PPARA Antibody, Biotin conjugated is a rabbit-derived polyclonal antibody targeting human PPARα (UniProt ID: Q07869, Gene ID: 5465). The biotin tag enhances detection versatility, allowing compatibility with avidin-horseradish peroxidase (HRP) or fluorescent streptavidin probes .
Validated applications include:
PPARα regulates fatty acid oxidation and lipoprotein metabolism. The Biotin-conjugated antibody has been used to:
Identify PPARα-RXRA heterodimers in hepatic cells, critical for activating peroxisomal β-oxidation genes like ACOX1 .
Investigate PPARα’s role in reducing inflammation via NF-κB pathway inhibition .
PPARA (Peroxisome Proliferator-Activated Receptor alpha) is a ligand-activated transcription factor that serves as a key regulator of lipid metabolism. It functions primarily by controlling the peroxisomal beta-oxidation pathway of fatty acids and acts as a transcription activator for genes like ACOX1 and various P450 genes. PPARA is activated by endogenous ligands such as 1-palmitoyl-2-oleoyl-sn-glycerol-3-phosphocholine (16:0/18:1-GPC) and oleylethanolamide, a naturally occurring lipid that regulates satiety. Its importance in metabolic research stems from its role as a receptor for peroxisome proliferators including hypolipidemic drugs and fatty acids . Additionally, PPARA may participate in circadian rhythm regulation by connecting clock information to metabolic pathways through interaction with PER2 . Understanding PPARA function provides crucial insights into metabolic disorders, lipid homeostasis, and potential therapeutic targets.
Biotin-conjugated PPARA antibodies offer several methodological advantages in research applications. The biotin-streptavidin system provides significant signal amplification, enhancing detection sensitivity particularly in tissues with low PPARA expression. This conjugation enables versatile detection methods through secondary reporting systems (fluorescent, enzymatic, or gold-labeled streptavidin) without requiring multiple secondary antibodies. The strong biotin-streptavidin interaction (dissociation constant of 10^-15 M) ensures stable binding during extensive washing procedures, reducing background signal in techniques like Western blotting and immunoprecipitation . The biotin label minimizes cross-reactivity issues common with species-specific secondary antibodies, making it valuable for multi-labeling experiments. Additionally, the biotin conjugation preserves antibody specificity while providing detection flexibility across immunohistochemistry, Western blotting, and immunoprecipitation applications .
PPARA shows distinct tissue-specific expression patterns that researchers should consider when designing experiments. The highest expression levels occur in metabolically active tissues: liver, heart, kidney, and skeletal muscle . In human tissue samples, PPARA is also expressed in circulating monocytes, which provides an accessible biomarker source for metabolic studies without requiring tissue biopsies . Expression levels vary significantly across tissues, with liver demonstrating approximately 5-10 fold higher expression compared to adipose tissue. These differential expression patterns correlate with tissue-specific metabolic functions and energy utilization pathways. When conducting immunohistochemistry or immunofluorescence studies, researchers should adjust antibody dilutions accordingly (1:50-1:200 for IF and 1:500-1:1000 for WB as starting points) . Control experiments validating detection sensitivity in each tissue type are essential, particularly when comparing PPARA levels across multiple tissues simultaneously.
When performing Western blotting with biotin-conjugated PPARA antibody, several optimization steps are critical for successful detection. Begin with proper sample preparation using a protease inhibitor cocktail and maintain cold conditions throughout extraction to prevent degradation of the approximately 52 kDa PPARA protein . For gel electrophoresis, a 10% polyacrylamide gel provides optimal resolution for the PPARA protein band. During transfer, use PVDF membranes rather than nitrocellulose for superior protein retention and signal-to-noise ratio with biotin-conjugated antibodies. For blocking, 5% BSA in TBST is preferable to milk-based blockers that can contain endogenous biotin and create background issues . The recommended starting dilution for the biotin-conjugated PPARA antibody is 1:500-1:1000 . For detection, use streptavidin-HRP (1:10,000-1:20,000) followed by enhanced chemiluminescence visualization. Include positive controls such as mouse adipose tissue extract, which shows clear PPARA expression at approximately 52 kDa . Be aware that some non-specific bands may appear, particularly in NIH-3T3 cell lysates, requiring careful interpretation .
For effective immunoprecipitation (IP) with biotin-conjugated PPARA antibody, protocol optimization is essential to maintain antibody functionality while leveraging the biotin conjugation. Begin with freshly prepared cell or tissue lysates using a gentle lysis buffer (typically 25mM Tris HCl pH 7.4, 150mM NaCl, 1% NP-40, 1mM EDTA, 5% glycerol) supplemented with protease inhibitors to preserve PPARA integrity . Pre-clear lysates with protein A/G beads (30 minutes at 4°C) to reduce non-specific binding. For the IP step, avoid directly conjugating biotin-labeled antibodies to streptavidin beads, as this may sterically hinder antigen recognition. Instead, first form antibody-antigen complexes by incubating 2-5 μg of biotin-conjugated PPARA antibody with 500-1000 μg of lysate (overnight at 4°C with gentle rotation) . Then add streptavidin-coated magnetic beads for 1-2 hours to capture the complexes. This sequential approach preserves the antibody's antigen-binding capacity while utilizing the biotin tag for purification. Perform at least 4-5 stringent washes with decreasing salt concentrations to minimize non-specific binding. For elution, gentle non-denaturing conditions or direct boiling in SDS sample buffer can be used, depending on downstream applications . When analyzing PPARA interactions with binding partners like RXRA, consider crosslinking approaches to stabilize transient protein-protein interactions prior to cell lysis.
For optimal immunofluorescence (IF) detection of PPARA using biotin-conjugated antibodies, proper fixation and processing are crucial to preserve both antigenicity and cellular architecture. For cultured cells, 4% paraformaldehyde (15 minutes at room temperature) provides superior epitope preservation compared to methanol fixation, which can denature the nuclear receptor structure . After fixation, a moderate permeabilization with 0.2% Triton X-100 (10 minutes) facilitates antibody access to nuclear PPARA without excessive extraction. For tissue sections, antigen retrieval is essential - use citrate buffer (pH 6.0) with heating to 95°C for 20 minutes followed by slow cooling to room temperature . The recommended antibody dilution range for IF applications is 1:50-1:200, requiring optimization for each specific tissue or cell type . For detection, use streptavidin conjugated to fluorophores like Alexa Fluor 488, 555, or 647 (typically at 1:500-1:1000 dilution). Include DAPI nuclear counterstain to confirm the expected nuclear localization pattern of PPARA. Given PPARA's function as a transcription factor, successful staining should show predominantly nuclear signal, with some cytoplasmic distribution depending on activation state . For multi-labeling experiments, the biotin-conjugated PPARA antibody can be combined with other primary antibodies of different species origin, followed by appropriate species-specific secondary antibodies and streptavidin-fluorophore detection.
Verifying antibody specificity is crucial for generating reliable data with biotin-conjugated PPARA antibodies. Implement a multi-faceted validation approach beginning with positive and negative tissue controls. Liver tissue shows high PPARA expression and serves as an excellent positive control, while tissues known to express minimal PPARA can serve as negative controls . For cellular systems, compare detection in wild-type cells versus PPARA knockout or siRNA-depleted cells. If knockout models are unavailable, competitive blocking experiments provide an alternative - pre-incubate the antibody with 5-10 fold excess of the immunizing peptide before application to samples, which should substantially reduce specific signals . Western blot analysis should reveal a distinct band at approximately 52 kDa corresponding to PPARA, with minimal non-specific bands . For immunofluorescence applications, co-localization studies with another validated PPARA antibody (from a different host species and recognizing a different epitope) can confirm staining patterns. Finally, functional validation through chromatin immunoprecipitation (ChIP) assays demonstrating PPARA binding to known target gene promoters (such as ACOX1) provides compelling evidence of specificity in the context of the protein's biological activity .
High background is a frequent challenge when using biotin-conjugated antibodies due to both technical and biological factors. Endogenous biotin present in many tissues and cells (particularly abundant in liver, kidney, and brain) can directly bind to detection reagents, creating false-positive signals. To address this, implement an endogenous biotin blocking step using a commercial avidin/biotin blocking kit before antibody application . Another common source of background is non-specific binding of the primary antibody. Optimize blocking conditions using 1-3% BSA (preferable to milk, which contains biotin) in TBS-T for 1-2 hours at room temperature. The antibody concentration significantly impacts background - start with the recommended dilution range (1:500-1:1000 for WB, 1:50-1:200 for IF) and adjust as needed . For streptavidin detection reagents, concentrations beyond 1:1000 can increase non-specific binding; titrate carefully and include a streptavidin-only control to assess background. Insufficient washing contributes substantially to background issues - implement 5-6 washes of 5-10 minutes each using 0.1% Tween-20 in appropriate buffer. For persistent background in immunohistochemistry, consider adding 0.1-0.3M NaCl to washing buffers to disrupt weak ionic interactions. Finally, sample over-fixation can create artifactual binding sites - limit fixation to recommended times (15 minutes for PFA fixation of cultured cells) and incorporate appropriate antigen retrieval methods for tissue sections .
Distinguishing between specific PPARA signals and artifacts in Western blotting requires systematic controls and careful interpretation. First, confirm the detected band appears at the expected molecular weight of approximately 52 kDa for PPARA . Include positive control samples such as mouse adipose tissue extract or human liver lysate known to express PPARA . For negative controls, use lysates from PPARA-knockout models or cells treated with PPARA-specific siRNA. Be aware that certain cell lines (e.g., NIH-3T3) may show non-specific bands with some PPARA antibodies . To confirm specificity, perform a peptide competition assay by pre-incubating the antibody with excess immunizing peptide, which should substantially reduce or eliminate specific bands while leaving non-specific bands largely unchanged . When evaluating PPARA levels across different samples, normalize to appropriate housekeeping proteins (β-actin, GAPDH, or preferably lamin B1 for nuclear proteins) and quantify using densitometry software. Post-translational modifications can alter PPARA migration patterns, potentially appearing as multiple bands or slight shifts from the expected 52 kDa - these are not necessarily artifacts but may represent biologically relevant modifications . For biotin-conjugated antibodies specifically, always include a streptavidin-only control lane to identify potential endogenous biotinylated proteins that might be misinterpreted as PPARA signal. Finally, validate key findings with an alternative PPARA antibody (different clone or from a different manufacturer) to confirm the observed pattern is antibody-independent and truly represents PPARA expression .
Chromatin immunoprecipitation using PPARA antibodies allows researchers to investigate direct DNA binding sites and transcriptional regulatory mechanisms. While biotin-conjugated antibodies present certain challenges in ChIP applications due to nuclear biotin content, they can be effectively utilized with proper experimental design. Begin with optimal crosslinking conditions - 1% formaldehyde for 10 minutes at room temperature preserves PPARA-DNA interactions without creating excessive crosslinks that might hinder epitope accessibility . For sonication, optimize conditions to generate DNA fragments between 200-500 bp, which is ideal for resolving specific PPARA binding sites while maintaining sufficient DNA content for downstream analysis. When using biotin-conjugated PPARA antibodies for ChIP, implement a pre-clearing step with unconjugated streptavidin beads to remove endogenous biotinylated nuclear proteins, followed by immunoprecipitation using 3-5 μg of antibody per chromatin sample from approximately 10^6 cells . For elution and reversal of crosslinks, standard ChIP protocols remain applicable. For downstream analysis, qPCR primers targeting known PPARA response elements in genes such as ACOX1, CPT1A, and HMGCS2 serve as positive controls, while primers targeting regions without predicted PPARA binding sites serve as negative controls. ChIP-seq approaches provide genome-wide binding profiles, revealing that PPARA predominantly binds DR1 elements (direct repeats separated by one nucleotide) approximately 70% of the time, with significant co-occupancy with RXR at approximately 80% of binding sites . For functional validation of identified binding sites, reporter gene assays incorporating the putative PPARA response elements can confirm their regulatory potential.
Tissue microarray analysis with PPARA antibodies requires special considerations to ensure accurate, reproducible results across multiple tissue samples. For biotin-conjugated PPARA antibodies specifically, implement strict biotin blocking steps using avidin/biotin blocking kits to minimize background from endogenous biotin, which varies significantly across tissue types . Antigen retrieval methods significantly impact PPARA detection - heat-induced epitope retrieval using citrate buffer (pH 6.0) typically yields optimal results for formalin-fixed, paraffin-embedded (FFPE) tissues. For TMA construction, include both positive control tissues (liver, heart, kidney) and negative control tissues with minimal PPARA expression for reference standards on each array slide . Standardize section thickness (4-5 μm is optimal) and storage conditions to minimize variability. The antibody dilution requires careful optimization - begin with 1:100 dilution for biotin-conjugated PPARA antibodies and adjust based on signal-to-noise ratio across different tissue types . For detection, streptavidin-HRP with DAB chromogen provides good signal stability for long-term storage and digital analysis. In scoring and interpreting TMA results, consider that PPARA demonstrates primarily nuclear staining with variable intensity depending on tissue type and metabolic state. Implement digital image analysis with appropriate nuclear recognition algorithms for quantitative assessment of staining intensity and distribution. For multi-marker studies, sequential multiplexing approaches can be employed to correlate PPARA expression with other metabolic regulators, though careful antibody stripping protocols are essential between rounds to prevent signal carryover .
Flow cytometry with PPARA antibodies enables single-cell resolution analysis of metabolic regulation across heterogeneous cell populations. Though primarily a nuclear receptor, PPARA demonstrates dynamic subcellular localization that correlates with metabolic states, making it valuable for flow cytometric profiling . For intracellular staining protocols, optimal fixation and permeabilization conditions are crucial - 4% paraformaldehyde (15 minutes) followed by permeabilization with 0.1% Triton X-100 or commercial permeabilization buffers designed for nuclear antigens provides consistent results. The biotin-conjugated PPARA antibody can be used at 1:50-1:100 dilution, with detection via streptavidin conjugated to bright fluorophores such as PE, APC, or BV421 . Include appropriate isotype controls and blocking of Fc receptors to minimize non-specific binding. For multi-parameter analysis, combine PPARA staining with surface markers defining specific cell populations and additional metabolic regulators like PGC-1α or CPT1A. This approach can identify metabolically distinct subpopulations within complex samples such as peripheral blood mononuclear cells or liver non-parenchymal cells. For validation, use cells treated with PPARA agonists (e.g., fenofibrate, WY-14,643) versus antagonists to confirm specificity of detected signals . When analyzing primary human samples, consider that PPARA expression in monocytes correlates with metabolic health indicators, making flow cytometric analysis of these accessible cells potentially valuable for translational studies . For data analysis, consider biaxial plotting of PPARA against other metabolic regulators or cell cycle markers to identify correlations between metabolic state and proliferative activity.
The activation state of PPARA significantly influences its detection characteristics with antibodies, requiring careful consideration when interpreting experimental results. PPARA undergoes conformational changes upon ligand binding that can alter epitope accessibility and subcellular localization patterns . Activated PPARA predominantly localizes to the nucleus through interaction with RXR heterodimeric partner, while inactive forms may show more diffuse cytoplasmic distribution. This differential localization affects fractionation efficiency in biochemical assays and staining patterns in microscopy applications. Post-translational modifications including phosphorylation, SUMOylation, and acetylation further modulate antibody recognition - phosphorylation at sites like Ser12, Ser21, and Ser76 can alter antibody binding efficiency by up to 40-60% depending on the epitope location relative to modification sites . When using biotin-conjugated PPARA antibodies specifically, consider that the bulky biotin moiety may differentially affect recognition of various PPARA conformational states. To properly interpret activation-dependent changes, implement parallel detection strategies: (1) subcellular fractionation with Western blotting to quantify nuclear/cytoplasmic distributions, (2) co-immunoprecipitation with RXRA to assess heterodimer formation characteristic of activated states, and (3) ChIP analysis to confirm DNA binding activity . Additionally, include experimental controls with known PPARA activators (fibrates, WY-14,643) and inhibitors to establish baseline detection patterns for different activation states. When quantifying PPARA levels across samples with potentially different activation states, consider that total protein levels may remain unchanged while functional activity varies substantially - complementary activity assays measuring target gene expression provide essential context for accurate interpretation .
Studying PPARA interactions with its binding partners using biotin-conjugated antibodies requires specialized approaches that leverage the biotin tag while addressing potential limitations. Co-immunoprecipitation (co-IP) represents a primary method, with several optimization strategies for biotin-conjugated antibodies. The sequential immunoprecipitation approach is particularly effective - first form PPARA-partner complexes using the biotin-conjugated antibody in solution, then capture these complexes with streptavidin beads . This approach preserves interaction interfaces that might be disrupted by direct antibody immobilization. For detecting transient interactions, implement in situ crosslinking with membrane-permeable crosslinkers like DSP (dithiobis[succinimidyl propionate]) prior to lysis. The biotin tag provides advantages for studying protein complexes through BioID or APEX2 proximity labeling when fused to PPARA, enabling identification of the broader interactome beyond direct binding partners . For confirming specific interactions, reciprocal co-IPs with antibodies against suspected partners (particularly RXRA, the primary heterodimerization partner) provide strong validation. Analytical techniques like AlphaScreen can quantify PPARA-partner interactions in solution using biotin-conjugated PPARA antibody paired with acceptor beads conjugated to partner-specific antibodies, providing quantitative binding parameters . For more complex interaction networks, implement tandem affinity purification approaches, using the biotin tag for initial capture followed by elution and secondary purification via another affinity tag on the partner protein. When analyzing PPARA-RXRA heterodimers specifically, gel shift assays (EMSA) using biotin-conjugated antibodies in supershift experiments can confirm the identity of complex components bound to labeled PPARE (PPAR response element) DNA probes .
Integrating multi-omics with PPARA antibody-based studies creates a comprehensive framework for understanding metabolic regulation networks. This integration requires strategic experimental design and sophisticated data analysis approaches. Begin with parallel sample processing for both antibody-based PPARA profiling and omics analyses (transcriptomics, proteomics, metabolomics) from the same biological specimens to ensure direct correlation capabilities. For transcriptomic integration, ChIP-seq using PPARA antibodies can be directly correlated with RNA-seq data to connect PPARA genomic binding events with resultant transcriptional changes. Implement peak-to-gene association algorithms that consider both proximal promoter binding and distal enhancer interactions through chromatin conformation data . For proteomics integration, immunoprecipitation using biotin-conjugated PPARA antibodies followed by mass spectrometry (IP-MS) identifies the PPARA interactome, which can be mapped against global proteome changes to distinguish direct from indirect effects. Metabolomic data provides functional validation of PPARA activity, particularly focusing on lipid metabolites within peroxisomal β-oxidation and fatty acid metabolism pathways .
| Data Integration Approach | Primary Methods | Key Insights |
|---|---|---|
| ChIP-seq + RNA-seq | Biotin-PPARA antibody ChIP-seq paired with RNA-seq | Identifies direct transcriptional targets versus secondary gene expression changes |
| IP-MS + Proteomics | Biotin-PPARA antibody immunoprecipitation with MS analysis | Maps PPARA protein interaction network and post-translational modifications |
| IF/IHC + Spatial Transcriptomics | Biotin-PPARA antibody tissue staining with spatial RNA sequencing | Correlates tissue-specific PPARA localization with regional gene expression patterns |
| Metabolomics + PPARA Target Validation | Targeted metabolite analysis of PPARA-regulated pathways | Confirms functional consequences of PPARA binding and transcriptional regulation |
For data integration, implement multivariate statistical approaches including principal component analysis and partial least squares discriminant analysis to identify correlated patterns across datasets. Network analysis algorithms can construct integrated regulatory networks with PPARA as a central node, connecting direct binding targets (from ChIP-seq), protein interactions (from IP-MS), and metabolic consequences (from metabolomics) . For translational relevance, correlate these integrated networks with clinical parameters in patient-derived samples, potentially using circulating monocytes as accessible surrogates for PPARA activity assessment . This multi-omics approach reveals not only the direct molecular actions of PPARA but also the broader systemic consequences of its activity across metabolic tissues and pathways.
Emerging technologies are significantly expanding the research applications of PPARA antibodies, including biotin-conjugated variants. Single-cell technologies represent a major advancement - single-cell CUT&Tag and CUT&RUN using biotin-conjugated PPARA antibodies enable mapping of PPARA genomic binding at single-cell resolution, revealing cell-specific regulatory patterns within heterogeneous tissues like liver and adipose . Spatial transcriptomics paired with PPARA immunofluorescence creates spatial-molecular maps connecting PPARA localization with regional gene expression patterns. Microfluidic antibody-based proteomic platforms are enabling high-throughput analysis of PPARA and interacting proteins across numerous samples simultaneously. CRISPR-based approaches for tagging endogenous PPARA with split-biotin ligase systems permit dynamic monitoring of PPARA interactions in living cells without exogenous antibody application . For therapeutic applications, antibody-based targeted delivery systems using anti-PPARA antibodies conjugated to nanoparticles can deliver PPARA modulators specifically to metabolically active tissues. High-content imaging platforms with automated image analysis algorithms are enhancing the quantitative assessment of PPARA subcellular distribution and co-localization patterns in response to metabolic perturbations . The integration of machine learning approaches with antibody-based detection is improving signal-to-noise discrimination and enabling more sensitive detection of subtle changes in PPARA expression or localization. These technological advances are collectively transforming PPARA antibody applications from simple detection tools to sophisticated probes for mapping complex regulatory networks in normal physiology and metabolic disorders.
Optimizing cross-species PPARA studies requires careful consideration of evolutionary conservation, antibody cross-reactivity, and species-specific metabolic differences. PPARA demonstrates approximately 91% amino acid sequence identity between human and mouse, and 88% between human and rat, with higher conservation in functional domains like the DNA-binding domain and lower conservation in regulatory regions . When selecting antibodies, prioritize those validated across target species - several commercial PPARA antibodies, including some biotin-conjugated variants, have confirmed reactivity with human, mouse, and rat samples . Epitope mapping is crucial - antibodies targeting highly conserved regions of PPARA (particularly the DNA-binding domain) tend to show better cross-species reactivity compared to those targeting variable N-terminal regions. For Western blotting applications, optimize loading amounts for different species, as baseline PPARA expression varies - mouse liver typically shows 2-3 fold higher PPARA expression compared to human liver samples . For immunohistochemistry applications, species-specific optimization of antigen retrieval conditions is essential - rodent tissues often require less aggressive retrieval compared to human FFPE samples.
When designing functional studies, consider species-specific PPARA activation responses - rodents demonstrate more pronounced hepatic peroxisome proliferation in response to PPARA agonists compared to humans. Control experiments should include species-specific positive controls (e.g., fenofibrate-treated tissue for each species) and negative controls (PPARA-null tissues when available). For transcriptional studies, recognize that while core PPARA target genes (ACOX1, CPT1A) are conserved across species, the broader transcriptional response differs substantially - in mice, approximately 40-50% more genes respond to PPARA activation compared to humans . When translating findings between models, prioritize consistency in metabolic state (fed/fasted) and circadian timing, as these factors significantly impact PPARA expression and activity across all species studied .
Validating novel PPARA antibodies, particularly biotin-conjugated variants, for specialized research applications requires a comprehensive, multi-parameter assessment strategy. Begin with fundamental validation steps: Western blotting should reveal a distinct band at approximately 52 kDa, with reduced or absent signal in PPARA-knockout or knockdown samples . Peptide competition assays provide essential specificity confirmation - pre-incubation with the immunizing peptide should abolish specific binding. For biotin-conjugated antibodies specifically, confirm that the conjugation process hasn't compromised antigen recognition by comparing detection patterns with the unconjugated antibody version . Application-specific validation is crucial - for ChIP applications, validate enrichment at established PPARA binding sites (ACOX1, CPT1A promoters) with minimal signal at negative control regions. For proximity ligation assays, confirm PPARA-RXRA interaction signals that increase upon treatment with PPARA agonists. For super-resolution microscopy applications, verify that the biotin-conjugated antibody provides sufficient signal density and specificity at the nanoscale level .