MAPT Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Development and Engineering

MAPT R-mAbs are generated through advanced pipelines involving:

  • Gene Cloning: Variable domains of immunoglobulin genes from hybridomas are cloned into plasmid vectors .

  • Subclass Switching: Engineered backbones allow subclass switching (e.g., IgG1 to IgG2a) for multiplex applications without altering specificity .

  • Validation: Functional testing via Western blot (WB), immunocytochemistry (ICC), and immunohistochemistry (IHC) .

For example, clone 2E9 targets the conserved epitope KDRVQSKIGSLDNITHVPGG (amino acids 347–366) present in all human tau isoforms , while RM461 detects tau phosphorylated at Ser396 .

Target Specificity and Epitope Mapping

MAPT R-mAbs exhibit distinct binding profiles:

CloneTarget EpitopePhosphorylation SiteSpecies ReactivityApplications
2E9 KDRVQSKIGSLDNITHVPGGNone (pan-tau)Human, Mouse, RatWB, ICC, IHC
RM461 Synthetic phospho-Ser396Ser396Human, Mouse, RatWB, IHC (1:50–1:200)
CSB-RA... Phospho-Thr231Thr231HumanWB, ELISA

Phospho-specific antibodies like RM461 enable precise detection of pathological tau aggregates in Alzheimer’s disease .

Neurodegenerative Disease Research

  • Detects hyperphosphorylated tau in brain tissues via IHC, aiding Alzheimer’s diagnostics .

  • Quantifies tau isoforms in WB analyses of cerebrospinal fluid .

Mechanistic Studies

  • Investigates tau’s role in microtubule stabilization and axonal transport .

  • Assesses phosphorylation-dependent tau aggregation using phospho-specific clones .

Key Parameters

Parameter2E9 RM461 CSB-RA...
Host SpeciesMouseRabbitRabbit
IsotypeIgG1IgGIgG
Dilution Range (WB)1:5,000–1:10,0001:1,000–1:2,0001:500–1:5,000
StorageLyophilized (-20°C)Liquid (-20°C)Liquid (-20°C)

Validation Data

  • 2E9: Bands at 48–67 kDa in human brain lysates .

  • RM461: Specificity confirmed via peptide blocking; no cross-reactivity with non-phospho tau .

Emerging Trends and Challenges

  • Subclass Engineering: IgG subclass-switched R-mAbs enable simultaneous multiplex labeling .

  • Therapeutic Potential: Neutralizing tau aggregation with R-mAbs is under exploration for tauopathies .

  • Reproducibility: Recombinant formats enhance transparency and reproducibility in research .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days of receiving your order. Delivery timelines may vary depending on the purchasing method and location. Please consult your local distributor for specific delivery times.
Synonyms
AI413597 antibody; AW045860 antibody; DDPAC antibody; FLJ31424 antibody; FTDP 17 antibody; G protein beta1/gamma2 subunit interacting factor 1 antibody; MAPT antibody; MAPTL antibody; MGC134287 antibody; MGC138549 antibody; MGC156663 antibody; Microtubule associated protein tau antibody; Microtubule associated protein tau isoform 4 antibody; Microtubule-associated protein tau antibody; MSTD antibody; Mtapt antibody; MTBT1 antibody; MTBT2 antibody; Neurofibrillary tangle protein antibody; Paired helical filament tau antibody; Paired helical filament-tau antibody; PHF tau antibody; PHF-tau antibody; PPND antibody; PPP1R103 antibody; Protein phosphatase 1, regulatory subunit 103 antibody; pTau antibody; RNPTAU antibody; TAU antibody; TAU_HUMAN antibody; Tauopathy and respiratory failure antibody; Tauopathy and respiratory failure, included antibody
Target Names
Uniprot No.

Target Background

Function
TAU/MAPT promotes microtubule assembly and stability, potentially playing a role in establishing and maintaining neuronal polarity. The C-terminus binds to axonal microtubules, while the N-terminus interacts with neural plasma membrane components, suggesting that tau acts as a linker protein between these structures. Axonal polarity is determined by the localization of TAU/MAPT within the neuronal cell, specifically in the region of the cell body defined by the centrosome. Short isoforms of TAU/MAPT facilitate cytoskeletal plasticity, while longer isoforms may preferentially contribute to cytoskeletal stabilization.
Gene References Into Functions
  1. Genetic manipulations involving Sirt3 have revealed that amyloid-beta increases levels of total tau and acetylated tau through modulation of Sirt3. PMID: 29574628
  2. Evidence suggests that both the small heat shock protein HspB1/Hsp27 and the constitutive chaperone Hsc70/HspA8 interact with tau, preventing tau-fibril/amyloid formation. Chaperones from different families play distinct but complementary roles in preventing the formation of tau-fibril/amyloid. (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. This peptide is present in hippocampal synaptosomal preparations from Alzheimer's disease subjects. PMID: 29508283
  4. A study reports the identification of 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 tau deficiency in mammary epithelial cells. PMID: 30142893
  7. This article presents key studies of tau in oligodendrocytes and highlights important studies of tau in neurons. The extensive research on tau in neurons has significantly advanced our 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 where tau acetylation disengages tau from microtubules (MT) and also promotes tau aggregation. Therefore, therapeutic approaches aimed at limiting tau K280/K281 acetylation could simultaneously restore MT stability and mitigate tau pathology in Alzheimer's disease and related tauopathies. PMID: 28287136
  11. In vitro neuroprotective effects of naringenin nanoemulsion against beta-amyloid toxicity have been demonstrated through the regulation of amyloidogenesis and tau phosphorylation. PMID: 30001606
  12. To confirm the neuroprotective role of 24-OH, in vivo experiments were conducted 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 the study series. This phenotype might aid in the differential diagnosis from other tauopathies and serve as a morphological indicator for genetic testing. Haplotype analysis results suggest a founder effect of the P301L mutation in this area. PMID: 28934750
  14. The study reports 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 consists of the PHF6* and PHF6 hexapeptide motifs, with 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 compared to FTLD-tau. PMID: 28984110
  16. The data indicates that 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 the primary cause 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. The 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, potentially serving as a proxy indicator of CARTS. PMID: 28281308
  20. Patients with Kii amyotrophic lateral sclerosis and parkinsonism-dementia complex (Kii ALS/PDC) exhibited 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 serve as a neuropathological marker for the condition. PMID: 28236345
  21. The study 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 the severity of progressive supranuclear palsy-related tau pathology. PMID: 29082658
  27. These observations indicate the ability of QUE to decrease tau protein hyperphosphorylation, thereby attenuating 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, providing 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 disease development. 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. The study confirmed that a Western diet did not exacerbate tau pathology in hTau mice. The researchers observed that voluntary treadmill exercise attenuated tau phosphorylation and reported that caloric restriction seems to exacerbate tau aggregation compared to control and obese hTau mice. PMID: 28779908
  36. The study showed a gradual accumulation of nuclear tau in human cells during aging and its general co-localization with the DAPI-positive heterochromatin. This seems to be related to aging pathologies (neurodegenerative or cancerous diseases), where nuclear AT100 decreases drastically, a condition that is 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. This is primarily 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. The study 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. The 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 significant 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. This is because 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 indicate 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 reveals inter- and intra-familial clinicopathologic heterogeneity of FTDP-17 due to MAPT p.P301L mutation, including globular glial tauopathy in one patient. PMID: 27859539

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 MAPT protein and why is it a target for recombinant monoclonal antibodies?

MAPT (Microtubule-Associated Protein Tau) is a protein primarily found in neurons that plays a crucial role in stabilizing microtubules and maintaining neuronal structure and function. It exists in multiple isoforms, with six subtypes identified, four of which are expressed under normal physiological conditions. MAPT binds to microtubules to promote their assembly and stability by crosslinking and bundling them together, while also regulating microtubule dynamics by promoting growth and preventing disassembly. Beyond structural roles, MAPT participates in signaling pathways controlling cell survival, growth, and differentiation .

The protein has become a significant research target because abnormal MAPT accumulation is directly linked to several neurodegenerative disorders, most notably Alzheimer's disease, as well as Parkinson's disease and frontotemporal dementia. In Alzheimer's disease, aberrant MAPT protein aggregation leads to neuronal dysfunction and cell death, resulting in cognitive decline. Recombinant monoclonal antibodies targeting MAPT provide researchers with standardized tools to investigate these pathological processes and potentially develop therapeutic interventions .

How are MAPT recombinant monoclonal antibodies produced?

MAPT recombinant monoclonal antibodies are developed using protein and DNA recombinant technology through a multi-step process:

  • Immunization: Mice are injected with synthetic peptides derived from human MAPT protein.

  • Cell extraction: After a specific duration, spleen cells containing B lymphocytes are aseptically extracted.

  • RNA isolation: Total RNA is isolated from these spleen cells.

  • cDNA synthesis: RNA is reverse-transcribed to create cDNA.

  • Gene amplification: The cDNA serves as a PCR template to amplify the MAPT antibody gene.

  • Vector integration: The amplified antibody gene is integrated into an expression vector.

  • Cell transfection: The vector is transfected into host cells (typically CHO cells).

  • Cell culture: Transfected cells are cultured to produce the antibody.

  • Purification: The recombinant antibody is purified from cell culture supernatant using affinity chromatography.

  • Validation: The purified antibody undergoes extensive validation testing before research use .

This recombinant approach offers significant advantages over traditional hybridoma technology, including better reproducibility, reduced batch-to-batch variation, and decreased reliance on animal-based production systems .

What are the key differences between recombinant monoclonal antibodies and traditional monoclonal antibodies?

ParameterRecombinant Monoclonal AntibodiesTraditional Monoclonal Antibodies
Production methodDNA recombinant technology in expression systemsHybridoma technology using mouse cells
ReproducibilityHigh; genetic sequence remains constantVariable; subject to genetic drift
Batch consistencyHighly consistentMay show batch-to-batch variation
CustomizationEasily engineered and modifiedLimited modification capabilities
Species adaptationCan be humanized or fully humanTypically murine or chimeric
Ethical considerationsReduced animal use after initial developmentContinuous animal use for production
Long-term stabilityHigh; sequence can be stored indefinitelyMedium; hybridomas can experience drift
StandardizationHigh; defined at genetic levelVariable; dependent on hybridoma stability
Production scalabilityHighly scalable in bioreactorsLimited by hybridoma growth characteristics

Recombinant antibodies represent a significant advancement in addressing reproducibility issues that have plagued traditional antibody production, while also aligning with efforts to reduce animal use in research .

What are the primary research applications for MAPT recombinant monoclonal antibodies?

MAPT recombinant monoclonal antibodies serve multiple critical functions in neurodegenerative disease research:

  • Detection and quantification: These antibodies enable precise identification and measurement of MAPT protein in various experimental systems, facilitating comparative studies of normal versus pathological conditions.

  • Structural and functional studies: They help elucidate the structure-function relationship of MAPT, particularly how different isoforms interact with microtubules and other cellular components.

  • Pathology research: These antibodies are instrumental in studying abnormal MAPT aggregation, phosphorylation, and other post-translational modifications associated with tauopathies.

  • Drug discovery: They serve as tools for screening potential therapeutic compounds that might prevent MAPT aggregation or promote clearance of pathological tau.

  • Biomarker development: They assist in developing diagnostic assays for measuring tau in cerebrospinal fluid or blood as potential biomarkers for neurodegenerative diseases.

  • Imaging studies: When appropriately labeled, these antibodies can visualize the distribution and aggregation of tau in tissue sections, providing insights into disease progression .

The high specificity of recombinant monoclonal antibodies makes them particularly valuable in distinguishing between different MAPT isoforms and post-translationally modified variants that may have distinct roles in disease pathogenesis.

What experimental techniques commonly utilize MAPT recombinant monoclonal antibodies?

MAPT recombinant monoclonal antibodies are versatile tools employed across multiple experimental techniques in neuroscience and neurodegeneration research:

  • Immunohistochemistry (IHC): For visualizing MAPT distribution in tissue sections with recommended dilutions typically ranging from 1:50 to 1:200, depending on antibody affinity and tissue preparation methods .

  • Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative measurement of MAPT levels in various biological samples including tissue lysates, cerebrospinal fluid, and cell culture supernatants .

  • Western blotting: For analyzing MAPT protein expression, isoform distribution, and post-translational modifications.

  • Immunoprecipitation: For isolating MAPT protein complexes to study protein-protein interactions.

  • Immunofluorescence microscopy: For subcellular localization studies and co-localization with other proteins.

  • Flow cytometry: For analyzing MAPT expression in neuronal cell populations.

  • Proximity ligation assays: For studying protein interactions involving MAPT in situ.

  • Surface plasmon resonance: For determining binding kinetics between MAPT and potential interacting partners.

Each technique requires specific optimization of antibody concentration, incubation conditions, and detection methods to achieve optimal results while minimizing background signal and non-specific binding .

How do MAPT recombinant monoclonal antibodies contribute to neurodegenerative disease research?

MAPT recombinant monoclonal antibodies have become essential tools in advancing our understanding of neurodegenerative diseases through multiple research avenues:

  • Disease mechanism elucidation: These antibodies help researchers investigate the molecular mechanisms underlying tauopathies by enabling visualization and quantification of pathological tau species.

  • Temporal and spatial progression mapping: By detecting different forms of MAPT, researchers can map how tau pathology spreads through the brain over time, contributing to the "prion-like" hypothesis of propagation.

  • Biomarker identification and validation: MAPT antibodies facilitate the development of diagnostic assays that measure specific tau species in biological fluids as potential biomarkers for disease diagnosis, progression monitoring, and treatment response assessment.

  • Therapeutic development: These antibodies can serve as prototypes for therapeutic antibodies or be used to evaluate the efficacy of tau-targeting therapies in preclinical models.

  • Structural studies: They help elucidate the conformational changes that occur when tau transitions from soluble to aggregated forms.

  • Animal model validation: MAPT antibodies are crucial for confirming that animal models recapitulate key aspects of human tau pathology.

Tilavonemab (ABBV-8E12/C2N-8E12), for example, is a humanized IgG4 monoclonal antibody that targets extracellular tau, initially developed as a potential therapeutic agent for progressive supranuclear palsy and early Alzheimer's disease. Though clinical trials have not shown sufficient efficacy for therapeutic use, the antibody remains valuable as a research tool for understanding tau pathology propagation mechanisms .

What storage conditions are optimal for maintaining MAPT recombinant monoclonal antibody activity?

Proper storage is critical for maintaining the functional integrity of MAPT recombinant monoclonal antibodies. The recommended storage conditions typically include:

  • Short-term storage (up to 2 weeks): 2-8°C under sterile conditions after reconstitution.

  • Long-term storage: -80°C for extended periods.

  • Avoid repeated freeze-thaw cycles: Multiple freeze-thaw cycles can lead to antibody degradation, denaturation, and loss of binding efficiency. Aliquoting the antibody upon receipt is strongly recommended.

  • Reconstitution medium: Typically phosphate-buffered saline (PBS, pH 7.4) without stabilizers or preservatives for maximum compatibility with biological systems.

  • Protection from light: For fluorophore-conjugated antibodies, protection from light exposure is essential to prevent photobleaching.

  • Sterility maintenance: Use of sterile techniques when handling the antibody to prevent microbial contamination.

  • Documentation: Maintain records of receipt date, lot number, aliquoting, and freeze-thaw cycles to track antibody usage and potential degradation .

Following these guidelines helps ensure consistent antibody performance across experiments and maximizes the usable lifespan of these valuable research reagents.

What validation steps should researchers perform before using MAPT recombinant monoclonal antibodies in critical experiments?

Before employing MAPT recombinant monoclonal antibodies in pivotal experiments, researchers should conduct comprehensive validation to ensure reliability and specificity:

  • Positive and negative controls: Test the antibody against samples known to express or lack MAPT, including knockout models when available.

  • Epitope verification: Confirm which MAPT epitope or isoform the antibody recognizes, particularly important given the six known tau isoforms.

  • Cross-reactivity assessment: Evaluate potential cross-reactivity with other microtubule-associated proteins or similar structural motifs.

  • Application-specific validation: Verify suitability for the intended application (IHC, ELISA, Western blot, etc.) as antibody performance can vary between applications.

  • Dilution series optimization: Determine optimal working concentrations through dilution series experiments to maximize signal-to-noise ratio.

  • Reproducibility testing: Repeat experiments to ensure consistent results across multiple trials.

  • Secondary antibody compatibility: Confirm appropriate secondary antibody selection to avoid species cross-reactivity issues.

  • Batch testing: When receiving a new lot, compare performance with previously validated lots.

  • Literature cross-reference: Review published literature using the same antibody to corroborate expected results and potential limitations.

  • Blocking optimization: Determine effective blocking conditions to minimize non-specific binding, particularly important for IHC applications.

What factors influence the stability and degradation of recombinant monoclonal antibodies during experimental use?

Several factors can impact the stability and performance of recombinant monoclonal antibodies during experimental procedures:

  • Temperature fluctuations: Exposure to temperature extremes can cause protein denaturation and loss of binding capacity.

  • pH variations: Deviation from optimal pH (typically 6.0-8.0) can alter antibody conformation and binding properties.

  • Oxidative stress: Methionine and tryptophan residues are particularly susceptible to oxidation, which can affect antibody function.

  • Proteolytic degradation: Contaminating proteases in samples can cleave antibodies, especially in the hinge region.

  • Aggregation: Protein aggregation, often triggered by improper handling or storage, can reduce effective antibody concentration and increase non-specific binding.

  • Chemical modifications: Exposure to certain chemicals or buffers can induce modifications like deamidation of asparagine and aspartate residues.

  • Light exposure: Particularly problematic for conjugated antibodies, leading to fluorophore photobleaching or photochemical damage.

  • Freeze-thaw cycles: Repeated freezing and thawing can cause physical stress leading to denaturation and aggregation.

  • Mechanical stress: Excessive vortexing or pipetting can induce shear forces that damage antibody structure.

  • Microbial contamination: Growth of microorganisms in antibody preparations can lead to degradation through microbial proteases.

Understanding these factors allows researchers to implement appropriate handling protocols to maximize antibody stability and experimental reliability .

How do post-translational modifications affect the functionality of MAPT recombinant monoclonal antibodies?

Post-translational modifications (PTMs) of recombinant monoclonal antibodies can significantly impact their functionality in research applications:

  • Glycosylation variations: Changes in glycosylation patterns can affect antibody stability, half-life, and Fc receptor binding. Recombinant antibodies produced in different expression systems (CHO cells, HEK293, etc.) may exhibit distinct glycosylation profiles that influence their performance.

  • Deamidation: Spontaneous deamidation of asparagine and glutamine residues, especially in complementarity-determining regions (CDRs), can alter antigen-binding affinity and specificity. This modification is time and pH-dependent, with higher rates observed at alkaline pH.

  • Oxidation: Methionine and tryptophan residues are susceptible to oxidation, which can modify the antibody's three-dimensional structure and potentially affect binding properties. This is particularly relevant for antibodies targeting conformational epitopes.

  • C-terminal lysine variability: Recombinant antibodies often exhibit heterogeneity in C-terminal lysine residues due to carboxypeptidase activity in expression systems, which may affect charge distribution and potentially binding characteristics.

  • N-terminal modifications: Pyroglutamate formation at N-terminal glutamine residues occurs spontaneously and can impact the antibody's isoelectric point and stability.

These modifications must be considered when analyzing experimental results and may require specific controls to account for their effects, particularly in quantitative applications where binding affinity directly impacts measurement accuracy .

What approaches can researchers use to address epitope masking issues when studying MAPT in tissue samples?

Epitope masking represents a significant challenge when studying MAPT in tissue samples, particularly in the context of neurodegenerative diseases where protein conformation and interactions may obstruct antibody binding sites. Researchers can employ several strategies to overcome these limitations:

  • Heat-induced epitope retrieval (HIER): Optimize heating conditions (temperature, duration, buffer composition) to break protein cross-links formed during fixation while preserving tissue morphology. Citrate buffer (pH 6.0) and Tris-EDTA buffer (pH 9.0) are commonly used, with selection based on specific antibody requirements.

  • Enzymatic epitope retrieval: Employ proteolytic enzymes (proteinase K, trypsin, pepsin) to expose hidden epitopes, though careful titration is necessary to prevent excessive tissue digestion.

  • Dual epitope retrieval approaches: Combine HIER with enzymatic methods for particularly challenging epitopes.

  • Formic acid pretreatment: Especially useful for revealing epitopes in amyloid and tau aggregates by partially disrupting beta-sheet structures.

  • Variable fixation protocols: Test different fixation durations and fixative compositions as overfixation often contributes to epitope masking.

  • Unfixed frozen tissue sections: Consider using frozen sections when fixation-induced epitope masking cannot be overcome.

  • Multiple antibody approach: Utilize antibodies recognizing different MAPT epitopes to comprehensively characterize tau pathology, as certain epitopes may be differentially accessible in various pathological states.

  • Pre-absorption controls: Perform pre-absorption with purified antigens to confirm specificity of observed staining patterns.

  • Denaturing conditions: Apply controlled denaturing conditions to unfold protein aggregates and expose hidden epitopes.

Systematic optimization of these approaches is essential for reliable detection of MAPT in different pathological contexts, particularly when studying aggregated forms of tau protein .

How can researchers troubleshoot cross-reactivity issues with MAPT recombinant monoclonal antibodies?

Cross-reactivity represents a significant challenge when working with MAPT recombinant monoclonal antibodies due to sequence homology with other microtubule-associated proteins and the existence of multiple tau isoforms. Researchers can implement the following troubleshooting strategies:

  • Comprehensive validation panel: Test antibody specificity against:

    • Recombinant MAPT isoforms

    • MAPT knockout models/samples

    • Closely related proteins

    • Species-specific variants

  • Epitope mapping: Precisely identify the binding epitope through techniques such as:

    • Peptide arrays

    • Hydrogen-deuterium exchange mass spectrometry

    • X-ray crystallography of antibody-antigen complexes

    • Alanine scanning mutagenesis

  • Pre-adsorption studies: Pre-incubate the antibody with purified recombinant MAPT protein before application to determine if specific staining is eliminated.

  • Multiple antibody verification: Compare staining/detection patterns using antibodies targeting different MAPT epitopes.

  • Modified blocking protocols: Optimize blocking solutions by incorporating:

    • Higher concentrations of blocking proteins (BSA, normal serum)

    • Commercial blocking reagents designed to reduce non-specific binding

    • Additives like Tween-20, Triton X-100, or gelatin

  • Western blot analysis: Confirm antibody specificity by checking for bands of appropriate molecular weight and absence of unexpected bands.

  • Titration experiments: Determine the minimum effective antibody concentration that maintains specific staining while reducing background.

  • Secondary antibody controls: Run secondary-only controls to identify potential background from secondary antibody interactions.

  • Isotype control experiments: Use isotype-matched control antibodies to identify Fc receptor-mediated non-specific binding.

Through systematic application of these troubleshooting approaches, researchers can significantly reduce cross-reactivity issues and increase confidence in their experimental results .

How do MAPT recombinant monoclonal antibodies compare to traditional antibodies in reproducibility studies?

Comparative analysis between MAPT recombinant monoclonal antibodies and traditional hybridoma-derived antibodies reveals significant differences in reproducibility metrics that directly impact research quality:

ParameterMAPT Recombinant Monoclonal AntibodiesTraditional Monoclonal AntibodiesImpact on Research
Sequence definitionCompletely defined at DNA levelOften undefined at molecular levelRecombinant antibodies can be regenerated precisely if lost
Batch-to-batch consistencyCV typically <5% in binding assaysCV often 10-30% between batchesLower experimental variability with recombinant antibodies
Epitope specificityHighly consistentMay drift over time with hybridoma passagesMore reliable target recognition across experiments
Long-term availabilityIndefinite (sequence can be stored)Limited by hybridoma viabilityEliminates reproducibility issues from antibody discontinuation
Antibody heterogeneityMinimalMedium to highFewer confounding variables in experimental systems
Post-translational modificationsControlled and characterizedVariable and often uncharacterizedBetter understanding of potential binding interferences
Expression system consistencyDefined cellular backgroundMay change with hybridoma healthMore predictable performance in applications

The defined molecular nature of recombinant antibodies addresses a fundamental challenge in the reproducibility crisis affecting biomedical research. When working with complex targets like MAPT with multiple isoforms and post-translational modifications, the enhanced reproducibility of recombinant antibodies provides significant advantages for longitudinal studies and cross-laboratory comparisons .

What emerging applications are being developed for MAPT recombinant monoclonal antibodies in neurodegenerative research?

MAPT recombinant monoclonal antibodies are at the forefront of several innovative research directions in neurodegenerative disease investigation:

  • Single-cell tau pathology mapping: Integration of highly specific MAPT antibodies with single-cell sequencing technologies to correlate tau pathology with cell-type-specific transcriptomic profiles, providing unprecedented resolution of vulnerable neural populations.

  • Tau seed amplification assays: Development of ultrasensitive diagnostic methods using MAPT antibodies to detect minute quantities of pathological tau seeds in biological fluids, potentially enabling early disease detection before clinical symptoms appear.

  • Intrabody applications: Engineering MAPT recombinant antibody fragments (scFvs) for intracellular expression to target tau in living neurons, opening new avenues for studying tau dynamics in real-time and potential therapeutic applications.

  • PET imaging development: Creating tau-specific antibody fragments for positron emission tomography tracer development, enabling non-invasive monitoring of tau pathology progression in living subjects.

  • Tau strain differentiation: Developing conformational-specific antibodies that can distinguish between different "strains" of pathological tau aggregates associated with distinct tauopathies.

  • Extracellular vesicle tau detection: Utilizing MAPT antibodies to study tau species transported in extracellular vesicles, investigating their potential role in pathology propagation.

  • Cryo-EM structural studies: Employing antibody labeling to facilitate structural determination of tau filaments using cryo-electron microscopy, advancing our understanding of aggregate formation.

  • Microfluidic-based tau assays: Creating lab-on-a-chip platforms incorporating MAPT antibodies for rapid, automated tau detection and characterization from minimal sample volumes.

These emerging applications highlight the continuing evolution of MAPT recombinant monoclonal antibodies as versatile tools in the fight against neurodegenerative diseases .

What are the current limitations of MAPT recombinant monoclonal antibodies in research applications?

Despite their advantages, MAPT recombinant monoclonal antibodies face several limitations that researchers should consider when designing experiments:

  • Conformational epitope recognition challenges: Recombinant antibodies may struggle to recognize complex conformational epitopes that form in pathological tau aggregates, particularly those dependent on post-translational modifications or specific folding patterns.

  • Species cross-reactivity limitations: Many MAPT antibodies are optimized for human tau detection but may have limited cross-reactivity with rodent or other animal models, complicating translational research.

  • Isoform specificity constraints: Achieving absolute specificity for individual tau isoforms remains challenging due to high sequence homology, potentially leading to overlapping detection that complicates isoform-specific research.

  • Blood-brain barrier penetration: For in vivo applications, the limited ability of full-sized antibodies to cross the blood-brain barrier restricts their utility in certain experimental paradigms.

  • Technical variability in phospho-epitope detection: Antibodies targeting phosphorylated tau epitopes may show variability depending on sample preparation methods and phosphatase activity in tissues.

  • Limited detection of oligomeric intermediates: Many antibodies fail to specifically distinguish between monomeric tau and potentially toxic oligomeric intermediates that may be crucial in disease pathogenesis.

  • Challenges in ultra-low abundance detection: Current antibody-based methods may lack sensitivity to detect the earliest pathological changes when tau species are present at extremely low concentrations.

  • Post-translational modification complexity: The extensive landscape of tau post-translational modifications (phosphorylation, acetylation, ubiquitination, etc.) creates a complex target environment that individual antibodies cannot fully capture.

Understanding these limitations allows researchers to design more effective experimental approaches, often combining multiple antibodies or complementary techniques to overcome individual shortcomings .

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.