TYMP Human

Thymidine Phosphorylase Human Recombinant
Shipped with Ice Packs
In Stock

Description

TYMP Human Recombinant fused with a 21 amino acid His tag at N-terminus produced in E.Coli is a single, non-glycosylated, polypeptide chain containing 493 amino acids (11-482 a.a.) and having a molecular mass of 51.3kDa. The TYMP is purified by proprietary chromatographic techniques.

Product Specs

Introduction
Thymidine phosphorylase precursor (TYMP) is a platelet-derived endothelial cell growth factor that catalyzes the formation of thymine and 2-deoxy-D-ribose-1-phosphate from thymidine and orthophosphate. TYMP is an angiogenic inducer that potently stimulates the growth of endothelial cells and induces chemotaxis. TYMP has a highly restricted target cell specificity, acting only on endothelial cells. Increased TYMP expression is found in various solid tumors and inflammatory diseases and is frequently associated with a poor prognosis. Mutations in the TYMP gene are linked to mitochondrial neurogastrointestinal encephalomyopathy.
Description
Recombinant human TYMP, fused with a 21 amino acid His tag at the N-terminus, is produced in E. coli. This single, non-glycosylated polypeptide chain contains 493 amino acids (11-482 a.a.) and has a molecular mass of 51.3 kDa. The TYMP protein is purified using proprietary chromatographic techniques.
Physical Appearance
Sterile, colorless solution.
Formulation
The TYMP solution (1 mg/ml) is supplied in 20 mM Tris-HCl buffer (pH 8.0), 1 mM DTT, and 10% glycerol.
Stability
Store at 4°C if the entire vial will be used within 2-4 weeks. For longer storage, store frozen at -20°C. It is recommended to add a carrier protein (0.1% HSA or BSA) for long-term storage. Avoid multiple freeze-thaw cycles.
Purity
Greater than 90.0% as determined by SDS-PAGE.
Synonyms
Thymidine phosphorylase, Gliostatin, Platelet-derived endothelial cell growth factor, PD-ECGF, TdRPase, TYMP, ECGF1, TP, MNGIE, MEDPS1, MTDPS1, PDECGF, hPD-ECGF.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MAPPAPGDFS GEGSQGLPDP SPEPKQLPEL IRMKRDGGRL SEADIRGFVA AVVNGSAQGA QIGAMLMAIR LRGMDLEETS VLTQALAQSG QQLEWPEAWR QQLVDKHSTG GVGDKVSLVL APALAACGCK VPMISGRGLG HTGGTLDKLE SIPGFNVIQS PEQMQVLLDQ AGCCIVGQSE QLVPADGILY AARDVTATVD SLPLITASIL SKKLVEGLSA LVVDVKFGGA AVFPNQEQAR ELAKTLVGVG ASLGLRVAAA LTAMDKPLGR CVGHALEVEE ALLCMDGAGP PDLRDLVTTL GGALLWLSGH AGTQAQGAAR VAAALDDGSA LGRFERMLAA QGVDPGLARA LCSGSPAERR QLLPRAREQE ELLAPADGTV ELVRALPLAL VLHELGAGRS RAGEPLRLGV GAELLVDVGQ RLRRGTPWLR VHRDGPALSG PQSRALQEAL VLSDRAPFAA PSPFAELVLP PQQ.

Q&A

What is the TYMP gene and what protein does it encode?

The TYMP gene, located on chromosome 22q13.33, encodes thymidine phosphorylase (TP), an enzyme involved in nucleoside metabolism in the salvage pathway of pyrimidine nucleosides . TP is also known as platelet-derived endothelial cell growth factor (PD-ECGF) and plays important roles in cell proliferation and angiogenesis . As a key enzyme in thymidine catabolism, TP catalyzes the reversible phosphorolysis of thymidine to thymine and 2-deoxy-D-ribose-1-phosphate. This reaction is critical for nucleotide homeostasis and cellular function, particularly in rapidly dividing cells.

What are the primary functions of human thymidine phosphorylase?

Human thymidine phosphorylase (HsTP) serves multiple functions in human physiology:

  • Nucleoside metabolism: HsTP catalyzes the phosphorolysis of thymidine, maintaining nucleotide balance .

  • Angiogenesis promotion: As PD-ECGF, it stimulates endothelial cell growth and new blood vessel formation .

  • Cell proliferation: It promotes cell division in various tissue types .

  • Epithelial to mesenchymal transition: TYMP enhances this cellular process, which is important in development and cancer progression .

  • Chemotherapeutic drug metabolism: TP serves as a rate-limiting enzyme in the 5-fluorouracil (5-FU) activation pathway, making it relevant for predicting chemotherapy efficacy .

These diverse functions make HsTP a subject of interest across multiple research areas, from basic cell biology to clinical applications in oncology and rare disease management.

How do mutations in the TYMP gene contribute to disease?

Mutations in the TYMP gene have been clearly linked to mitochondrial neurogastrointestinal encephalomyopathy (MNGIE), a rare autosomal recessive disorder . MNGIE is characterized by severe neurological and gastrointestinal complications, including progressive external ophthalmoplegia, peripheral neuropathy, and gastrointestinal dysmotility .

The pathophysiological mechanism involves:

  • Loss of thymidine phosphorylase activity due to mutations

  • Resultant accumulation of thymidine and deoxyuridine in plasma and tissues

  • Mitochondrial DNA instability from nucleoside imbalance

  • Progressive mitochondrial dysfunction

  • Tissue-specific manifestations, particularly in metabolically active tissues

This connection between TYMP mutations and MNGIE presents opportunities for targeted therapeutic approaches, including enzyme replacement therapy, which researchers are actively developing to address the high unmet medical need associated with this condition .

What are the current approaches for optimizing recombinant expression of human thymidine phosphorylase?

Optimizing recombinant expression of HsTP presents significant challenges for researchers, as the human enzyme expresses poorly in bacterial systems. Current methodological approaches include:

  • N-terminus truncation design: Phylogenetic and structural analysis can guide rational design of N-terminus-truncation constructs with improved expression profiles . This approach accounts for conserved domains while eliminating regions that may impede bacterial expression.

  • Expression system selection: While E. coli remains the most common expression system due to its simplicity and cost-effectiveness, mammalian cell lines (HEK293, CHO) or insect cell systems (baculovirus) may yield better results for HsTP expression, despite higher costs and complexity.

  • Codon optimization: Adapting the coding sequence to match the codon usage bias of the expression host can significantly improve translation efficiency and protein yield.

  • Fusion partners: Adding solubility-enhancing tags (SUMO, MBP, GST) can improve folding and stability of the recombinant protein.

  • Culture condition optimization: Adjusting temperature, induction timing, and media composition can dramatically improve yield of correctly folded protein.

For researchers pursuing therapeutic applications, it is critical to utilize the human enzyme as a starting scaffold rather than bacterial orthologs (like EcTP), as the latter can elicit adverse immune responses when used clinically .

What are the challenges and solutions in PEGylation of human thymidine phosphorylase?

Challenges:

  • Inefficient conjugation: HsTP shows poor PEGylation efficiency compared to other therapeutic proteins

  • Activity loss: PEGylation often negatively affects HsTP's catalytic activity

  • Site-specificity: Random PEGylation can modify catalytically important residues

Methodological solutions:

  • Site-directed mutagenesis: Introducing non-catalytic lysine residues at strategic surface locations can provide alternative PEGylation sites that don't compromise activity.

  • N-terminal PEGylation: Selective modification of the N-terminus can be achieved by carefully controlling reaction pH, as the N-terminal amine has a lower pKa than lysine side chains.

  • Cysteine-specific PEGylation: Engineering free cysteine residues on the protein surface allows for maleimide-based PEGylation chemistry with high specificity.

  • Enzymatic approaches: Using transglutaminase or other enzymes to catalyze site-specific PEG attachment.

  • PEG size and branching optimization: Testing various PEG architectures (linear vs. branched) and molecular weights to balance pharmacokinetic improvement with retention of enzymatic activity.

These approaches require careful structural analysis of HsTP to identify modification sites that maintain protein folding, stability, and catalytic function while achieving the desired pharmacokinetic profile .

How can researchers effectively distinguish between TYMP expression in normal tissues versus cancer tissues?

Distinguishing TYMP expression patterns between normal and cancerous tissues requires rigorous methodological approaches:

Recommended methods:

  • Immunohistochemistry (IHC) with quantitative analysis:

    • Using validated anti-TYMP antibodies with appropriate controls

    • Employing digital pathology and algorithm-based quantification

    • Scoring systems that account for both staining intensity and percentage of positive cells

  • RNA-seq and qRT-PCR:

    • Normalized expression analysis using appropriate housekeeping genes

    • Careful microdissection of tissues to avoid contamination

    • Comparison with matched normal adjacent tissue from the same patient

  • Tissue microarray (TMA) analysis:

    • Creating representative TMAs with multiple cores per case

    • Including various control tissues

    • Blinded scoring by multiple pathologists

  • Single-cell RNA sequencing:

    • Resolving cell type-specific TYMP expression patterns

    • Identifying which cells within the tumor microenvironment express TYMP

    • Comparing with single-cell data from normal tissues

  • Multi-omics integration:

    • Correlating TYMP expression with methylation status

    • Assessing copy number variations affecting TYMP

    • Connecting expression patterns with proteomic data

TYMP overexpression has been documented in several human cancers, including lung cancer, head and neck cancer, esophageal squamous cell carcinoma, gastric carcinoma, and colorectal cancer . This differential expression may contribute to worse prognosis in some cancer types, highlighting the importance of accurate quantification methods.

How does TYMP expression influence cancer prognosis and treatment response?

TYMP expression has significant implications for both cancer prognosis and treatment response, making it an important target for translational research:

Prognostic implications:
TYMP overexpression has been significantly associated with worse prognosis in several human cancers . This correlation likely stems from TYMP's roles in:

  • Promoting angiogenesis through its PD-ECGF activity

  • Enhancing tumor cell proliferation

  • Facilitating epithelial-to-mesenchymal transition (EMT)

  • Contributing to invasive phenotypes and metastatic potential

Treatment response relationships:

TYMP plays a dual role in treatment response:

  • Chemosensitivity indicator: Since TYMP encodes thymidine phosphorylase, an important rate-limiting enzyme in the 5-fluorouracil (5-FU) activation pathway, its expression levels can predict response to certain chemotherapies . Quantification of TYMP expression has proven useful in predicting efficacy of:

    • 5-Fluorouracil (5-FU)

    • Capecitabine (a 5-FU prodrug)

    • Other pyrimidine analogs

  • Resistance mechanism: In some contexts, TYMP upregulation can contribute to treatment resistance through:

    • Enhanced DNA damage repair

    • Promotion of cancer stem cell phenotypes

    • Increased survival signaling

Methodological approaches for researchers:

To effectively study TYMP's role in treatment response, researchers should:

These approaches can help clarify the complex relationship between TYMP expression and treatment outcomes, potentially leading to more personalized treatment strategies.

What methodologies are being developed for enzyme replacement therapy using human thymidine phosphorylase?

Enzyme replacement therapy (ERT) using human thymidine phosphorylase represents an important therapeutic approach for MNGIE. Current methodological developments include:

Production strategies:

  • Protein engineering for improved expression:

    • Development of N-terminus truncation constructs with significantly improved expression in bacterial systems

    • Codon optimization for enhanced translation efficiency

    • Signal sequence modifications for improved secretion in eukaryotic systems

  • PEGylation optimization:

    • Strategic introduction of PEGylation sites through mutagenesis

    • Testing various PEG architectures and molecular weights

    • Developing methods that preserve catalytic activity

  • Alternative formulations:

    • Liposomal encapsulation

    • Nanoparticle delivery systems

    • Red blood cell encapsulation

Delivery considerations:

  • Administration routes:

    • Intravenous infusion protocols

    • Slow-release subcutaneous formulations

    • Targeted delivery approaches

  • Dosing strategies:

    • Enzyme kinetics-based dosing calculations

    • Pharmacokinetic/pharmacodynamic modeling

    • Biomarker-guided dose adjustments

  • Immunogenicity reduction:

    • Using human enzyme rather than bacterial orthologs to reduce immunogenicity

    • Tolerization protocols

    • Immunomodulatory co-therapies

Clinical trial design considerations:

  • Patient stratification based on mutation type and disease severity

  • Appropriate endpoints (biochemical normalization, clinical improvement)

  • Long-term safety monitoring

  • Quality of life assessments

These methodological developments aim to overcome the limitations of current approaches and provide effective treatment options for patients with MNGIE, addressing the significant unmet medical need in this rare disease population .

How can researchers analyze the relationship between TYMP mutations and MNGIE phenotypes?

Analyzing the relationship between TYMP mutations and MNGIE phenotypes requires sophisticated methodological approaches spanning genomics, biochemistry, and clinical assessment:

Genotype-phenotype correlation methodologies:

  • Comprehensive mutation analysis:

    • Whole gene sequencing including promoter and intronic regions

    • Copy number variation detection

    • RNA analysis to detect splicing defects

    • Methylation analysis

  • Functional characterization of variants:

    • In vitro enzyme activity assays for each variant

    • Protein stability and half-life studies

    • Subcellular localization analysis

    • Structural modeling of mutations

  • Clinical severity scoring systems:

    • Standardized neurological assessment tools

    • Gastrointestinal symptom quantification

    • Quality of life measurements

    • Timed functional tests

  • Biomarker analysis:

    • Plasma thymidine and deoxyuridine levels

    • Mitochondrial DNA copy number and deletion analysis

    • Metabolomic profiling

    • Tissue-specific mitochondrial function assays

Data integration approaches:

  • Machine learning algorithms to identify patterns between specific mutations and clinical features

  • Network analysis of biochemical pathways affected by different mutations

  • Longitudinal studies tracking disease progression with different mutation types

  • Multi-center data sharing to increase cohort size for rare variants

Practical research considerations:

  • Patient registries should collect standardized genetic and clinical data

  • Functional assays should be performed under consistent conditions

  • Analysis should account for potential genetic modifiers and environmental factors

  • Both homozygous and compound heterozygous genotypes should be characterized

By applying these methodologies, researchers can establish more precise relationships between specific TYMP mutations and clinical manifestations, potentially allowing for personalized treatment approaches and more accurate prognostication for MNGIE patients.

What novel technologies are being applied to study TYMP function and regulation?

Cutting-edge technologies are transforming our understanding of TYMP function and regulation:

  • CRISPR-Cas9 genome editing:

    • Generation of isogenic cell lines with TYMP knockouts or specific mutations

    • Creation of physiologically relevant disease models

    • Base editing to introduce specific point mutations without double-strand breaks

    • CRISPRi/CRISPRa for reversible modulation of TYMP expression

  • Single-cell technologies:

    • Single-cell RNA sequencing to identify cell type-specific TYMP expression patterns

    • Single-cell proteomics to correlate TYMP protein levels with phenotypic changes

    • Spatial transcriptomics to map TYMP expression within tissue architecture

    • Multi-omics integration at single-cell resolution

  • Advanced imaging techniques:

    • Live-cell imaging with fluorescently tagged TYMP to track subcellular localization

    • Super-resolution microscopy for nanoscale visualization of TYMP interactions

    • Intravital microscopy to study TYMP in living tissues

    • Correlative light and electron microscopy for ultrastructural context

  • Computational approaches:

    • Molecular dynamics simulations of TYMP structure and substrate interactions

    • AI-based prediction of TYMP binding partners and regulatory networks

    • Systems biology modeling of TYMP in nucleoside metabolism pathways

    • Natural language processing to extract TYMP-related insights from literature

  • Organoid and microfluidic systems:

    • Patient-derived organoids to study TYMP in 3D tissue-like structures

    • Organ-on-chip models integrating multiple cell types

    • Microfluidic platforms for high-throughput screening of TYMP modulators

    • Bioprinted tissues with controlled TYMP expression patterns

These technologies allow researchers to study TYMP at unprecedented resolution and in more physiologically relevant contexts, potentially revealing new functions and regulatory mechanisms that could inform therapeutic development.

How can researchers investigate the role of TYMP in the tumor microenvironment?

Investigating TYMP's role in the tumor microenvironment requires specialized methodological approaches:

Experimental methods for spatial and cellular analysis:

  • Multiplexed immunohistochemistry/immunofluorescence:

    • Simultaneous visualization of TYMP with immune cell markers, endothelial markers, and cancer cell markers

    • Digital quantification of spatial relationships between TYMP-expressing cells and other cell types

    • Correlation with clinical outcomes

  • Single-cell RNA sequencing of tumor ecosystems:

    • Cell type identification and quantification in TYMP-high vs. TYMP-low tumors

    • Trajectory analysis to understand cellular differentiation states

    • Receptor-ligand interaction analysis to identify TYMP-mediated intercellular communication

  • Spatial transcriptomics:

    • Mapping TYMP expression within the architectural context of tumors

    • Identification of spatial gradients and niches

    • Correlation with hypoxic regions, vascular structures, and immune infiltrates

  • Co-culture systems:

    • Cancer cells with stromal components (fibroblasts, immune cells, endothelial cells)

    • Manipulation of TYMP expression in specific cell types

    • Assessment of bidirectional signaling and phenotypic changes

  • In vivo models with cell type-specific TYMP manipulation:

    • Conditional knockout/overexpression in specific cell lineages

    • Bioluminescent/fluorescent reporting systems

    • Intravital microscopy for real-time visualization

Functional assessment approaches:

  • Angiogenesis assays:

    • Tube formation assays with conditioned media from TYMP-expressing cells

    • Chorioallantoic membrane assays

    • Matrigel plug assays with controlled TYMP expression

  • Immune function analysis:

    • T cell proliferation and activation in the presence of TYMP-expressing cells

    • Macrophage polarization assays

    • Immune checkpoint expression in response to TYMP signaling

  • Metabolic profiling:

    • Extracellular flux analysis of different cell types after TYMP exposure

    • Isotope tracing to track nucleoside metabolism across cell types

    • Metabolite profiling of tumor microenvironments with varying TYMP levels

These methodological approaches can help researchers unravel the complex role of TYMP in shaping the tumor microenvironment, potentially identifying new therapeutic targets and strategies for cancer treatment.

What are the current challenges in developing diagnostics based on TYMP expression or activity?

Developing diagnostics based on TYMP expression or activity faces several methodological challenges that researchers must address:

Technical challenges and solutions:

  • Standardization of measurement methods:

    • Challenge: Variability in antibody specificity and sensitivity across different detection platforms

    • Solution: Development of reference standards and calibration materials for TYMP quantification

    • Approach: Multi-center validation studies to establish reproducible protocols

  • Sample type and preparation considerations:

    • Challenge: TYMP expression and activity can vary significantly based on sample handling

    • Solution: Standardized collection, processing, and storage protocols

    • Approach: Stability studies to determine optimal preservation methods

  • Distinguishing isoforms and post-translational modifications:

    • Challenge: Different TYMP variants and modifications may have distinct functional implications

    • Solution: Development of isoform-specific detection methods

    • Approach: Mass spectrometry-based approaches for detailed protein characterization

  • Reference ranges and threshold determination:

    • Challenge: Establishing clinically meaningful cutoffs for normal vs. pathological TYMP levels

    • Solution: Large-scale studies in diverse populations to establish reference ranges

    • Approach: ROC curve analysis to optimize sensitivity and specificity for specific clinical applications

Clinical application challenges:

  • Context-dependent interpretation:

    • Challenge: TYMP's significance varies across different cancer types and stages

    • Solution: Development of cancer-specific interpretation algorithms

    • Approach: Machine learning models incorporating multiple biomarkers and clinical variables

  • Prognostic vs. predictive value differentiation:

    • Challenge: Distinguishing TYMP's role as a general prognostic marker from its ability to predict specific treatment responses

    • Solution: Well-designed clinical trials with appropriate control groups

    • Approach: Multivariate analysis controlling for known prognostic factors

  • Point-of-care testing development:

    • Challenge: Translating complex TYMP assays into rapid, accessible formats

    • Solution: Simplification of detection methods while maintaining accuracy

    • Approach: Lateral flow immunoassays or electrochemical biosensors for TYMP detection

  • Integration with existing diagnostic workflows:

    • Challenge: Adding TYMP testing to established clinical pathways

    • Solution: Demonstration of added clinical value beyond current standards

    • Approach: Health economic studies and implementation science research

Addressing these challenges requires interdisciplinary collaboration between basic scientists, clinical researchers, and diagnostic development specialists to translate TYMP biology into clinically useful diagnostic tools.

Product Science Overview

Structure and Mechanism

Thymidine Phosphorylase is a dimeric protein composed of two identical subunits, each consisting of 440 amino acids . The enzyme has a unique S-shaped structure with a deep cavity that serves as the binding site for thymine, thymidine, and phosphate . The catalytic mechanism involves the binding of phosphate before thymidine, followed by the release of 2-deoxyribose-1-phosphate after the nitrogenous base .

Biological Significance

TP is involved in several metabolic pathways, including purine and pyrimidine metabolism . It is particularly important in the context of mitochondrial neurogastrointestinal encephalomyopathy (MNGIE), a rare metabolic disorder caused by mutations in the TYMP gene . MNGIE is characterized by severe gastrointestinal and neurological symptoms due to the accumulation of toxic metabolites .

Recombinant Expression

The recombinant expression of human TP (HsTP) has been a focus of research due to its therapeutic potential . However, expressing HsTP in Escherichia coli (E. coli) has proven challenging due to poor expression levels and inefficient PEGylation, a chemical modification crucial for enhancing the enzyme’s stability and serum persistence . Researchers have employed various strategies to improve the recombinant expression and PEGylation efficiency of HsTP, including phylogenetic and structural analysis, as well as rational surface engineering .

Therapeutic Applications

The therapeutic potential of recombinant HsTP lies in its application for enzyme replacement therapy (ERT) in MNGIE patients . Unlike bacterial enzymes, which can elicit adverse immune responses, human recombinant enzymes are less likely to be recognized as foreign by the immune system . This makes HsTP a promising candidate for developing safer and more effective treatments for MNGIE and other related disorders .

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.