TDG Human

Thymine-DNA Glycosylase Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biochemical Properties of TDG Human

TDG Human is a recombinant protein expressed in E. coli as a single polypeptide chain (amino acids 1–410) with a molecular mass of 48.4 kDa . Key features include:

PropertyDescriptionSource
Expression HostE. coli
Purity>85% (SDS-PAGE)
Formulation20 mM Tris-HCl (pH 8.0), 0.1 M NaCl, 0.4 M Urea, 10% glycerol
His-TagN-terminal 23-amino acid fusion for purification
Stability4°C (2–4 weeks), -20°C (long-term) with 0.1% BSA/HSA carrier protein

TDG is sterility-filtered and colorless, optimized for laboratory research .

Biological Function

TDG belongs to the TDG/mug DNA glycosylase family and excises mismatched or oxidized bases to prevent mutagenesis. Key activities include:

  • G/T Mismatch Repair: Hydrolyzes thymine from G·T mismatches, restoring G·C pairs .

  • Uracil/5-Bromouracil Removal: Removes uracil or 5-bromouracil from G·U mispairs .

  • Oxidized Cytosine Processing: Excises 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC) in CpG contexts, aiding active DNA demethylation .

TDG’s activity is context-dependent, favoring CpG sequences and double-stranded DNA .

Substrate Specificity and Catalytic Efficiency

TDG exhibits broad but species-specific substrate spectra. Comparative data for human TDG (hTDG), Drosophila Thd1p, and S. pombe Thp1p highlight its versatility:

SubstrateHuman TDGThd1pThp1pKey Notes
G·UHighModerateHighUniversal high-efficiency substrate
G·TModerateLowNoneHuman-specific processing
G·5caCModerateNoneNoneCritical for demethylation
G·FUHighHighHighProcesses 5-fluorouracil in all contexts

Table 1: Substrate Processing Efficiency of TDG Orthologs

Human TDG uniquely excises 5-methylcytosine derivatives (e.g., 5caC) with moderate efficiency, while bacterial homologs like E. coli Mug lack this activity .

Clinical and Therapeutic Implications

TDG’s dysregulation is linked to cancer progression:

  • Colon Cancer: Overexpression inhibits migration/invasion via the DNMT3A-TIMP2 axis, reducing metastasis .

  • Glioma: Upregulated TDG correlates with poor prognosis; knockdown suppresses tumor growth .

Cancer TypeTDG EffectMechanismSource
ColonInhibits migration/invasionDNMT3A-TIMP2 axis regulation
GliomaPromotes tumorigenesisTNC-mediated signaling pathways

Table 2: TDG’s Role in Cancer

Research Findings and Future Directions

  • DNA Demethylation: TDG-mediated excision of 5fC/5caC drives active demethylation, critical for cell differentiation .

  • Therapeutic Targeting: Inhibiting TDG in gliomas may disrupt tumor growth, while enhancing its activity in colon cancer could suppress metastasis .

Product Specs

Introduction
Thymine-DNA glycosylase (TDG) is a member of the TDG/mug DNA glycosylase family. It is a nuclear protein responsible for correcting G/T mismatches to G/C pairs in DNA. This correction is achieved by hydrolyzing the bond between the DNA backbone and the mispaired thymine. Additionally, TDG removes uracil and 5-bromouracil mispaired with guanine. This enzyme plays a crucial role in protecting cells from mutations caused by the natural deamination of 5-methylcytosine and cytosine.
Description
Recombinant human TDG, expressed in E. coli, is a single polypeptide chain consisting of 433 amino acids (residues 1-410) with a molecular weight of 48.4 kDa. A 23 amino acid His-tag is fused to the N-terminus for purification using proprietary chromatographic methods.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The TDG solution (0.5mg/ml) is supplied in 20mM Tris-HCl buffer (pH 8.0), containing 0.1M NaCl, 0.4M Urea, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 85% using SDS-PAGE analysis.
Synonyms
Thymine-DNA Glycosylase, G/T Mismatch-Specific Thymine DNA Glycosylase, EC 3.2.2.29.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMEAENAG SYSLQQAQAF YTFPFQQLMA EAPNMAVVNE QQMPEEVPAP APAQEPVQEA PKGRKRKPRT TEPKQPVEPK KPVESKKSGK SAKSKEKQEK ITDTFKVKRK VDRFNGVSEA ELLTKTLPDI LTFNLDIVII GINPGLMAAY KGHHYPGPGN HFWKCLFMSG LSEVQLNHMD DHTLPGKYGI GFTNMVERTT PGSKDLSSKE FREGGRILVQ KLQKYQPRIA VFNGKCIYEI FSKEVFGVKV KNLEFGLQPH KIPDTETLCY GMPSSSARCA QFPRAQDKVH YYIKLKDLRD QLKGIERNMD VQEVQYTFDL QLAQEDAKKM AVKEEKYDPG YEAAYGGAYG ENPCSSEPCG FSSNGLIESV ELRGESAFSG IPNGQWMTQS FTDQIPSFSN HCGTQEQEEE SHA.

Q&A

What is human Thymine DNA Glycosylase and what is its primary function?

Human Thymine DNA Glycosylase (hTDG) is a DNA repair enzyme belonging to the uracil DNA glycosylase superfamily. Its primary function involves recognizing mismatched pyrimidine bases such as uracil and thymine in G- U and G- T pairs, then cleaving their glycosylic bonds to initiate the base excision repair (BER) pathway. hTDG employs a base-flipping mechanism to identify damaged bases in double-stranded DNA, facilitating their replacement through BER . More recently, hTDG has been recognized for its critical role in active DNA demethylation, particularly in recognizing and excising oxidized forms of 5-methylcytosine, which represents a distinct but equally important function .

How does the structure of hTDG's catalytic domain contribute to its function?

The catalytic domain of hTDG (hTDG cat, residues 111-308) contains the active site that specifically recognizes target bases through precise structural interactions. Crystal structures have revealed that hTDG cat can form complexes with DNA containing various modified bases, including 5-carboxylcytosine (5caC) . The specificity of recognition depends on the configuration of the active site, particularly involving residues like Asn140, which is crucial for catalytic activity. When this residue is mutated (as in the N140A mutant), the enzyme retains binding capabilities but loses catalytic function, making such mutations valuable for structural studies . The enzyme's architecture enables base-flipping, where the target nucleotide is rotated out of the DNA helix and into the active site pocket for processing.

What distinguishes hTDG from other DNA glycosylases in the uracil DNA glycosylase superfamily?

While other members of the uracil DNA glycosylase superfamily primarily focus on removing uracil from DNA, hTDG exhibits broader substrate specificity. Beyond processing mismatched thymine and uracil, hTDG uniquely recognizes oxidized derivatives of 5-methylcytosine, specifically 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC) . This expanded substrate range connects hTDG to epigenetic regulation pathways not served by other family members. The enzyme shows different binding affinities for various substrates, with apparent binding affinity following the order: G- AP > G- 5caC > G- 5fC > G- U > G- T . This hierarchy of substrate preference differentiates hTDG from other glycosylases and reflects its specialized biological roles.

How does hTDG recognize and process mismatched DNA bases?

hTDG recognizes mismatched bases through a sophisticated structure-specific mechanism. The enzyme scans DNA and identifies destabilized regions containing mismatches like G- T or G- U. Upon recognition, hTDG employs base-flipping to rotate the target base out of the DNA helix and into its catalytic pocket . Inside this pocket, specific amino acid residues form hydrogen bonds with the flipped base, stabilizing it for glycosylic bond cleavage. The enzyme's active site contains a water molecule positioned for nucleophilic attack on the C1′ of the deoxyribose sugar, resulting in hydrolysis of the N-glycosylic bond. This process creates an abasic site (AP site) that signals subsequent BER pathway enzymes to complete the repair .

What experimental approaches are most effective for analyzing hTDG's glycosylase activity?

Several methodologies have proven effective for investigating hTDG's glycosylase activity:

  • Single turnover experiments: These assess the enzyme's catalytic efficiency against different substrates by measuring the rate of base excision under conditions where the enzyme concentration exceeds substrate concentration .

  • Electrophoretic mobility shift assays (EMSA): These evaluate binding affinities between hTDG and various DNA substrates by detecting mobility shifts when protein-DNA complexes form .

  • Crystallographic studies: X-ray crystallography of hTDG-DNA complexes provides atomic-level insights into substrate recognition and catalytic mechanisms. Using catalytically inactive mutants (e.g., N140A) can stabilize complexes for structural studies .

  • Fluorescence-based assays: These monitor real-time base excision using fluorescently labeled DNA substrates, allowing kinetic analyses of enzyme activity.

  • Activity assays with synthetic oligonucleotides: Using defined DNA duplexes containing specific modifications (G- T, G- U, G- 5caC, etc.) enables comparative analysis of substrate preferences .

How do mutations in the hTDG active site affect its substrate specificity and catalytic efficiency?

Mutations in hTDG's active site can dramatically alter both substrate specificity and catalytic function. The N140A mutation exemplifies this effect, as it abolishes catalytic activity while preserving DNA binding capabilities . This mutation has been instrumental in structural studies by stabilizing enzyme-substrate complexes. Other active site mutations can modify substrate preferences; for example, alterations to residues that interact with specific functional groups on the target bases may enhance recognition of certain substrates while diminishing others. Research has shown that single amino acid substitutions can shift the enzyme's preference between substrates like 5caC and 5fC, highlighting the precision of substrate recognition . These structure-function relationships provide critical insights for understanding the molecular basis of hTDG activity.

What role does hTDG play in active DNA demethylation pathways?

hTDG serves as a critical component in active DNA demethylation pathways, particularly in mammals. While plants employ direct 5mC glycosylases for demethylation, mammals utilize a more complex pathway involving TET (ten-eleven translocation) family dioxygenases and hTDG . In this pathway, 5-methylcytosine (5mC) is first oxidized by TET enzymes to 5-hydroxymethylcytosine (5hmC), then further to 5-formylcytosine (5fC) and finally to 5-carboxylcytosine (5caC) . hTDG specifically recognizes and excises 5fC and 5caC, creating an abasic site that is subsequently processed by the BER pathway to install an unmethylated cytosine . This TET-mediated oxidation followed by TDG-mediated excision constitutes a complete active demethylation cycle, essential for epigenetic reprogramming during development and transcriptional regulation.

How does hTDG distinguish between 5-methylcytosine derivatives during demethylation?

hTDG exhibits remarkable selectivity among 5-methylcytosine derivatives through precise structural recognition. Research has demonstrated that hTDG efficiently excises 5fC and 5caC but cannot excise 5hmC or 5mC . This selectivity stems from specific interactions between the enzyme's active site and the oxidized groups on cytosine derivatives. Crystal structures of hTDG bound to DNA containing 5caC reveal that the carboxyl group of 5caC forms specific hydrogen bonds within the active site, facilitating recognition .

Binding affinity studies show that hTDG cat(N140A) preferentially binds to dsDNA containing G- 5caC over G- 5fC, G- U, and G- T, indicating that the carboxyl group contributes significantly to binding stability . Despite stronger binding to 5caC, kinetic studies indicate that 5fC is processed more efficiently, suggesting that catalysis and binding are optimized differently for these substrates . These subtle distinctions enable hTDG to precisely target specific cytosine derivatives in the demethylation pathway.

What evidence connects hTDG to developmental processes and transcriptional regulation?

Several lines of evidence firmly establish hTDG's role in development and transcriptional regulation:

  • Embryonic lethality: Studies have shown that TDG is essential for mouse embryonic development, with TDG knockout resulting in embryonic lethality .

  • Transcriptional control: TDG has been implicated in transcriptional regulation through its interaction with transcription factors and its role in maintaining promoter hypomethylation at developmentally regulated genes .

  • Epigenetic reprogramming: During periods of genome-wide epigenetic reprogramming, such as in early embryogenesis and primordial germ cell development, active demethylation involving TDG is particularly important.

  • Gene expression patterns: The spatial and temporal expression patterns of TDG correlate with developmental transitions requiring epigenetic plasticity.

These connections highlight why TDG's demethylation activity cannot be explained solely by its uracil/thymine glycosylase function, pointing to its broader role in epigenetic regulation during development .

What are the optimal conditions for expressing and purifying recombinant hTDG for structural studies?

For successful structural studies of hTDG, researchers typically focus on the catalytic domain (hTDG cat, residues 111-308) rather than the full-length protein, as the catalytic domain yields better crystallization results . The recommended expression and purification protocol includes:

  • Expression system: Escherichia coli BL21(DE3) with a pET vector system containing the hTDG catalytic domain sequence with an N-terminal His-tag.

  • Induction conditions: IPTG induction at lower temperatures (16-18°C) for 16-20 hours to enhance soluble protein expression.

  • Purification steps:

    • Nickel affinity chromatography using an imidazole gradient

    • Tag cleavage with TEV protease (if tag removal is desired)

    • Ion-exchange chromatography (typically Q-Sepharose)

    • Size-exclusion chromatography for final polishing

  • Buffer optimization: The final buffer typically contains 20 mM Tris-HCl pH 7.5, 100-150 mM NaCl, 1 mM DTT, and 5% glycerol to maintain protein stability .

For crystallography studies of protein-DNA complexes, using catalytically inactive mutants such as hTDG cat(N140A) is advisable to prevent substrate processing, enabling stable complex formation .

How can researchers effectively design DNA substrates to study hTDG's activity on different modified bases?

Effective DNA substrate design for hTDG studies requires careful consideration of several factors:

  • Oligonucleotide length: 23-mers have been successfully used in crystallographic and biochemical studies, providing sufficient flanking sequence around the target base .

  • Target base positioning: Placing the target base (T, U, 5fC, or 5caC) centrally within the sequence maximizes enzyme accessibility.

  • Base pairing: Ensuring the modified base is paired with guanine (creating G- T, G- U, G- 5fC, or G- 5caC pairs) is crucial, as hTDG specifically recognizes mismatches with G .

  • Modified base incorporation: Chemical synthesis methods for incorporating modified bases like 5caC, or using non-hydrolyzable analogs (β-F-5caC with 2′-fluoro substitution) for structural studies .

  • Fluorescent labeling: For activity assays, incorporating fluorescent labels at termini that won't interfere with enzyme binding.

  • Control substrates: Including properly matched G- C pairs and other modifications (G- 5mC, G- 5hmC) as negative controls .

The synthesis and purification of these specialized oligonucleotides often require HPLC purification and mass spectrometry verification to ensure substrate purity and identity.

What crystallization techniques are most successful for obtaining high-resolution structures of hTDG-DNA complexes?

Successful crystallization of hTDG-DNA complexes typically employs the following approaches:

  • Protein preparation: Using the catalytic domain (hTDG cat) or catalytically inactive mutants (hTDG cat(N140A)) to form stable complexes with DNA .

  • DNA complex formation: Pre-forming the protein-DNA complex by incubating purified hTDG with synthetic DNA duplexes containing the target modification at a slight molar excess of DNA.

  • Crystallization methods: Vapor diffusion techniques (hanging or sitting drop) have yielded successful crystals, with drops containing 1-2 μL of protein-DNA complex mixed with an equal volume of reservoir solution .

  • Optimization strategies:

    • Screening various DNA lengths and overhangs

    • Testing different protein:DNA ratios

    • Varying precipitant concentrations

    • Applying seeding techniques to improve crystal quality

  • Cryoprotection: Careful optimization of cryoprotectant conditions to prevent ice formation during flash-freezing.

  • Data collection: Using synchrotron radiation sources for high-resolution diffraction data collection, often employing strategies to minimize radiation damage .

This approach has successfully yielded crystal structures of hTDG complexed with various DNA substrates, including those containing 5caC and fluorinated analogs, at resolutions sufficient to reveal atomic-level interactions .

How do the dual binding modes of thio-digalactosides with human galectins inform the development of selective inhibitors?

Research on thio-digalactosides (TDGs) has revealed fascinating insights into their interactions with human galectins. TDGs, including TDG itself, TD139, and TAZTDG, demonstrate dual binding modes when interacting with human galectins-1, -3, and -7 . These galectins contain carbohydrate-recognition domains comprised of five binding subsites (A-E). Crystallographic studies have shown that the fluorophenyl group of TAZTDG preferentially binds to subsite B in galectin-3 but favors subsite E in galectins-1 and 7 .

This differential binding results from novel interactions between an arginine within subsite E of the galectins and arene groups in the ligands. These structure-function relationships explain how binding potency can be improved from μM to nM ranges . The dual binding modes offer critical insights for developing selective inhibitors targeting specific galectin types, which is valuable for therapeutic applications in cancer immunotherapy and fibrotic disease treatment. Researchers can exploit these structural differences to design inhibitors with enhanced selectivity profiles by modifying aryl substitutions that preferentially interact with specific galectin subsites .

How does the catalytic mechanism of hTDG differ when processing different substrates (G- T vs. G- 5caC)?

The catalytic mechanism of hTDG exhibits subtle but significant differences when processing different substrates:

These mechanistic differences have significant implications for understanding hTDG's dual roles in DNA repair and active demethylation, and may provide opportunities for selectively modulating specific activities through targeted inhibitor design.

What are the implications of hTDG's role in cytosine demethylation for understanding cancer epigenetics and potential therapeutic approaches?

hTDG's critical involvement in active DNA demethylation pathways has profound implications for cancer epigenetics and emerging therapeutic strategies:

  • Epigenetic dysregulation: Aberrant DNA methylation patterns are hallmarks of cancer. As a key enzyme in active demethylation, hTDG dysfunction may contribute to methylation abnormalities observed in various cancers .

  • Transcriptional deregulation: hTDG helps maintain appropriate methylation status at promoter regions. Its dysfunction could lead to inappropriate silencing of tumor suppressor genes or activation of oncogenes through altered methylation patterns .

  • Therapeutic targeting: Modulating TDG activity represents a potential strategy for epigenetic therapy. Unlike targeting DNA methyltransferases (which prevents new methylation), enhancing TDG activity could actively remove existing inappropriate methylation marks.

  • Biomarker potential: The expression levels or activity of TDG and related demethylation pathway components could serve as biomarkers for certain cancer types or predict responsiveness to epigenetic therapies.

  • Synthetic lethality: Cancer cells with defects in certain DNA repair pathways might become more dependent on TDG-mediated repair, suggesting potential synthetic lethal interactions that could be therapeutically exploited.

Understanding these connections provides a framework for developing novel epigenetic therapies that specifically target the active demethylation pathway in cancer contexts.

How do binding affinities compare across different hTDG substrates and what does this reveal about specificity?

DNA SubstrateRelative Binding AffinityExcision ActivityImplications for Function
G- APHighestN/A (product)Strong product retention limits turnover
G- 5caCVery HighEfficientKey substrate in demethylation pathway
G- 5fCHighMost EfficientPreferred substrate in demethylation
G- UModerateEfficientClassical DNA repair substrate
G- TModerateModerateClassical DNA repair substrate
G- 5hmCLowNoneNot a substrate for excision
G- 5mCVery LowNoneNot a substrate for excision
G- CVery LowNoneNormal base pair, not a substrate

This hierarchy of binding affinities, determined through EMSA studies with hTDG cat(N140A), reveals that hTDG has evolved to strongly recognize both its reaction products (G- AP) and its epigenetically relevant substrates (G- 5caC) . The stronger binding to 5caC compared to 5fC, despite 5fC being processed more efficiently, suggests that binding affinity and catalytic efficiency are optimized independently. This pattern supports hTDG's specialized role in the demethylation pathway while maintaining its classical DNA repair functions.

What methodological approaches can distinguish between the dual functions of hTDG in DNA repair versus active demethylation?

Research QuestionExperimental ApproachControls/VariablesExpected Results
Substrate preferenceSingle turnover kineticsCompare excision rates of G- T vs. G- 5caCFaster processing of demethylation substrates suggests prioritization
Biological contextChIP-seqMap hTDG binding sites genome-wideEnrichment at either damage sites or developmental regulatory regions
Protein interactionsCo-immunoprecipitationPull down with TET enzymes vs. BER factorsDifferential interaction partners in repair vs. demethylation
Mutational analysisStructure-guided mutationsTarget residues specific to each substrateMutations affecting only one substrate class
Cell-type specificityExpression profilingCompare proliferating vs. differentiating cellsHigher expression during differentiation suggests demethylation role
Developmental timingConditional knockoutTissue-specific or temporal TDG deletionPhenotypes revealing primary function in specific contexts

These complementary approaches allow researchers to distinguish between hTDG's dual functions and determine which role predominates in different biological contexts. By analyzing both the biochemical properties and cellular contexts of hTDG activity, researchers can gain insights into how this single enzyme serves these distinct yet mechanistically related functions .

What structural features contribute to hTDG's recognition of 5caC versus other cytosine derivatives?

Structural FeatureInteraction with 5caCComparison to Other Cytosine FormsFunctional Significance
Carboxyl group at C5Forms hydrogen bonds with active site residuesAbsent in C, 5mC; different electronegativity than 5hmC, 5fCPrimary recognition determinant
Base flipping mechanismStabilizes extruded 5caC in active siteSimilar for all substratesEssential for base inspection
Active site water positioningCoordinates nucleophilic attackSimilar position across substratesCritical for catalysis
Protein conformational changesSpecific to 5caC bindingDiffers among substratesRegulates catalytic efficiency
DNA backbone distortionCharacteristic bend with 5caCDifferent degrees with other substratesAffects enzyme-substrate stability
Stacking interactionsSpecific aromatic interactions with 5caCDifferent electronic properties with other modificationsContributes to binding energy

Crystal structures of hTDG in complex with DNA containing 5caC or its analogs have revealed these specific recognition features . The carboxyl group of 5caC forms critical hydrogen bonds within the enzyme active site that are not possible with unmodified cytosine or 5mC, explaining hTDG's selectivity in the demethylation pathway. These structural insights provide a molecular basis for understanding how a single enzyme can exhibit such precise substrate discrimination.

What emerging technologies could enhance our understanding of hTDG's role in tissue-specific epigenetic reprogramming?

Several cutting-edge technologies hold promise for elucidating hTDG's tissue-specific functions:

  • Single-cell epigenomics: Techniques like single-cell ATAC-seq and single-cell bisulfite sequencing could reveal cell-type-specific activities of hTDG during development and disease progression.

  • CRISPR-based epigenome editing: Targeted recruitment of hTDG to specific genomic loci using dCas9-TDG fusions could help define its site-specific functions in various cellular contexts.

  • Base-resolution mapping of 5fC and 5caC: Advanced sequencing techniques that can directly detect 5fC and 5caC at base resolution would provide insights into the dynamics of TDG-mediated demethylation.

  • Cryo-EM studies: Applied to larger complexes of hTDG with partner proteins like TET enzymes and transcription factors could reveal higher-order regulatory mechanisms.

  • Integrative multi-omics approaches: Combining methylome, transcriptome, and TDG binding data across developmental time points could elucidate temporal dynamics of demethylation.

  • Organoid models: Patient-derived organoids could be used to study hTDG function in human tissue contexts that better recapitulate in vivo conditions.

These approaches could significantly advance our understanding of how hTDG contributes to tissue-specific epigenetic regulation during development and in disease contexts.

How might artificial intelligence and computational approaches accelerate the development of selective TDG inhibitors or modulators?

Artificial intelligence and computational methods offer several promising avenues for TDG inhibitor development:

  • Structure-based virtual screening: Using crystal structures of hTDG-DNA complexes to virtually screen large compound libraries for potential inhibitors that target specific binding modes .

  • Machine learning for activity prediction: Training ML models on existing biochemical data to predict the activity of novel compounds against hTDG, accelerating the identification of promising candidates.

  • Molecular dynamics simulations: Exploring the conformational dynamics of hTDG-substrate interactions to identify transient binding pockets or allosteric sites not evident in static crystal structures.

  • Quantum mechanical calculations: Detailed modeling of the reaction mechanism to design transition state analogs as potential inhibitors.

  • Network analysis of protein-protein interactions: Identifying key interaction partners that could be targeted to indirectly modulate hTDG activity in specific contexts.

  • AI-driven compound optimization: Using generative models to suggest structural modifications that enhance selectivity and pharmacokinetic properties of lead compounds.

These computational approaches could significantly accelerate the development of chemical probes and potential therapeutic agents targeting hTDG, providing valuable tools for basic research and potential clinical applications.

What research gaps remain in understanding the interplay between TETs and TDG in the complete demethylation pathway?

Despite significant advances, several critical knowledge gaps remain in understanding the TET-TDG demethylation pathway:

  • Spatial and temporal coordination: How TET enzymes and TDG are recruited to specific genomic loci in a coordinated manner remains poorly understood .

  • Regulatory mechanisms: The factors controlling the relative activities of TETs and TDG in different cellular contexts need further investigation.

  • Rate-limiting steps: Whether oxidation by TETs or excision by TDG represents the rate-limiting step in active demethylation remains controversial.

  • Protein complexes: The composition of multi-protein complexes involving TETs and TDG, and how these complexes affect function, requires further characterization.

  • Feedback mechanisms: Potential feedback loops between TDG activity and TET recruitment or activity have not been fully explored.

  • Alternative pathways: The extent to which alternative demethylation pathways complement or compete with the TET-TDG pathway remains unclear.

  • Metabolic influences: How cellular metabolism affects TET activity (through α-ketoglutarate levels) and subsequently impacts TDG function needs further study.

Addressing these research gaps would provide a more comprehensive understanding of active DNA demethylation and potentially reveal new therapeutic targets for diseases involving epigenetic dysregulation.

Product Science Overview

Introduction

Thymine-DNA glycosylase (TDG) is a crucial enzyme involved in the base excision repair (BER) pathway, which is responsible for maintaining genomic stability by correcting DNA base mismatches. This enzyme is particularly significant in the context of human health, as it plays a vital role in preventing mutations that could lead to diseases such as cancer.

Structure and Function

TDG belongs to the TDG/mug DNA glycosylase family and is characterized by its ability to remove thymine moieties from G/T mismatches. It achieves this by hydrolyzing the carbon-nitrogen bond between the sugar-phosphate backbone of DNA and the mispaired thymine . Additionally, TDG can excise uracil and 5-bromouracil from mispairings with guanine, albeit with lower activity .

The enzyme operates through a base-flipping mechanism, where the target base is flipped out of the DNA helix and into the enzyme’s active site. This allows TDG to recognize and bind to the substrate, catalyzing the hydrolysis of the N-glycosidic bond and generating an abasic site (AP-site) in the DNA backbone .

Biological Significance

TDG plays a central role in the cellular defense against genetic mutations. It is particularly important in countering the effects of spontaneous deamination of 5-methylcytosine and cytosine, which can lead to G/T and G/U mismatches, respectively . By excising these mismatched bases, TDG helps to maintain the integrity of the genetic code and prevent the accumulation of mutations that could potentially lead to cancer and other genetic disorders.

Recombinant Human TDG

Recombinant human TDG is produced using recombinant DNA technology, which involves inserting the human TDG gene into a suitable expression system, such as bacteria or yeast. This allows for the large-scale production of the enzyme, which can then be purified and used for various research and therapeutic applications .

Recombinant TDG is particularly valuable in studying the enzyme’s structure and function, as well as its role in DNA repair mechanisms. It also holds potential for therapeutic applications, such as enhancing DNA repair in cancer cells to improve the efficacy of certain treatments.

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.