DOT5 Antibody refers to immunological tools designed to detect the DOT5 protein, a nuclear thioredoxin peroxidase in Saccharomyces cerevisiae (baker’s yeast). Initially identified as a disruptor of telomeric silencing (DOT), this enzyme plays dual roles in oxidative stress response and chromatin regulation .
DOT5 (UniProt ID: P40553) is a thiol-specific peroxidase that reduces hydrogen peroxide and organic hydroperoxides, acting as an antioxidant during oxidative stress . Key features:
Property | Detail |
---|---|
Gene ID | 854805 (YIL010W) |
Aliases | Disrupter of telomere silencing protein 5, Peroxiredoxin DOT5 |
Localization | Nucleus |
Substrate Preference | Alkyl-hydroperoxides over hydrogen peroxide |
DOT5 detoxifies reactive oxygen species (ROS), ensuring cell survival during oxidative stress. Unlike cytosolic thioredoxin peroxidases, it preferentially reduces alkyl-hydroperoxides in the nucleus .
DOT5 disrupts telomeric silencing independently of its peroxidase activity, influencing chromatin structure and gene repression near telomeres .
It modulates telomere length during post-diauxic growth, balancing replication and maintenance mechanisms .
Protocol: Antibody dilution of 1:1,000 detects DOT5 in yeast nuclear extracts.
Controls: Use DOT5-deficient strains to confirm specificity .
Localization: Staining reveals nuclear distribution of DOT5, critical for oxidative stress response .
Studies show DOT5’s telomeric-silencing activity does not depend on peroxidase function, indicating distinct enzymatic and regulatory roles .
While DOT5 is yeast-specific, its mechanisms highlight conserved pathways in eukaryotic stress response and genome stability. Antibody-based studies in model organisms inform broader oxidative stress research .
Epigenetic Studies: Probe telomere silencing dynamics in yeast models.
Oxidative Stress Models: Assess nuclear ROS detoxification.
Protein Localization: Confirm nuclear compartmentalization via IF .
KEGG: sce:YIL010W
STRING: 4932.YIL010W
The tau oligomer-specific monoclonal antibody (TOMA) is a novel antibody developed specifically to recognize tau oligomers with high specificity. What distinguishes this antibody from others is its unique ability to bind exclusively to tau oligomers without recognizing monomeric functional tau or mature metastable neurofibrillary tangles (NFTs). This specificity makes it an ideal tool for immunotherapy and research focused on understanding the role of tau oligomers in neurodegenerative tauopathies. The antibody has demonstrated high affinity for tau oligomers and can effectively sequester tau oligomer toxicity in vitro, suggesting potential therapeutic applications beyond basic research .
Unlike antibodies directed against NFT-associated tau phosphoepitopes or those used in active vaccination using phosphorylated tau fragments, TOMA specifically targets the oligomeric form of tau. This distinction is crucial because growing evidence suggests that large metastable tau aggregates such as NFTs may not be causally linked to tauopathy phenotypes in animal models. TOMA's specificity addresses a key concern in tau immunotherapy approaches - the risk of targeting functional endogenous tau protein, which could induce autoimmunity or other complications. By focusing exclusively on the pathological oligomeric forms of tau, TOMA offers a more targeted approach for both research and potential therapeutic applications .
Research using the TOMA antibody has provided direct evidence supporting a critical role for tau oligomers in disease progression. Studies in the P301L mouse model of tauopathy demonstrated that a single dose of TOMA administered either intravenously or intracerebroventricularly was sufficient to reverse both locomotor and memory deficits for up to 60 days. Importantly, this therapeutic effect coincided with rapid reduction of tau oligomers but not phosphorylated NFTs or monomeric tau. These findings validate tau oligomers as a legitimate target for treating Alzheimer's disease and other neurodegenerative tauopathies, challenging earlier focuses on NFTs as the primary toxic species .
The development of effective tau oligomer antibodies requires careful preparation of tau oligomer antigens. Based on established protocols, recombinant tau (tau-441 (2N4R) 45.9 kDa) is first treated with 8M urea to obtain monomeric tau, followed by overnight dialysis against 1× PBS buffer (pH 7.4). Samples are then adjusted to 1 mg/ml with PBS, with aliquots of tau monomer stored at -20°C. To generate tau oligomers, 300 μl of the tau stock (1 mg/ml) is mixed with 700 μl of 1× PBS, yielding a final concentration of 0.3 mg/ml. This mixture is incubated at room temperature for 1 hour on an orbital shaker. The resulting tau oligomers are then purified using fast protein liquid chromatography (Superdex 200 HR 10/30 column). This methodology ensures production of consistent, high-quality tau oligomers suitable for generating specific antibodies .
For researchers developing tau oligomer-specific antibodies, the following immunization protocol has proven effective: 2-month-old BALB/c mice are immunized with tau oligomers mixed with an equal volume of saline or Freund's complete adjuvant. The initial intraperitoneal injection consists of 100 μl of a 1:1 (antigen:adjuvant) mixture, delivering approximately 20 μg per mouse. A second injection with Freund's incomplete adjuvant follows two weeks later, with subsequent boosts administered after 28, 47, 60, 80, and 103 days. Prior to hybridoma fusion, mice receive daily booster injections for four consecutive days. This systematic immunization schedule ensures robust antibody production with high specificity for tau oligomers .
Thorough validation of tau oligomer antibodies should follow a multi-step process. First, screen serial dilutions of animal sera using ELISA with plates coated with 50 or 200 ng of tau oligomers, Aβ oligomers, or α-synuclein oligomers to assess specificity and potential cross-reactivity with other amyloid oligomers. Follow this with dot blot analysis using a panel of proteins: tau monomer, multiple preparations of tau oligomers, Aβ oligomers, tau fibrils, and Aβ fibrils. Selected clones should then undergo Western blot testing using in vitro prepared samples and dot blot analysis with brain homogenates. Final validation should include testing with human and mouse brain samples. Antibody isotype and light chain composition (κ or λ) should be determined using a commercially available mouse monoclonal antibody isotyping kit. This comprehensive validation ensures antibody specificity before proceeding to experimental applications .
When designing passive immunization studies with tau oligomer antibodies like TOMA, researchers should consider several critical factors. First, determine appropriate administration routes - both intracerebroventricular and intravenous administration have proven effective in mouse models, with doses of 1 μg/animal and 30 μg/animal respectively showing efficacy. Age-matched controls should include both wild-type mice receiving saline and transgenic mice receiving non-specific IgG antibodies (e.g., anti-rhodamine) at equivalent concentrations. For intracerebroventricular administration, precise stereotactic coordinates are essential: -2.06 mm caudal to bregma, 1.7 mm lateral to midline, at a depth of 2.5 mm, with injection rates of 0.5 μl/min. For intravenous administration, researchers should use appropriate restrainers and warm the tail to dilate veins before injection into the lateral tail vein. Quality control is paramount - all antibody preparations must be endotoxin-free (confirmed using Limulus amebocyte lysate assay), with batches containing ≥3 ng/ml endotoxin being purified or rejected for in vivo use .
To track antibody biodistribution in tau pathology models, researchers should employ biotinylation techniques. Specifically, antibodies (e.g., 2 mg of TOMA) can be biotinylated using EZ-link Sulfo-NHS-SS Biotinylation kit following manufacturer's instructions. Following intravenous administration of 30 μg of biotinylated antibody (with non-biotinylated antibody as control), blood samples should be collected at multiple timepoints (pre-injection, 30 minutes, 1 hour, 24 hours, and 1 week post-injection) to track peripheral circulation. Terminal collection points (e.g., 2, 6, and 24 hours, and 1 week post-injection) should include deep anesthesia followed by transcardial perfusion with 1× PBS before tissue harvest. For comprehensive analysis, divide brain samples for both biochemical and histological examination - homogenize one hemisphere in PBS containing protease inhibitors (centrifuged at 10,000 rpm for 10 minutes at 4°C) for biochemical analysis, while preparing the other hemisphere for histological examination (10 μm sagittal sections). Brain sections can be stained with streptavidin-horseradish peroxidase to visualize biotinylated antibody localization .
The efficacy of peripherally administered tau antibodies in affecting central nervous system pathology involves several potential mechanisms. Research with TOMA antibody suggests that protection is mediated by both extracellular clearance and rapid peripheral clearance mechanisms. When administered intravenously, anti-tau oligomer antibodies appear to cross the blood-brain barrier, similar to other IgG antibodies observed in P301L mice. Once in the brain, these antibodies likely bind to extracellular tau oligomers, preventing their uptake by neurons and their prion-like spreading between cells. Additionally, the peripheral sink hypothesis may apply - by binding tau in the peripheral circulation, antibodies could alter the equilibrium of tau species between the CNS and periphery, effectively drawing more tau out of the brain. The rapid reduction of tau oligomers following antibody administration, without corresponding changes in phosphorylated NFTs or monomeric tau, suggests a specific targeting and clearance mechanism for the oligomeric species implicated as the primary toxic entities in tauopathies .
To enhance the therapeutic potential of tau oligomer antibodies like TOMA for clinical applications, several strategic modifications should be considered. First, humanization of the antibody sequence would be essential to minimize immunogenicity while preserving the critical oligomer-binding epitope. Second, engineering the antibody to have enhanced blood-brain barrier penetration, potentially through reducing size (using Fab or scFv fragments) or incorporating receptor-mediated transcytosis targeting moieties (such as transferrin receptor binding domains), could improve CNS bioavailability. Third, optimizing the antibody's pharmacokinetic profile through Fc engineering could extend half-life while maintaining appropriate effector functions. Fourth, development of bispecific antibodies that simultaneously target tau oligomers and recruit clearance mechanisms (e.g., microglial phagocytosis) might enhance therapeutic efficacy. Finally, structure-guided optimization of binding affinity and specificity based on detailed epitope mapping would ensure maximum selectivity for pathological tau oligomers while avoiding interference with physiological tau functions .
Integration of tau oligomer antibodies into multi-target therapeutic approaches represents a promising frontier in neurodegenerative disease research. Such strategies could combine TOMA-like antibodies with complementary interventions targeting different aspects of disease pathology. Potential combination approaches include: (1) dual targeting of tau and amyloid-β pathologies through co-administration with Aβ-directed antibodies, addressing both major protein aggregation pathways in Alzheimer's disease; (2) pairing with anti-inflammatory agents to simultaneously clear tau oligomers while dampening neuroinflammatory responses; (3) combination with neurotrophic factors or their mimetics to both remove toxic tau species and promote neuronal recovery and synaptic function; (4) integration with metabolism-enhancing drugs that improve neuronal energetics while tau oligomer burden is reduced; and (5) use alongside blood-brain barrier modulators to enhance CNS delivery of the tau oligomer antibody itself. Such multi-modal approaches require careful design of preclinical studies with appropriate controls to distinguish additive from synergistic effects and to identify potential antagonistic interactions between therapeutic modalities .
Advancing our understanding of tau oligomer antibodies' mechanisms requires several methodological innovations. First, development of in vivo tau oligomer imaging techniques would allow real-time tracking of antibody engagement with targets and subsequent clearance dynamics. Second, single-cell transcriptomics and proteomics applied to various CNS cell populations following antibody treatment could reveal cell type-specific responses and identify key molecular pathways involved in therapeutic effects. Third, advanced intravital microscopy techniques could visualize antibody-oligomer interactions in live animals, providing insights into the kinetics and localization of these interactions. Fourth, development of more physiologically relevant human in vitro models, such as 3D organoids derived from patient iPSCs, would better recapitulate human tau biology and potentially identify species-specific aspects of antibody function. Finally, computational approaches integrating systems biology with structural biology could model the complex dynamics of antibody-mediated tau oligomer clearance and predict optimal antibody properties for therapeutic efficacy. These methodological advances would collectively enhance our mechanistic understanding of how tau oligomer antibodies exert their beneficial effects and guide rational optimization of their therapeutic potential .