques:
Advanced structural biology approaches:
Cryo-electron microscopy for tau filament structures
NMR spectroscopy for dynamic structural ensembles
Single-molecule FRET for conformational dynamics
AlphaFold and other AI-based structural prediction tools
Spatial multi-omics integration:
Spatial transcriptomics combined with tau pathology mapping
Imaging mass spectrometry for spatial PTM profiling
Multiplexed protein imaging with cellular resolution
Integrated multi-modal data analysis frameworks
Advanced cellular models:
Patient-derived iPSC neurons with isogenic controls
Brain organoids with region-specific identity
Assembloids combining multiple brain regions
Microfluidic organ-on-chip platforms with circulatory components
In vivo approaches:
PET tracers with improved specificity for tau conformers
Optogenetic control of tau expression or aggregation
Live imaging of tau dynamics in intact organisms
Tau-targeting antisense oligonucleotides
Computational and AI-enhanced analysis:
Machine learning for prediction of tau aggregation properties
Network analysis of tau interactomes
Molecular dynamics simulations of tau-membrane interactions
Large-scale virtual screening for tau-targeting compounds
These emerging methodologies will help address current knowledge gaps regarding the complex biology of MAPT Human 383a.a., including the molecular mechanisms underlying isoform-specific functions, the structural basis of pathological aggregation, and the development of more targeted therapeutic approaches for tauopathies.
To comprehensively understand the complex biology of MAPT Human 383a.a., researchers should implement an integrated experimental framework that combines multiple methodological approaches:
Multi-scale experimental integration:
Molecular-level: Structural studies, binding assays, PTM mapping
Cellular-level: Live-cell imaging, functional assays, interactome analysis
Tissue-level: Organoids, brain slices, regional vulnerability assessment
Organism-level: Behavioral phenotyping, in vivo imaging, biomarker studies
Technical integration strategies:
Correlated light and electron microscopy (CLEM)
Integrated multi-omics approaches (proteomics, transcriptomics, metabolomics)
Combined functional and structural analyses
Computational integration of diverse experimental datasets
Temporal dimension incorporation:
Longitudinal studies across disease progression
Time-resolved molecular and cellular analyses
Developmental trajectory mapping
Aging-related changes in tau biology
Cross-validation framework:
Verification across multiple model systems
Orthogonal technique confirmation
Translation between in vitro and in vivo findings
Correlation with human patient data
Data integration methodology:
Bayesian integration of multiple data sources
Network-based data fusion approaches
Machine learning for pattern recognition across datasets
Knowledge graph construction from diverse evidence types
Integrated approaches are particularly valuable when studying the complex interactions between MAPT Human 383a.a. and various biological systems. For example, research has demonstrated that P301L mutant tau affects not only neuronal function but also vascular and immune processes , highlighting the need for multi-system analysis approaches that can capture these diverse pathological mechanisms.
Microtubule-Associated Protein Tau (MAPT), commonly referred to as Tau protein, is a critical component in the stabilization and assembly of microtubules in neuronal cells. The specific variant, Tau 383 a.a., is a human recombinant protein consisting of 383 amino acids. This variant is often used in research to understand its role in neurodegenerative diseases, particularly Alzheimer’s disease.
Tau protein exists in several isoforms, which are produced through alternative splicing of the MAPT gene. The Tau 383 a.a. variant, also known as 0N4R, lacks N-terminal inserts and contains four microtubule-binding repeats . This structure allows it to interact effectively with microtubules, promoting their stability and assembly .
Tau proteins are primarily found in neurons, where they bind to microtubules and promote their assembly and stability. This function is crucial for maintaining the structure and function of axons. The Tau 383 a.a. variant is particularly important in research due to its involvement in the formation of neurofibrillary tangles, a hallmark of Alzheimer’s disease .
In neurodegenerative diseases like Alzheimer’s, Tau proteins become hyperphosphorylated, leading to their detachment from microtubules and subsequent aggregation into paired helical filaments and neurofibrillary tangles . These aggregates disrupt neuronal function and contribute to cell death. Studying the Tau 383 a.a. variant helps researchers understand the mechanisms behind these pathological changes and develop potential therapeutic interventions.
Recombinant Tau 383 a.a. is widely used in various research applications, including: