The HPRT1 antibody is a specialized immunological reagent designed to detect hypoxanthine phosphoribosyltransferase 1 (HPRT1), a critical enzyme in the purine salvage pathway. This pathway salvages purine bases for nucleotide synthesis, essential for DNA repair and cellular proliferation . HPRT1 antibodies are widely used in research to study HPRT1's expression, localization, and role in diseases such as cancer, Lesch-Nyhan syndrome, and gout .
HPRT1 antibodies are available in monoclonal and polyclonal forms, with varying host species, clonal specificity, and applications:
HPRT1 antibodies are utilized across diverse experimental workflows:
Prognostic Marker: High HPRT1 expression correlates with poor survival in lung adenocarcinoma (LUAD), head-neck squamous cell carcinoma (HNSC), and oral squamous cell carcinoma (OSCC) .
Chemoresistance: HPRT1 promotes cisplatin resistance in OSCC by activating the PI3K/AKT pathway and upregulating MMP1 .
Immunotherapy Link: HPRT1 expression positively correlates with PD-1 levels in HNSC, suggesting a role in immune checkpoint regulation .
Cell Proliferation: Knockdown of HPRT1 reduces migration, invasion, and proliferation in HNSC and nasopharyngeal carcinoma (NPC) cells .
Drug Targets: HPRT1 interacts with Topo I and MEK inhibitors, highlighting its potential as a therapeutic target .
Specificity: Antibodies are validated using siRNA knockdown, knockout cell lines, and recombinant protein controls .
Cross-Reactivity: Confirmed in human, mouse, and rat samples .
Citations: Over 40 peer-reviewed publications cite HPRT1 antibodies for cancer research, metabolic studies, and neurological disorders .
HPRT1 antibodies enable researchers to:
HPRT1 (also known as hypoxanthine-guanine phosphoribosyltransferase or HGPRTase) is a transferase enzyme that catalyzes the conversion of hypoxanthine to inosine monophosphate and guanine to guanosine monophosphate via transfer of the 5-phosphoribosyl group from 5-phosphoribosyl 1-pyrophosphate. This enzyme plays a central role in the generation of purine nucleotides through the purine salvage pathway . Beyond its metabolic functions, HPRT1 has emerged as an important research target due to its involvement in several pathological conditions. Mutations in HPRT1 result in Lesch-Nyhan syndrome (LNS) or gout . Additionally, recent research indicates HPRT1 is overexpressed in various cancers including nasopharyngeal carcinoma, breast cancer, endometrial cancer, lung cancer, and prostate cancer, where it's associated with poor prognosis . This dual role in metabolic disorders and cancer makes HPRT1 a compelling research target.
Researchers have access to both monoclonal and polyclonal HPRT1 antibodies from various sources, with different host species and target epitopes:
The choice between these options depends on the specific experimental requirements and applications. Monoclonal antibodies offer high specificity to a single epitope, while polyclonals may provide higher sensitivity by binding multiple epitopes .
HPRT1 antibodies have been validated for multiple research applications, as documented across various suppliers and publications:
The versatility of these applications allows researchers to study HPRT1 expression, localization, interactions, and modifications in various experimental contexts .
When selecting an HPRT1 antibody, consider these methodological factors:
Experimental application: Different antibodies perform optimally in specific applications. For example, some HPRT1 antibodies are specifically validated for Western blot but may not work well for IHC .
Species reactivity: Confirm the antibody recognizes HPRT1 from your species of interest. Many HPRT1 antibodies react with human, mouse, and rat samples, but cross-reactivity should be verified .
Antibody type:
Validation data: Review the antibody's validation data, including Western blot images showing the expected 24-28 kDa band and positive controls in relevant tissues .
Epitope location: Consider whether your research requires targeting a specific region of HPRT1. Some antibodies target full-length protein (AA 1-218), while others target specific domains .
KO/KD validation: For critical research, select antibodies validated in knockout/knockdown systems to ensure specificity .
The optimal choice depends on your specific experimental goals, target species, and detection system.
Understanding HPRT1's molecular characteristics is essential for effective antibody selection:
Molecular weight: HPRT1 has a calculated molecular weight of approximately 25 kDa, with observed molecular weight in Western blot typically between 24-28 kDa . This information is crucial for verifying antibody specificity.
Cellular localization: HPRT1 is predominantly cytoplasmic , so antibodies for immunofluorescence should effectively detect cytoplasmic signals.
Sequence conservation: HPRT1 is highly conserved across species, explaining why many antibodies show cross-reactivity between human, mouse, and rat samples .
Important domains: The protein contains catalytic domains involved in substrate binding and enzyme activity. Antibodies targeting different regions may have different effects on protein function in experimental settings .
Post-translational modifications: Consider whether your research questions involve detecting modified forms of HPRT1, as some antibodies may have differential recognition of phosphorylated or other modified forms .
Expression levels: HPRT1 expression varies across tissues and is notably elevated in certain cancer types, potentially requiring antibodies with different sensitivity ranges depending on your experimental system .
These molecular characteristics should guide antibody selection to ensure optimal detection and specificity for your particular research focus.
For optimal Western blot results with HPRT1 antibodies, follow these methodological guidelines:
Sample preparation:
Electrophoresis and transfer:
Use 10-12% SDS-PAGE gels for optimal resolution around 24-28 kDa
Transfer proteins to PVDF or nitrocellulose membranes using standard methods
Antibody dilutions and incubation:
Detection and analysis:
Studies have shown that HPRT1 protein levels are significantly elevated in cancer cells compared to normal controls, with approximately 47% of cancer patients showing upregulation . This should be considered when interpreting Western blot results from clinical samples.
For successful immunohistochemical detection of HPRT1 in tissue sections:
Tissue preparation:
Antigen retrieval (critical step):
Blocking and antibody incubation:
Detection systems:
Scoring and interpretation:
Research has demonstrated that HPRT1 staining patterns differ between normal and cancer tissues, with increased intensity and cytoplasmic localization in tumors, particularly in nasopharyngeal carcinoma and other cancers .
For high-quality immunofluorescence detection of HPRT1:
Cell preparation:
Fixation and permeabilization:
Fix cells with 4% paraformaldehyde for 15 minutes at room temperature
Permeabilize with 0.1-0.5% Triton X-100 for 5-10 minutes
Alternative: Fix and permeabilize simultaneously with methanol at -20°C for 10 minutes
Blocking and antibody incubation:
Secondary antibody and nuclear counterstaining:
Use fluorophore-conjugated secondary antibodies appropriate for your microscopy setup
Counterstain nuclei with DAPI or Hoechst
Mount with anti-fade mounting medium
Imaging and analysis:
Research has shown that HPRT1 expression patterns can vary between normal and cancer cells, with potential diagnostic and prognostic implications . For quantitative analysis, standardized image acquisition and analysis parameters should be established.
To investigate HPRT1 protein interactions, consider these methodological approaches:
Co-immunoprecipitation (Co-IP):
Use 0.5-4.0 μg of HPRT1 antibody per 1-3 mg of total protein lysate
Optimize lysis buffers to preserve protein interactions (typically RIPA or NP-40 based)
Validate interactions by reverse Co-IP using antibodies against suspected interacting partners
Western blot analysis of immunoprecipitates to confirm interactions
Proximity Ligation Assay (PLA):
Use dilutions of 1:400-1:800 of HPRT1 antibody
Combine with antibodies against suspected interaction partners
Visualize interactions as fluorescent dots representing proteins within 40 nm proximity
Quantify signal to assess interaction strength
Immunofluorescence co-localization:
Pull-down assays with recombinant proteins:
Use recombinant HPRT1 as bait
Confirm interactions using HPRT1 antibodies in Western blot
Identify novel interacting partners by mass spectrometry
Recent research suggests potential interactions between HPRT1 and proteins involved in cancer-related pathways, including CyclinD1, CyclinE, MDR1, MMP-2, and MMP-9 . These interactions may contribute to HPRT1's role in cancer progression and chemoresistance, making them important targets for investigation.
To maintain optimal activity and specificity of HPRT1 antibodies, follow these storage and handling guidelines:
Storage temperature:
Buffer composition:
Working dilutions and temporary storage:
Prepare working dilutions immediately before use
Working dilutions can typically be stored at 4°C for up to one week
For longer storage of working dilutions, add stabilizing proteins (e.g., 0.1-1% BSA)
Antibody stability:
Shipping and temporary storage:
HPRT1 antibodies are typically shipped with ice packs
Upon receipt, transfer immediately to -20°C for long-term storage
Brief storage at 4°C is acceptable for antibodies in glycerol buffer but should be minimized
Proper storage and handling ensure consistent antibody performance across experiments, which is particularly important for quantitative studies examining HPRT1 expression in cancer research and other applications .
Multiple bands in HPRT1 Western blots may occur for several reasons, requiring different troubleshooting approaches:
Post-translational modifications:
HPRT1 may undergo phosphorylation or other modifications resulting in shifted bands
Compare with literature to identify if additional bands correspond to known modified forms
Use phosphatase treatment of samples to determine if additional bands are phosphorylated forms
Proteolytic degradation:
Add protease inhibitors to lysis buffer (complete protease inhibitor cocktail)
Keep samples cold during preparation
Reduce sample handling time before adding SDS sample buffer
Look for lower molecular weight bands that may represent degradation products
Non-specific binding:
Splice variants:
Compare band patterns with known splice variant molecular weights
Consult literature or databases for HPRT1 splice variants
Validate with RT-PCR or other methods if splice variants are suspected
Cross-reactivity:
Test the antibody on knockout or knockdown samples if available
Compare results with a different HPRT1 antibody targeting a different epitope
Perform peptide competition assays to confirm specificity
The expected molecular weight for HPRT1 is 24-28 kDa . Any additional bands should be carefully evaluated against these potential causes.
To enhance signal-to-noise ratio in HPRT1 immunohistochemistry:
Optimize antigen retrieval:
Titrate primary antibody:
Optimize blocking conditions:
Increase blocking duration (1-2 hours)
Test different blocking agents (normal serum, BSA, commercial blockers)
Consider dual blocking with serum followed by protein block
Reduce non-specific binding:
Add 0.1-0.3% Triton X-100 to antibody diluent
Increase washing steps (5-6 washes of 5 minutes each)
Use TBS-T instead of PBS-T if high background persists
Detection system considerations:
Use polymer-based detection systems for improved signal-to-noise ratio
Optimize DAB development time (monitor under microscope)
Consider amplification systems for weak signals
Tissue-specific considerations:
For tissues with high endogenous peroxidase, increase H₂O₂ quenching time
For high-biotin tissues, use biotin blocking kits before antibody incubation
For tissues with high background, consider adding 5% non-fat milk to antibody diluent
In research studying HPRT1 in cancer tissues, researchers have successfully used these optimization steps to achieve clear distinction between HPRT1-positive and negative samples, enabling accurate scoring and classification .
Understanding potential sources of false results is critical for accurate HPRT1 detection:
Common causes of false positives:
Cross-reactivity with similar proteins:
Endogenous enzyme activity:
In IHC: Ensure complete quenching of endogenous peroxidase (3% H₂O₂, 10-15 minutes)
In IF: Use appropriate controls to distinguish autofluorescence from specific signal
Non-specific binding of secondary antibodies:
Common causes of false negatives:
Inadequate antigen retrieval:
Antibody degradation:
Suboptimal protein extraction:
For Western blot: Use appropriate lysis buffers (RIPA or NP-40 based)
Include protease inhibitors to prevent degradation
Optimize protein extraction protocol for your specific sample type
Epitope masking or alteration:
Consider issues with fixation duration in tissue samples
Test multiple antibodies targeting different epitopes
For modified forms, use specific antibodies designed to detect phosphorylated or other modified forms
Rigorous validation of HPRT1 antibody specificity is essential for robust research findings:
Genetic validation approaches:
Biochemical validation methods:
Positive and negative controls:
Cross-platform validation:
Confirm findings across multiple techniques (e.g., WB, IHC, IF)
Compare protein detection with mRNA expression data
Correlate with functional assays where possible
Literature comparison:
Compare your results with published data on HPRT1 expression patterns
Reference the molecular weight and localization patterns reported in literature
Note any discrepancies and investigate potential causes
Research has shown that HPRT1 knockdown leads to decreased expression of specific proteins including CyclinD1, CyclinE, MDR1, MMP-2, and MMP-9 . Validation studies should include assessment of these known downstream effects to confirm antibody specificity and biological relevance.
A comprehensive set of controls ensures reliable interpretation of HPRT1 antibody experiments:
Positive controls:
Cell lines with confirmed HPRT1 expression:
Tissue samples with known HPRT1 expression:
Recombinant protein controls:
Purified recombinant HPRT1 protein
Cells transfected with HPRT1 expression constructs
Negative controls:
Technical negative controls:
Secondary antibody only (omit primary antibody)
Isotype control antibody (matched isotype, irrelevant specificity)
Pre-immune serum (for polyclonal antibodies)
Biological negative controls:
Experimental validation controls:
Loading controls for Western blot:
Housekeeping proteins (β-actin, GAPDH, tubulin)
Total protein staining (Ponceau S, SYPRO Ruby)
Staining controls for IHC/IF:
Including appropriate controls is particularly important in cancer research, where HPRT1 expression differences between normal and malignant tissues are being evaluated for diagnostic and prognostic purposes .
HPRT1 antibodies have become valuable tools for investigating its emerging role in cancer progression:
Expression analysis in clinical samples:
IHC analysis of tumor microarrays (TMAs) to correlate HPRT1 expression with:
Tumor stage and grade
Patient survival outcomes
Metastatic potential
Western blot quantification comparing tumor vs. adjacent normal tissues
Research has demonstrated HPRT1 overexpression in multiple cancer types, including nasopharyngeal carcinoma, where high expression correlates with poor prognosis
Functional studies using knockout/knockdown approaches:
Mechanistic investigations:
Analyze expression of downstream targets after HPRT1 manipulation:
Study signaling pathway alterations:
Tumor microenvironment interactions:
These approaches have revealed that HPRT1 silencing significantly reduces cancer cell proliferation and metastatic potential, suggesting its potential as a therapeutic target .
Investigating HPRT1's role in chemoresistance requires specialized methodological approaches:
Expression analysis in resistant vs. sensitive cells:
Manipulation of HPRT1 expression:
Mechanistic studies:
Signaling pathway analysis:
Combination treatment approaches:
Test synergy between HPRT1 inhibition and chemotherapeutic agents
Evaluate combination indices to quantify synergistic effects
Monitor biomarkers of response using HPRT1 antibodies
Research has shown that HPRT1 knockdown significantly reduces MDR1 expression in nasopharyngeal carcinoma cells, suggesting a potential mechanism for its involvement in chemoresistance . This finding opens avenues for developing strategies to overcome treatment resistance in cancer patients.
Exploring the connections between HPRT1 and the tumor immune microenvironment requires sophisticated analytical approaches:
Multiplex immunohistochemistry/immunofluorescence:
Correlation analysis with immune signatures:
Functional immunological assays:
Co-culture HPRT1-manipulated cancer cells with immune cells
Measure immune cell activation, cytokine production, and cytotoxicity
Evaluate immune checkpoint molecule expression (PD-L1, CTLA-4)
Immunomodulator correlation analysis:
Integration with TMB and MSI analyses:
Investigating post-translational modifications (PTMs) of HPRT1 requires specialized techniques and antibody applications:
Phosphorylation-specific analysis:
Immunoprecipitation with general HPRT1 antibodies (0.5-4.0 μg per 1-3 mg lysate)
Western blot with phospho-specific antibodies
Alternative: immunoblotting with general HPRT1 antibodies after:
Phosphatase treatment
Phos-tag SDS-PAGE for mobility shift detection
Mass spectrometry identification of phosphorylation sites
Ubiquitination detection:
Immunoprecipitate HPRT1 using validated antibodies
Western blot with anti-ubiquitin antibodies
Include proteasome inhibitors in lysis buffer (MG132)
Consider denaturing conditions to disrupt protein interactions
Acetylation analysis:
Immunoprecipitate HPRT1
Probe with anti-acetylated lysine antibodies
Include deacetylase inhibitors in lysis buffer
Mass spectrometry validation of acetylation sites
SUMOylation detection:
Lysates prepared with SUMO protease inhibitors (NEM)
Immunoprecipitate HPRT1
Western blot with anti-SUMO antibodies
Validate with mass spectrometry
Glycosylation analysis:
Treat samples with glycosidases (PNGase F, O-glycosidase)
Analyze mobility shifts on Western blots
Lectin blotting after HPRT1 immunoprecipitation
Mass spectrometry-based approaches:
Immunoprecipitate HPRT1 using highly specific antibodies
Trypsin digest and LC-MS/MS analysis
Targeted multiple reaction monitoring (MRM) for specific modifications
Compare modification patterns between:
Normal vs. cancer cells
Drug-sensitive vs. resistant cells
While specific PTMs of HPRT1 have not been extensively characterized in the provided research materials, these approaches provide a methodological framework for future investigations that may reveal how modifications regulate HPRT1's role in cancer progression and chemoresistance .
HPRT1's emerging role in cancer progression positions it as a potential biomarker, with antibodies serving as key tools in biomarker development:
Research has demonstrated that high HPRT1 expression correlates with poor prognosis in nasopharyngeal carcinoma and other cancers , supporting its potential utility as a prognostic biomarker. The standardized use of validated antibodies is essential for consistent biomarker assessment across laboratories and clinical settings.
Research on HPRT1 in nasopharyngeal carcinoma (NPC) requires specialized methodological approaches:
Expression analysis in clinical samples:
IHC on NPC tissue microarrays using standardized protocols:
RT-qPCR and Western blot analysis:
Functional studies using knockdown approaches:
Molecular mechanism investigations:
Prognostic analysis:
Research has demonstrated that HPRT1 is significantly upregulated in NPC compared to normal nasopharyngeal epithelium, and its silencing reduces proliferation, migration, and invasion capabilities of NPC cells . These findings establish HPRT1 as a promising target for further investigation in NPC pathogenesis and potential therapeutic development.
Investigating HPRT1 in Lesch-Nyhan syndrome (LNS) requires specialized approaches focusing on loss-of-function scenarios:
Mutation analysis and functional consequences:
Immunohistochemistry to assess HPRT1 protein expression:
Western blot analysis:
Enzymatic activity assays correlated with protein expression:
Measure HPRT enzyme activity in patient samples
Correlate activity with protein levels detected by antibodies
Western blot using HPRT1 antibodies to detect:
Wild-type protein (24-28 kDa)
Potential mutant forms with altered mobility
Cellular models of LNS:
Neural implications of HPRT1 deficiency:
iPSC-derived neural cultures:
Immunofluorescence to assess HPRT1 in different neural cell types
Counterstain with neural markers (TUJ1, GFAP, etc.)
Brain organoid models:
Section and stain for HPRT1 expression patterns
Correlate with developmental and functional markers
Purine metabolism analysis coupled with protein detection:
Measure purine metabolites in patient samples
Correlate with HPRT1 protein expression quantified by Western blot
Analyze salvage pathway components in relation to HPRT1 levels
While the provided search results focus more on HPRT1's role in cancer, these methodological approaches leverage HPRT1 antibodies to investigate the fundamental mechanisms underlying Lesch-Nyhan syndrome, where HPRT1 deficiency leads to severe neurological and behavioral abnormalities .
Exploring HPRT1's emerging connection to cancer immunotherapy response requires integrative approaches:
Correlation of HPRT1 expression with immunotherapy biomarkers:
Immune microenvironment characterization:
Functional studies in immunocompetent models:
Knockdown or overexpress HPRT1 in cancer cells
Implant into immunocompetent mice
Treat with immune checkpoint inhibitors
Monitor:
Tumor growth
Immune infiltration
Response to immunotherapy
Cell-based immune response assays:
Co-culture HPRT1-modified cancer cells with:
T cells
NK cells
Dendritic cells
Measure immune cell activation and cancer cell killing
Assess effect of HPRT1 on antigen presentation
Immunomodulator relationship analysis:
Research has shown that HPRT1 expression correlates with immune cell infiltration patterns in various cancers, suggesting potential involvement in immunotherapy response mechanisms . These methodological approaches provide a framework for investigating how HPRT1 might influence response to immune checkpoint inhibitors and other immunotherapeutic strategies.
For comprehensive investigation of HPRT1 in breast cancer:
Research indicates that HPRT1 RNA levels are significantly elevated in breast cancer tissues, particularly in basal cells and triple-negative breast cancer, suggesting its involvement in cancer progression through positive regulation of genes associated with cancer pathways . These protocols provide a structured approach to further elucidate HPRT1's role in breast cancer biology and potential therapeutic targeting.
When applying HPRT1 antibodies to neurodegenerative disease research:
Optimal tissue preparation for neural tissues:
Post-mortem tissue considerations:
Perfusion-fixed animal tissues:
Transcardial perfusion with 4% paraformaldehyde
Post-fixation (4-24 hours depending on tissue size)
Cryoprotection and sectioning protocols
Cell type-specific analysis in neural tissues:
Double immunofluorescence with:
Regional analysis of HPRT1 expression:
Purine metabolism context in neurodegeneration:
Connect HPRT1 expression to purine metabolism:
Double labeling with purine metabolism enzymes
Correlation with purine metabolite levels
Investigate HPRT1 in oxidative stress responses:
Co-labeling with oxidative stress markers
Relationship to antioxidant systems
Technical adaptations for neural tissue:
Models for studying HPRT1 in neurodegeneration:
While HPRT1 is traditionally studied in Lesch-Nyhan syndrome , these methodological approaches facilitate investigation of its potential roles in other neurodegenerative conditions, leveraging the specificity and versatility of available HPRT1 antibodies.