PPARA Antibody,FITC conjugated

Shipped with Ice Packs
In Stock

Description

Definition and Target Biology

PPARA (peroxisome proliferator-activated receptor alpha) is a ligand-activated transcription factor regulating fatty acid oxidation, lipid transport, and anti-inflammatory pathways . The FITC-conjugated PPARA antibody combines specificity for PPARA with fluorescein isothiocyanate (FITC), enabling fluorescent detection in assays like flow cytometry or immunofluorescence .

Key Uses

  • ELISA: Direct quantification of PPARA in biological samples using fluorescent readouts .

  • Hypothetical Adaptations:

    • Western Blot (WB): Unconjugated PPARA antibodies show WB efficacy at 1:500–1:1,000 dilutions , suggesting FITC versions could be optimized similarly.

    • Immunofluorescence (IF): Demonstrated in unconjugated formats for cellular localization studies .

Performance Considerations

  • Conjugation Efficiency: Site-specific FITC conjugation (e.g., via engineered residues) enhances consistency and activity, as shown in CAR-T cell studies using analogous FITC-tagged antibodies .

  • Cross-Reactivity: Broad species reactivity (human, mouse, rat) ensures utility in diverse models .

FITC vs. Other Conjugates

ConjugateProduct CodeApplicationSensitivity
FITCCSB-PA018421LC01HUELISAHigh (fluorometric detection)
HRPCSB-PA018421LB01HUELISAHigh (colorimetric detection)
BiotinCSB-PA018421LD01HUELISAMedium (requires streptavidin step)

Research Findings and Validation

  • Functional Studies: PPARA modulates lipid metabolism by activating β-oxidation pathways and suppresses inflammation via NF-κB inhibition . The FITC conjugate enables real-time tracking of PPARA dynamics in these processes.

  • Technical Validation:

    • CAR-T Cell Studies: Site-specific FITC conjugation (e.g., at engineered p-azidophenylalanine residues) improves pseudoimmunological synapse formation, enhancing assay precision .

    • Dose Dependency: FITC-conjugated switches show activity proportional to concentration, allowing precise control in experimental systems .

Optimization Guidelines

  • Titration: Mandatory for all applications due to variability in sample preparation and equipment .

  • Interference Checks: Excess free FITC or unlabeled antibodies can inhibit detection, requiring rigorous wash steps .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receiving them. Delivery times may vary depending on the shipping method and destination. For specific delivery times, please contact your local distributor.
Synonyms
hPPAR antibody; MGC2237 antibody; MGC2452 antibody; NR1C1 antibody; Nuclear receptor subfamily 1 group C member 1 antibody; OTTHUMP00000197740 antibody; OTTHUMP00000197741 antibody; Peroxisome proliferative activated receptor alpha antibody; Peroxisome proliferator activated receptor alpha antibody; Peroxisome proliferator-activated receptor alpha antibody; PPAR antibody; PPAR-alpha antibody; ppara antibody; PPARA_HUMAN antibody; PPARalpha antibody
Target Names
Uniprot No.

Target Background

Function
PPARα (Peroxisome Proliferator-Activated Receptor Alpha) is a ligand-activated transcription factor that plays a pivotal role in regulating lipid metabolism. It is activated by the endogenous ligand 1-palmitoyl-2-oleoyl-sn-glycerol-3-phosphocholine (16:0/18:1-GPC). Additionally, PPARα is activated by oleoylethanolamide, a naturally occurring lipid that regulates satiety. PPARα serves as a receptor for peroxisome proliferators, such as hypolipidemic drugs and fatty acids. It regulates the peroxisomal β-oxidation pathway of fatty acids and acts as a transcription activator for the ACOX1 and P450 genes. Transactivation activity requires heterodimerization with RXRA and is antagonized by NR2C2. PPARα may be essential for the propagation of clock information to metabolic pathways regulated by PER2.
Gene References Into Functions
  1. Ubiquitination modification, through the coordinated action of PAQR3 with HUWE1, plays a critical role in regulating the activity of hepatic PPARα in response to starvation. PMID: 29331071
  2. Circulated eosinophilic expression of PPARα protein is reduced in metabolic syndrome. PMID: 29699951
  3. TNFα differentially regulates the levels of PPARα, LXRα, and LXRβ binding to the apoA-I gene promoter in THP-1 cells. These findings suggest a novel tissue-specific mechanism for TNFα-mediated regulation of the apoA-I gene in monocytes and macrophages, indicating that endogenous ApoA-I might be positively regulated in macrophages during inflammation. PMID: 29442267
  4. MAR1 ameliorates LPS-induced atherosclerotic reactions via PPARα-mediated suppression of inflammation and ER stress. PMID: 29971543
  5. Data suggest that, in hepatocytes, MIRN34A plays roles in regulating mitochondrial remodeling and lipid metabolism, including the development/prevention of non-alcoholic fatty liver disease; MIRN34A appears to act via AMPK/PPARα signal transduction. (MIRN34A = microRNA 34a; AMPK = AMP-activated protein kinase; PPARα = peroxisome proliferator activated receptor alpha) PMID: 29197627
  6. miR-214 overexpression inhibits glioma cell growth in vitro and in vivo by inducing cell cycle arrest in G0/G1. Collectively, these data uncover a novel role for a PPARα-miR-214-E2F2 pathway in controlling glioma cell proliferation. PMID: 29862267
  7. Improvements in metabolic and neurodegenerative diseases are often attributed to anti-inflammatory effects of PPAR activation. (Review) PMID: 29799467
  8. circRNA_0046366, which demonstrated expression loss in HepG2-based hepatocellular steatosis, exerts an antagonistic effect on miR-34a activity. miR-34a inactivation abrogates its inhibitory role against PPARα. PMID: 29391755
  9. We first reported that the FOMX1 pathway is the most upregulated and the PPARα pathway is the most downregulated pathway in Triple Negative Breast Cancers (TNBCs). These two pathways could be simultaneously targeted in further studies. Additionally, the pathway classifier we performed in this study provided insight into the TNBC heterogeneity. PMID: 29301506
  10. Polymorphism of PPARA is associated with late onset of type 2 diabetes mellitus. PMID: 28292576
  11. results demonstrated that OEA exerts anti-inflammatory effects by enhancing PPARα signaling, inhibiting the TLR4-mediated NF-κB signaling pathway, and interfering with the ERK1/2-dependent signaling cascade (TLR4/ERK1/2/AP-1/STAT3), suggesting that OEA may be a therapeutic agent for inflammatory diseases. PMID: 27721381
  12. data suggested that miR-19a negatively controlled the autophagy of hepatocytes attenuated in D-GalN/LPS-stimulated hepatocytes via regulating NBR2 and AMPK/PPARα signaling. PMID: 28586153
  13. The minor allele of rs1800206 and rs1805192 from PPAR A and PPAR G and its interaction were associated with increased Breast Cancer risk. PMID: 28669518
  14. High concentrations of DINCH urinary metabolites activate human PPAR-α. PMID: 29421333
  15. PPARα is overexpressed in primary glioblastoma. PMID: 27926792
  16. these results suggest that the E2F1/miR19a/PPARα feedback loop is critical for glioma progression. PMID: 27835866
  17. Data conclude that the ER-stress mediated reduction in apoA-I transcription could be partly mediated via the inhibition of PPARα mRNA expression and activity. In addition, BET inhibition increased apoA-I transcription, even if PPARα production and activity were decreased. Both BET inhibition and PPARα activation ameliorate the apoA-I lowering effect of ER-stress and are therefore interesting targets to elev... PMID: 28012209
  18. Results demonstrated that PPARα directly inhibited Glut1 mRNA expression resulting in influx of glucose in cancer cells. PMID: 27918085
  19. PPARα and LXRα may be mediators by which omega3PUFA attenuate bile acid-induced hepatocellular injury. PMID: 26756785
  20. Our results support an important association between rs1800206 minor allele of PPAR alpha and diabetic retinopathy, and the interaction analysis also shown a combined effect of Leu162 allele-abdominal obesity interaction on diabetic retinopathy. PMID: 26671228
  21. Taken together, our data suggest that eupatilin inhibits TNFα-induced MMP-2/-9 expression by suppressing NF-κB and MAPKAP-1 pathways via PPARα. Our findings suggest the usefulness of eupatilin for preventing skin aging. PMID: 28899779
  22. Hepatic PARP1 activation inhibits FAO pathway upregulation through poly(ADP-ribosyl)ation of PPARα, worsening hepatic steatosis and inflammatory responses associated with overnutrition. PMID: 27979751
  23. Aleglitazar protects cardiomyocytes against hyperglycaemia-induced apoptosis by combined activation of both peroxisome proliferator-activated receptor-alpha and peroxisome proliferator-activated receptor-gamma. PMID: 28111985
  24. Study reports a molecular mechanism by which glucocorticoid-induced PPARα expression negatively affects the activity of PPARγ and downregulates BCO1 gene expression. Results explicate novel aspects of local glucocorticoid:retinoid interactions that may contribute to alveolar tissue remodeling in chronic lung diseases that affect children and, possibly, adults. PMID: 28732066
  25. Interference with PLIN2 and PPARα resulted in major alterations in gene expression, especially affecting lipid, glucose, and purine metabolism. PMID: 27308945
  26. PPARα and FXR function coordinately to integrate liver energy balance. PMID: 28287408
  27. This study showed that oleoylethanolamine and palmitoylethanolamine have endogenous roles and potential therapeutic applications in conditions of intestinal hyperpermeability and inflammation. PMID: 27623929
  28. An association was found with PPARα polymorphism and patients with nicotine dependency and schizophrenia. PMID: 27624431
  29. PPAR agonists have shown to have anti proliferative effect in squamous cell carcinoma of the head and neck. PMID: 27896820
  30. Results show that PPARα is downregulated and SREBP-1c is upregulated in steatosis L-02 cells. These changes increase lipid synthesis and reduce lipid disposal, which ultimately lead to hepatic steatosis. PMID: 27270405
  31. Perfluoroalkyl acids addition to activating PPARα as a primary target, also have the potential to activate CAR, PPARγ, and ERα as well as suppress STAT5B. PMID: 28558994
  32. the metabolic events, controlled by PPARs, occurring during neuronal precursor differentiation, the glucose and lipid metabolism was investigated. PMID: 27860527
  33. The CYP2E1-PPARα axis may play a role in ethanol-induced neurotoxicity via the alteration of the genes related with synaptic function. PMID: 28385499
  34. Studies indicate that natural dietary compounds, including nutrients and phytochemicals, are Peroxisome proliferator-activated receptor alpha (PPARα) ligands or modulators. PMID: 27863018
  35. Genome-wide comparison of the inducible transcriptomes of nuclear receptors CAR, PXR and PPARα in primary human hepatocytes has been presented. PMID: 26994748
  36. Hepatitis B virus increases the expression of alpha-mannosidases both in vitro and in vivo via activation of the PPARα pathway by its envelope protein. PMID: 27920474
  37. These observations candidate PPARs as new biomarkers of follicle competence opening new hypotheses on controlled ovarian stimulation effects on ovarian physiology. PMID: 26332656
  38. PPARα activation plays defensive and compensative roles by reducing cellular toxicity associated with fatty acids and sulfuric acid. PMID: 27644403
  39. PPARα/γ agonist, elafibranor resolves nonalcoholic steatohepatitis without worsening fibrosis. PMID: 26874076
  40. The effects of fenofibrate on nonalcoholic fatty liver disease in the context of PPAR-α activation was studied. PMID: 27930988
  41. PPARA polymorphism is associated with the risk of coronary heart disease. PMID: 27512842
  42. Telmisartan improved the hyperglycemia-induced cardiac fibrosis through the PPARδ/STAT3 pathway. PMID: 27519769
  43. A modest relationship was found between PPARα and AIP expression, both being significantly higher in the presence of pre-operative somatostatin analogues in somatotropinoma patients. PMID: 26872613
  44. Fenofibrate inhibited atrial metabolic remodeling in atrial fibrillation (AF) by regulating the PPAR-α/sirtuin 1/PGC-1α pathway indicating a novel therapeutic strategy for AF. PMID: 26787506
  45. PPAR delta + 294TT genotype frequency in the Chinese Han population was higher than in the Chinese Uyghur population and may affect the risk of ischemic stroke. PMID: 26814631
  46. PPARα functions as an E3 ubiquitin ligase to induce Bcl2 ubiquitination and degradation, leading to increased cancer cell sensitivity in response to chemotherapy drugs. PMID: 26556865
  47. There was no statistically significant difference in the distribution of PPARα Leu162Val polymorphism between the ischemic stroke patients and controls in the Han ethnic group. PMID: 26671025
  48. results support an important association between rs1800206 minor allele (V) of PPAR alpha and lower CRP level; the interaction analysis showed a combined effect between rs1800206 and rs135539 on the lower CRP level. PMID: 26497374
  49. PPAR-γ and PTEN expressions are related to the clinical parameters and prognosis of renal cell carcinoma. PMID: 26722456
  50. Describe a renoprotective role of fenofibrate in albumin bound fatty acid associated tubular toxicity, involving the transcriptional activation of PPARα and suppression of NF-kB. PMID: 26617775

Show More

Hide All

Database Links

HGNC: 9232

OMIM: 170998

KEGG: hsa:5465

STRING: 9606.ENSP00000262735

UniGene: Hs.103110

Protein Families
Nuclear hormone receptor family, NR1 subfamily
Subcellular Location
Nucleus.
Tissue Specificity
Skeletal muscle, liver, heart and kidney. Expressed in monocytes.

Q&A

What is PPARA and what cellular functions does it regulate?

PPARA (peroxisome proliferator-activated receptor alpha) is a ligand-activated transcription factor that serves as a key regulator of lipid metabolism in cells. It is activated by several ligands including the endogenous 1-palmitoyl-2-oleoyl-sn-glycerol-3-phosphocholine (16:0/18:1-GPC) and oleylethanolamide, a naturally occurring lipid that regulates satiety. PPARA functions primarily as a receptor for peroxisome proliferators including hypolipidemic drugs and fatty acids .

PPARA's principal function involves regulating the peroxisomal beta-oxidation pathway of fatty acids. At the molecular level, it acts as a transcription activator for genes such as ACOX1 and cytochrome P450, with transactivation activity requiring heterodimerization with RXRA (Retinoid X Receptor Alpha) . This activity is antagonized by NR2C2 (Nuclear Receptor Subfamily 2 Group C Member 2). Additionally, PPARA may play a role in transmitting circadian clock information to metabolic pathways regulated by PER2 (Period Circadian Regulator 2) .

What applications are suitable for FITC-conjugated PPARA antibodies?

FITC-conjugated PPARA antibodies are versatile reagents suitable for multiple research applications, particularly those requiring fluorescent detection. Based on manufacturer recommendations, the primary applications include:

ApplicationRecommended DilutionNotes
Flow Cytometry1:20-1:100Optimal for intracellular detection
Immunofluorescence1:50-1:200Works well with paraffin-embedded samples (IHC-P)

These antibodies have been validated for reactivity with human, mouse, and rat samples . When selecting applications, researchers should consider that FITC has an excitation maximum around 495 nm and emission maximum around 519 nm, making it compatible with standard FITC filter sets on most fluorescence microscopes and flow cytometers.

How can I validate the specificity of FITC-conjugated PPARA antibodies?

Validating antibody specificity is crucial for ensuring reliable experimental results. For FITC-conjugated PPARA antibodies, several approaches are recommended:

  • Knockout/Knockdown Validation: Use PPARA knockout or knockdown samples as negative controls. Publications using knockout validation are available and can serve as reference points for expected results .

  • Western Blot Analysis: Though not the primary application for FITC-conjugated antibodies, unconjugated versions of the same antibody clone can be used in Western blot to confirm that the antibody detects a protein of the expected molecular weight (approximately 52 kDa for PPARA) .

  • Competition Assays: Pre-incubate the antibody with recombinant PPARA protein before staining to confirm that the signal is abolished when the antibody's binding sites are occupied.

  • Multi-antibody Verification: Compare staining patterns with other validated PPARA antibodies targeting different epitopes to confirm consistent localization patterns.

  • Positive Control Samples: Use cell lines known to express PPARA, such as C2C12 or U-937 cells, which have been confirmed to express detectable levels of PPARA in previous studies .

What are the optimal storage conditions for maintaining FITC-conjugated antibody activity?

To maintain optimal activity of FITC-conjugated PPARA antibodies, proper storage conditions are essential. The following guidelines should be followed:

What troubleshooting approaches should I consider for weak FITC signals?

When encountering weak fluorescent signals with FITC-conjugated PPARA antibodies, consider the following troubleshooting approaches:

  • Optimize Antibody Concentration: Titrate the antibody concentration. The recommended dilution ranges (1:20-1:100 for flow cytometry, 1:50-1:200 for immunofluorescence) should be used as starting points for optimization .

  • Increase Exposure Time: For microscopy applications, longer exposure times may help detect weak signals, though this should be balanced against increasing background autofluorescence.

  • Enhance Permeabilization: Since PPARA is an intracellular nuclear receptor, ensure adequate cell permeabilization to allow antibody access to the target.

  • Evaluate Fixation Methods: Different fixation protocols can affect epitope accessibility. Consider testing both paraformaldehyde and methanol-based fixation methods.

  • Check for Photobleaching: FITC is susceptible to photobleaching; minimize sample exposure to light during processing and use anti-fade mounting media for microscopy applications.

  • Verify Sample Quality: Ensure that your samples are freshly prepared and properly processed to maintain PPARA expression levels.

  • Consider Signal Amplification: For very low abundance targets, consider using a secondary amplification system compatible with FITC detection.

How does site-specific conjugation of FITC to PPARA antibodies improve experimental outcomes?

Site-specific conjugation of fluorophores like FITC to antibodies offers significant advantages over traditional chemical conjugation methods, particularly for critical applications requiring precise quantification and optimal binding activity. Recent advances using CRISPR/Cas9 genomic editing to introduce specific conjugation sites in antibody-producing hybridoma cell lines have revolutionized this field .

Site-specific conjugation methods can produce antibodies with almost doubled specific targeting compared to chemically conjugated antibodies. In mouse models, site-specifically conjugated antibodies showed substantially improved target tissue accumulation (particularly in the lung) with concomitant reduction in non-specific uptake in the liver and spleen . This improved targeting specificity occurs because:

  • Preservation of Antigen-Binding Regions: Site-specific methods ensure that conjugation does not occur within or near the complementarity-determining regions (CDRs), preserving the antibody's affinity and specificity.

  • Controlled Orientation: By attaching FITC at defined locations, the antibody maintains optimal orientation on the target, minimizing steric hindrance that can impair binding.

  • Batch-to-Batch Consistency: Site-specific conjugation produces more homogeneous products with consistent fluorophore-to-antibody ratios, enhancing experimental reproducibility.

  • Reduced Off-Target Binding: The more controlled nature of site-specific conjugation minimizes alterations to the antibody's surface properties that can contribute to non-specific binding.

What enzymatic approaches can be used for site-specific conjugation of FITC to PPARA antibodies?

Sortase-mediated conjugation represents an elegant enzymatic approach for site-specific modification of antibodies, including the attachment of FITC. This method offers several advantages for researchers working with PPARA antibodies:

  • Mechanism of Action: Sortase A, a bacterial transpeptidase, recognizes the LPETGG motif (sortase tag) and cleaves between threonine and glycine, then forms a new peptide bond with oligoglycine-containing molecules such as GGGK-FITC .

  • Optimization Strategy: The efficiency of sortase-mediated conjugation can be modulated by adjusting several parameters:

ParameterOptimization RangeEffect
mAb:GGGK-FITC Ratio1:2 to 1:1014.1% to 17.6% conjugation efficiency
Sortase Concentration50-200 μMHigher concentrations increase reaction speed
Reaction Time1-16 hoursLonger times increase yield
Temperature25-37°CHigher temperatures accelerate reaction
Calcium Concentration5-10 mMRequired for sortase activity
  • Conjugation Analysis: Successful conjugation can be verified using fluorescence scanning of SDS-PAGE gels and quantified by HPLC with fluorescence detection. These methods allow precise determination of the conjugation efficiency .

  • Payload Enhancement: While conjugation efficiencies of approximately 14-18% may seem modest, multiple FITC molecules can be incorporated onto a single GGG-peptide to enhance the fluorophore-to-antibody ratio without additional conjugation sites .

How can CRISPR/Cas9 genomic editing be utilized to develop hybridoma cell lines producing site-specifically modifiable PPARA antibodies?

CRISPR/Cas9 genomic editing offers a revolutionary approach to engineer hybridoma cell lines that produce antibodies with built-in conjugation sites. This strategy represents a significant advance over traditional methods requiring antibody sequencing and recombinant expression. The process involves:

  • Tag Insertion Strategy: CRISPR/Cas9 can be used to incorporate specialized tags (e.g., sortase tag LPETGG) at the C-terminal end of the CH3 domain of the heavy chain in hybridoma cells . This location is optimal as it:

    • Is distant from the antigen-binding regions

    • Allows accessibility for enzymatic conjugation

    • Minimizes potential interference with antibody function

  • Verification of Successful Editing: Successfully edited hybridoma clones can be verified through:

    • PCR screening and sequencing of the modified genomic region

    • SDS-PAGE analysis showing the expected size shift of the heavy chain

    • Western blot detection of inserted tags (e.g., FLAG tag) under reducing conditions

    • Functional testing of antibody binding to confirm target specificity is maintained

  • Advantages Over Traditional Methods: This approach eliminates several expensive and time-consuming steps:

    • No need to sequence variable regions

    • Avoids cloning into producer cell lines

    • Maintains the original glycosylation pattern and post-translational modifications

    • Provides consistent site-specific conjugation capacity for all antibody molecules

  • Scalability: The engineered hybridoma cells can be expanded to produce larger quantities of modifiable antibodies using standard hybridoma culture techniques.

What strategies minimize background when using FITC-conjugated PPARA antibodies in lipid-rich tissues?

Detecting PPARA in lipid-rich tissues presents unique challenges due to high autofluorescence and potential non-specific binding to lipids. Advanced strategies to minimize background include:

  • Tissue Processing Optimization:

    • Remove lipids using delipidation protocols with careful temperature control to preserve epitopes

    • Employ antigen retrieval methods optimized for nuclear receptors

    • Use thinner tissue sections (4-5 μm) to reduce autofluorescence

  • Signal Enhancement and Background Reduction:

    • Apply Sudan Black B (0.1-0.3%) treatment for 10-20 minutes to quench lipofuscin autofluorescence

    • Use specialized mounting media containing anti-fading agents to preserve FITC signal while reducing background

    • Consider tyramide signal amplification (TSA) to enhance specific signals relative to background

  • Optimized Controls:

    • Include PPARA knockout tissue sections as negative controls

    • Use competing unlabeled antibodies to confirm specificity

    • Employ isotype-matched FITC-conjugated control antibodies to identify non-specific binding

  • Advanced Imaging Techniques:

    • Utilize spectral unmixing to separate FITC signal from autofluorescence

    • Implement time-gated detection to exploit the longer fluorescence lifetime of FITC compared to autofluorescence

    • Consider confocal microscopy with precisely adjusted pinhole settings to reduce out-of-focus fluorescence

How can multiplexed imaging be performed using FITC-conjugated PPARA antibodies alongside other markers?

Multiplexed imaging allows simultaneous visualization of PPARA alongside other cellular components to understand its interactions and contextualize its function. Advanced multiplexing with FITC-conjugated PPARA antibodies can be achieved through:

  • Fluorophore Selection for Multiplexing:

    • Pair FITC (excitation: 495 nm, emission: 519 nm) with fluorophores having minimal spectral overlap

    • Recommended combinations include:

      • FITC + Texas Red/Cy3 + Cy5/Alexa 647

      • FITC + TRITC + Cy5 + DAPI

  • Sequential Staining Protocols:

    • For co-localization with other nuclear receptors (e.g., RXRA, PPARA's dimerization partner):

      • Apply FITC-conjugated PPARA antibody first

      • Block remaining binding sites

      • Apply differently-labeled antibodies against other targets

    • Use zenon labeling technology for same-species antibodies to prevent cross-reactivity

  • Staining Controls for Multiplexed Imaging:

    • Single-color controls to establish proper compensation settings

    • Fluorescence-minus-one (FMO) controls to identify spillover

    • Cross-adsorbed secondary antibodies (if using indirect detection for other targets) to prevent cross-reactivity

  • Advanced Analysis Methods:

    • Use co-localization analysis software to quantify spatial relationships between PPARA and other proteins

    • Employ machine learning algorithms for automated identification of co-expression patterns

    • Consider proximity ligation assays to verify protein-protein interactions at the sub-cellular level

How does the cellular localization of PPARA change under different metabolic conditions, and how can this be monitored with FITC-conjugated antibodies?

PPARA localization and expression changes in response to various metabolic states and ligand activation. FITC-conjugated PPARA antibodies can be used to monitor these dynamic changes through:

  • Monitoring Nuclear Translocation:

    • PPARA primarily functions as a nuclear transcription factor, but its distribution between cytoplasm and nucleus can shift under different conditions

    • Use confocal microscopy with FITC-conjugated PPARA antibodies to quantify nuclear:cytoplasmic ratio changes following treatment with:

      • Natural ligands (fatty acids, 16:0/18:1-GPC)

      • Synthetic agonists (fibrates)

      • Fasting conditions or high-fat feeding

  • Quantitative Analysis Methods:

    • Implement automated image analysis to measure:

      • Mean nuclear FITC intensity

      • Nuclear area occupied by PPARA

      • Co-localization with other nuclear factors like RXRA

    • Use flow cytometry with appropriate permeabilization to quantify total cellular PPARA levels across large cell populations

  • Time-course Experiments:

    • Design pulse-chase experiments to track PPARA dynamics following stimulation

    • Consider live-cell imaging approaches using cell-permeable FITC-conjugated nanobodies against PPARA

    • Correlate localization changes with downstream gene activation (ACOX1, P450 genes)

  • Tissue-specific Differences:

    • Compare PPARA localization patterns across tissues with different metabolic profiles (liver, heart, skeletal muscle)

    • Monitor changes during developmental stages or disease progression

    • Correlate with tissue-specific expression of co-factors that influence PPARA activity

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.