PPARD (Peroxisome Proliferator-Activated Receptor Delta) is a member of the PPAR family, which regulates gene networks linked to metabolic homeostasis, energy balance, and inflammation . It is notably implicated in diseases such as diabetes, obesity, atherosclerosis, and cancer . PPARD monoclonal antibodies are critical tools for studying its expression, localization, and function in these pathways.
PPARD monoclonal antibodies are produced using hybridoma technology, where mouse spleen cells immunized with PPARD-derived antigens are fused with myeloma cells to generate immortalized antibody-producing clones . Key clones include:
Clone | Host | Applications | Reactivity | Source |
---|---|---|---|---|
1D7 | Mouse | IHC-p | Human, Mouse, Rat | |
1G4 | Mouse | WB, ELISA | Human | |
1B10E1 | Mouse | WB, ChIP, ELISA | Human, Pig | |
PCRP-PPARD-1E3 | Mouse | IHC, IF | Human |
These antibodies are typically unconjugated and purified via Protein A/G affinity chromatography .
PPARD monoclonal antibodies are utilized across diverse experimental workflows:
Western Blot (WB): Detects PPARD at ~54 kDa in human, mouse, and rat tissues (e.g., heart, skeletal muscle) .
Immunohistochemistry (IHC): Localizes PPARD in nuclei of cells from brain, ovary, and tumor tissues .
Enzyme-Linked Immunosorbent Assay (ELISA): Quantifies PPARD expression in serum or lysates .
Chromatin Immunoprecipitation (ChIP): Identifies PPARD-DNA interactions in metabolic gene regulation .
PPARD activation enhances fatty acid oxidation and mitochondrial biogenesis. Studies using PPARD monoclonal antibodies (e.g., clone 1B10E1) revealed its role in ameliorating diabetic kidney disease via anti-inflammatory mechanisms .
Elevated PPARD expression in colorectal cancer cells correlates with tumor progression. Antibodies like 1D7 have been used to validate PPARD as a biomarker and therapeutic target .
PPARD monoclonal antibodies (e.g., Sc-7197) demonstrated that PPARD induces slow-twitch muscle fiber formation, linking muscle activity to metabolic adaptation .
Species Specificity: Some clones (e.g., 1G4) exhibit limited reactivity beyond humans .
Validation: Optimal dilution ratios must be empirically determined for each application .
Safety: Sodium azide-containing buffers require careful handling .
Advancements in humanized monoclonal antibody engineering could reduce immunogenicity in therapeutic contexts . Additionally, bispecific PPARD-targeting antibodies are under exploration for dual-pathway modulation in metabolic disorders .
PPARD, also known as NR1C2, FAAR, and Nuclear receptor subfamily 1 group C member 2, is a member of the peroxisome proliferator-activated receptor (PPAR) family. It functions as a nuclear hormone receptor that binds peroxisome proliferators and regulates the size and number of peroxisomes produced by cells. PPARD mediates numerous biological processes and has been implicated in the development of several chronic diseases, including diabetes, obesity, atherosclerosis, and cancer. It serves as a potent inhibitor of ligand-induced transcription activity of PPAR alpha and PPAR gamma and may function as an integrator of transcription repression and nuclear receptor signaling .
Research interest in PPARD has intensified due to its ubiquitous expression with maximal levels in placenta and skeletal muscle, and its elevated expression in certain cancer types, particularly colorectal cancer cells. This elevation can be repressed by adenomatosis polyposis coli (APC), a tumor suppressor protein related to the APC/beta-catenin signaling pathway . PPARD monoclonal antibodies are therefore essential tools for investigating these mechanisms in basic and translational research settings.
PPARD monoclonal antibodies are primarily used in immunohistochemistry (IHC) applications, where they enable visualization of PPARD expression patterns in tissue samples. Based on the available product information, PPARD antibodies have been validated for:
Immunohistochemistry-paraffin (IHC-p): Used to detect PPARD in formalin-fixed, paraffin-embedded tissue sections
Western Blot (WB): Multiple publications have reported successful use in detecting PPARD protein in cell and tissue lysates
Immunofluorescence (IF): At least one publication has reported successful application for fluorescent detection of PPARD
ELISA: Some antibodies have been validated for enzyme-linked immunosorbent assay applications
Different antibody clones (such as 1D7 and 2F9) have been optimized for specific applications, and researchers should select the appropriate clone based on their experimental requirements .
For long-term storage, PPARD monoclonal antibodies should be stored at -20°C, where they typically remain stable for one year after shipment . For frequent use and short-term storage (up to one month), antibodies can be stored at 4°C to avoid repeated freeze-thaw cycles that may compromise antibody integrity and performance .
Most PPARD monoclonal antibodies are supplied in a buffer containing stabilizers:
Typically in phosphate-buffered saline (PBS)
With 50% glycerol as a cryoprotectant
Including protein protectants (0.5% BSA or similar)
When handling the antibody, it's important to:
Aliquot upon initial thawing to minimize freeze-thaw cycles
Briefly centrifuge the vial before opening to collect all liquid at the bottom
Avoid contamination by using clean pipette tips
Return to appropriate storage conditions immediately after use
Optimal dilution ratios for PPARD monoclonal antibodies vary by application and specific antibody clone. Based on the product information provided, the following dilutions are recommended as starting points:
For IHC applications:
For Western Blot applications:
These values provide initial guidelines, but the optimal working concentration should be determined by each researcher through titration experiments in their specific experimental system. Factors such as tissue type, fixation method, detection system, and incubation conditions can all influence the optimal antibody concentration .
When working with challenging tissue samples, several optimization strategies can improve PPARD detection:
Antigen retrieval optimization: The search results indicate that for some PPARD antibodies, TE buffer pH 9.0 is recommended for antigen retrieval, though citrate buffer pH 6.0 can be used as an alternative . For tissues with high lipid content or dense extracellular matrix, extended retrieval times may be necessary.
Signal amplification approaches: For tissues with low PPARD expression, consider employing:
Polymer-based detection systems
Tyramide signal amplification
Extended primary antibody incubation (overnight at 4°C)
Background reduction techniques: To reduce non-specific binding:
Increase blocking time (2-3 hours)
Use a combination of different blocking agents (serum, BSA, casein)
Perform longer washing steps with 0.1% Tween-20 in PBS
Include a peroxidase quenching step (if using HRP-based detection)
Consider titrating the antibody to identify the optimal concentration that gives specific signal with minimal background
Validation controls: Always include positive control tissues known to express PPARD (such as human brain, mouse heart tissue, mouse skeletal muscle tissue) and negative controls (primary antibody omission and/or isotype controls) .
Rigorous validation of PPARD antibody specificity is crucial for reliable research outcomes. A comprehensive validation approach should include:
Multiple detection methods: Confirm PPARD detection using at least two independent methods (e.g., IHC and Western blot) .
Knockout/knockdown controls: The gold standard for antibody validation includes:
Testing the antibody in PPARD-knockout tissues/cells
Using PPARD-knockdown samples (siRNA or shRNA) compared to controls
Observing the expected reduction or absence of signal
Blocking peptide experiments: Pre-incubate the antibody with a specific blocking peptide (recombinant PPARD protein) to demonstrate signal specificity. The search results mention that blocking peptides can be purchased to validate antibody specificity .
Multiple antibody comparison: Test multiple PPARD antibodies with different epitopes (e.g., compare monoclonal antibodies from different clones like 1D7 and 2F9) .
Molecular weight verification: In Western blot applications, verify that the detected band aligns with the expected molecular weight of PPARD (calculated MW: ~50 kDa, observed MW: ~54 kDa) .
The literature presents contradictory findings regarding PPARD's role in cancer, which researchers must carefully consider when designing experiments and interpreting results. The table in the search results demonstrates this complexity, showing that PPARD can exhibit both pro-tumorigenic and anti-tumorigenic effects depending on context .
Key contradictions include:
Tumor growth effects:
Pro-tumorigenic: In multiple models including LLC1 tumors, SW480 cells, and Apc(Min/+) mice, PPARD agonists like GW501516 increased tumor growth .
Anti-tumorigenic: In azoxymethane-induced colon tumors, GW0742 (PPARD agonist) decreased tumor growth. In transgenic hepatitis B virus mice, GW0742 reduced hepatic tumor foci .
Cancer type specificity:
Colorectal cancer: Some studies show PPARD promotes tumorigenesis, while others show anti-tumorigenic effects .
Melanoma: PPARD agonists (GW0742, GW501516) decreased proliferation in multiple melanoma cell lines .
Breast cancer: Mixed effects observed depending on cell line (inhibited proliferation in MCF-7 but not in MDA-MB-231) .
To address these discrepancies, researchers should:
Carefully design experiments with appropriate positive and negative controls
Consider the specific cellular context and cancer type
Document experimental conditions thoroughly (cell density, passage number, reagent concentrations)
Use multiple complementary approaches to validate findings
Consider the role of PPARD in different cell populations within the tumor microenvironment
Examine potential differences in downstream signaling pathways
PPARD functions within complex signaling networks, and multi-parameter analysis can provide deeper insights into its regulatory mechanisms. Advanced researchers can integrate PPARD antibody detection with other techniques:
Multiplex immunofluorescence/immunohistochemistry:
Co-stain tissues for PPARD alongside other pathway components (e.g., APC, β-catenin)
Use spectral unmixing to resolve multiple fluorophores
Employ tyramide signal amplification for detecting low-abundance signaling proteins
Combine with nuclear markers to confirm PPARD nuclear localization
Proximity ligation assay (PLA):
Detect protein-protein interactions between PPARD and potential binding partners
Validate transcriptional complexes involving PPARD
Investigate post-translational modifications of PPARD
ChIP-seq integration:
Combine PPARD protein detection with chromatin immunoprecipitation sequencing to map genomic binding sites
Correlate PPARD protein levels with target gene expression
Identify cell-type-specific regulatory mechanisms
Phospho-protein analysis:
Examine the phosphorylation status of PPARD and downstream effectors
Correlate PPARD activation with signaling pathway activity
Study the impact of therapeutic agents on PPARD-dependent signaling
Researchers often encounter situations where PPARD protein levels do not directly correlate with its transcriptional activity. To address this complexity:
Combine protein detection with functional assays:
Use luciferase reporter assays with PPARD-responsive elements
Measure expression of known PPARD target genes by qRT-PCR
Assess metabolic changes associated with PPARD activation (fatty acid oxidation, glucose utilization)
Characterize post-translational modifications:
Use phospho-specific antibodies to detect activation status
Examine SUMOylation, ubiquitination, and acetylation patterns
Correlate modifications with transcriptional activity
Analyze protein-protein interactions:
Investigate co-repressor/co-activator recruitment
Examine interactions with other nuclear receptors (PPARα, PPARγ)
Study competitive binding of ligands and antagonists
Consider ligand availability and metabolism:
Measure endogenous PPARD ligand levels
Evaluate the impact of metabolic changes on PPARD activity
Assess the influence of inflammatory mediators on PPARD function
Common challenges when using PPARD antibodies in IHC include:
High background staining:
Weak or absent staining:
Problem: Insufficient antigen detection despite PPARD expression.
Solutions: Verify antibody reactivity with your species of interest (human, mouse, rat confirmed for most antibodies), optimize antigen retrieval (try both citrate buffer pH 6.0 and TE buffer pH 9.0), increase antibody concentration, extend incubation time, or employ signal amplification systems .
Variable staining intensity:
Problem: Inconsistent results between experiments or tissue regions.
Solutions: Standardize fixation protocols, control tissue processing time, use automated staining platforms if available, include internal positive controls, and normalize staining time and temperature.
Nuclear versus cytoplasmic localization:
Problem: Unexpected subcellular localization pattern.
Solutions: Verify fixation quality (overfixation can mask nuclear antigens), optimize permeabilization, compare with literature reports of PPARD localization in your specific tissue/cell type, and consider using cell fractionation followed by Western blot to confirm localization .
Cancer heterogeneity presents significant challenges for interpreting PPARD expression data. Advanced analytical approaches include:
Spatial heterogeneity analysis:
Systematic sampling across different tumor regions
Quantitative image analysis with spatial statistics
Correlation of PPARD expression with histopathological features (tumor grade, invasion front, necrotic areas)
Single-cell approaches:
Integration with single-cell RNA sequencing data
Multiplex immunofluorescence to analyze co-expression patterns
Cell-type specific analysis (tumor cells vs. stromal/immune components)
Temporal dynamics consideration:
Serial sampling during disease progression
Analysis of primary tumors versus metastases
Treatment-induced changes in PPARD expression
Functional correlation:
Link expression patterns to proliferation markers (Ki-67)
Correlate with markers of metabolic activity
Associate with treatment resistance phenotypes
The conflicting data in the literature regarding PPARD's role in cancer (promoting or inhibiting tumor growth depending on context) highlights the importance of considering tumor heterogeneity in experimental design and data interpretation .
PPARD plays crucial roles in metabolic regulation, making its antibodies valuable tools for studying cancer metabolism:
Metabolic pathway correlation:
Co-stain for PPARD and key metabolic enzymes (e.g., FASN, ACC, CPT1)
Correlate PPARD expression with glucose transporters (GLUTs) and lactate transporters (MCTs)
Examine relationship with mitochondrial markers in different cancer models
Microenvironmental adaptation:
Analyze PPARD expression in hypoxic versus normoxic regions (co-stain with HIF-1α)
Correlate with nutrient stress markers
Investigate expression changes in response to metabolic inhibitors
Therapeutic implications:
Monitor PPARD expression changes following metabolic-targeted therapies
Assess PPARD as a biomarker for response to metabolic interventions
Explore combinatorial approaches targeting PPARD and metabolic vulnerabilities
Lipid metabolism integration:
Combine PPARD immunodetection with lipid droplet staining
Correlate with fatty acid oxidation capacity
Investigate membrane lipid composition changes
The search results indicate that PPARD has been studied in various cancer contexts with different metabolic profiles, including colorectal, prostate, breast, and lung cancers .
Advanced techniques for investigating PPARD protein interactions in their native context include:
Proximity Ligation Assay (PLA):
Enables visualization of protein-protein interactions (<40 nm proximity)
Can detect interactions between PPARD and co-regulators in fixed cells/tissues
Provides spatial information about interaction sites within cells
Quantifiable by digital image analysis
FRET/FLIM microscopy:
Förster Resonance Energy Transfer combined with Fluorescence Lifetime Imaging
Requires fluorophore-conjugated antibodies or expression of fluorescent fusion proteins
Provides dynamic information about protein interactions
Can detect conformational changes in protein complexes
In situ hybridization with immunodetection:
Correlate PPARD protein localization with target gene transcription
RNAscope technology combined with immunofluorescence
Spatial transcriptomics integration with protein detection
Mass spectrometry imaging:
Emerging technique for spatial proteomics
Can map PPARD interaction networks across tissue regions
Combines with traditional immunohistochemistry for validation
These approaches can help resolve contradictions in the literature by revealing context-specific interactions that may explain differential PPARD functions in various cancer types and experimental models .
PPARD belongs to the nuclear receptor superfamily, and understanding its relationship with other family members is critical:
Cross-regulation with other PPARs:
Shared co-regulator networks:
Many nuclear receptors compete for the same co-activators/co-repressors
PPARD-specific effects must be distinguished from general nuclear receptor signaling
Validation using multiple approaches (genetic, pharmacological, immunological)
Ligand specificity considerations:
Cross-reactivity of synthetic ligands between PPAR subtypes
Natural ligands may have multiple targets
Combine ligand studies with specific antibody detection for pathway validation
Therapeutic implications:
PPARD agonists and antagonists have been developed for metabolic diseases
Cancer applications require careful consideration of context-dependent effects
Combination approaches targeting multiple nuclear receptors may offer advantages
The table in the search results highlights the complexity of PPARD functions in cancer, showing both pro- and anti-tumorigenic effects depending on context, similar to the context-dependent roles observed with other nuclear receptors .
Integrating protein-level data with genomic and transcriptomic information provides a comprehensive understanding of PPARD biology:
ChIP-seq correlation:
Map PPARD genomic binding sites using ChIP-seq
Correlate binding patterns with protein expression in matching samples
Identify cell type-specific regulatory elements
Multi-omic integration:
Perform integrated analysis of PPARD protein levels, mRNA expression, and target gene activation
Identify discordances that suggest post-transcriptional regulation
Reveal feedback mechanisms and regulatory circuits
Single-cell multi-modal analysis:
Emerging technologies allow simultaneous detection of proteins and mRNAs in single cells
Reveals heterogeneity in PPARD expression and activity at cellular resolution
Identifies rare cell populations with unique PPARD regulation
Patient-derived models:
Validate PPARD antibody performance in patient-derived xenografts and organoids
Correlate with genomic alterations in the same samples
Develop personalized approaches based on PPARD status
These integrative approaches can help resolve contradictions in the literature by revealing the molecular contexts that determine whether PPARD promotes or inhibits cancer progression in specific settings .