HPRT1 Human

Hypoxanthine-Guanine Phosphoribosyltransferase Human Recombinant
Shipped with Ice Packs
In Stock

Description

Enzymatic Activity

HPRT1 catalyzes the conversion of hypoxanthine to inosine monophosphate (IMP) and guanine to guanosine monophosphate (GMP) via transfer of a 5-phosphoribosyl group from 5-phosphoribosyl 1-pyrophosphate (PRPP) . This reaction is central to purine nucleotide synthesis, enabling efficient DNA/RNA production.

Protein Architecture

HPRT1 forms a tetramer composed of four identical subunits (218 amino acids each). Key functional domains include:

  • Active site: β-α-β motif for substrate binding and catalysis.

  • Subunit interfaces: Critical for tetramer stability and enzyme activity .

Lesch-Nyhan Syndrome (LNS)

Caused by severe HPRT1 deficiency (enzyme activity <1.5%), LNS manifests as:

  • Hyperuricemia: Uric acid accumulation leading to gouty arthritis and kidney stones.

  • Neurological dysfunction: Self-mutilation, intellectual disability, and involuntary movements .

  • Over 200 mutations identified, including missense, nonsense, and frameshift variants .

HPRT-Related Gout (Kelley-Seegmiller Syndrome)

Partial enzyme deficiency (1.5–20% activity) results in:

  • Uric acid overproduction: Kidney stones and gout.

  • No neurological symptoms .

DiseaseEnzyme ActivityKey MutationsClinical Features
Lesch-Nyhan Syndrome<1.5%Nonsense (e.g., c.151C>T), frameshiftHyperuricemia, neurobehavioral deficits
HPRT-Related Gout1.5–20%Missense, splice variantsGout, kidney stones

Metabolic Regulation

Metformin upregulates HPRT1 to enhance purine anabolism and reduce oxidative stress (ROS) in osteoblasts, potentially mitigating osteoporosis . This suggests a role for HPRT1 in age-related metabolic disorders.

Oncological Significance

Overexpression of HPRT1 is linked to poor prognosis in head and neck squamous cell carcinoma (HNSCC):

Genetic Testing

  • LOVD Database: 41 public variants documented, including pathogenic and benign mutations .

  • qRT-PCR: Quantifies HPRT1 mRNA levels for diagnostic confirmation .

Biotechnological Relevance

HPRT1-deficient myeloma cells are used in hybridoma production via HAT medium selection, enabling monoclonal antibody development .

Future Directions

  • Gene Therapy: Targeting HPRT1 mutations to restore enzyme function in LNS.

  • Oncology: Investigating HPRT1 inhibitors as therapeutic agents in cancers with elevated expression .

Product Specs

Introduction
The enzyme HPRT1 plays a crucial role in the synthesis of purine nucleotides via the purine salvage pathway. Its primary function is to recycle purines from broken-down DNA for new purine synthesis. This is accomplished through its catalytic activity in the reaction between guanine and phosphoribosyl pyrophosphate, resulting in the formation of GMP.
Description
Recombinant Human HPRT1, produced in E. coli, is a single, non-glycosylated polypeptide chain. It comprises 238 amino acids (with positions 1 through 218 being the active amino acids) and has a molecular weight of 26.7 kDa. A 20 amino acid His-Tag is fused to the N-terminus of the HPRT1. The protein is purified using proprietary chromatographic techniques.
Physical Appearance
A clear and colorless solution that has been sterilized by filtration.
Formulation
This solution of Human HPRT1 is supplied in a buffer of 20mM Tris HCl at pH 8.0, with 20% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined by SDS-PAGE analysis and is consistently greater than 95%.
Synonyms
Hypoxanthine-Guanine Phosphoribosyltransferase , EC 2.4.2.8, HGPRT, HGPRTase, HPRT, HPRT1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MATRSPGVVI SDDEPGYDLD LFCIPNHYAE DLERVFIPHG LIMDRTERLA RDVMKEMGGH HIVALCVLKG GYKFFADLLD YIKALNRNSD RSIPMTVDFI RLKSYCNDQS TGDIKVIGGD DLSTLTGKNV LIVEDIIDTG KTMQTLLSLV RQYNPKMVKV ASLLVKRTPR SVGYKPDFVG FEIPDKFVVG YALDYNEYFR DLNHVCVISE TGKAKYKA.

Q&A

What is the primary function of HPRT1 in human cellular metabolism?

HPRT1 (hypoxanthine phosphoribosyltransferase 1) is an enzyme that plays a crucial role in the purine salvage pathway, allowing cells to recycle purines - essential building blocks of DNA and RNA. This recycling process is significantly more energy-efficient and time-effective than de novo purine synthesis, making it a metabolically advantageous pathway. Through this mechanism, HPRT1 ensures cells maintain sufficient supplies of nucleotide precursors for DNA and RNA production without the energetic costs associated with new synthesis . The enzyme specifically catalyzes the conversion of hypoxanthine to inosine monophosphate and guanine to guanosine monophosphate, key intermediates in nucleotide metabolism.

How is the HPRT1 gene organized genomically?

The HPRT1 gene is located on the X chromosome and is inherited in an X-linked recessive manner. It contains only one functional messenger RNA transcript that encodes the hypoxanthine-guanine phosphoribosyltransferase enzyme . As one of the most extensively studied genes in human genetics, its structure has been well-characterized. For genomic DNA amplification, researchers have successfully used specific primers: Forward GTGAAAAGGACCCCACGAAG and reverse CAAATTATGAGGTGCTGGAAGGA. For cDNA amplification, standard primers include: Forward CAAAGATGGTCAAGGTCGCA and reverse ACAGTTTAGGAATGCAGCAACT . Understanding this genomic organization is essential for designing primers for mutation analysis and expression studies.

What methodologies are most reliable for quantifying HPRT1 expression?

Multiple complementary approaches are recommended for accurate HPRT1 quantification:

  • Quantitative RT-PCR:

    • SYBR green qPCR assay performed on platforms such as the ABI PRISM 7700 cycler

    • PCR-efficiency-corrected -ΔΔCt method for analysis

    • Normalization to established housekeeping genes like GAPDH (FW: 5-TGCACCACCAACTGCTTAG C-3; REV: 5-GGCATGGACTGTGGTCATGAG-3)

  • Protein Detection Methods:

    • Immunohistochemistry for tissue specimen analysis, particularly valuable when comparing expression between tumor and adjacent normal tissues

    • Western blotting for quantitative protein assessment

  • Transcriptomic Analysis:

    • RNA-sequencing to assess HPRT1 expression within the broader transcriptional landscape

    • Comparative analysis across tissue types or disease states

For maximum reliability, researchers should employ multiple methodologies with appropriate technical and biological replicates to account for measurement variability and individual variation.

How do different HPRT1 mutations correlate with Lesch-Nyhan syndrome severity?

More than 200 distinct mutations in the HPRT1 gene have been associated with Lesch-Nyhan syndrome (LNS) . These mutations produce a spectrum of phenotypic manifestations that correlate with residual enzyme activity levels:

Phenotype CategoryClinical FeaturesTypical Residual Enzyme ActivityCommon Mutation Types
Classic Lesch-Nyhan DiseaseComplete triad: hyperuricemia, severe neurological dysfunction, self-injurious behavior<1% of normal activityNonsense mutations, large deletions, frameshift mutations
HGprt-related Neurological Dysfunction (HND)Hyperuricemia with variable neurological manifestations but no self-injury1-5% of normal activityMissense mutations, splicing defects
HGprt-related Hyperuricemia (HRH)Isolated hyperuricemia without neurological manifestations>5% of normal activityMissense mutations affecting catalytic efficiency

What is the significance of HPRT1 expression alterations in cancer progression?

HPRT1, once considered solely a housekeeping gene, has emerged as a significant factor in cancer biology based on consistent findings across multiple cancer types:

These findings collectively suggest HPRT1 may represent both a valuable prognostic biomarker and potential therapeutic target across multiple cancer types.

How does HPRT1 interact with the tumor immune microenvironment?

Emerging evidence suggests HPRT1 plays previously unrecognized roles in modulating tumor-immune interactions:

  • Immunomodulatory Associations:

    • Correlation analysis demonstrates relationships between HPRT1 expression and various immunomodulatory genes, including immunoinhibitors, immunostimulators, and major histocompatibility complex (MHC) molecules

    • Comprehensive analysis using the integrated repository portal for tumor-immune system interactions database (TISIDB) reveals complex relationships with immune regulatory networks

  • Immune Cell Recruitment:

    • HPRT1 has been shown to promote lung cancer formation specifically by inducing neutrophil recruitment, suggesting direct influence on tumor-associated immune cell populations

    • This neutrophil recruitment mechanism may contribute to tumor progression through immunosuppressive activities

  • Potential Immunotherapeutic Relevance:

    • HPRT1 has been validated as a target antigen for selective cell-mediated killing in prostate cancer contexts

    • This suggests potential applications in adoptive cell therapy or vaccine development

Although our understanding of HPRT1's immunomodulatory functions remains incomplete, these findings indicate important roles in shaping the tumor immune microenvironment that warrant further investigation, particularly for developing novel immunotherapeutic strategies.

What techniques are optimal for analyzing HPRT1 mutations and their functional consequences?

A comprehensive approach to HPRT1 mutation analysis should integrate multiple complementary methodologies:

  • Genomic Analysis:

    • Whole-exome sequencing (WES) provides comprehensive mutation detection across the entire gene

    • Targeted PCR amplification followed by Sanger sequencing for confirming specific variants

    • Next-generation sequencing approaches for detecting low-frequency mosaic mutations

  • RNA Analysis:

    • RT-PCR with specific primers to detect abnormal splicing or expression changes

    • Quantitative RT-PCR for measuring relative expression levels of normal versus mutant transcripts

    • RNA-seq for comprehensive transcriptome analysis, including potential splicing variants

  • Functional Assessment:

    • Enzyme activity assays measuring conversion of hypoxanthine to IMP

    • Metabolomic analysis of purine pathway intermediates

    • Cell-based assays measuring sensitivity to 6-thioguanine (cells lacking HPRT1 activity are resistant)

  • Protein Structure Analysis:

    • In silico modeling of mutation effects on protein structure

    • Biochemical kinetics studies to determine effects on enzyme activity parameters

    • X-ray crystallography or cryo-EM for direct structural visualization

For female patients with heterozygous mutations, X-chromosome inactivation analysis is critical, as demonstrated by the methylation-sensitive restriction enzyme approach using HhaI and analysis of the androgen receptor (AR) locus . This approach can determine whether the mutant or wild-type allele is preferentially expressed.

How should X-chromosome inactivation be addressed in female HPRT1 research subjects?

X-chromosome inactivation (XCI) patterns significantly complicate the analysis of X-linked genes like HPRT1 in female subjects. A methodical approach includes:

  • XCI Pattern Analysis:

    • Utilize methylation-sensitive restriction enzymes like HhaI to distinguish active from inactive X chromosomes

    • Analyze the methylation status of polymorphic loci such as the CAG repeat in the first exon of the human androgen receptor (AR) gene

    • Compare patterns across different tissue types when possible, as XCI can be tissue-specific

  • Experimental Protocol:

    • Extract genomic DNA from relevant tissues (blood, fibroblasts, etc.)

    • Amplify without HhaI digestion to establish baseline polymorphic alleles

    • Digest separate aliquot with HhaI before amplification

    • Compare pre- and post-digestion patterns to determine XCI ratio

  • Interpretation Framework:

    • Random XCI: Both alleles detectable after digestion in approximately equal amounts

    • Non-random XCI: Predominance of one allele after digestion

    • Complete skewing: Only one allele detectable after digestion, as observed in a case study where only the AR1 maternal allele was amplifiable after HhaI digestion

  • Implications for Analysis:

    • In cases of skewed XCI, expression analysis may not reflect genotype

    • Functional consequences of heterozygous mutations may mimic homozygous patterns

    • Tissue-specific XCI patterns may lead to tissue-specific phenotypes

This methodological approach is essential for accurate interpretation of HPRT1 mutations in female patients and research subjects, particularly in cases where clinical manifestations seem inconsistent with heterozygous carrier status.

What bioinformatic approaches are most valuable for HPRT1 cancer research?

Multi-layered bioinformatic strategies have proven effective for understanding HPRT1's role in cancer:

These computational approaches have revealed that HPRT1 and its associated genes are enriched in cancer-related pathways including DNA replication and cell cycle regulation, providing important insights for translational research.

How might HPRT1 serve as a therapeutic target in oncology?

Multiple lines of evidence suggest HPRT1 as a promising therapeutic target:

  • Differential Expression Profile:

    • Significant overexpression in multiple cancer types compared to normal tissues provides a potential therapeutic window

    • Expression correlates with aggressive clinicopathological features, suggesting targeting could affect more advanced disease

  • Drug Response Associations:

    • Patients with HPRT1 overexpression demonstrate predictable sensitivity patterns:

      • Increased resistance to abiraterone

      • Enhanced sensitivity to tozasertib and teniposide

      • Responsiveness to DNA topoisomerase I (Topo I) and mitogen-activated extracellular signal-regulated kinase (MEK) inhibitors

  • Immunotherapeutic Potential:

    • Validation as a target antigen for selective cell-mediated killing in prostate cancer models

    • Correlations with immunomodulatory pathways suggest potential combination strategies with immune checkpoint inhibitors

  • Pathway Integration:

    • Association with DNA damage repair mechanisms in lung cancer suggests synthetic lethality approaches may be effective

    • Cell cycle regulatory roles provide opportunities for combination with existing cell cycle inhibitors

  • Targeted Therapy Development Considerations:

    • Direct enzyme inhibition could disrupt purine salvage pathway

    • Antisense oligonucleotides or siRNA approaches to reduce expression

    • Antibody-drug conjugates targeting surface-expressed HPRT1

While early in development, these multiple mechanisms suggest HPRT1-targeted therapies could address an important unmet need across several challenging cancer types.

What methodological considerations are critical for studying HPRT1 enzyme activity?

Accurate assessment of HPRT1 enzyme activity requires careful methodological considerations:

  • Sample Preparation:

    • Standardized protocols for tissue homogenization or cell lysis to preserve enzyme activity

    • Subcellular fractionation to isolate cytosolic components where HPRT1 primarily functions

    • Immediate processing or appropriate stabilization to prevent activity loss

  • Activity Assay Selection:

    • Radiochemical assays measuring conversion of [14C]-labeled hypoxanthine to IMP

    • Spectrophotometric methods following NADH oxidation in coupled reactions

    • HPLC-based assays quantifying reaction products

    • Selection based on required sensitivity and available equipment

  • Normalization Approaches:

    • Total protein content (Bradford or BCA assay)

    • Cell number for cultured cell studies

    • Activity of reference enzymes unrelated to purine metabolism

  • Controls and Validation:

    • Positive controls from samples with known HPRT1 activity

    • Negative controls using HPRT1-deficient samples or specific inhibitors

    • Linearity verification across sample dilutions

    • Cross-validation using multiple methodological approaches

  • Interpretation Framework:

    • Correlation of enzyme activity with protein and mRNA expression levels

    • Relation to clinical or phenotypic parameters

    • Comparison to established reference ranges for the specific methodology

Since residual enzyme activity is the primary determinant of phenotypic severity in HPRT1-related disorders , standardized and accurate activity measurements are essential for both research applications and clinical correlations.

What experimental systems are most appropriate for modeling HPRT1 functions?

Several experimental models have proven valuable for investigating different aspects of HPRT1 biology:

  • Patient-Derived Primary Cells:

    • Skin fibroblasts provide accessible material for studying enzyme activity and expression

    • Lymphoblastoid cell lines enable longer-term studies and larger-scale experiments

    • These models maintain patient-specific genetic backgrounds and modification patterns

  • Cancer Cell Line Models:

    • Panels of cancer cell lines with varying HPRT1 expression levels

    • CRISPR-engineered isogenic lines with HPRT1 knockout or specific mutations

    • Useful for studying oncogenic functions and therapeutic targeting

  • Neuronal Models:

    • Induced pluripotent stem cells (iPSCs) differentiated to neuronal lineages

    • Primary neuronal cultures

    • Critical for understanding neurological manifestations of Lesch-Nyhan syndrome

  • In Vivo Systems:

    • Mouse models with targeted Hprt1 modifications

    • Xenograft models for cancer applications

    • Allow whole-organism studies of metabolic and physiological consequences

  • Selection Systems:

    • HPRT1 deficiency confers resistance to 6-thioguanine and sensitivity to HAT medium

    • This property enables powerful selection strategies in gene editing applications

    • Particularly valuable for creating isogenic cell models

The selection of appropriate experimental systems should be guided by the specific research question, with consideration for tissue relevance, accessibility, ease of manipulation, and physiological context. Multiple complementary models often provide the most comprehensive insights.

How does HPRT1 interact with DNA damage response pathways?

Emerging evidence indicates HPRT1's previously unrecognized roles in DNA damage response:

  • Cancer-Specific Mechanisms:

    • HPRT1 promotes lung cancer formation partly through modulation of DNA damage repair mechanisms

    • This suggests context-dependent functions beyond canonical purine salvage

  • Pathway Interconnections:

    • Co-expression analysis reveals enrichment of HPRT1-associated genes in DNA replication and repair pathways

    • This network position suggests potential roles in genomic stability maintenance

  • p53 Regulatory Relationships:

    • p53 has been shown to significantly affect HPRT1 expression in cancer cells

    • This connection to a master regulator of DNA damage response suggests integration into broader damage sensing networks

  • Metabolic-DNA Damage Intersections:

    • Purine metabolism disruption may influence nucleotide pool balance

    • Imbalanced nucleotide pools are known to increase DNA replication errors and damage

  • Experimental Approaches:

    • Comet assays to measure DNA strand breaks in HPRT1-modified systems

    • γH2AX foci quantification to assess double-strand break formation and resolution

    • Analysis of HPRT1 interactions with key DNA repair proteins through co-immunoprecipitation or proximity ligation assays

This emerging research area suggests HPRT1 may function at the critical interface between metabolism and genome maintenance, with important implications for both cancer biology and genetic disorders.

What are the non-canonical functions of HPRT1 in cellular physiology?

Research has revealed several unexpected functions of HPRT1 beyond its classical role in purine salvage:

  • Cell Cycle Regulation:

    • HPRT1 plays a crucial role in modulating the cell cycle

    • Enrichment analysis shows HPRT1 and its associated genes involved in cell cycle control pathways

    • This regulatory function may explain its influence on cancer progression

  • Immune System Modulation:

    • HPRT1 can induce neutrophil recruitment in lung cancer contexts

    • Correlation with multiple immunomodulatory pathways suggests broader immune regulatory functions

    • Potential roles in shaping tumor immune microenvironment

  • Surface Expression:

    • HPRT1 has been detected on the surface of certain cancer cells

    • This unexpected localization enables its function as a target antigen for cell-mediated killing

    • Suggests possible receptor or signaling functions

  • Developmental Functions:

    • The severe neurological manifestations of Lesch-Nyhan syndrome suggest critical roles in neural development

    • These effects cannot be fully explained by purine metabolism disruption alone

  • Metabolic Integration:

    • Beyond direct purine recycling, HPRT1 may serve as a metabolic sensor integrating purine metabolism with other cellular processes

    • This integrative function could explain its diverse effects when dysregulated

These non-canonical functions represent important areas for future research, particularly for understanding the complex phenotypes associated with HPRT1 mutations and its roles in cancer progression.

Product Science Overview

Function and Mechanism

HGPRT catalyzes the conversion of hypoxanthine to inosine monophosphate (IMP) and guanine to guanosine monophosphate (GMP) . This reaction involves the transfer of a 5-phosphoribosyl group from 5-phosphoribosyl 1-pyrophosphate (PRPP) to the purine base . The enzyme’s activity is vital for maintaining the balance of purine nucleotides in the cell, which are the building blocks of DNA and RNA .

Role in Disease

Mutations in the HPRT1 gene can lead to severe metabolic disorders. One such disorder is Lesch-Nyhan syndrome, a rare genetic condition characterized by neurological and behavioral abnormalities, as well as the overproduction of uric acid . This syndrome results from a deficiency of HGPRT, leading to the accumulation of hypoxanthine and guanine, which are then converted to uric acid . Another condition associated with HGPRT deficiency is Kelley-Seegmiller syndrome, which also involves hyperuricemia and gout .

Recombinant HGPRT

Recombinant HGPRT refers to the enzyme produced through recombinant DNA technology, which involves inserting the HPRT1 gene into a suitable expression system to produce the enzyme in large quantities. This recombinant enzyme is used in various research applications, including studies on purine metabolism, drug development, and understanding the molecular basis of diseases associated with HGPRT deficiency .

Research and Applications

Research on HGPRT has provided significant insights into the purine salvage pathway and its role in cellular metabolism. Studies have shown that HGPRT is essential for the survival of cells that rely heavily on purine salvage, such as certain types of cancer cells . Additionally, HGPRT inhibitors are being explored as potential therapeutic agents for treating diseases like cancer and parasitic infections .

In conclusion, Hypoxanthine-Guanine Phosphoribosyltransferase is a critical enzyme in purine metabolism, and its recombinant form has become an invaluable tool in biomedical research. Understanding its function and the consequences of its deficiency has paved the way for advancements in the diagnosis and treatment of 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.