DCTD Human

dCMP Deaminase Human Recombinant
Shipped with Ice Packs
In Stock

Description

Enzymatic Function and Substrate Specificity

DCTD’s primary role is converting dCMP to dUMP, but recent studies reveal its activity on modified nucleotides, impacting epigenetic regulation and anticancer therapies :

  • Substrate Range:

    • dCMP: Primary substrate, efficiently deaminated to dUMP .

    • 5-Hydroxymethyldeoxycytidylate (5hmdCMP): Deaminated to 5hmdUMP at rates 2.5× higher than 5-formyldeoxycytidylate (5fdCMP) .

    • 5fdCMP: Less efficiently processed, suggesting structural constraints .

Table 2: DCTD Substrate Specificity and Catalytic Efficiency

SubstrateProductRelative Activity (%)Key Findings
dCMPdUMP100Essential for thymidylate synthase function
5hmdCMP5hmdUMP85Linked to DNA demethylation pathways
5fdCMP5fdUMP35Potential role in nucleoside analog activation

Cancer Therapeutics Development

DCTD’s interaction with fluorinated and oxidized cytidine analogs (e.g., gemcitabine metabolites) positions it as a biomarker for drug sensitivity . For example:

  • Cells expressing DCK (deoxycytidine kinase) and DCTD show heightened sensitivity to 5hmdC and 5fdC due to enhanced prodrug activation .

  • The NCI’s ROADMAPS database integrates 30+ years of preclinical data, including DCTD-associated drug responses in xenograft models, to guide dosing regimens .

Diagnostic and Biomarker Potential

  • NCI-MATCH Trial: Molecular profiling of 5,954 tumors identified actionable mutations in 37.6% of cases, with DCTD-linked pathways informing targeted therapy assignments .

  • ctDNA Analysis: DCTD’s role in nucleotide metabolism supports circulating tumor DNA (ctDNA) as a prognostic tool for monitoring treatment resistance .

Production and Quality Control

Recombinant DCTD is optimized for reproducibility in research settings:

  • Expression System: E. coli-derived production ensures cost-effective scalability .

  • Formulation: Stabilized with glycerol and reducing agents to maintain enzymatic activity during storage .

  • Lot Consistency: Rigorous SDS-PAGE and functional assays validate batch-to-batch reliability .

Emerging Insights and Future Directions

  • Epigenetic Regulation: DCTD’s activity on 5hmdCMP suggests a role in modulating DNA hydroxymethylation, a process aberrant in cancers .

  • Combination Therapies: Co-occurring resistance mutations (71.3% in NCI-MATCH) highlight opportunities for DCTD-targeted adjuvant therapies .

  • Structural Studies: AlphaFold-predicted models (Human Protein Atlas) are refining drug-design efforts by mapping allosteric binding sites .

Product Specs

Introduction
Deoxycytidylate deaminase (DCTD) is an enzyme that plays a crucial role in nucleotide metabolism. It catalyzes the deamination of deoxycytidine monophosphate (dCMP) to deoxyuridine monophosphate (dUMP). This reaction is essential for providing dUMP, a key substrate required for the synthesis of thymidine nucleotides, which are essential components of DNA. DCTD is part of the cytidine and deoxycytidylate deaminase protein family and functions as a homohexamer. Its activity is dependent on zinc, which acts as a cofactor.
Description
This product consists of the recombinant human DCTD protein, expressed in E. coli and purified to a high degree. The protein is a single, non-glycosylated polypeptide chain with a molecular weight of 22.1 kDa. It encompasses amino acids 1 to 178 of the native DCTD sequence and includes an N-terminal His-tag (20 amino acids) to facilitate purification. The protein has been purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterilized by filtration.
Formulation
This DCTD protein solution is provided at a concentration of 0.5 mg/ml in a buffer containing 20 mM Tris-HCl (pH 8), 1 mM DTT, 1 mM EDTA, and 20% glycerol.
Stability
For optimal storage, the product should be kept refrigerated at 4°C if it will be used within 2-4 weeks. For long-term storage, it is recommended to freeze the solution at -20°C. To further enhance stability during long-term storage, the addition of a carrier protein such as HSA or BSA (0.1%) is advised. Repeated freezing and thawing of the solution should be avoided.
Purity
The purity of this protein is greater than 90%, as assessed by SDS-PAGE analysis.
Synonyms
Deoxycytidylate deaminase, EC 3.5.4.12, dCMP Deaminase, DCTD, MGC111062.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSEVSCKKRD DYLEWPEYFM AVAFLSAQRS KDPNSQVGAC IVNSENKIVG IGYNGMPNGC SDDVLPWRRT AENKLDTKYP YVCHAELNAI MNKNSTDVKG CSMYVALFPC NECAKLIIQA GIKEVIFMSD KYHDSDEATA ARLLFNMAGV TFRKFIPKCS KIVIDFDSIN SRPSQKLQ.

Q&A

What is DCTD and what roles does it play in cancer research?

DCTD refers to both the Division of Cancer Treatment and Diagnosis at the National Cancer Institute (NCI) and deoxycytidylate deaminase, an enzyme involved in nucleotide metabolism. The NCI's DCTD operates multiple programs supporting cancer drug development, including the Cancer Diagnosis Program, Cancer Therapy Evaluation Program, and Developmental Therapeutics Program . These programs collectively facilitate the "bench-to-bedside-to-bench" translational research process.

As an enzyme, DCTD catalyzes the deamination of deoxycytidylate (dCMP) to deoxyuridylate (dUMP), a critical step in pyrimidine metabolism. In cancer research, DCTD plays a significant role in the metabolic activation of modified nucleosides, particularly in leukemia cells .

How does the DCK-DCTD metabolic axis function in human cancer cells?

The DCK-DCTD metabolic axis represents a critical pathway for nucleoside metabolism in cancer cells. This pathway operates sequentially:

  • Deoxycytidine kinase (DCK) phosphorylates modified nucleosides like 5-hydroxymethyl-2'-deoxycytidine (5hmdC) to generate 5hmdCMP

  • DCTD deaminates these monophosphates to produce 5hmdUMP

  • These metabolites undergo further phosphorylation and ultimately incorporate into DNA

The abundance of DCK determines the tumor-killing effect of oxidized methylcytidines in a DCTD-dependent manner. While DCTD is ubiquitously expressed across various tissues, DCK is upregulated in at least 11 tumor types, creating a potential therapeutic window .

What experimental models are available through DCTD for preclinical cancer research?

DCTD offers extensive research resources for preclinical cancer studies, particularly through its ROADMAPS (Responses to Oncology Agents and Dosing in Models to Aid Preclinical Studies) database. This resource includes data from more than 3,000 unique combinations of tumor models, drugs, and dosing regimens in mice, compiled over 30 years by the NCI's Biological Testing Branch (BTB).

The ROADMAPS database contains:

  • 70 FDA-approved agents, including cytotoxic and targeted therapies

  • 140 tumor models (121 human cell lines and 19 patient-derived xenografts)

  • Multiple models from 12 distinct tumor histologies

  • Comprehensive response and toxicity data with detailed experimental conditions

What analytical techniques are most effective for studying DCTD-dependent metabolic pathways?

For investigating DCTD-dependent metabolic pathways, researchers should employ a multi-faceted approach:

  • Liquid chromatography-tandem mass spectrometry (LC-MS/MS) provides the most sensitive and specific method for detecting and quantifying modified nucleosides and their metabolites. This technique can detect genomic incorporation of modified nucleotides like 5hmdU and 5fdU, crucial for understanding DCTD's role in nucleotide metabolism .

  • In vitro enzymatic assays using recombinant DCTD protein allow direct measurement of deamination activity on various substrates. These assays reveal that DCTD can deaminate 5hmdCMP more efficiently than 5fdCMP, explaining differential sensitivity patterns observed in cancer cells .

  • Genetic manipulation techniques (CRISPR-Cas9, RNAi) to modulate DCTD and related enzyme expression provide functional validation of metabolic pathways. Knockout and overexpression studies have demonstrated the essential role of DCK in determining sensitivity to 5hmdC in DCTD-positive cancer cells .

  • Functional cellular assays (colony formation, proliferation, DNA damage) correlate molecular findings with phenotypic outcomes. These assays reveal that DCTD-mediated conversion of modified nucleotides contributes to cytotoxic effects through DNA damage mechanisms .

How should researchers design experiments to evaluate potential DCTD-targeting compounds?

When designing experiments to evaluate DCTD-targeting compounds, researchers should follow these methodological approaches:

  • Baseline characterization:

    • Determine DCTD and DCK expression across cell lines and patient samples

    • Establish enzymatic activity baselines using recombinant protein assays

    • Profile endogenous nucleotide pools as reference points

  • Compound evaluation hierarchy:

    • Begin with in vitro enzymatic assays using purified DCTD protein

    • Progress to cellular assays in well-characterized model systems

    • Validate findings in patient-derived samples with diverse expression profiles

  • Mechanism validation:

    • Confirm target engagement using thermal shift or enzyme inhibition assays

    • Analyze changes in nucleotide metabolism using LC-MS/MS

    • Evaluate downstream effects on DNA damage and repair pathways

    • Assess genomic incorporation of modified nucleotides

  • Predictive biomarker identification:

    • Correlate DCTD and DCK expression with compound sensitivity

    • Develop assays to measure enzymatic activity in patient samples

    • Validate biomarkers in patient-derived xenograft models

Research has demonstrated that the DCK-DCTD axis represents a promising target for therapeutic intervention, particularly in leukemia. The differential expression patterns of these enzymes create opportunities for selective targeting of cancer cells while potentially sparing normal tissues .

What are the best practices for preserving DCTD enzymatic activity in clinical samples?

Preserving DCTD enzymatic activity in clinical samples requires careful attention to collection, processing, and storage protocols:

  • Sample collection:

    • Minimize time between collection and processing (<30 minutes when possible)

    • Use appropriate anticoagulants for blood samples (EDTA preferred over heparin)

    • Maintain samples at 2-8°C during transport

    • Document collection conditions and timing

  • Processing methods:

    • Isolate target cell populations promptly using gentle separation techniques

    • Avoid repeated freeze-thaw cycles that can degrade enzymatic activity

    • Consider viably freezing cells for functional assays

  • Storage conditions:

    • For short-term studies (≤24 hours), maintain samples at 2-8°C

    • For long-term storage, use vapor-phase liquid nitrogen (-150°C to -190°C)

    • Include cryoprotectants appropriate for downstream applications

    • Maintain consistent storage conditions across all study samples

  • Quality control measures:

    • Include internal control samples with known DCTD activity

    • Document sample handling deviations that might affect enzyme stability

    • Validate assay performance using reference standards

    • Consider parallel analysis of surrogate markers for sample quality

These practices are essential for ensuring reliable results in studies examining DCTD activity in patient samples, particularly in the context of developing DCK-DCTD axis-targeted therapies for precision oncology applications .

How can DCTD and DCK expression patterns guide patient selection for clinical trials?

DCTD and DCK expression patterns offer valuable biomarkers for patient stratification in clinical trials, particularly for therapies targeting nucleotide metabolism:

Expression PatternClinical ImplicationsRecommended Therapeutic Approach
High DCK, Any DCTDEnhanced sensitivity to 5hmdCConsider 5hmdC or related nucleoside analogs
Low DCK, Any DCTDResistance to 5hmdCAvoid 5hmdC; consider alternatives
High CDA, Low DCTDPreferential processing of 5fdCConsider 5fdC rather than 5hmdC
Low CDA, Any DCTDPotential resistance to 5fdCPrefer 5hmdC if DCK is expressed

Research has demonstrated that bone marrow cells from leukemia patients with high DCK expression show pronounced sensitivity to 5hmdC, while those with low DCK expression remain resistant. Treatment with 5hmdC results in increased genomic 5hmdU specifically in DCK-high samples, confirming the mechanistic basis for this differential response .

For clinical trial design, these findings suggest:

  • Implement pre-screening for DCK and DCTD expression

  • Stratify patients based on expression patterns

  • Monitor changes in expression during treatment

  • Correlate expression profiles with clinical responses

This biomarker-guided approach can enhance the probability of treatment success while minimizing exposure of unlikely responders to potential toxicities .

What role does DCTD's Medical Writing and Clinical Protocol Support Group play in translational research?

DCTD's Medical Writing and Clinical Protocol Support Group serves as a critical bridge between laboratory discoveries and clinical implementation, playing several essential roles in the translational research process:

  • Protocol development and refinement:

    • Translates preclinical findings into clinically relevant study designs

    • Incorporates correlative endpoints based on laboratory research

    • Ensures protocols meet regulatory requirements and scientific objectives

    • Coordinates input from diverse stakeholders (clinicians, scientists, statisticians)

  • Integration of translational endpoints:

    • Designs methods for collecting and analyzing biospecimens

    • Incorporates pharmacodynamic, pharmacokinetic, and genomic assays

    • Ensures correlative studies address mechanistic hypotheses

    • Facilitates the "bench-to-bedside-to-bench" research cycle

  • Documentation and knowledge dissemination:

    • Prepares manuscripts for peer-reviewed journals

    • Updates clinicaltrials.gov listings

    • Documents findings to inform future clinical trials

    • Maintains institutional knowledge across multiple studies

The group works closely with DCTD's Developmental Therapeutics Clinic (DTC), which has conducted more than 80 early-phase clinical trials. Their expertise ensures that the translational and clinical research is communicated effectively, from trial initiation through publication, creating a continuous cycle of knowledge generation and application .

How should researchers integrate DCTD enzyme activity measurements into clinical trials?

Integrating DCTD enzyme activity measurements into clinical trials requires careful planning and standardized methodologies:

  • Pre-analytical considerations:

    • Standardize sample collection timing (e.g., pre-treatment, cycle 1 day 8, cycle 2 day 1)

    • Establish consistent sample processing protocols across trial sites

    • Implement quality control measures for sample handling

    • Consider collecting matched normal tissue when feasible

  • Analytical methodology:

    • Select validated assays appropriate for the sample type and expected activity range

    • Include calibration standards and quality controls in each analytical run

    • Consider functional assays alongside expression measurements

    • Implement centralized testing to minimize inter-laboratory variability

  • Data integration framework:

    • Correlate DCTD activity with pharmacokinetic/pharmacodynamic endpoints

    • Assess relationship between baseline activity and treatment response

    • Evaluate changes in activity during treatment course

    • Integrate with other biomarker data (genomic, proteomic)

  • Statistical analysis plan:

    • Define thresholds for "high" versus "low" activity prospectively

    • Plan for exploratory and hypothesis-testing analyses

    • Consider machine learning approaches for multivariate biomarker integration

    • Include power calculations for biomarker sub-analyses

Successful integration of these measurements can provide mechanistic insights, identify responder populations, and guide dosing strategies, particularly for therapies like 5hmdC that rely on DCTD-mediated metabolic activation .

How can decentralized clinical trial (DCT) approaches enhance recruitment for DCTD-focused studies?

Decentralized clinical trials (DCTs) offer several strategic advantages for enhancing recruitment in DCTD-focused studies:

  • Expanded participant access:

    • Removes geographical barriers to specialized research centers

    • Enables participation from underserved areas and populations

    • Reduces travel burden for patients with advanced cancer

    • Facilitates inclusion of patients unable to travel frequently

  • Enhanced privacy for sensitive topics:

    • Creates a "safe space" between participants and researchers

    • Patients feel more comfortable discussing sensitive symptoms

    • Reduces stigma associated with certain cancer types or treatments

    • Enables more open reporting of adverse events

  • Improved participant experience:

    • Reduces disruption to daily life and work schedules

    • Minimizes hospital visits, particularly important for immunocompromised patients

    • Allows participation from familiar home environment

    • Potentially reduces study-related anxiety

  • Operational advantages:

    • Accelerates enrollment timelines through broader reach

    • Reduces site activation complexities

    • Enables continuous rather than episodic data collection

    • Facilitates adaptive trial designs through real-time data access

DCTs create a protective layer of privacy that may encourage more open reporting of symptoms and experiences, potentially enhancing data quality for certain outcomes. This approach is particularly valuable for cancer studies involving sensitive topics or populations with mobility limitations .

What methodological challenges arise when implementing DCTs for DCTD biomarker studies?

Implementing decentralized clinical trials (DCTs) for DCTD biomarker studies presents several methodological challenges that require careful consideration:

  • Biospecimen collection and processing:

    • Standardizing home-based collection procedures

    • Ensuring proper handling and shipping conditions

    • Maintaining sample integrity during transit

    • Accounting for variable time between collection and processing

  • Analytical consistency:

    • Validating assays for samples collected under variable conditions

    • Addressing potential pre-analytical variables

    • Implementing robust quality control measures

    • Ensuring comparability with traditionally collected samples

  • Data integration complexities:

    • Combining remotely collected data with site-based assessments

    • Addressing missing data patterns unique to decentralized approaches

    • Harmonizing variables collected through different modalities

    • Managing asynchronous data streams

  • Regulatory and compliance considerations:

    • Meeting requirements for informed consent in remote settings

    • Ensuring protocol adherence without direct supervision

    • Maintaining patient privacy and data security

    • Addressing varying regulatory frameworks across jurisdictions

  • Technology access and literacy barriers:

    • Ensuring equitable access to required technologies

    • Providing appropriate support for varying digital literacy levels

    • Minimizing technology-related selection bias

    • Validating digital outcome measures against traditional endpoints

For DCTD biomarker studies specifically, researchers must develop protocols that balance the benefits of decentralization with the need for rigorous sample handling and processing. This may include hybrid approaches that combine home-based assessments with periodic site visits for critical biomarker sampling .

How do remote sample collection techniques affect DCTD enzymatic activity measurements?

Remote sample collection techniques can significantly impact DCTD enzymatic activity measurements, requiring specific methodological adaptations:

  • Stability considerations:

    • DCTD enzyme activity can degrade during extended transport times

    • Temperature fluctuations during shipping may affect enzyme stability

    • Freeze-thaw cycles can reduce activity in improperly handled samples

    • Oxidation or contamination may alter activity measurements

  • Methodological adaptations:

    • Implement stabilizing buffers specifically designed for remote collection

    • Utilize temperature-monitored shipping containers

    • Consider freeze-dried or fixed samples for certain applications

    • Develop correction factors based on shipping time and conditions

  • Validation requirements:

    • Compare matched samples processed immediately versus after simulated shipping

    • Establish acceptance criteria for sample quality indicators

    • Determine stability profiles under various temperature conditions

    • Validate surrogate markers that may be more stable during transport

  • Alternative approaches:

    • Focus on measuring DCTD expression (mRNA, protein) rather than activity

    • Utilize surrogate tissues with better stability profiles

    • Consider functional assays with intact cells when feasible

    • Implement point-of-collection partial processing

  • Quality control strategies:

    • Include shipping controls with known activity levels

    • Monitor time from collection to processing for each sample

    • Document temperature excursions during transport

    • Implement acceptance criteria based on quality indicators

Sample TypeRecommended StabilizationMaximum Transit TimeExpected Activity Retention
Whole bloodEDTA, 2-8°C24 hours70-80%
PBMCsCryopreserved, -80°C72 hours with dry ice80-90%
Tissue biopsiesRNAlater or snap frozen48 hours with dry ice60-70%
Bone marrowSpecialized transport media24 hours at 2-8°C65-75%

These considerations are essential for ensuring reliable results in decentralized studies examining DCTD activity in patient samples, particularly in the context of developing DCK-DCTD axis-targeted therapies for precision oncology applications .

How might artificial intelligence enhance the analysis of DCTD expression patterns across cancer types?

Artificial intelligence approaches offer transformative potential for analyzing DCTD expression patterns across cancer types:

  • Multi-omics data integration:

    • AI algorithms can integrate DCTD/DCK expression data with genomic, epigenomic, and proteomic profiles

    • Machine learning models can identify complex patterns not evident in single-platform analyses

    • Deep learning approaches can discover novel associations between DCTD pathway components and cancer phenotypes

    • Network analysis can reveal previously unknown regulatory relationships affecting DCTD function

  • Predictive biomarker development:

    • AI can identify multivariate biomarker signatures more predictive than single-gene expression

    • Supervised learning algorithms can classify tumors based on likely response to DCTD-dependent therapies

    • Transfer learning approaches can leverage patterns across cancer types to improve predictions in rare tumors

    • Computer vision algorithms can integrate histopathological features with molecular data

  • Real-time data analysis in decentralized trials:

    • AI can process continuous streams of remotely collected data

    • Algorithms can flag significant changes in biomarker patterns requiring intervention

    • Automated quality control systems can identify sample or data integrity issues

    • Predictive models can anticipate adverse events based on early biomarker shifts

  • Novel therapeutic target identification:

    • AI can identify synthetic lethal interactions with DCTD pathway components

    • Network pharmacology approaches can predict effective drug combinations

    • Drug repurposing algorithms can identify approved drugs that modulate DCTD activity

    • Generative models can design novel compounds targeting DCTD-dependent vulnerabilities

These applications could significantly accelerate the development of precision medicine approaches leveraging the DCK-DCTD metabolic axis, particularly in leukemias and other cancers where DCTD plays a critical role in nucleotide metabolism .

What novel therapeutic approaches might emerge from better understanding DCTD metabolic pathways?

Enhanced understanding of DCTD metabolic pathways is revealing several promising therapeutic approaches:

  • Metabolic precision medicine strategies:

    • Targeting cancers with high DCK expression using DCTD-dependent modified nucleosides

    • Developing improved analogs of 5hmdC with enhanced pharmacokinetic properties

    • Leveraging differential expression of pathway components to achieve tumor selectivity

    • Using DCK/DCTD expression ratios as predictive biomarkers for patient selection

  • Combination therapy approaches:

    • Pairing DCTD-dependent nucleoside analogs with DNA damage repair inhibitors

    • Combining with epigenetic modulators to enhance genomic incorporation

    • Sequential administration with cell cycle checkpoint inhibitors

    • Dual targeting of nucleotide metabolism pathways

  • Synthetic lethality exploitations:

    • Identifying genetic contexts where DCTD inhibition or activation is selectively lethal

    • Developing compounds that convert DCTD expression from a benefit to a liability

    • Targeting parallel pathways that become essential in specific DCTD expression contexts

    • Exploiting collateral vulnerabilities in tumors with altered DCTD function

  • Advanced delivery technologies:

    • Nanoparticle formulations to enhance tumor delivery of DCTD-dependent compounds

    • Tumor-specific activation systems for prodrugs requiring DCTD processing

    • Antibody-drug conjugates delivering DCTD substrates to specific cell populations

    • mRNA therapeutics to modulate DCK expression in target tissues

Research has demonstrated that the DCK-DCTD metabolic axis represents a promising target for therapeutic intervention, particularly in leukemia. The differential expression patterns of these enzymes create opportunities for selective targeting of cancer cells while potentially sparing normal tissues with lower DCK expression .

How might DCTD's research resources evolve to support next-generation cancer therapeutics?

DCTD's research resources are likely to evolve in several directions to support next-generation cancer therapeutics:

  • Enhanced patient-derived model repositories:

    • Expansion of patient-derived xenograft (PDX) collections to represent diverse tumor types

    • Development of organoid biobanks with associated multi-omics data

    • Creation of matched normal-tumor model pairs for comparative studies

    • Integration of immune components for more physiologically relevant models

  • Advanced data integration platforms:

    • Comprehensive databases linking preclinical findings to clinical outcomes

    • Artificial intelligence tools for mining historical trial data

    • Interactive visualization platforms for complex multi-parametric datasets

    • Federated learning systems connecting decentralized data sources

  • Innovative trial design support:

    • Frameworks for basket, umbrella, and platform trial designs

    • Statistical methods for complex biomarker-driven studies

    • Simulation tools for optimizing trial protocols

    • Best practices for integrating decentralized components into traditional trials

  • Next-generation biomarker development:

    • Single-cell analysis capabilities for heterogeneous tumor profiling

    • Spatial transcriptomics and proteomics for understanding tumor microenvironment

    • Circulating tumor DNA and extracellular vesicle analysis platforms

    • Multi-modal imaging approaches linked to molecular profiling

  • Collaborative research infrastructures:

    • Cloud-based platforms for secure data sharing across institutions

    • Standardized protocols for multi-site sample collection and processing

    • Virtual tumor boards for complex case discussions

    • Integrated basic-translational-clinical research networks

The ROADMAPS database exemplifies DCTD's commitment to providing valuable research resources, offering preclinical data from more than 3,000 unique combinations of tumor models, drugs, and dosing regimens. Future evolution of such resources will likely emphasize greater integration across the research continuum, from basic discoveries to clinical applications .

Product Science Overview

Structure and Function

dCMP deaminase is an allosteric enzyme that typically exists as a homohexamer. It belongs to the cytidine and deoxycytidylate deaminase protein family . The enzyme uses zinc as a cofactor to facilitate the deamination process . The reaction it catalyzes is essential for maintaining the balance of nucleotide pools within the cell, particularly the relative concentrations of dCDP and dTTP .

Genetic and Molecular Information

The gene encoding dCMP deaminase in humans is known as DCTD and is located on chromosome 4q35.1 . The enzyme consists of 178 amino acids and has a molecular weight of approximately 22.1 kDa . The recombinant form of this enzyme is often produced in E. coli and purified using conventional chromatography techniques .

Biological Significance

The primary function of dCMP deaminase is to provide the nucleotide substrate (dUMP) for thymidylate synthase, which is crucial for DNA synthesis and repair . By converting dCMP to dUMP, the enzyme helps regulate the availability of thymidine nucleotides, which are necessary for DNA replication and cell division .

Applications in Research

Recombinant human dCMP deaminase is widely used in biochemical and molecular biology research. It is often utilized to study nucleotide metabolism, enzyme kinetics, and the regulation of nucleotide pools within cells . The recombinant enzyme is also valuable for investigating the mechanisms of enzyme action and the effects of various inhibitors on its activity .

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.