TYMS Human

Thymidylate Synthetase Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Function and Mechanism

TYMS catalyzes the reductive methylation of deoxyuridine monophosphate (dUMP) to deoxythymidine monophosphate (dTMP), the sole de novo pathway for thymidylate synthesis . The reaction equation is:

5,10-methylenetetrahydrofolate + dUMPdihydrofolate + dTMP\text{5,10-methylenetetrahydrofolate + dUMP} \rightleftharpoons \text{dihydrofolate + dTMP}

This process is essential for DNA replication and repair, with dTMP subsequently phosphorylated to dTTP for incorporation into DNA . TYMS also auto-regulates its translation and influences cell-cycle progression via interactions with p53 and other oncogenic pathways .

Clinical Significance and Oncogenic Role

TYMS overexpression is linked to tumorigenesis, drug resistance, and poor prognosis in multiple cancers . Key findings include:

Cancer TypeClinical Impact
Hepatocellular CarcinomaAssociated with advanced TNM stage and reduced survival (AUC = 0.72 at 1 year)
Pancreatic Neuroendocrine TumorsLinked to Men1 mutations and chemoresistance
Lung AdenocarcinomaCorrelates with tumor-infiltrating CD4+ cells and metastatic potential

Therapeutic Targeting and Inhibitors

TYMS is a primary target for antimetabolites like 5-FU, which is metabolized to 5-FdUMP, forming a stable ternary complex with TYMS and folate cofactors to block dTMP synthesis . Recent advances include:

  • Combination Therapies: Leucovorin (LV) enhances 5-FU efficacy by stabilizing folate cofactors .

  • Novel Inhibitors: AAV-based therapies and nonclassical antifolates (e.g., AG337) show promise in overcoming resistance .

InhibitorMechanismStatus
5-Fluorouracil (5-FU)Competitive binding to TYMS active siteFDA-approved
AG337Nonclassical antifolate inhibiting TS-folate bindingPreclinical
TYMS shRNAReduces tumor incidence in GEMM modelsExperimental

Research Models and Experimental Data

  • Mouse Models: hTS/Ink4a/Arf mice exhibit enhanced lymphoma and histiocytic sarcoma incidence (31% vs. 15.4% in controls) .

  • In Vitro Studies: TYMS knockdown reduces proliferation in cervical carcinoma (by 60%) and glioblastoma (by 45%) .

  • Structural Insights: X-ray crystallography reveals intermolecular interactions (e.g., Arg174/Arg175) stabilizing the dUMP-TYMS complex .

Product Specs

Introduction
Thymidylate synthase is an enzyme that catalyzes the methylation of deoxyuridylate to deoxythymidylate. This process utilizes 5,10-methylenetetrahydrofolate as a cofactor and is essential for maintaining the dTMP (thymidine-5-prime monophosphate) pool necessary for DNA replication and repair. Thymidylate synthase is a significant target for cancer chemotherapy, serving as the primary site of action for the drug 5-fluoro-2-prime-deoxyuridine and various folate analogs.
Description
Recombinant Human Thymidylate synthase, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 333 amino acids (with a sequence spanning from amino acid 1 to 313) and possesses a molecular weight of 37.8 kDa. This Thymidylate synthase variant includes a 20 amino acid His-Tag fused at its N-terminus and undergoes purification using proprietary chromatographic methods.
Physical Appearance
Clear, colorless solution, sterilized by filtration.
Formulation
The Thymidylate synthase solution is provided at a concentration of 1mg/ml and is formulated in a buffer containing 20mM Tris-HCl (pH 8.0), 1mM DTT, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, freezing at -20°C is recommended. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freezing and thawing cycles should be avoided.
Purity
Purity is determined to be greater than 95.0% using SDS-PAGE analysis.
Synonyms
TMS, EC 2.1.1.45, HST422, Thymidylate synthase, TSase, TS, TYMS, MGC88736.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MPVAGSELPR RPLPPAAQER DAEPRPPHGE LQYLGQIQHI LRCGVRKDDR TGTGTLSVFG MQARYSLRDE FPLLTTKRVF WKGVLEELLW FIKGSTNAKE LSSKGVKIWD ANGSRDFLDS LGFSTREEGD LGPVYGFQWR HFGAEYRDME SDYSGQGVDQ LQRVIDTIKT NPDDRRIIMC AWNPRDLPLM ALPPCHALCQ FYVVNSELSC QLYQRSGDMG LGVPFNIASY ALLTYMIAHI TGLKPGDFIH TLGDAHIYLN HIEPLKIQLQ REPRPFPKLR ILRKVEKIDD FKAEDFQIEG YNPHPTIKME MAV.

Q&A

What is the molecular function of human TYMS and why is it important in cellular metabolism?

TYMS catalyzes the methylation of deoxyuridylate to deoxythymidylate using 10-methylenetetrahydrofolate (methylene-THF) as a cofactor. This reaction is essential for maintaining the dTMP (thymidine-5-prime monophosphate) pool critical for DNA replication and repair . As a key enzyme in the de novo thymidylate synthesis pathway, TYMS plays a vital role in nucleotide metabolism and genomic stability. Methodologically, TYMS activity can be assessed through enzyme assays measuring the conversion of dUMP to dTMP, often using spectrophotometric or radioactive labeling techniques.

The enzyme exists as a protein of approximately 30-35kDa in humans and has been extensively characterized:

CharacteristicDetail
Gene SymbolTYMS
Entrez Gene ID7298
SynonymsHST422, TMS, TS
Gene TypeProtein-coding
OrganismHomo sapiens (human)
Isoelectric Point6.8
Primary FunctiondUMP to dTMP conversion

How is TYMS gene expression regulated in normal cellular processes versus cancer states?

TYMS expression varies inversely with that of a naturally occurring antisense transcript, mitochondrial enolase superfamily member 1 (GeneID:55556), as cell growth progresses from late-log to plateau phase . In cancer, TYMS exhibits pronounced expression across multiple tumor types compared to normal tissues .

Research approaches for studying this differential expression include:

  • RNA-Seq analysis of cancer vs. normal tissue from TCGA and GTEx databases

  • Pathological stage correlation analysis using GEPIA2 platforms

  • Protein-level validation through immunohistochemistry from Human Protein Atlas

  • Promoter analysis to identify cancer-specific transcription factors

What are the key polymorphisms in the TYMS gene and how do researchers investigate their functional significance?

The TYMS gene contains 28-bp tandem repeat sequences or VNTR (variable numbers of tandem repeats) in the 5′-untranslated region (5′-UTR). The most common variants include double (2R) and triple (3R) repeat sequences, with a single G/C nucleotide polymorphism in the triple repeat sequence giving rise to 3Rc or 3Rg structures .

When investigating these polymorphisms, researchers should:

  • Use the native human TYMS Kozak sequence rather than non-native sequences like firefly luciferase Kozak, as this can significantly affect translational efficiency results

  • Employ multiple experimental approaches including transient transfection, antibiotic-selected pools of transfected cells, and stably transfected clones

  • Validate findings across different cell types relevant to the research question

  • Consider both transcriptional and translational effects, as contradictory results exist in the literature

How do TYMS genetic variations correlate with clinical outcomes in cancer treatment?

Clinical studies examining TYMS polymorphisms and treatment outcomes have produced inconsistent results. A recent Blue Cross Blue Shield report concluded that VNTR determination alone could not reliably predict 5-FU toxicity .

When evaluating contradictory literature, researchers should consider:

  • Study design limitations (retrospective vs. prospective)

  • Sample size and statistical power

  • Treatment protocol variations

  • Patient population heterogeneity

  • Potential confounding factors like microsatellite instability (MSI) status, which affects 5-FU response independently

Methodologically, clinical researchers should employ multivariate models that incorporate multiple genetic and clinical factors rather than focusing on single polymorphisms.

How does TYMS expression vary across different cancer types and what are the implications for targeted therapy?

Research using TCGA data demonstrates that TYMS expression is significantly elevated in multiple cancer types compared to normal tissues:

Cancer TypeTYMS Expression StatusP-value
BLCA, BRCA, CESCHigher than normalP<0.01
CHOL, COAD, ESCAHigher than normalP<0.01
GBM, HNSC, KIRCHigher than normalP<0.01
KIRP, LIHC, LUADHigher than normalP<0.01
LUSC, PCPG, STADHigher than normalP<0.01
THCA, UCECHigher than normalP<0.01
KICH, PRAD, TGCTSimilar to normalP>0.05

Furthermore, TYMS expression correlates positively with pathological stages in ACC, KICH, LIHC, and TGCT, while showing negative correlations in COAD, LUSC, and OV . These expression patterns can guide the selection of cancer types most likely to respond to TYMS-targeting therapies.

What mechanisms underlie cancer cell resistance to TYMS inhibitors and how can researchers investigate these pathways?

TYMS has been extensively studied as a target for cancer chemotherapeutic agents, particularly 5-fluorouracil (5-FU) and its derivatives . Resistance mechanisms include:

  • TYMS gene amplification leading to increased enzyme expression

  • Mutations affecting drug binding sites

  • Alterations in folate metabolism pathways

  • Enhanced DNA repair mechanisms

  • Changes in drug metabolism or cellular uptake

To investigate these mechanisms, researchers should employ:

  • Cell line models with acquired resistance through gradual drug exposure

  • CRISPR-Cas9 gene editing to introduce or correct specific mutations

  • Combined genomic and proteomic profiling to identify pathway alterations

  • Functional assays measuring TYMS catalytic activity in resistant models

  • Drug combination studies to overcome specific resistance mechanisms

What are the optimal approaches for studying TYMS protein interactions and how do they inform our understanding of its cellular functions?

TYMS protein interactions can be studied through multiple complementary approaches:

  • Binding ELISA assays: The research literature describes a binding ELISA method to detect the interaction between TYMS and AST (Aspartate Aminotransferase) . This technique involves:

    • Coating microtiter wells with one protein (e.g., AST)

    • Adding serially diluted partner protein (TYMS)

    • Incubating with anti-TYMS polyclonal antibody

    • Detection with HRP-labeled secondary antibody

    • Quantification through colorimetric substrate reaction

  • Additional methods include:

    • Co-immunoprecipitation with mass spectrometry identification

    • Yeast two-hybrid screening for systematic interaction mapping

    • Proximity labeling techniques (BioID, APEX2) for in vivo interaction studies

    • Structural biology approaches to characterize binding interfaces

Methodologically, researchers should employ multiple complementary techniques and include appropriate controls to confirm specificity of interactions.

What considerations are important when designing experiments to evaluate TYMS inhibitors as cancer therapeutics?

When evaluating TYMS inhibitors, researchers should implement a multi-level experimental approach:

  • Biochemical characterization:

    • Enzyme inhibition kinetics (Ki, IC50 determination)

    • Binding affinity measurements (isothermal titration calorimetry, surface plasmon resonance)

    • Structural studies (X-ray crystallography, cryo-EM) to elucidate binding modes

  • Cellular models:

    • Dose-response studies in multiple cell lines

    • Time-course experiments to determine optimal treatment duration

    • Combination studies with other therapeutic agents

    • Assessment of dTMP pool depletion and DNA damage markers

  • Mechanistic validation:

    • Target engagement assays in intact cells

    • Genetic approaches (TYMS overexpression, knockdown) to confirm specificity

    • Rescue experiments with thymidine supplementation

    • Investigation of downstream pathways activated by TYMS inhibition

  • Translational considerations:

    • Correlation with TYMS expression levels across cell lines

    • Assessment of genetic markers (TYMS polymorphisms) and response

    • Evaluation in 3D cultures and patient-derived models

    • Pharmacokinetic and pharmacodynamic studies in vivo

How can researchers effectively analyze TYMS expression and genetic variation data from public databases?

Researchers can leverage multiple public databases to analyze TYMS expression patterns:

Methodological best practices include:

  • Using log2-transformed expression values (TPM+1) for analysis

  • Applying appropriate statistical tests with multiple testing correction

  • Validating findings across multiple independent datasets

  • Integrating expression data with clinical parameters for meaningful correlations

What statistical approaches are most appropriate for analyzing conflicting data regarding TYMS polymorphisms and their clinical relevance?

When faced with conflicting data on TYMS polymorphisms and clinical outcomes, researchers should employ:

  • Meta-analytical approaches:

    • Fixed or random effects models depending on heterogeneity assessment

    • Subgroup analyses stratified by ethnicity, cancer type, and treatment regimen

    • Publication bias assessment using funnel plots and Egger's test

    • Sensitivity analyses excluding outlier studies

  • Bayesian methods:

    • Prior probability incorporation based on functional evidence

    • Network meta-analysis for comparing multiple polymorphisms

    • Probabilistic assessment of clinical significance

  • Machine learning techniques:

    • Random forest or support vector machines for multi-marker integration

    • Feature selection to identify most predictive polymorphisms

    • Cross-validation to assess predictive performance

    • Ensemble methods combining multiple algorithms

  • Causal inference methods:

    • Mendelian randomization to establish causal relationships

    • Mediation analysis to identify intermediate phenotypes

    • Directed acyclic graphs to visualize confounding relationships

These approaches should be complemented by functional validation studies to establish biological plausibility for statistically significant associations.

What emerging technologies are transforming our understanding of TYMS function and regulation?

Recent technological advances are revolutionizing TYMS research:

  • CRISPR-Cas9 genome editing:

    • Precise modification of endogenous TYMS locus

    • Introduction of specific polymorphisms to study functional effects

    • Creation of reporter knock-ins for real-time expression monitoring

    • High-throughput screening of genetic modifiers

  • Single-cell technologies:

    • Analysis of TYMS expression heterogeneity within tumors

    • Correlation with cell cycle states and differentiation

    • Multi-omics integration at single-cell resolution

    • Spatial transcriptomics to map TYMS expression in tissue context

  • Structural biology advances:

    • Cryo-EM for dynamic structural states of TYMS complexes

    • Hydrogen-deuterium exchange mass spectrometry for conformational changes

    • Integrative structural modeling combining multiple data types

    • In-cell NMR for studying TYMS in native environments

  • Systems biology approaches:

    • Metabolic flux analysis of nucleotide synthesis pathways

    • Network modeling of TYMS interactions with related enzymes

    • Multi-scale modeling linking molecular mechanisms to cellular phenotypes

    • In silico drug screening and rational inhibitor design

How might TYMS research contribute to precision medicine approaches in cancer treatment?

TYMS research has significant potential to advance precision oncology through:

  • Biomarker development:

    • Integrated genetic panels combining TYMS polymorphisms with other predictive markers

    • Expression-based stratification for TYMS inhibitor sensitivity

    • Dynamic monitoring of TYMS activity during treatment

    • Liquid biopsy approaches for non-invasive assessment

  • Novel therapeutic strategies:

    • Dual-targeting approaches combining TYMS inhibition with complementary pathways

    • Development of inhibitors targeting specific TYMS variants or conformational states

    • Delivery technologies for selective targeting to tumor cells

    • Immunotherapeutic approaches targeting TYMS as a tumor-associated antigen

  • Treatment personalization:

    • Algorithm development for optimizing 5-FU dosing based on TYMS genotype

    • Machine learning models integrating multiple biomarkers for response prediction

    • Adaptive treatment strategies guided by molecular monitoring

    • Prevention of toxicity through prophylactic interventions in high-risk patients

  • Combination therapy rationales:

    • Synthetic lethality approaches exploiting TYMS inhibition vulnerabilities

    • Rationalized drug combinations based on pathway interactions

    • Sequential therapy designs to overcome resistance mechanisms

    • Integration with immunotherapy based on TYMS-mediated immune effects

Product Science Overview

Introduction

Thymidylate synthetase, also known as thymidylate synthase (TS), is a crucial enzyme in the de novo synthesis of thymidylate (dTMP), an essential precursor for DNA replication and repair. This enzyme catalyzes the conversion of deoxyuridine monophosphate (dUMP) to deoxythymidine monophosphate (dTMP) using 5,10-methylenetetrahydrofolate as a cofactor . Thymidylate synthetase is highly conserved across various species, including humans, and plays a pivotal role in maintaining the balance of deoxynucleotide pools within the cell .

Structure and Function

Thymidylate synthetase is a homodimeric enzyme, meaning it consists of two identical subunits. Each subunit contains an active site where the catalytic reaction occurs. The enzyme’s structure is highly conserved, with bacterial and mammalian TSases sharing remarkable similarities . The active site of thymidylate synthetase binds to both dUMP and the folate cofactor, facilitating the transfer of a methyl group to form dTMP .

Biological Significance

The activity of thymidylate synthetase is critical for DNA synthesis and cell proliferation. Inhibition or dysregulation of this enzyme can lead to an imbalance in deoxynucleotide pools, resulting in DNA damage and cell death . Due to its essential role in DNA synthesis, thymidylate synthetase is a target for chemotherapeutic agents, particularly in the treatment of cancer . Overexpression of thymidylate synthetase has been associated with resistance to certain chemotherapeutic drugs, such as 5-fluorouracil (5-FU), making it a significant focus of cancer research .

Recombinant Thymidylate Synthetase

Human recombinant thymidylate synthetase is produced using recombinant DNA technology, which involves inserting the human TS gene into a suitable expression system, such as Escherichia coli or yeast. This allows for the large-scale production of the enzyme for research and therapeutic purposes. Recombinant thymidylate synthetase retains the same structural and functional properties as the native enzyme, making it a valuable tool for studying enzyme kinetics, drug interactions, and the development of new therapeutic agents .

Clinical and Research Applications

Thymidylate synthetase is a key target in cancer therapy due to its role in DNA synthesis. Inhibitors of this enzyme, such as 5-FU and its derivatives, are commonly used in the treatment of various cancers, including colorectal, breast, and gastric cancers . Research on thymidylate synthetase also focuses on understanding the mechanisms of drug resistance and developing new strategies to overcome it. Additionally, the enzyme is used in biochemical studies to elucidate the molecular mechanisms of DNA synthesis and repair .

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.