PRTFDC1 produced in E.Coli is a single, non-glycosylated polypeptide chain containing 248 amino acids (1-225 a.a.) and having a molecular mass of 28.1kDa.
PRTFDC1 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.
Phosphoribosyltransferase domain-containing protein 1, PRTFDC1, HHGP.
MGSSHHHHHH SSGLVPRGSH MGSMAGSSEE APDYGRGVVI MDDWPGYDLN LFTYPQHYYG DLEYVLIPHG IIVDRIERLA KDIMKDIGYS DIMVLCVLKG GYKFCADLVE HLKNISRNSD RFVSMKVDFI RLKSYRNDQS MGEMQIIGGD DLSTLAGKNV LIVEDVVGTG RTMKALLSNI EKYKPNMIKV ASLLVKRTSR SDGFRPDYAG FEIPNLFVVG YALDYNEYFR DLNHICVINE HGKEKYRV.
PRTFDC1 (Phosphoribosyl Transferase Domain Containing 1) is a protein-coding gene that enables protein homodimerization activity and is predicted to be involved in purine ribonucleoside salvage pathways . The gene produces a protein with low, barely detectable phosphoribosyltransferase activity in vitro. It has binding affinity for GMP, IMP, and alpha-D-5-phosphoribosyl 1-pyrophosphate (PRPP), though it is not expected to significantly contribute to purine metabolism or GMP salvage under normal physiological conditions . Gene Ontology (GO) annotations related to PRTFDC1 include protein homodimerization activity and magnesium ion binding .
The PRTFDC1 gene is located on chromosome 10, specifically at position 10:25226229-25231340 according to PCR validation data . Previous GeneCards identifiers for this genomic location include GC10M025141, GC10M025177, and GC10M024799 . This chromosomal location information is particularly important when designing genomic studies or when investigating potential regulatory elements in the region surrounding the gene.
At least three isoforms of PRTFDC1 are known to exist . While the complete functional differences between these isoforms have not been fully characterized in the provided sources, molecular studies of the protein indicate that all variants contain the phosphoribosyltransferase domain. Different isoforms may exhibit tissue-specific expression patterns or possess altered enzymatic efficiencies, but detailed comparative analyses of their specific functions would require targeted studies comparing their biochemical properties, cellular localization, and interaction partners.
PRTFDC1 is highly homologous to hypoxanthine phosphoribosyltransferase (HPRT1) and may have arisen from a gene duplication event of a common ancestor gene . This evolutionary relationship explains their structural similarities and partially overlapping functions. HPRT1 is considered an important paralog of PRTFDC1 . The divergence between these genes likely resulted in functional specialization, with HPRT1 maintaining stronger phosphoribosyltransferase activity compared to the weaker activity observed in PRTFDC1. This evolutionary context is important for understanding the potential redundancy and complementary roles these genes may play in purine metabolism.
Recent studies have shown that PRTFDC1 can act as a tumor suppressor gene, particularly in oral squamous cell carcinomas (OSCC) . The tumor suppressive function of PRTFDC1 has been demonstrated through several experimental approaches. CpG islands in the PRTFDC1 promoter can become hypermethylated in ovarian cancers and OSCC, leading to gene silencing . When PRTFDC1 expression is experimentally restored in OSCC cells, a significant inhibition of cell growth is observed in colony-formation assays . Conversely, knockdown of PRTFDC1 expression in OSCC cells that normally express the gene promotes cell growth .
These findings suggest that PRTFDC1 may regulate cell proliferation pathways, potentially through its involvement in nucleotide metabolism or through other cellular signaling mechanisms that remain to be fully characterized. The epigenetic silencing of PRTFDC1 through promoter hypermethylation represents a potential biomarker for cancer detection and prognosis, particularly in oral and ovarian cancers.
PRTFDC1 has been associated with Lesch-Nyhan Syndrome , a rare genetic disorder typically caused by mutations in the HPRT1 gene. Given that PRTFDC1 is a paralog of HPRT1 and shares similar domain structure, it may play a compensatory or modulatory role in the pathophysiology of this syndrome. The relationship likely stems from the involvement of both genes in purine metabolism pathways, though PRTFDC1's specific contribution to the disease mechanism requires further investigation.
Methodologically, researchers investigating this relationship should consider:
Conducting genetic association studies to identify potential PRTFDC1 variants in Lesch-Nyhan patients without HPRT1 mutations
Examining potential interactions between PRTFDC1 and HPRT1 proteins
Investigating whether PRTFDC1 expression changes in response to HPRT1 dysfunction
Developing animal models with both PRTFDC1 and HPRT1 modifications to assess combined effects
The regulation of PRTFDC1 expression involves epigenetic mechanisms, particularly DNA methylation. The CpG islands in the PRTFDC1 promoter can become hypermethylated in certain cancer types, leading to gene silencing . This epigenetic control suggests that PRTFDC1 expression may be dynamically regulated during development and in response to cellular stress or disease states.
To comprehensively study PRTFDC1 regulation, researchers should:
Perform chromatin immunoprecipitation (ChIP) assays to identify transcription factors binding to the PRTFDC1 promoter
Use bisulfite sequencing to map methylation patterns across the promoter region in different cell types
Investigate the effects of histone modifications on PRTFDC1 expression using ChIP-seq
Examine potential microRNA regulation using prediction algorithms followed by luciferase reporter assays
For reliable detection of PRTFDC1 expression in human samples, researchers have several validated methodological options:
RT-qPCR Analysis: PrimePCR assays for PRTFDC1 have been validated with high specificity (100%) and efficiency (98%) . The validated assay (qHsaCIP0032857) can detect multiple transcript variants (ENST00000320152, ENST00000376378, ENST00000376376, ENST00000358336) . When performing RT-qPCR, researchers should use intron-spanning primers to avoid genomic DNA amplification, as the gDNA Cq (39.53) is significantly higher than the cDNA Cq (21.95) .
ELISA: Commercially available ELISA kits can detect human PRTFDC1 protein . When using this approach, optimization of sample dilutions is crucial for accurate quantification.
Western Blotting: While not specifically mentioned in the search results, antibody-based detection of PRTFDC1 would typically employ GST-tagged recombinant proteins as positive controls.
For comprehensive expression analysis, combining nucleic acid and protein detection methods is recommended to account for potential post-transcriptional regulation.
To effectively investigate PRTFDC1's tumor suppressive function, a multi-faceted experimental approach is recommended:
Expression Modulation Studies:
Overexpression experiments using validated expression vectors containing the PRTFDC1 coding sequence in cancer cell lines with low endogenous expression
RNA interference (siRNA or shRNA) to knockdown PRTFDC1 in cells with high endogenous expression
CRISPR-Cas9 gene editing to create isogenic cell lines with PRTFDC1 knockout
Functional Assays:
Methylation Analysis:
Bisulfite sequencing of the PRTFDC1 promoter region in tumor vs. normal samples
Treatment with demethylating agents (5-aza-2′-deoxycytidine) to restore expression
In vivo Studies:
Xenograft models using cell lines with modulated PRTFDC1 expression
Patient-derived xenografts to maintain tumor heterogeneity
This comprehensive approach allows for validation of findings across multiple experimental systems and provides stronger evidence for PRTFDC1's role in tumor suppression.
When working with recombinant PRTFDC1 proteins, several important considerations should guide experimental design:
Protein Stability: Recombinant PRTFDC1 proteins should be stored at -80°C and aliquoted to avoid repeated freeze-thaw cycles which could compromise protein activity .
Buffer Compatibility: The storage buffer (typically 50 mM Tris-HCl, 10 mM reduced Glutathione, pH 8.0) may affect downstream applications and should be considered when designing binding or activity assays.
Tag Influence: Common tags such as GST can affect protein folding, activity, and interactions. Control experiments with tag-only proteins should be included to account for potential tag-specific effects.
Shelf Life: Best results are typically obtained when using the protein within three months from the date of receipt .
Quality Control: Verification of protein purity and integrity by SDS-PAGE is recommended before use in sensitive applications .
Functional Validation: Given PRTFDC1's low phosphoribosyltransferase activity, sensitive assays may be required to detect enzymatic function. Controls with the more active HPRT1 paralog can provide a reference point for activity levels.
When interpreting PRTFDC1 expression data in cancer studies, researchers should consider several contextual factors:
Tissue-Specific Expression Patterns:
Compare expression levels to matched normal tissue rather than relying on absolute values
Consider the normal expression pattern of PRTFDC1 across different tissue types as a baseline
Epigenetic Context:
Isoform-Specific Analysis:
Functional Correlation:
Statistical Considerations:
A comprehensive interpretation should integrate expression data with functional studies and clinical outcomes to establish the biological and clinical significance of PRTFDC1 alterations in specific cancer types.
Due to the evolutionary relationship and functional similarity between PRTFDC1 and HPRT1, several confounding factors must be considered:
Sequence Homology Issues:
Functional Redundancy:
HPRT1 may compensate for PRTFDC1 deficiency, masking phenotypes
Double knockdown experiments may be necessary to reveal functions
Differential Expression:
HPRT1 and PRTFDC1 may have different tissue-specific expression patterns
Changes in one gene may affect expression of the other through feedback mechanisms
Pathway Complexity:
Both genes function within complex purine metabolism pathways
Effects attributed to PRTFDC1 might be indirect through alterations in nucleotide pools
Evolutionary Conservation:
Cross-species comparisons must account for different evolutionary trajectories of these paralogs
Model organism studies should verify the presence and function of both genes
To address these confounding factors, researchers should:
Use multiple detection methods with validated specificity
Include controls for both genes in expression and functional studies
Consider compensatory mechanisms in data interpretation
Design experiments that can distinguish direct from indirect effects
PRTFDC1 promoter hypermethylation has been observed in ovarian cancers and oral squamous cell carcinomas (OSCC) , suggesting potential utility as a cancer biomarker. To develop this as a clinically relevant biomarker, researchers should consider the following methodological approach:
Biomarker Validation Strategy:
Conduct large-scale methylation analysis across diverse cancer cohorts
Compare methylation patterns in tumor vs. adjacent normal tissue
Correlate methylation status with clinical outcomes (survival, treatment response)
Determine sensitivity and specificity metrics for diagnostic applications
Sample Collection Considerations:
Evaluate methylation in readily accessible samples (liquid biopsies, saliva for OSCC)
Assess concordance between tissue methylation and detection in circulating cell-free DNA
Analytical Method Development:
Optimize methylation-specific PCR protocols for clinical laboratory implementation
Develop standardized cutoff values for positive/negative results
Ensure reproducibility across different testing platforms
Clinical Integration:
Determine the appropriate clinical context for testing (screening, diagnosis, prognosis)
Develop testing algorithms that incorporate PRTFDC1 methylation with other biomarkers
Conduct prospective clinical validation studies
Therapeutic Implications:
Investigate whether PRTFDC1 methylation status predicts response to demethylating agents
Explore targeted approaches to restore PRTFDC1 expression in hypermethylated tumors
This systematic approach would establish whether PRTFDC1 methylation can serve as a reliable and clinically actionable biomarker in specific cancer types.
Based on the understanding that PRTFDC1 functions as a tumor suppressor gene that is often silenced through promoter hypermethylation , several therapeutic strategies could be developed:
Epigenetic Therapy:
DNA methyltransferase inhibitors (DNMTi) like 5-azacytidine or decitabine to reverse hypermethylation
Combination with histone deacetylase inhibitors (HDACi) for synergistic reactivation of silenced genes
Targeted delivery systems to enhance specificity for cancer cells
Gene Therapy Approaches:
Viral vector-mediated PRTFDC1 gene delivery to restore expression
CRISPR-based epigenetic editing to specifically demethylate the PRTFDC1 promoter
mRNA therapeutics for transient expression in tumor cells
Metabolic Pathway Modulation:
Targeting purine metabolism pathways to exploit vulnerabilities created by PRTFDC1 deficiency
Synthetic lethal approaches focusing on parallel metabolic pathways
Combination Therapies:
Integrating PRTFDC1-targeted approaches with conventional chemotherapy
Sequencing strategies to prime tumors for enhanced response to immunotherapy
Biomarker-Guided Treatment:
Using PRTFDC1 methylation status to select patients for epigenetic therapy
Monitoring PRTFDC1 expression as a pharmacodynamic marker of treatment efficacy
Each approach requires preclinical validation in appropriate model systems before advancing to clinical investigation. The optimal strategy may vary depending on cancer type, molecular context, and patient-specific factors.
Despite growing recognition of PRTFDC1's importance, several significant knowledge gaps remain:
Detailed Biochemical Function:
The precise substrates and reaction kinetics of PRTFDC1 remain poorly characterized
While it has low phosphoribosyltransferase activity in vitro , its actual physiological activity and regulation need further investigation
Structural studies comparing PRTFDC1 and HPRT1 could reveal differences in substrate binding and catalysis
Cellular Signaling Roles:
Beyond its enzymatic function, potential roles in cellular signaling pathways remain unexplored
How PRTFDC1 contributes to growth suppression independent of its predicted enzymatic activity needs clarification
Protein interaction networks of PRTFDC1 have not been comprehensively mapped
Isoform-Specific Functions:
Physiological Relevance:
Regulatory Mechanisms:
Beyond promoter methylation, other regulatory mechanisms controlling PRTFDC1 expression and function are poorly understood
Post-translational modifications that might regulate PRTFDC1 activity have not been extensively characterized
Addressing these knowledge gaps will require multidisciplinary approaches combining biochemistry, structural biology, cell signaling, and in vivo models.
Single-cell analysis technologies offer powerful new approaches to address unresolved questions about PRTFDC1:
Cell Type-Specific Expression Patterns:
Single-cell RNA sequencing (scRNA-seq) can reveal cell populations with distinct PRTFDC1 expression levels
This approach could identify specific cell types where PRTFDC1 plays critical roles in normal and disease states
Spatial transcriptomics can map PRTFDC1 expression within tissue architecture
Tumor Heterogeneity Analysis:
Characterizing PRTFDC1 expression across diverse cell populations within tumors
Identifying resistant subpopulations with altered PRTFDC1 methylation or expression
Tracking clonal evolution of PRTFDC1 alterations during disease progression
Epigenetic Profiling:
Single-cell ATAC-seq can map chromatin accessibility at the PRTFDC1 locus
Single-cell bisulfite sequencing can reveal heterogeneity in PRTFDC1 promoter methylation
Integrating these data with expression analysis to establish direct regulatory relationships
Protein-Level Analysis:
Single-cell proteomics and phosphoproteomics to assess PRTFDC1 protein levels and modifications
Mass cytometry (CyTOF) with PRTFDC1-specific antibodies to track protein expression in relation to other cellular markers
Metabolic Analysis:
Single-cell metabolomics to correlate PRTFDC1 expression with purine metabolite levels
Identifying metabolic signatures associated with PRTFDC1 function or deficiency
These technologies would enable researchers to move beyond bulk tissue analysis to understand the context-specific functions of PRTFDC1 at unprecedented resolution, potentially revealing new therapeutic opportunities and biomarker applications.
PRTFDC1 is a protein-coding gene that has been associated with several biological processes and molecular functions. The protein encoded by this gene has low, barely detectable phosphoribosyltransferase activity in vitro. It binds guanosine monophosphate (GMP), inosine monophosphate (IMP), and alpha-D-5-phosphoribosyl 1-pyrophosphate (PRPP) . Despite its structural similarity to hypoxanthine-guanine phosphoribosyltransferase (HPRT), PRTFDC1 is not expected to contribute significantly to purine metabolism or GMP salvage .
The structure of PRTFDC1 has been determined at a resolution of 1.7 Å with bound GMP . The overall structure and GMP binding mode are very similar to that observed for HPRT. However, the catalytic efficiency of PRTFDC1 towards hypoxanthine and guanine is significantly lower than that of HPRT . This low activity is attributed to the presence of a glycine residue in the position of the proposed catalytic aspartate of HPRT .
PRTFDC1 is predicted to be involved in the purine ribonucleoside salvage pathway, although its exact biological function remains unclear . It has been suggested that PRTFDC1 may play a role in certain physiological conditions and diseases. For instance, it has been associated with auditory neuropathy, autosomal dominant 3, and physiological polycythemia . Additionally, recent studies have indicated a potential involvement of PRTFDC1 in the development of ovarian cancer .