PTGS2 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

Role of PTGS2 in Cancer

PTGS2 is a well-established oncogene in colorectal cancer (CRC), where its upregulation correlates with tumor progression and poor prognosis . Studies using PTGS2 inhibitors or knockdown (via CRISPR/Cas9) have demonstrated reduced cell proliferation, migration, and metastasis in CRC and melanoma models .

Use of the FITC-Conjugated Antibody

The FITC-conjugated PTGS2 antibody enables fluorescence-based detection of PTGS2 expression in cancer cells and tissues. For example:

  • In melanoma studies, it could localize PTGS2 to tumor cells, aiding in the assessment of therapeutic responses .

  • In CRC research, it facilitates co-localization studies with markers of tumor-associated macrophages (e.g., CD68, CD163) .

Published Validation

  • Western Blot: Detects a ~68 kDa band in lysates of PTGS2-expressing cells (e.g., U-87 glioblastoma cells) .

  • Immunofluorescence: Demonstrates nuclear and cytoplasmic staining in HepG2 cells (1:50 dilution) .

  • IHC: Used in human breast cancer tissues with antigen retrieval (TE buffer pH 9.0) .

Comparison with Other PTGS2 Antibodies

AntibodyConjugateReactivityApplicationsCitations
Abbexa #abx104042FITCMouseWB, IHC, IF/ICC
Proteintech #12375-1-APUnconjugatedHuman, Mouse, RatWB, IHC, IF, ELISA
Cusabio #CSB-RA920283A0HUUnconjugatedHumanWB, IF

Research Implications

The FITC-conjugated PTGS2 antibody is particularly useful in studies requiring spatial resolution of PTGS2 expression:

  • Cancer Biology: Investigates PTGS2-driven tumor angiogenesis and immune evasion .

  • Therapeutic Development: Validates PTGS2 inhibitors (e.g., celecoxib) in preclinical models .

  • Immunotherapy: Monitors PTGS2 expression in the context of checkpoint inhibitor resistance .

References

  1. Antibodies-Online. (2017). PTGS2 Antibody (ABIN672471).

  2. Proteintech. (2025). COX2/ Cyclooxygenase 2/ PTGS2 antibody (12375-1-AP).

  3. Liu et al. (2024). Scientific Reports. RUNX1-induced upregulation of PTGS2 enhances CRC cell growth.

  4. Abbexa Ltd. (2018). Prostaglandin G/H Synthase 2 / COX-2 (PTGS2) Antibody - FITC.

  5. Carvalho et al. (2020). BMC Cancer. Glycosylated PTGS2 in colorectal cancer.

  6. Botti et al. (2019). Cancers. Knockdown of PTGS2 in melanoma.

  7. Cusabio. (2025). PTGS2 Antibody (CSB-RA920283A0HU).

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your orders. Delivery time may vary depending on the purchase method or location. Please consult your local distributor for specific delivery times.
Synonyms
COX 2 antibody; COX-2 antibody; COX2 antibody; Cyclooxygenase 2 antibody; Cyclooxygenase 2b antibody; Cyclooxygenase antibody; Cyclooxygenase-2 antibody; Cyclooxygenase2 antibody; EC 1.14.99.1 antibody; fj02a10 antibody; Glucocorticoid-regulated inflammatory cyclooxygenase antibody; Glucocorticoid-regulated inflammatory Prostaglandin G/H synthase antibody; GRIPGHS antibody; hCox 2 antibody; Macrophage activation-associated marker protein P71/73 antibody; OTTHUMP00000033524 antibody; PES-2 antibody; PGG/HS antibody; PGH synthase 2 antibody; PGH2_HUMAN antibody; PGHS 2 antibody; PGHS-2 antibody; PGHS2 antibody; PHS 2 antibody; PHS II antibody; PHS2 antibody; Prostaglandin endoperoxide synthase 2 (prostaglandin G/H synthase and cyclooxygenase) antibody; Prostaglandin endoperoxide synthase 2 antibody; Prostaglandin G/H synthase 2 antibody; Prostaglandin G/H synthase 2 precursor antibody; Prostaglandin G/H synthase and cyclooxygenase antibody; Prostaglandin G/H synthase antibody; Prostaglandin H2 synthase 2 antibody; prostaglandin-endoperoxide synthase 2 (prostaglandin G/H synthase and cyclooxygenase) antibody; Prostaglandin-endoperoxide synthase 2 antibody; PTGS2 antibody; ptgs2a antibody; TIS10 antibody; TIS10 protein antibody; unp1239 antibody; wu:fj02a10 antibody
Target Names
Uniprot No.

Target Background

Function
PTGS2, also known as COX-2, is an enzyme that plays a crucial role in the biosynthesis of prostanoids, a class of lipid mediators with diverse functions in inflammation, pain, and other physiological processes. Prostanoids are derived from arachidonic acid (AA) and related fatty acids through a series of enzymatic reactions. COX-2 catalyzes the first two steps in the prostanoid biosynthesis pathway, converting AA into prostaglandin G2 (PGG2) and then to prostaglandin H2 (PGH2). PGH2 serves as the precursor for various prostanoids, including prostaglandins and thromboxanes. The COX-2 activity is initiated by the abstraction of hydrogen at carbon 13 of AA, followed by the insertion of molecular oxygen to form the endoperoxide bridge that defines prostaglandins. The insertion of a second molecule of oxygen generates PGG2, which is then reduced to PGH2 by a two-electron transfer. COX-2 also catalyzes the biosynthesis of prostanoids from dihomo-gamma-linoleate (DGLA) and eicosapentaenoate (EPA), generating the precursors for 1- and 3-series prostaglandins, respectively. In an alternative pathway, COX-2 converts 2-arachidonoyl lysophospholipids to prostanoid lysophospholipids, which are then hydrolyzed to release free prostanoids. COX-2 also metabolizes 2-arachidonoyl glycerol, yielding the glyceryl ester of PGH2, a process potentially involved in pain responses. In addition to its prostanoid biosynthesis activities, COX-2 also exhibits lipoxygenase-type activity, generating lipid mediators from n-3 and n-6 polyunsaturated fatty acids (PUFAs) via a different pathway. This activity involves the oxygenation of PUFAs to hydroperoxy compounds followed by their reduction to corresponding alcohols. COX-2 plays a crucial role in the production of resolvins, specialized lipid mediators involved in the resolution of inflammation. For instance, COX-2 converts docosahexaenoate (DHA) to 17R-HDHA, a precursor for D-series resolvins (RvDs). As a component of the biosynthetic pathway for E-series resolvins (RvEs), COX-2 converts EPA to 18S-HEPE, which is further metabolized to generate 18S-RvE1 and 18S-RvE2. In vascular endothelial cells, COX-2 converts docosapentaenoate (DPA) to 13R-HDPA, a precursor for 13-series resolvins (RvTs), which activate macrophage phagocytosis during bacterial infections. In activated leukocytes, COX-2 contributes to the oxygenation of hydroxyeicosatetraenoates (HETE) to diHETES (5,15-diHETE and 5,11-diHETE). During neuroinflammation, COX-2 is involved in neuronal secretion of specialized pro-resolving mediators (SPMs) like 15R-lipoxin A4, which regulates phagocytic microglia.
Gene References Into Functions
  1. SND1 could serve as a potential biomarker for therapeutic strategies utilizing COX2 inhibitors. PMID: 30365124
  2. Research has shown that mRNA and protein levels of COX2 and HER2 are elevated in colorectal cancer (CRC) compared to the adjacent tissues. COX2 protein levels and nuclear COX2 expression correlate with a poor prognosis for CRC patients. Furthermore, COX2 expression is positively associated with HER2 expression. PMID: 29873317
  3. PTGS2 polymorphisms are linked to advanced liver fibrosis in patients with HCV mono-infection and HCV/HIV co-infection. PMID: 30139224
  4. An underappreciated cellular interaction between follicular dendritic cells and B cells leads to COX-2 expression during immune inflammatory responses. PMID: 29241029
  5. Neuronal SphK1 acetylates COX2 and contributes to the pathogenesis in Alzheimer's disease patients and in a transgenic mouse model. PMID: 29662056
  6. miR-137 suppresses the proliferation and invasion of retinoblastoma cells by targeting the COX-2/PGE2 signaling pathway. PMID: 29945115
  7. Dual regulation of YAP and COX-2 might lead to the identification of promising therapeutic strategies for HCC patients. PMID: 29505957
  8. COX-2 expression is positively correlated with the recurrence and a poor prognosis of patients with nasopharyngeal carcinoma. PMID: 29956730
  9. COX-2 is an essential factor for dengue virus replication. PMID: 28317866
  10. Lysophosphatidylcholine induces COX-2-mediated IL-6 expression. NADPH oxidase/Reactive Oxygen Species are involved in Lysophosphatidylcholine-induced COX-2 expression. PMID: 30229288
  11. Findings suggest a crucial role for ATF6alpha in establishing and maintaining cellular senescence in normal human fibroblasts through the upregulation of a COX2/PGE2 intracrine pathway. PMID: 28803844
  12. High PTGS2 expression is associated with Breast Carcinoma. PMID: 30051683
  13. Research suggests that COX-2 gene rs5275 variant contributes to nasopharyngeal carcinoma risk in a Chinese population. PMID: 30087034
  14. COX2 and YAP1 signaling pathways are interconnected to induce SOX2 expression, cancer stem cell enrichment, and acquired resistance to chemotherapy in urothelial carcinoma of the bladder. PMID: 29180467
  15. No significant association between COX-2 8473 T > C polymorphism and cancer risk was observed. PMID: 30143023
  16. Studies have shown that TLR4 and COX-2 are upregulated in prostate cancer (PCa) tissues; silencing of TLR4 or COX-2 inhibits PCa cell proliferation, migration, and invasion. PMID: 30098292
  17. Research indicates a novel role for cyclooxygenase-2 (COX-2) in mediating the TGFbeta effects on breast cancer stem cells (BCSC) properties and suggests that targeting the COX-2 pathway may be beneficial for treating triple-negative breast cancer by eliminating BCSCs. PMID: 28054666
  18. The effects of miR-101 inhibition on tumor growth were suppressed by COX-2 inhibition. PMID: 29404887
  19. Low PTGS2 expression is associated with Invasive Breast Carcinoma. PMID: 28808873
  20. rs2243250 (IL4) and rs5275 (PTGS2) were found to be significantly associated with shorter renal cell cancer-specific survival (CSS). PMID: 28117391
  21. Meta-analysis has demonstrated that COX-2 rs5275 and rs689466 polymorphism significantly decrease the risk of lung cancer in Asians but not in Caucasians, indicating that COX-2 could serve as a potential diagnostic marker for lung cancer. PMID: 30170377
  22. Meta-analysis of the association between 765G/C polymorphism and periodontitis in the Chinese population. PMID: 29514641
  23. Through downregulation of COX-2 expression in SGC-7901 and MGC-803 cells. PMID: 29901169
  24. Patients with high COX-2 expression in baseline tumor biopsies had a lower response to treatment of pathology compared to patients with lower COX-2 expression in baseline tumor biopsies. PMID: 29893307
  25. Studies have investigated the association between integrin subunit alpha 2 (GPIa) and prostaglandin-endoperoxide synthase 2 (COX-2) genetic polymorphisms in Chinese ischemic stroke patients with or without aspirin resistance. PMID: 28948649
  26. COX-2 was significantly associated with a lower 5-year disease-free survival (DFS) rate. PMID: 29559247
  27. The polymorphism in the COX2 gene is associated with an increased susceptibility to colorectal cancer, specifically rectosigmoid tumors. PMID: 29257846
  28. Research has demonstrated the unregulated expression of ANXA1 and COX-2 in precursor lesions of esophageal and stomach cancers. PMID: 29254791
  29. The cytotoxicity induced by EB1 gene knockdown was due to the activation and generation of reactive oxygen species by p38 mitogen-activated protein kinase. This signaling cascade, however, not nuclear factor-kappaB-mediated signaling, induced the expression of cyclooxygenase-2, a key effector of apoptotic death. PMID: 29484424
  30. High COX2 expression is associated with Ras and BRAF mutations in Hepatocellular Carcinoma. PMID: 28188432
  31. High COX2 expression is associated with ovarian cancer cell migration and invasion. PMID: 28677781
  32. Findings suggest that lower transcript levels of PTGS2 in cumulus cells may be involved in the impairment of oocyte quality, suggesting a possible mechanism involved in disease-related infertility. PMID: 28734688
  33. Results indicate a significant correlation in Japan between the COX-2 1195 G-carrier genotype and intestinal metaplasia in histological and endoscopic findings based on the Kyoto classification in H. pylori-infected gastric mucosa. PMID: 28946145
  34. Activated Ras, protumorigenic COX-2, and Notch1 play roles in the onset of papillary mucinous neoplasm. PMID: 27381829
  35. TGF-beta1 increased the COX-2 and PGE2 level of cultured pulp cells. The effect of TGF-beta1 on COX-2 protein expression was associated with ALK5/Smad2/3 and MEK/ERK pathways. PMID: 28779848
  36. Culinary herbs and spices prevent the growth of HCA-7 colorectal adenocarcinoma cancer cells and inhibit their COX-2 expression. PMID: 28934138
  37. The medical use of COX inhibitors in glioblastoma treatment has been limited due to their well-documented vascular toxicity and inconsistent outcomes from recent human studies. Prostaglandin E2 (PGE2) has emerged as a principal mediator for COX-2 cascade-driven gliomagenesis. PMID: 28718447
  38. COX2 inversely regulated Notch1 in gastric cancer and partially depended on the Notch1 signaling pathway in altering the expression of Snail. PMID: 28586004
  39. Contribution maps from three techniques suggest that both the benzenesulfonyl group and the central five-membered ring—having a high-electronegativity functional group or atom or having a substituent hydrogen bonding acceptor—contribute positively to the selective inhibition of COX-2. PMID: 27145042
  40. Findings demonstrate that COX-2 and p-Akt1 play an important combined role during melanoma progression and are associated with highly metastatic tumors and survival rates in patients with MM. PMID: 28604419
  41. Results suggest that higher COX-2 expression may be a negative prognostic factor in conjunctival melanoma. Further studies can address the potential use of anti-COX-2 drugs as adjuvant therapy for this disease. PMID: 29297092
  42. Activation of ERK1/2 signaling was required for hCG-induced upregulation of SPRY2 expression. Further, SPRY2 knockdown attenuated the AREG-induced COX-2 expression and PGE2 production by inhibiting AREG-activated ERK1/2 signaling. PMID: 27539669
  43. COX-2 was elevated in glioma tissues, and its expression was negatively correlated with the levels of miR-128. These findings may establish miR-128 as a new potential target for the treatment of patients with gliomas. PMID: 29524580
  44. Post-transcriptional regulation of COX-2 mRNA translation by SGs indicates a role in IL-1beta-mediated catabolic response that could be therapeutically targeted in Osteoarthritis. PMID: 27271770
  45. Results show that in influenza A viruses (IAV)-infected cells, COX-2 expression is regulated. While the protein is induced at early time points of infection, via recognition of IAV vRNA by RIG-I, COX-2 expression is reduced again during ongoing replication due to destabilization of its mRNA by IAV-induced TTP. PMID: 27265729
  46. Research highlights the role of COX-2 in constitutive IDO1 expression by human tumors and supports the use of COX-2 inhibitors to improve the efficacy of cancer immunotherapy by reducing constitutive IDO1 expression, which contributes to the lack of T-cell infiltration in "cold" tumors, which fail to respond to immunotherapy. PMID: 28765120
  47. The Chinese population with the GG genotype of COX-2 gene polymorphism rs689466 has a higher risk of developing post-traumatic osteomyelitis. PMID: 28682162
  48. 4-Hydroxy-2-nonenal is a natural inducer of COX-2 in atherosclerosis. (Review) PMID: 28192229
  49. RhoA and COX-2 are upregulated in early gastric cancer tissues, which facilitate the proliferation and migration of gastric cancer cells. PMID: 28624843
  50. LXR gene expression was significantly increased in obese children with obstructive sleep apnea-hypopnea syndrome (OSAHS). The severity of OSAHS was positively correlated with COX-2 levels. PMID: 28676625

Show More

Hide All

Database Links

HGNC: 9605

OMIM: 600262

KEGG: hsa:5743

STRING: 9606.ENSP00000356438

UniGene: Hs.196384

Protein Families
Prostaglandin G/H synthase family
Subcellular Location
Microsome membrane; Peripheral membrane protein. Endoplasmic reticulum membrane; Peripheral membrane protein. Nucleus inner membrane; Peripheral membrane protein. Nucleus outer membrane; Peripheral membrane protein.

Q&A

What is PTGS2 and why is it important to study?

PTGS2 is an inducible enzyme that catalyzes the conversion of arachidonic acid to prostaglandin H2, which is subsequently converted into various prostaglandins by downstream enzymes. Unlike its constitutively expressed counterpart PTGS1 (COX-1), PTGS2 is induced in response to inflammatory stimuli such as cytokines, growth factors, and cellular stress . PTGS2 plays crucial roles in inflammation, pain, fever, angiogenesis, and tumor growth, making it a significant target for research across multiple disease models .

What applications are FITC-conjugated PTGS2 antibodies suitable for?

FITC-conjugated PTGS2 antibodies are particularly suitable for flow cytometry, immunocytochemistry/immunofluorescence (ICC/IF), and can also be used for Western blot applications . The fluorescent properties of FITC make these antibodies especially valuable for applications requiring direct visualization of PTGS2 expression in cells or tissues without the need for secondary antibody incubation steps.

How should FITC-conjugated PTGS2 antibodies be stored for optimal performance?

FITC-conjugated antibodies require special storage considerations to maintain their fluorescent properties. They should be aliquoted and stored in the dark at 2-8°C, protected from prolonged exposure to light . Repeated freeze/thaw cycles should be avoided. Before opening, it is recommended to spin the vial, and the antibody solution should be gently mixed before use .

What controls should be included when using PTGS2 antibodies in experiments?

For rigorous experimental design, include both positive and negative controls. For positive controls, use cell lines or tissues known to express PTGS2, such as macrophages stimulated with IL-1β . For negative controls, use samples where PTGS2 is not expressed or is knocked down. Additionally, an isotype control (matching the host species and immunoglobulin class) should be included to control for non-specific binding, especially in flow cytometry and immunofluorescence applications.

How can I quantify PTGS2 protein expression in tissue lysates?

Western blot analysis can effectively quantify the glycosylated form of PTGS2 (gPTGS2) in tissue lysates. In a study examining colorectal cancer specimens, researchers detected gPTGS2 in 96/100 CRC samples with a median of 156.86 pg and a range of 0.00–1515.64 pg of protein in 30 μg of tissue lysate . This method demonstrated high reproducibility with a Pearson's correlation of r = 0.907 (p < 0.0000000000000000000000217) when replicated .

How can I differentiate between tumor-derived and stroma-derived PTGS2 in cancer tissue samples?

Distinguishing tumor-derived from stroma-derived PTGS2 requires specialized immunohistochemistry (IHC) approaches. Research has shown that tumor and stromal cells contribute differently to total PTGS2 levels in tissue samples. To differentiate:

  • Perform IHC using anti-PTGS2 antibodies on serial tissue sections

  • Score tumor epithelial-derived and stroma-derived fractions separately

  • For more specific quantification of macrophage-derived PTGS2, implement multiplex IHC:

    • Use consecutive destaining, stripping, and reprobing techniques on the same tissue slices

    • Test specific marker combinations (e.g., CD68–iNOS–PTGS2 for M1 macrophages or Arg1–MRC1–CD163–PTGS2 for M2 macrophages)

Studies have demonstrated a moderate correlation (Pearson coefficient 0.422, p = 0.0000586) between CD68/PTGS2 and a weaker correlation (0.316, p = 0.00324) for CD163/PTGS2 in colorectal cancer tissues .

What methodological approaches can address the influence of photobleaching on FITC-conjugated PTGS2 antibody experiments?

FITC is susceptible to photobleaching, which can affect experimental outcomes, particularly in long-duration imaging studies. To mitigate photobleaching effects:

  • Minimize exposure to excitation light during sample preparation and imaging

  • Use anti-fade mounting media containing anti-photobleaching agents

  • Adjust imaging parameters to use minimal excitation light intensity without compromising signal detection

  • Consider time-series correction algorithms if quantitative analysis is required

  • For extended imaging sessions, use reference standards to normalize signal intensity across time points

How can I optimize multiplex immunofluorescence protocols to study PTGS2 co-localization with macrophage markers?

Optimizing multiplex immunofluorescence for PTGS2 and macrophage markers requires:

  • Sequential antibody staining with careful ordering:

    • Begin with the lowest abundance target (often PTGS2)

    • Follow with higher abundance targets (CD68, CD163)

  • Implement proper tyramide signal amplification (TSA) for each target

  • Complete antibody stripping between rounds using optimized buffer systems

  • Use spectral unmixing to reduce fluorophore bleed-through

  • Include single-stained controls for each antibody to facilitate accurate spectral unmixing

Research has shown that quantifying cells expressing these antigens in overlapping areas of equal extension can validate co-localization observations, with correlation coefficients of 0.422 for CD68/PTGS2 and 0.316 for CD163/PTGS2 in colorectal cancer tissues .

What are the key considerations when using WGCNA (Weighted Gene Co-expression Network Analysis) to identify PTGS2 as a biomarker in disease states?

WGCNA is a powerful bioinformatic approach for identifying disease-associated gene modules. When using WGCNA to study PTGS2:

  • Optimize the soft-threshold power parameter (e.g., power=13, scale-free R² = 0.85 has been effective in arteriovenous fistula studies)

  • Identify modules with highest correlation to disease phenotype (e.g., blue and red modules)

  • Intersect hub genes from relevant modules with oxidative stress-related differentially expressed genes (OSDEGs)

  • Validate findings using independent datasets and experimental approaches

  • Confirm PTGS2 expression patterns in clinical samples using the FITC-conjugated antibodies

This approach has successfully identified PTGS2 as an essential biomarker in arteriovenous fistulas (AVFs) failure in hemodialysis patients .

How can I investigate the role of inflammatory mediators like IL-1β in PTGS2 induction using FITC-conjugated antibodies?

To study IL-1β-mediated PTGS2 induction:

  • Experimental design:

    • Culture target cells (e.g., fibroblasts, cancer cell lines) in serum-free conditions for 48 hours

    • Treat with IL-1β (0.1 ng/mL is typically effective) for 24 hours

    • Include appropriate controls (untreated, other cytokines like IL8/CXCL8 at 10 ng/mL)

  • Detection approach:

    • Process cells for flow cytometry using FITC-conjugated PTGS2 antibodies

    • Analyze median fluorescence intensity as a measure of PTGS2 expression

    • Alternatively, prepare lysates for Western blotting to quantify total PTGS2 protein

  • Validation strategies:

    • Confirm IL-1β specificity using neutralizing antibodies or receptor antagonists

    • Use PTGS2 inhibitors (e.g., NS398) as negative controls

    • Perform dose-response and time-course studies to characterize induction kinetics

TreatmentPTGS2 Induction Relative to Controlp-value
IL-1β (0.1 ng/mL)+++ (>10-fold)<0.001
IL8/CXCL8 (10 ng/mL)+ (2-3 fold)<0.05
PGE2 (100 nM)++ (5-6 fold)<0.01
EGF (10 ng/mL)++ (4-5 fold)<0.01

Note: Table represents typical response patterns based on research literature

What steps should be taken when FITC-conjugated PTGS2 antibody shows weak or no signal?

When encountering weak signals with FITC-conjugated PTGS2 antibodies:

  • Evaluate antibody viability:

    • Check storage conditions (light exposure, temperature fluctuations)

    • Verify expiration date

    • Test antibody using a known positive control

  • Optimize protocol parameters:

    • Increase antibody concentration (titrate from 1:100 to 1:50 or higher)

    • Extend incubation time (4°C overnight instead of 1-2 hours)

    • Improve permeabilization for intracellular targets

    • Optimize fixation (overfixation can mask epitopes)

  • Enhance detection sensitivity:

    • Use higher gain settings on flow cytometer/microscope

    • Apply signal amplification methods

    • Reduce background with additional blocking steps

  • Consider target expression levels:

    • Stimulate cells with IL-1β (0.1 ng/mL for 24h) to induce PTGS2 expression

    • Verify PTGS2 expression using alternative methods (qPCR, Western blot)

How can I resolve high background issues when using FITC-conjugated PTGS2 antibodies in tissue sections?

High background is a common challenge with fluorescent antibodies in tissue sections. To address this:

  • Implement rigorous blocking:

    • Use 5-10% serum from the same species as the secondary antibody

    • Add 0.1-0.3% Triton X-100 for permeabilization

    • Include background-reducing agents (e.g., 0.1% BSA, 0.05% Tween-20)

  • Optimize antibody dilution:

    • Perform titration experiments to identify optimal antibody concentration

    • Test dilutions ranging from 1:50 to 1:500

  • Reduce autofluorescence:

    • Treat sections with Sudan Black B (0.1-0.3% in 70% ethanol)

    • Apply copper sulfate solution (10mM in 50mM ammonium acetate buffer)

    • Use commercial autofluorescence quenchers

  • Improve washing procedures:

    • Increase wash duration and frequency

    • Use PBS with 0.05-0.1% Tween-20

    • Perform washes on orbital shaker

The specificity of PTGS2 antibodies is crucial, as research shows they do not cross-react with COX-1, ensuring specific detection of PTGS2 protein .

What are effective strategies for analyzing PTGS2 expression heterogeneity in tumor samples using FITC-conjugated antibodies?

Tumor heterogeneity presents unique challenges for PTGS2 analysis. Effective strategies include:

  • Spatial analysis approaches:

    • Implement whole-slide imaging to capture entire tissue sections

    • Use computational segmentation to distinguish tumor from stromal regions

    • Apply hot-spot analysis to identify regions of highest PTGS2 expression

  • Heterogeneity quantification methods:

    • Calculate coefficients of variation across multiple regions

    • Apply spatial statistics (Moran's I, Geary's C) to characterize distribution patterns

    • Use H-score methodology, considering both intensity and percentage of positive cells

  • Multi-parameter analysis:

    • Combine PTGS2-FITC with markers for specific cell populations

    • Include proliferation markers to correlate with tumor aggressiveness

    • Incorporate hypoxia markers to evaluate microenvironmental influence

  • Validation approaches:

    • Compare findings across multiple tumor regions

    • Correlate with RNA expression data from microdissected regions

    • Validate with alternative detection methods (e.g., RNAscope)

Research has demonstrated that the correlation coefficient of tumor PTGS2 compared with stromal PTGS2 was 0.334 (Spearman's rank, p < 0.001), suggesting distinct mechanisms of PTGS2 induction in different cell populations within the same sample .

How should PTGS2 expression data be normalized for comparative analysis across different experimental conditions?

Proper normalization is essential for valid comparisons of PTGS2 expression:

  • Western blot quantification:

    • Normalize to housekeeping proteins (β-actin, GAPDH, β-tubulin)

    • Use total protein normalization methods (Ponceau S, REVERT staining)

    • Include recombinant PTGS2 protein standards for absolute quantification

  • Flow cytometry analysis:

    • Normalize to isotype control (matched IgG-FITC)

    • Calculate fold change relative to unstimulated controls

    • Use standardized beads to calibrate fluorescence intensity

  • Immunohistochemistry/immunofluorescence:

    • Score relative to internal positive controls

    • Implement digital pathology algorithms for consistent quantification

    • Consider ratio of tumor to stromal expression for comprehensive evaluation

  • qPCR validation:

    • Use multiple reference genes validated for stability in your experimental system

    • Apply geometric averaging of multiple reference genes (GeNorm approach)

    • Calculate relative expression using the 2^-ΔΔCt method

Research has demonstrated that gPTGS2 can be reliably quantified in tissue lysates with high sensitivity, showing nearly undetectable levels in normal mucosa (median = 0.00 pg) compared to significant expression in colorectal cancer tissues (median = 156.86 pg) .

What statistical approaches are most appropriate for analyzing PTGS2 expression correlation with clinical outcomes?

For correlating PTGS2 expression with clinical outcomes:

  • Univariate analyses:

    • Kaplan-Meier survival analysis with log-rank test for time-to-event outcomes

    • Cox proportional hazards models for calculating hazard ratios

    • ROC curve analysis to determine optimal cutoff values for PTGS2 expression

  • Multivariate analyses:

    • Multiple Cox regression incorporating established prognostic factors

    • Propensity score matching to reduce confounding

    • Competing risk analysis when multiple outcome events are possible

  • Advanced modeling approaches:

    • Machine learning algorithms for pattern recognition

    • Random forest models for identifying variable importance

    • Nomogram development to predict individual patient outcomes

  • Validation strategies:

    • Internal validation using bootstrap or cross-validation

    • External validation with independent patient cohorts

    • Time-dependent ROC curves to assess predictive accuracy

Research has identified PTGS2 as a potential biomarker for arteriovenous fistulas failure through WGCNA analysis, suggesting its utility in predicting clinical outcomes .

How can I develop a multiplex flow cytometry panel incorporating FITC-conjugated PTGS2 antibody for comprehensive immune cell phenotyping?

Developing a comprehensive multiplex flow cytometry panel requires:

  • Strategic panel design:

    • Assign FITC to PTGS2 based on expected expression level (reserve brighter fluorophores for lower-expressed targets)

    • Select compatible fluorophores with minimal spectral overlap

    • Include markers for major immune cell populations (CD3, CD4, CD8, CD19, CD14, CD56)

    • Add activation/functional markers (HLA-DR, CD69, cytokines)

  • Optimization steps:

    • Perform single-stain controls for compensation

    • Titrate each antibody individually

    • Test fluorescence minus one (FMO) controls

    • Validate on known positive and negative populations

  • Analysis strategy:

    • Implement hierarchical gating to identify major populations

    • Use dimensionality reduction techniques (tSNE, UMAP) for visualization

    • Apply clustering algorithms to identify novel PTGS2+ subpopulations

    • Correlate PTGS2 expression with functional parameters

  • Quality control measures:

    • Include viability dye to exclude dead cells

    • Monitor instrument performance with tracking beads

    • Standardize protocols for consistent results across experiments

How can PTGS2 antibodies be used to evaluate the efficacy of cyclooxygenase inhibitors in preclinical models?

PTGS2 antibodies provide valuable tools for evaluating COX inhibitor efficacy:

  • Experimental design approach:

    • Establish baseline PTGS2 expression in target tissues/cells

    • Administer COX inhibitors at various doses and durations

    • Include selective (e.g., NS398) and non-selective inhibitors for comparison

    • Collect samples at strategic timepoints to assess acute and chronic effects

  • Readout parameters:

    • Measure PTGS2 protein levels by Western blot or flow cytometry

    • Assess prostaglandin production by ELISA or mass spectrometry

    • Evaluate downstream signaling pathway activation

    • Monitor phenotypic changes (inflammation, cell proliferation)

  • In vivo model considerations:

    • Use FITC-conjugated antibodies for flow cytometry of dissociated tissues

    • Perform ex vivo imaging of intact tissues

    • Consider pharmacokinetic/pharmacodynamic relationships

    • Correlate PTGS2 inhibition with physiological outcomes

Research has shown that NS398, a PTGS2 inhibitor, affects hemodynamics, smooth muscle cell proliferation, migration, and oxidative stress in mouse arteriovenous fistula models, demonstrating the utility of these approaches .

What are the key considerations when designing experiments to investigate the relationship between PTGS2 expression and macrophage polarization?

Investigating PTGS2 in macrophage polarization requires:

  • Macrophage polarization protocol:

    • Derive macrophages from primary monocytes or cell lines

    • Induce M1 polarization (IFN-γ + LPS) and M2 polarization (IL-4 + IL-13)

    • Include unstimulated (M0) macrophages as baseline controls

    • Validate polarization with established markers (CD80/CD86 for M1; CD163/CD206 for M2)

  • PTGS2 assessment strategy:

    • Use FITC-conjugated PTGS2 antibodies for flow cytometry

    • Perform time-course analysis to track expression dynamics

    • Quantify both percentage of positive cells and expression intensity

    • Correlate with functional readouts (cytokine production, phagocytic activity)

  • Mechanistic investigations:

    • Apply selective PTGS2 inhibitors to determine functional consequences

    • Use siRNA/shRNA approaches for genetic validation

    • Rescue experiments with prostaglandin supplementation

    • Investigate upstream regulators and downstream effectors

Research has revealed correlations between PTGS2 and macrophage markers (CD68, CD163) in colorectal cancer tissues, with Pearson correlation coefficients of 0.422 and 0.316 respectively, supporting the biological relevance of these investigations .

How can FITC-conjugated PTGS2 antibodies be integrated into live-cell imaging studies to monitor dynamic regulation of PTGS2 expression?

Integrating FITC-conjugated PTGS2 antibodies into live-cell imaging requires:

  • Cell preparation considerations:

    • Use membrane-permeabilizing agents that maintain cell viability

    • Consider chimeric antibody fragments with enhanced cell penetration

    • Optimize antibody concentration to minimize potential functional interference

    • Implement nuclear or membrane staining for cell identification

  • Imaging parameters:

    • Use spinning disk or light sheet microscopy for reduced phototoxicity

    • Establish minimal laser power settings that maintain adequate signal-to-noise ratio

    • Implement environmental controls (temperature, CO2, humidity)

    • Design time-lapse intervals to capture relevant dynamics while minimizing exposure

  • Analysis approaches:

    • Track single-cell PTGS2 expression over time

    • Correlate with morphological changes or co-expressed markers

    • Implement automated image analysis pipelines for unbiased quantification

    • Use photobleaching correction algorithms for extended imaging sessions

  • Validation strategies:

    • Compare with fixed-time point analyses

    • Confirm specificity with PTGS2 knockout controls

    • Verify that antibody binding doesn't alter normal PTGS2 function

    • Correlate imaging results with biochemical assays

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 2024 Thebiotek. All Rights Reserved.