HPDL Antibody

Shipped with Ice Packs
In Stock

Description

Biological Function of HPDL

HPDL is an iron-containing enzyme involved in metabolic reprogramming and oxidative stress mitigation. Recent studies highlight its overexpression in 21 cancer types, including pancreatic ductal adenocarcinoma (PAAD) and lung adenocarcinoma (LUAD), where it correlates with poor prognosis and influences immunotherapy response . Conversely, reduced HPDL expression occurs in glioblastoma (GBM) and renal cancers . Genetic variants in HPDL are also linked to hereditary spastic paraplegia, suggesting broader neurological implications .

Therapeutic Implications

  • Immunotherapy prediction: High HPDL levels correlate with improved response to anti-PD1/PD-L1 therapies in melanoma (GSE78220 cohort) and bladder cancer (IMvigor210 trial) .

  • Functional studies: HPDL knockdown suppresses LUAD cell proliferation and migration in vitro, suggesting therapeutic targeting potential .

Neurological Research Findings

HPDL localizes to mitochondria in neurons, and biallelic mutations cause hereditary spastic paraplegia characterized by motor neuron degeneration. Key evidence includes:

  • Subcellular localization: HPDL resides in mitochondrial matrices, protected from proteinase K digestion unless membranes are disrupted .

  • Animal models: HPDL-deficient mice exhibit progressive motor deficits, mirroring human disease phenotypes .

Key Research Databases and Tools

Tool/DatabaseApplicationKey Insight
TCGA/GTExPancancer transcriptomic analysisHPDL overexpression in 21 malignancies
TIDEImmune cell infiltration analysisLinks HPDL to cytotoxic T-cell recruitment
CTRP/PRISMDrug sensitivity screeningIdentifies HPDL-targeting small molecules

Limitations and Future Directions

While HPDL antibodies enable critical discoveries, challenges remain:

  • Specificity: Polyclonal antibodies may cross-react with homologous proteins .

  • Therapeutic validation: No FDA-approved HPDL inhibitors exist despite preclinical promise .

  • Neurological mechanisms: HPDL’s exact role in mitochondrial function requires further study .

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery times may vary based on the shipping method and location. For specific delivery timelines, please consult your local distributor.
Synonyms
4 HPPD L antibody; 4-hydroxyphenylpyruvate dioxygenase-like antibody; 4-hydroxyphenylpyruvate dioxygenase-like protein antibody; Gloxd1 antibody; Glyoxalase domain containing 1 antibody; Glyoxalase domain-containing protein 1 antibody; Hpdl antibody; HPDL_HUMAN antibody
Target Names
HPDL
Uniprot No.

Target Background

Function
This antibody may exhibit dioxygenase activity.
Database Links

HGNC: 28242

KEGG: hsa:84842

STRING: 9606.ENSP00000335060

UniGene: Hs.162717

Protein Families
4HPPD family
Subcellular Location
Mitochondrion.

Q&A

What is HPDL protein and why is it significant for research?

HPDL (4-Hydroxyphenylpyruvate Dioxygenase-Like) is a 371 amino acid protein with sequence similarity to 4-hydroxyphenylpyruvate dioxygenase. Recent research has revealed that HPDL contains a mitochondrial localization signal and localizes to mitochondria, suggesting its involvement in mitochondrial metabolism . The protein has gained significant research interest following discoveries that bi-allelic pathogenic HPDL variants cause progressive, pediatric-onset spastic movement disorders with variable clinical presentation ranging from severe neonatal-onset neurodevelopmental delay to milder adolescent-onset hereditary spastic paraplegia . Despite its clinical importance, HPDL's precise function remains incompletely characterized, with some evidence suggesting it may have dioxygenase activity .

What applications are HPDL antibodies most commonly used for?

HPDL antibodies are primarily utilized in several key laboratory techniques:

  • Western Blotting (WB): For detection of HPDL protein expression levels in cell or tissue lysates, with typical recommended concentrations ranging from 0.04-0.4 μg/mL

  • Immunocytochemistry/Immunofluorescence (ICC/IF): For visualization of HPDL subcellular localization, typically using concentrations of 0.25-2 μg/mL

  • Immunohistochemistry (IHC): For studying HPDL expression patterns in tissue sections

  • Immunoprecipitation (IP): For isolation of HPDL and its interacting partners

  • ELISA: For quantitative measurement of HPDL in solution

These applications enable researchers to study HPDL expression patterns, subcellular localization, protein interactions, and potential functional roles in normal physiology and disease states.

How should researchers select the appropriate HPDL antibody for their experiments?

Selection of an appropriate HPDL antibody should be guided by several experimental considerations:

  • Epitope recognition: Different antibodies recognize distinct regions of HPDL. For example, some antibodies target the C-terminal region (aa 253-302), while others target mid-regions (aa 150-300) . Choose based on:

    • The protein domain you wish to study

    • Potential post-translational modifications

    • Protein interaction sites that might mask certain epitopes

  • Species reactivity: HPDL antibodies demonstrate variable cross-reactivity across species. Some show high homology and reactivity with human (100%), dog (92%), and mouse/bovine (85%) HPDL . Verify reactivity with your experimental model organism.

  • Clonality consideration:

    • Polyclonal antibodies offer broader epitope recognition but may have batch-to-batch variability

    • Monoclonal antibodies provide higher specificity for particular epitopes and greater consistency

  • Validated applications: Confirm the antibody has been validated for your specific application, as performance can vary significantly between techniques .

What are the recommended protocols for visualizing HPDL's subcellular localization?

Based on published research, the following immunofluorescence protocol has been successfully employed for visualizing HPDL's subcellular localization:

  • Cell preparation:

    • Culture cells on coverslips to 70-80% confluence

    • Fix with 4% paraformaldehyde (PFA) for 15 minutes at room temperature

    • Permeabilize with 0.1% Triton X-100 for 10 minutes

  • Immunostaining procedure:

    • Block with 5% normal serum for 1 hour

    • Incubate with primary HPDL antibody at 0.25-4 μg/mL concentration overnight at 4°C

    • Wash 3× with PBS

    • Apply fluorophore-conjugated secondary antibody for 1 hour at room temperature

    • Counterstain nuclei with DAPI

    • Mount with anti-fade mounting medium

  • Co-localization markers:

    • Include mitochondrial markers (e.g., MitoTracker or Tom20) to confirm mitochondrial localization

    • Consider nuclear markers to distinguish nucleoplasmic staining

This approach has revealed that HPDL localizes to both nucleoplasm and mitochondria in human cell lines, including CACO-2 cells . When analyzing results, focus on the distinctive pattern of mitochondrial staining, which typically appears as a reticular network throughout the cytoplasm.

How should researchers troubleshoot non-specific binding when using HPDL antibodies in Western blotting?

When encountering non-specific binding in Western blotting with HPDL antibodies, implement the following troubleshooting strategy:

  • Optimize blocking conditions:

    • Increase blocking time to 2 hours

    • Test alternative blocking agents (5% BSA may be more effective than milk for some phospho-epitopes)

    • Consider specialized blocking buffers if standard options fail

  • Antibody validation steps:

    • Perform antibody titration (0.04-0.4 μg/mL range is recommended for HPDL antibodies)

    • Include positive controls (CACO-2 and PC-3 cell lysates have demonstrated reliable HPDL expression)

    • Run a negative control with HPDL-depleted samples (siRNA knockdown)

  • Wash optimization:

    • Increase wash duration and number (5 washes of 5 minutes each)

    • Add 0.05-0.1% Tween-20 to wash buffer to reduce non-specific interactions

  • Membrane handling:

    • Ensure complete protein transfer

    • Cut membranes to minimize antibody consumption and improve signal-to-noise ratio

    • Consider using PVDF membranes instead of nitrocellulose for better protein retention

  • Expected band pattern:

    • The predicted molecular weight of HPDL is 39 kDa

    • Verify any unexpected bands against known HPDL isoforms or post-translational modifications

How can HPDL antibodies be utilized to investigate HPDL's role in mitochondrial metabolism?

To investigate HPDL's putative role in mitochondrial metabolism, researchers can employ HPDL antibodies in the following advanced approaches:

  • Subcellular fractionation and immunoblotting:

    • Isolate mitochondrial, cytosolic, and nuclear fractions using differential centrifugation

    • Perform Western blotting with HPDL antibody on each fraction

    • Include markers for each compartment (e.g., VDAC for mitochondria, GAPDH for cytosol)

    • Quantify relative HPDL distribution to determine primary localization

  • Proximity labeling combined with immunoprecipitation:

    • Express HPDL fused to a promiscuous biotin ligase (BioID or TurboID)

    • Perform proximity labeling to identify proteins in close proximity to HPDL

    • Use HPDL antibodies for immunoprecipitation to validate interactions

    • Analyze interacting partners for enrichment of metabolic enzymes

  • Functional mitochondrial assays:

    • Use siRNA or CRISPR to deplete HPDL in cell models

    • Apply HPDL antibodies to confirm knockdown efficiency

    • Measure changes in:

      • Oxygen consumption rate (OCR)

      • Mitochondrial membrane potential

      • ROS production

      • ATP synthesis

  • Metabolomic profiling:

    • Compare metabolite profiles between HPDL-depleted and control cells

    • Use HPDL antibodies for immunofluorescence to correlate alterations with HPDL subcellular distribution

    • Focus on pathways related to amino acid metabolism, given HPDL's sequence similarity to 4-hydroxyphenylpyruvate dioxygenase

This multifaceted approach can help elucidate whether HPDL possesses dioxygenase activity and identify specific metabolic pathways in which it participates.

What strategies can be employed to study the relationship between HPDL protein levels and neurodegenerative disease progression?

To investigate the relationship between HPDL protein levels and neurodegenerative disease progression, researchers can implement the following methodological approaches:

  • Patient-derived cell models:

    • Establish fibroblast cultures from patients with bi-allelic HPDL variants

    • Generate iPSCs and differentiate into relevant neural cell types

    • Use HPDL antibodies to quantify protein levels via Western blotting

    • Correlate HPDL levels with clinical severity (studies have shown reduced HPDL levels in fibroblasts from more severely affected individuals)

  • Tissue microarray analysis:

    • Develop tissue microarrays from patient CNS samples

    • Perform immunohistochemistry with HPDL antibodies

    • Quantify region-specific expression changes

    • Compare with clinical and pathological features

  • Animal model characterization:

    • Generate HPDL knockout or knockin models recapitulating human mutations

    • Apply HPDL antibodies to track protein expression across development

    • Correlate protein levels with onset and progression of motor symptoms

    • Analyze mitochondrial function in affected neural tissues

  • Mechanistic studies:

    • Investigate whether HPDL loss affects mitochondrial morphology or dynamics

    • Assess impact on oxidative phosphorylation complexes

    • Evaluate markers of mitochondrial stress and quality control

    • Test whether HPDL restoration rescues disease phenotypes

These approaches can help establish whether HPDL deficiency causes neurodegeneration through mitochondrial dysfunction and identify potential therapeutic targets for HPDL-related disorders.

How should researchers interpret conflicting results between different antibody-based techniques when studying HPDL?

When faced with conflicting results between different antibody-based techniques, researchers should implement the following systematic resolution strategy:

  • Technique-specific considerations:

    TechniqueCommon IssuesResolution Approach
    Western BlotBand size discrepanciesVerify using knockout controls; check for post-translational modifications
    ImmunofluorescenceDivergent localization patternsValidate with multiple antibodies targeting different epitopes; use GFP-tagged constructs for confirmation
    IP-MSDifferent interacting partnersCompare stringency conditions; validate key interactions with reciprocal co-IP
    IHCVariable tissue expressionOptimize antigen retrieval; compare with mRNA expression data
  • Antibody validation status assessment:

    • Review validation data for each antibody used

    • Check if antibodies recognize different epitopes that might be differentially accessible

    • Consider using orthogonal approaches (e.g., CRISPR knockout controls, tagged HPDL expression)

  • Biological vs. technical variation analysis:

    • Determine if differences reflect true biological variation or technical limitations

    • Examine if conflicting results occur in different cell types or conditions

    • Test whether HPDL mutations or modifications might affect epitope recognition

  • Integration with non-antibody techniques:

    • Compare antibody-based results with mRNA expression data

    • Use CRISPR/Cas9 genome editing to introduce epitope tags

    • Consider targeted mass spectrometry approaches

By systematically addressing these factors, researchers can determine whether discrepancies reflect genuine biological complexity or technical limitations of specific antibodies or methods.

What quality control measures are essential when working with HPDL antibodies in protein interaction studies?

When conducting protein interaction studies with HPDL antibodies, implement these essential quality control measures:

  • Antibody specificity validation:

    • Perform immunoprecipitation followed by mass spectrometry to confirm primary pull-down of HPDL

    • Include HPDL-depleted samples as negative controls

    • Verify by Western blotting that the antibody recognizes a band of expected size (39 kDa)

  • Interaction stringency assessment:

    • Perform parallel immunoprecipitations with increasing salt concentrations (150-500 mM NaCl)

    • Compare detergent conditions (e.g., NP-40, Triton X-100, CHAPS) to distinguish membrane-dependent interactions

    • Include appropriate blocking of non-specific binding sites with pre-immune serum

  • Reciprocal confirmation strategy:

    • Validate key interactions by reverse immunoprecipitation

    • Confirm co-localization by immunofluorescence

    • Perform proximity ligation assay (PLA) to verify interactions occur in intact cells

  • Controls hierarchy:

    • Isotype-matched control antibodies

    • Pre-immune serum controls

    • Input sample analysis (typically 5-10% of IP material)

    • Beads-only controls to identify non-specific binding to matrix

  • Biological relevance filtration:

    • Focus on interactions consistent with HPDL's mitochondrial localization

    • Prioritize interactions with proteins implicated in pathways related to HPDL function

    • Cross-reference with known mitochondrial protein databases

Following these quality control measures will help distinguish genuine HPDL-interacting proteins from background contaminants, yielding higher confidence interaction datasets.

How can HPDL antibodies be leveraged to study the role of this protein in neurodegenerative disease models?

HPDL antibodies can be strategically employed to investigate neurodegenerative disease mechanisms through these methodological approaches:

  • Cellular phenotyping in disease models:

    • Generate neuronal models using patient-derived iPSCs with HPDL mutations

    • Apply HPDL antibodies to:

      • Quantify protein levels and correlate with disease severity

      • Track subcellular localization changes during neuronal differentiation

      • Measure alterations in mitochondrial morphology and function

    • Compare neurons with different bi-allelic HPDL variants to establish genotype-phenotype correlations

  • Pathway analysis in affected tissues:

    • Use HPDL antibodies for immunohistochemistry on brain sections from affected individuals

    • Perform co-staining with:

      • Cell-type specific markers

      • Mitochondrial dysfunction indicators

      • Markers of neuroinflammation and stress response

    • Analyze regional vulnerability patterns

  • Therapeutic screening framework:

    • Develop high-content screening assays using HPDL antibodies to:

      • Measure HPDL stabilization by small molecules

      • Detect restoration of proper subcellular localization

      • Identify compounds that rescue mitochondrial defects

    • Validate hits using secondary assays for functional recovery

  • In vivo model development and analysis:

    • Generate transgenic models expressing HPDL variants

    • Use HPDL antibodies to:

      • Validate model fidelity through protein expression analysis

      • Track age-dependent changes in HPDL expression and localization

      • Correlate biochemical alterations with behavioral phenotypes

    • Test genetic or pharmacological interventions that target HPDL-related pathways

This comprehensive approach enables researchers to establish mechanistic links between HPDL dysfunction and neurological disease, potentially identifying novel therapeutic targets.

What advanced techniques can be combined with HPDL antibodies to investigate post-translational modifications of this protein?

To comprehensively characterize HPDL post-translational modifications (PTMs), researchers can combine HPDL antibodies with these advanced techniques:

  • Phosphorylation analysis workflow:

    • Immunoprecipitate HPDL using validated antibodies

    • Perform phospho-specific Western blotting or mass spectrometry

    • Compare phosphorylation under different cellular conditions:

      • Mitochondrial stress

      • Cell cycle phases

      • Differentiation states

    • Validate findings using phospho-mutant HPDL constructs

  • Ubiquitination and degradation pathway investigation:

    • Treat cells with proteasome inhibitors (MG132) or lysosomal inhibitors

    • Immunoprecipitate HPDL and probe for ubiquitin

    • Perform cycloheximide chase experiments with HPDL antibody detection

    • Identify E3 ligases responsible for HPDL regulation

  • Redox modification characterization:

    • Given HPDL's mitochondrial localization, investigate redox-sensitive modifications

    • Use redox proteomics approaches with HPDL antibodies to detect:

      • Cysteine oxidation

      • S-nitrosylation

      • Glutathionylation

    • Correlate modifications with mitochondrial ROS production

  • PTM cross-talk mapping strategy:

    • Generate a comprehensive PTM map using complementary approaches:

      • Targeted mass spectrometry following HPDL immunoprecipitation

      • Peptide arrays probed with modification-specific antibodies

      • Proximity labeling to identify modifying enzymes

    • Develop a temporal model of how different PTMs regulate HPDL function

These advanced approaches will help elucidate how post-translational modifications regulate HPDL stability, localization, and function, potentially revealing how dysregulation of these processes contributes to disease pathogenesis.

How can HPDL antibodies be utilized in developing potential biomarkers for neurodegenerative diseases?

HPDL antibodies can facilitate biomarker development for neurodegenerative diseases through these methodological approaches:

  • Clinical sample analysis strategy:

    • Develop sensitive ELISA or immunoassays using HPDL antibodies

    • Apply to:

      • CSF samples from patients with movement disorders

      • Plasma/serum exosomes containing mitochondrial proteins

      • Tissue biopsies (e.g., skin fibroblasts) from affected individuals

    • Compare HPDL levels between patients with bi-allelic HPDL variants and other neurodegenerative conditions

  • Correlation with disease progression:

    • Conduct longitudinal studies measuring HPDL protein levels

    • Track changes in relation to:

      • Clinical severity scores

      • Neuroimaging findings

      • Other biochemical markers

    • Establish whether HPDL levels predict disease onset in pre-symptomatic carriers

  • Multiplexed biomarker panel development:

    • Combine HPDL antibody-based detection with other mitochondrial proteins

    • Create a fingerprint of mitochondrial dysfunction

    • Include markers for:

      • Oxidative stress

      • Mitochondrial DNA damage

      • Mitochondrial dynamics

  • Companion diagnostic potential:

    • Develop antibody-based assays to:

      • Identify patients with HPDL-related disorders

      • Monitor treatment response

      • Stratify patients for clinical trials

    • Validate assays across multiple clinical centers

This systematic approach can help determine whether HPDL protein levels or post-translational modifications serve as useful biomarkers for diagnosis, prognosis, or treatment monitoring in patients with movement disorders or other neurodegenerative conditions.

What considerations are critical when using HPDL antibodies to validate gene therapy approaches for HPDL-related disorders?

When validating gene therapy approaches for HPDL-related disorders, researchers should address these critical considerations when using HPDL antibodies:

By addressing these considerations, researchers can develop robust validation strategies for gene therapy approaches targeting HPDL-related disorders, ensuring appropriate expression, localization, and function of the therapeutic protein.

How might HPDL antibodies contribute to understanding the broader mitochondrial disease landscape?

HPDL antibodies can advance our understanding of the mitochondrial disease landscape through these innovative research approaches:

  • Mitochondrial interactome mapping:

    • Use HPDL antibodies for immunoprecipitation followed by mass spectrometry

    • Identify HPDL interaction partners within the mitochondrial network

    • Compare interactomes between:

      • Normal and disease states

      • Different cell types with varied metabolic profiles

      • Developmental stages

    • Construct protein interaction networks to position HPDL within mitochondrial pathways

  • Comparative mitochondrial pathology:

    • Apply HPDL antibodies in tissue studies across various mitochondrial diseases

    • Analyze whether HPDL expression or localization changes in:

      • Primary mitochondrial disorders (e.g., Leigh syndrome)

      • Secondary mitochondrial dysfunction (e.g., Parkinson's, ALS)

      • Age-related mitochondrial decline

    • Identify common versus disease-specific patterns

  • Metabolic flux analysis integration:

    • Combine HPDL protein level quantification with:

      • Metabolomic profiling

      • Stable isotope tracing

      • Oxygen consumption measurements

    • Correlate HPDL levels with specific metabolic alterations

    • Identify potential metabolic signatures of HPDL dysfunction

  • Single-cell resolution studies:

    • Apply HPDL antibodies in single-cell protein analysis techniques

    • Characterize cell-to-cell variability in:

      • HPDL expression levels

      • Mitochondrial content and morphology

      • Metabolic states

    • Identify particularly vulnerable cell populations

This systematic application of HPDL antibodies can help position HPDL-related disorders within the broader context of mitochondrial diseases, potentially revealing common therapeutic targets or disease mechanisms.

What novel methodological approaches combining HPDL antibodies with advanced imaging techniques can reveal about mitochondrial dynamics?

Novel methodologies combining HPDL antibodies with advanced imaging techniques can provide unprecedented insights into mitochondrial dynamics through these approaches:

  • Super-resolution microscopy implementation:

    • Apply HPDL antibodies in STORM or PALM super-resolution microscopy

    • Achieve 20-30 nm resolution to:

      • Precisely map HPDL within mitochondrial subcompartments

      • Visualize colocalization with other mitochondrial proteins at nanoscale

      • Identify potential HPDL-containing protein complexes

    • Compare localization patterns in healthy versus diseased cells

  • Live-cell imaging integration:

    • Develop split-GFP complementation systems with HPDL

    • Use HPDL antibodies to validate expression and localization

    • Track in real-time:

      • HPDL recruitment to mitochondria

      • Changes during mitochondrial stress responses

      • Dynamics during mitochondrial fission/fusion events

  • Correlative light and electron microscopy (CLEM):

    • Combine immunofluorescence using HPDL antibodies with electron microscopy

    • Achieve both molecular specificity and ultrastructural context

    • Analyze HPDL distribution in relation to:

      • Cristae morphology

      • Contact sites with other organelles

      • Areas of mitochondrial division or fusion

  • Expansion microscopy application:

    • Physically expand cellular structures using polymer embedding

    • Apply HPDL antibodies after expansion

    • Achieve super-resolution imaging on standard microscopes

    • Visualize HPDL distribution within expanded mitochondrial structures

  • Functional imaging correlation:

    • Combine HPDL immunolabeling with:

      • Mitochondrial membrane potential indicators

      • ROS sensors

      • ATP production measures

    • Correlate HPDL levels or distribution with functional mitochondrial parameters

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 2025 TheBiotek. All Rights Reserved.