PTGDS Antibody

Shipped with Ice Packs
In Stock

Description

Fundamentals of PTGDS Biology

PTGDS, or Prostaglandin D Synthase, is a glutathione-independent enzyme that catalyzes the conversion of prostaglandin H2 (PGH2) to prostaglandin D2 (PGD2) . This protein plays diverse physiological roles, functioning primarily as a neuromodulator and trophic factor within the central nervous system . Additionally, PTGDS participates in smooth muscle contraction/relaxation processes and serves as a potent inhibitor of platelet aggregation .

The protein is preferentially expressed in the brain and represents the most abundant protein in cerebrospinal fluid . Scientific evidence suggests PTGDS plays crucial roles in:

  • Development and maintenance of various biological barriers (blood-brain, blood-retina, blood-aqueous humor, and blood-testis)

  • Regulation of non-rapid eye movement (NREM) sleep

  • Beta-Amyloid chaperoning with potential implications in Alzheimer's disease pathology

PTGDS is recognized by multiple synonyms in scientific literature, including L-PGDS, LPGDS, PDS, PGD2, PGDS, PGDS2, Beta-trace protein, and Cerebrin-28 .

Technical Applications of PTGDS Antibodies

PTGDS antibodies demonstrate utility across multiple experimental platforms, with varying optimal conditions depending on the specific antibody and application.

Western Blot Analysis

Western blotting represents a primary application for PTGDS antibodies, with product-specific recommended dilutions:

  • OriGene (TA321242): 1:500-1:2000 dilution, with positive controls identified in HeLa and Raji cell lysates

  • Bio-Techne (MAB10099): 0.5 μg/mL concentration, successfully detecting PTGDS in human heart and rat brain tissue lysates

  • Novus Biologicals (NBP1-81291): 0.04-0.4 μg/ml concentration range

Western blot analyses typically detect PTGDS as a band of approximately 21-26 kDa, with experimental conditions generally employing reducing conditions . Successful detection has been reported in various tissue types including human heart, rat brain, human testis, and overexpression systems using HEK293T cells .

Immunohistochemical Applications

Immunohistochemistry (IHC) applications enable spatial visualization of PTGDS expression in tissue sections:

  • OriGene (TA321242): 1:50-1:200 dilution, with human cervical cancer serving as a positive control

  • Bio-Techne (MAB10099): 5 μg/mL concentration, successfully localizing PTGDS in human brain caudate nucleus

  • Novus Biologicals (NBP1-81291): 1:20-1:50 dilution for both standard IHC and paraffin-embedded sections

For paraffin-embedded tissue sections, heat-induced epitope retrieval using basic pH buffer is generally recommended . PTGDS typically localizes to the cytoplasm and nuclei of neurons in brain tissue samples , while showing negative staining in hepatocytes of human liver samples .

Specialized Research Applications

Beyond standard applications, PTGDS antibodies have been utilized in specialized research contexts:

  • Quantitative analysis of neuronal PTGDS expression using confocal microscopy and digital image analysis software

  • Co-immunoprecipitation studies to identify protein-protein interactions, revealing association between PTGDS and MYH9

  • Comparative expression analysis between different biological specimens, such as investigating sex-based differences in PTGDS levels

Western Blot Protocol Recommendations

Based on documented successful detection of PTGDS :

  1. Prepare tissue lysates from PTGDS-expressing samples

  2. Separate proteins using SDS-PAGE under reducing conditions

  3. Transfer to PVDF membrane

  4. Block with appropriate buffer (e.g., Immunoblot Buffer Group 1)

  5. Probe with primary PTGDS antibody (e.g., 0.5 μg/mL MAB10099)

  6. Wash and incubate with species-appropriate HRP-conjugated secondary antibody

  7. Develop using chemiluminescent detection system

  8. Identify PTGDS band at approximately 21-26 kDa

Immunohistochemistry Protocol Outline

For optimal PTGDS detection in paraffin-embedded tissue sections :

  1. Perform heat-induced epitope retrieval using Antigen Retrieval Reagent-Basic

  2. Block endogenous peroxidase activity and non-specific binding

  3. Incubate with primary PTGDS antibody (concentration per manufacturer recommendations)

  4. Wash and incubate with appropriate detection system (e.g., Anti-Mouse IgG HRP Polymer)

  5. Develop with DAB (brown) and counterstain with hematoxylin (blue)

  6. Visualize PTGDS staining in appropriate cellular compartments (cytoplasm and nuclei in neurons)

PTGDS Quantification in Tissue Samples

For quantitative analysis of PTGDS expression :

  1. Stain tissue sections with PTGDS antibody and appropriate cell-type markers

  2. Image using confocal microscopy with standardized acquisition settings

  3. Analyze images using specialized software (e.g., Olympus CellSens)

  4. Quantify mean gray intensity values across sufficient cell numbers (hundreds per specimen)

  5. Apply statistical analysis to compare expression levels between experimental groups

Sex-Based Differences in PTGDS Expression

Research utilizing PTGDS antibodies has revealed important biological sex differences:

  • PTGDS protein expression is significantly higher in dorsal root ganglion (DRG) neurons obtained from female organ donors compared to male donors

  • These findings contribute to understanding sex-specific differences in prostaglandin signaling pathways with potential implications for pain perception and therapeutic approaches

PTGDS in Cancer Biology

PTGDS antibody-based investigations have uncovered critical roles in cancer progression:

  • Enhanced PTGDS expression has been documented in diffuse large B-cell lymphoma (DLBCL) specimens

  • High PTGDS expression correlates with unfavorable therapeutic outcomes and poor prognosis in DLBCL patients

  • Patients with PTGDS-positive DLBCL demonstrate significantly reduced progression-free survival (29 months versus 55 months)

  • Functional studies show PTGDS knockdown inhibits cancer cell proliferation, induces apoptosis, arrests cell cycle progression, and reduces invasive capability

  • Mechanistically, PTGDS interacts with MYH9 protein and regulates the Wnt-β-catenin-STAT3 signaling pathway in DLBCL cells

  • PTGDS inhibition sensitizes DLBCL cells to chemotherapeutic agents (adriamycin and bendamustine) by promoting DNA damage

PTGDS in Neurological Research

PTGDS antibodies have facilitated important neurological discoveries:

  • PTGDS acts as a Beta-Amyloid chaperone with potential implications in Alzheimer's disease pathogenesis

  • The protein plays a role in regulating non-rapid eye movement sleep, as demonstrated through studies with transgenic mouse models

  • As the most abundant protein in cerebrospinal fluid, PTGDS serves as a diagnostic marker for cerebrospinal fluid rhinorrhea in head trauma assessment

Experimental Controls and Validation

Recommended positive controls for validating PTGDS antibody performance:

  • Human brain tissue (particularly for OriGene TA321242 and Thermo Fisher PA1-46023)

  • Human heart tissue and rat brain tissue (especially for Bio-Techne MAB10099)

  • Human testis tissue (validated for Novus Biologicals NBP1-81291)

  • PTGDS-overexpressing HEK293T cells

Appropriate negative controls include:

  • Vector-only transfected HEK293T lysate

  • Human liver hepatocytes (for Novus Biologicals NBP1-81291)

Future Research Directions

PTGDS antibodies continue to enable important scientific advances, with several promising research directions:

  • Therapeutic targeting of PTGDS in malignancies, particularly DLBCL, where inhibition may sensitize cancer cells to conventional chemotherapeutics

  • Further investigation of sex-specific differences in PTGDS expression and function across various tissues and biological contexts

  • Exploration of PTGDS as a biomarker or therapeutic target in neurological disorders, including Alzheimer's disease

  • Development of next-generation PTGDS antibodies with enhanced specificity, sensitivity, and versatility for research and diagnostic applications

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery times may vary depending on the purchase method or location. Please consult your local distributors for specific delivery time estimates.
Synonyms
Beta trace protein antibody; Beta-trace protein antibody; Cerebrin 28 antibody; Cerebrin-28 antibody; glutathione independent PGD synthase antibody; Glutathione independent PGD synthetase antibody; Glutathione-independent PGD synthase antibody; L PGDS antibody; Lipocalin type prostaglandin D synthase antibody; Lipocalin-type prostaglandin-D synthase antibody; LPGDS antibody; PDS antibody; PGD2 antibody; PGD2 synthase antibody; PGDS antibody; PGDS2 antibody; prostaglandin D synthase antibody; prostaglandin D2 synthase (21 kDa; brain) antibody; prostaglandin D2 synthase (brain) antibody; prostaglandin D2 synthase 21kDa (brain) antibody; Prostaglandin D2 synthase antibody; prostaglandin H2 D isomerase antibody; Prostaglandin-D2 synthase antibody; Prostaglandin-H2 D-isomerase antibody; PTGDS antibody; PTGDS_HUMAN antibody; RP23-47P18.11-006 antibody
Target Names
PTGDS
Uniprot No.

Target Background

Function
PTGDS (Prostaglandin D Synthase) is an enzyme that catalyzes the conversion of PGH2 to PGD2, a prostaglandin implicated in smooth muscle contraction/relaxation and a potent inhibitor of platelet aggregation. PTGDS is involved in various CNS functions, including sedation, NREM sleep, and PGE2-induced allodynia, and may contribute to anti-apoptotic processes in oligodendrocytes. This enzyme binds small non-substrate lipophilic molecules, such as biliverdin, bilirubin, retinal, retinoic acid, and thyroid hormone. It potentially acts as a scavenger for harmful hydrophobic molecules and serves as a secretory retinoid and thyroid hormone transporter. PTGDS is likely involved in the development and maintenance of the blood-brain, blood-retina, blood-aqueous humor, and blood-testis barriers. It is believed to play significant roles in both the maturation and maintenance of the central nervous system and the male reproductive system. PTGDS is also involved in PLA2G3-dependent maturation of mast cells. PLA2G3, secreted by immature mast cells, acts on nearby fibroblasts upstream of PTGDS to synthesize PGD2. PGD2, in turn, promotes mast cell maturation and degranulation via PTGDR.
Gene References Into Functions

Related Research and Applications

  • High lipocalin-type prostaglandin D synthase expression is associated with Spontaneous intracranial hypotension. PMID: 29621631
  • Measurement of serum BTP can be a reliable tool for detecting kidney function in neonates. PMID: 29421771
  • Our findings suggest decreased NK cell-mediated eosinophil regulation, possibly through an increased level of PGD2, as a previously unrecognized link between PG dysregulation and eosinophilic inflammation in CRS. PMID: 27271931
  • These observations suggest that tumor cell-derived inflammatory cytokines increase L-PGDS expression and subsequent PGD2 production in the tumor endothelial cells (ECs). This PGD2 acts as a negative regulator of the tumorigenic changes in tumor ECs. PMID: 29124765
  • The cut-off value for betaTP in the diagnosis and follow-up of cerebrospinal fluid leaks should be modified depending on the type of secretion (sample type), for a better diagnostic accuracy. PMID: 27614217
  • Serum levels of beta trace protein and beta 2 microglobulin can be used to predict residual kidney function in hemodialysis patients. PMID: 26924065
  • BTP and B2M levels are less affected than serum by amputation, and should be considered for future study of GFR estimation. PMID: 26800100
  • Hematopoietic prostaglandin D synthase defines a proeosinophilic pathogenic effector human T(H)2 cell subpopulation with enhanced function. PMID: 26431580
  • levels of L-PGDS in cervicovaginal secretions of pregnant women at different stages of parturition correlate with preterm birth. PMID: 25964109
  • Given that uPGDS levels fall after treatment of LN, uPGDS may be used to monitor the efficacy of therapy. It can also differentiate patients with active nephritis and active non-renal lupus. PMID: 26211517
  • PTGDS mRNA expression was down-regulated in rapid-cycling bipolar disorder patients in a euthymic, depressive, and manic/hypomanic state compared with healthy control subjects. PMID: 25522430
  • Expression of MR and prostaglandin D2 synthase is strongly correlated in adipose tissues from obese patients. PMID: 25966493
  • involved in pathogenesis of androgenetic alopecia [review]. PMID: 24521203
  • These data indicate that scalp is spatially programmed via mast cell prostaglandin D-synthase distribution in a manner reminiscent of the pattern seen in androgenetic alopecia. PMID: 24438498
  • These results suggest that L-PGDS acted as a scavenger of biliverdin, which is a molecule not found in normal CSF. PMID: 25005874
  • Among NSTE-ACS patients, BTP and CysC were superior to conventional renal parameters for predicting MB, and improved clinical stratification for hemorrhagic risk. PMID: 23698027
  • Structural and dynamic insights into substrate binding and catalysis of human lipocalin prostaglandin D synthase. PMID: 23526831
  • betaTP, beta2M, CysC, and creatinine differ in their associations with demographic and clinical factors, suggesting variation in their non-glomerular filtration rate determinants. PMID: 23335043
  • All-cause and cardiovascular mortality are strongly associated with beta-trace protein marker levels and beta-microglobulin in a representative sample of US adults. PMID: 23518194
  • The results we obtained from mice led to our investigation of PTGDS in 29 human cryptorchid patients but we failed to find any mutation that supported an involvement of this gene in human testicular descent. PMID: 23076868
  • no overall evidence of an association between wireless phone use and serum concentrations of beta-trace protein. PMID: 22989106
  • Elevated plasma level of beta-trace protein associated with all cause of death in patients with acute coronary syndrome. PMID: 22818840
  • The serum level of beta-trace protein is an independent predictor of death and cardiovascular disease mortality in incident hemodialysis patients. PMID: 22745274
  • Low PTGDS expression is associated with testis cancer. PMID: 22960220
  • L-PGDS expression may contribute to the restricted proliferation of epidermal melanocytes, but conversely its overexpression may reflect the dysregulated proliferation of melanoma cells. PMID: 22299829
  • The gene coding for PTGDS was found to be more expressed in patients with attention deficit hyperactivity disorder (ADHD) relative to patients with bipolar disorder indicating a possible link with the differential etiology of ADHD. PMID: 22370065
  • Data suggest that L-PGDS binds small lipophilic ligands with both high-affinity and low-affinity interactions; molecular models are proposed from studies that include binding of hemin, biliverdin, and bilirubin. PMID: 22677050
  • Ascites and pleural effusion contain high concentrations of beta-TP that exceed the levels in corresponding plasma. PMID: 21501068
  • Urinary PGDS, not ZA2G, may serve as a biomarker for active LN and upon validation in larger studies, may become the non-invasive test to evaluate the disease activity in future management of LN. PMID: 22498882
  • NM 000954 NM 000954. PMID: 22342541
  • These results demonstrate that L-PGDS protected against neuronal cell death by scavenging reactive oxygen species without losing its ligand-binding function. PMID: 22248185
  • Beta-trace protein can be used as an alternative diagnostic tool to detect moderate or severe glomerular filtrate rate reduction in patients after liver transplantation. PMID: 21745310
  • Two genes involved in cardiovascular diseases, ADORA1 and PTGDS, were differentially up-regulated in epicardial adipose tissue compared to mediastinal and subcutaneous adipose tissue. PMID: 21603615
  • Lipocalin-type prostaglandin D synthase is associated with coronary vasospasm and vasomotor reactivity in response to acetylcholine. PMID: 21325722
  • Proteomic profiling of cerebrospinal fluid identifies prostaglandin D2 synthase as a putative biomarker for pediatric medulloblastoma. PMID: 21271676
  • RBP-4, lipocalin-2 and L-PGDS do not regulate insulin sensitivity in healthy men. Rather their expression seemed to reflect inflammatory activity and were inversely correlated with alcohol intake and serum HDL levels. PMID: 21104585
  • Report the presence of L-PGDS in the COX-2-expressing cells in the mucosa of active ulcerative colitis patients in parallel with disease activity. PMID: 21163901
  • It may be feasible to test for perilymphatic fluid fistula using PTGDS in samples from the tympanic cavity. PMID: 21192373
  • Met64 seems to function as a kinetic clamp, pushing the thiol group of Cys65 close to the site of nucleophilic attack during catalysis. PMID: 20667974
  • Using RT-PCR we demonstrated that L-PGDS gene expression decreased proportionately with tumor progression in lung cancer. PMID: 20144489
  • L-PGDS may fine-tune the retinoic acid signaling in melanocytes. PMID: 20403807
  • This study included 62 persons aged 18-30 years and cell phone exposure. EMF emissions may down-regulate the synthesis of beta-trace protein. PMID: 20596612
  • protein levels in nasal fluids can serve for diagnosis of cerebrospinal fluid leak. PMID: 19958607
  • This beta-trace protein based formula was found to estimate GFR with reasonable precision. PMID: 19949816
  • The expression of H-PGDS in human dendritic cells (DCs) and the regulatory mechanisms by which DCs produce prostaglandin D2, is demonstrated. PMID: 20008150
  • As the CSF is in contact with axons and mitochondria of the optic nerve, we postulate that a change in the concentration of CSF protein such as L-PGDS could exercise a harmful effect on these structures. PMID: 19598000
  • Pronounced eosinophilia and Th2 cytokine release in human lipocalin-type prostaglandin D synthase transgenic mice. PMID: 11751991
  • Study performed in patients with kidney function ranging from normal to advanced renal failure suggests that serum beta-trace protein is an indicator of impaired glomerular filtration rate. PMID: 12900000
  • The circadian lipocalin-type PGDS pattern and its suppression by total sleep deprivation indicate an interaction of the prostaglandin D system and human sleep regulation. PMID: 15453544
  • Shear stress induces l-PGDS expression by transcriptional activation through the AP-1 binding site. PMID: 15718494
Database Links

HGNC: 9592

OMIM: 176803

KEGG: hsa:5730

STRING: 9606.ENSP00000360687

UniGene: Hs.446429

Protein Families
Calycin superfamily, Lipocalin family
Subcellular Location
Rough endoplasmic reticulum. Nucleus membrane. Golgi apparatus. Cytoplasm, perinuclear region. Secreted. Note=Detected on rough endoplasmic reticulum of arachnoid and menigioma cells. Localized to the nuclear envelope, Golgi apparatus, secretory vesicles and spherical cytoplasmic structures in arachnoid trabecular cells, and to circular cytoplasmic structures in meningeal macrophages and perivascular microglial cells. In oligodendrocytes, localized to the rough endoplasmic reticulum and nuclear envelope. In retinal pigment epithelial cells, localized to distinct cytoplasmic domains including the perinuclear region. Also secreted.
Tissue Specificity
Abundant in the brain and CNS, where it is expressed in tissues of the blood-brain barrier and secreted into the cerebro-spinal fluid. Abundantly expressed in the heart. In the male reproductive system, it is expressed in the testis, epididymis and prosta

Q&A

What is PTGDS and why is it significant in research?

PTGDS (Prostaglandin D2 Synthase) is a 21 kDa glycoprotein enzyme that catalyzes the conversion of prostaglandin H2 (PGH2) to prostaglandin D2 (PGD2) . Its significance extends beyond this enzymatic function, as it serves multiple roles in the central nervous system, including involvement in sedation, NREM sleep, and PGE2-induced allodynia . PTGDS also functions as a lipocalin that binds small lipophilic molecules such as biliverdin, bilirubin, retinal, retinoic acid, and thyroid hormones, potentially acting as both a scavenger for harmful hydrophobic molecules and a transporter for these compounds . Notably, PTGDS has emerged as a critical player in cancer biology, with differential expression patterns observed across various cancer types, making it an important target for cancer research .

Which applications are most suitable for PTGDS antibody detection?

PTGDS antibodies have been validated for multiple experimental applications, with the most commonly used techniques being:

ApplicationSuitabilityCommon ProtocolsSpecial Considerations
Western Blot (WB)HighStandard protein detection at ~21 kDaMay detect glycosylated forms at higher molecular weights
Immunohistochemistry (IHC)HighBoth paraffin-embedded (IHC-p) and frozen sectionsOptimization of antigen retrieval is critical
Immunocytochemistry (ICC)Medium-HighStandard cell fixation protocolsWorks well with formaldehyde fixation
Immunofluorescence (IF)Medium-HighStandard IF protocolsSecondary antibody selection crucial for sensitivity
ELISAMediumDirect and sandwich ELISA formatsValidation required for quantitative analysis

For most research applications, Western blot and IHC represent the most robust methods for PTGDS detection, with consistent results reported across multiple studies . The choice of application should be guided by specific research questions, with consideration given to the cellular localization of PTGDS (both intracellular and secreted forms) .

What is the difference between PTGDS and other prostaglandin synthases in experimental contexts?

PTGDS belongs to the lipocalin family and is distinguished from other prostaglandin synthases by several key characteristics:

  • PTGDS (also known as L-PGDS or lipocalin-type PTGDS) differs structurally from H-PGDS (hematopoietic PTGDS), despite catalyzing the same reaction .

  • Unlike other prostaglandin synthases that typically function only as enzymes, PTGDS serves dual roles as both an enzyme and a lipid transporter .

  • PTGDS is uniquely regulated by glycosylation, which affects its cellular localization, half-life, and biological functions .

When designing experiments targeting PTGDS specifically, researchers should be aware of potential cross-reactivity with other prostaglandin synthases. Using antibodies that target unique epitopes of PTGDS (particularly those in the lipocalin domain rather than the catalytic site) can help ensure specificity .

How does PTGDS expression correlate with clinical outcomes in cancer research?

Recent investigations have revealed complex and sometimes contradictory roles for PTGDS across different cancer types:

Cancer TypePTGDS ExpressionClinical CorrelationReference
Diffuse Large B-cell Lymphoma (DLBCL)OverexpressedPoor prognosis, reduced progression-free survival (29 months vs. 55 months)
Malignant MelanomaOverexpressedAssociated with progression
Ovarian CarcinomaOverexpressedAssociated with progression
Hepatocellular AdenomaOverexpressedAssociated with progression
Prostate CancerDown-regulatedTumor suppressor function
Lung CancerDown-regulatedTumor suppressor function
Gastric CancerDown-regulatedTumor suppressor function

In DLBCL specifically, high PTGDS expression correlates with the germinal center B-cell (GCB) subtype, elevated sialic acid levels, and unfavorable therapeutic efficacy . Interestingly, while PTGDS expression is higher in the GCB subtype of DLBCL, it is negative in normal germinal centers, suggesting a specific oncogenic role in lymphoma cells rather than normal B cells .

These contradictory expression patterns across cancer types highlight the context-specific nature of PTGDS function and underscore the importance of using multiple antibody-based detection methods to accurately assess PTGDS status in cancer tissues.

What molecular mechanisms underlie PTGDS function in cancer progression?

PTGDS promotes cancer progression through multiple interconnected molecular pathways:

  • Wnt-β-catenin-STAT3 Signaling Axis: In DLBCL, PTGDS interacts with myosin heavy chain 9 (MYH9), which in turn regulates the Wnt-β-catenin-STAT3 pathway . PTGDS inhibition reduces MYH9 expression, leading to decreased activation of this oncogenic pathway through altered ubiquitination and degradation of GSK3-β .

  • Cell Cycle Regulation: PTGDS knockdown induces G0/G1 cell cycle arrest and decreases expression of cell cycle regulators like Cyclin D1 and CDK2 .

  • Apoptotic Pathway Modulation: PTGDS inhibition increases pro-apoptotic proteins (Bax, cleaved caspase-3, caspase-9, and PARP) while reducing anti-apoptotic proteins like Bcl-xl .

  • DNA Damage Response: PTGDS inhibition enhances DNA damage and sensitizes cancer cells to chemotherapeutic agents like adriamycin and bendamustine .

  • Glycosylation-Dependent Functions: The glycosylation status of PTGDS affects its nuclear translocation, protein half-life, and proliferative effects in cancer cells .

Understanding these mechanisms is essential for researchers designing experiments to target PTGDS or its downstream pathways for potential therapeutic interventions.

How does glycosylation affect PTGDS function and antibody detection?

Glycosylation represents a critical post-translational modification that significantly impacts PTGDS biology:

  • Functional Impact: Low glycosylation of PTGDS has been associated with nuclear translocation, prolonged half-life, and increased cell proliferation in DLBCL . This suggests that differently glycosylated forms of PTGDS may have distinct biological functions.

  • Detection Considerations: When using antibodies for PTGDS detection, researchers should consider that:

    • Different glycoforms may appear at variable molecular weights in Western blots (ranging from ~21 kDa for unglycosylated forms to higher molecular weights for glycosylated variants) .

    • Some antibodies may preferentially recognize specific glycoforms, potentially leading to incomplete detection of the total PTGDS pool .

    • Deglycosylation treatments prior to analysis may be necessary to accurately compare total PTGDS levels across different samples .

  • Clinical Correlations: In DLBCL patients, elevated sialic acid (a type of glycosylation) correlates with high PTGDS expression and unfavorable therapeutic outcomes , suggesting that monitoring both PTGDS expression and its glycosylation status may provide more comprehensive prognostic information.

Researchers should select antibodies that recognize conserved epitopes present in all PTGDS forms or use multiple antibodies targeting different regions to ensure comprehensive detection of all glycoforms relevant to their research question.

What are the optimal sample preparation techniques for PTGDS antibody applications?

Different applications require specific sample preparation approaches to maximize PTGDS detection:

ApplicationSample TypeRecommended PreparationCritical Steps
Western BlotCell/Tissue LysatesRIPA buffer with protease inhibitorsInclude phosphatase inhibitors when studying phosphorylation-dependent interactions
Western BlotCulture SupernatantsTCA precipitation or ultrafiltrationConcentration step essential for secreted PTGDS
IHCFFPE TissuesHeat-induced epitope retrieval (citrate buffer pH 6.0)Adequate deparaffinization and antigen retrieval crucial
ICC/IFFixed Cells4% paraformaldehyde fixation, 0.1% Triton X-100 permeabilizationGentle permeabilization to preserve cellular structures
Co-IPProtein ComplexesNon-denaturing lysis buffers (e.g., Pierce™ Co-IP Kit)Maintain native protein interactions during extraction

For co-immunoprecipitation studies investigating PTGDS interaction partners (such as MYH9), gentle extraction conditions are essential to preserve protein-protein interactions . When analyzing multiple parameters simultaneously, optimizing sample preparation to be compatible with all downstream applications will provide the most consistent results.

How can researchers validate PTGDS antibody specificity?

Ensuring antibody specificity is critical for obtaining reliable results. Recommended validation strategies include:

  • Positive and Negative Controls:

    • Positive controls: Cell lines with known PTGDS expression (based on literature or genomic data)

    • Negative controls: PTGDS knockdown cells using siRNA or CRISPR-Cas9

    • Tissue controls: Normal brain tissue (high expression) versus tissues known to lack PTGDS

  • Multiple Detection Methods:

    • Comparing results from different applications (WB, IHC, IF)

    • Using antibodies targeting different epitopes of PTGDS

  • Recombinant Protein Controls:

    • Peptide competition assays with the immunizing peptide

    • Comparing detection of recombinant PTGDS with and without glycosylation

  • Molecular Weight Verification:

    • Confirming the expected ~21 kDa band for unglycosylated PTGDS

    • Accounting for higher molecular weight bands representing glycosylated forms

For research focused on specific PTGDS functions or interactions, additional validation may be required to ensure the antibody can effectively detect the relevant protein pool (e.g., nuclear versus secreted PTGDS).

What factors influence antibody selection for different PTGDS research questions?

Selecting the appropriate PTGDS antibody depends on several research-specific factors:

  • Epitope Specificity:

    • For studies on PTGDS enzymatic activity: Antibodies targeting the catalytic domain

    • For lipocalin transport function: Antibodies recognizing the lipocalin domain

    • For protein interaction studies: Antibodies that do not interfere with binding sites

  • Species Reactivity:

    • Human-specific studies: Antibodies validated on human samples

    • Translational research: Antibodies with cross-reactivity to relevant model organisms (mouse, rat, etc.)

  • Clonality:

    • Polyclonal antibodies: Better for detection of denatured proteins or multiple epitopes

    • Monoclonal antibodies: Superior specificity for single epitopes and reduced batch variation

  • Application Compatibility:

    • For multiparameter analyses: Antibodies validated across multiple applications

    • For specialized techniques: Antibodies specifically optimized for the intended application

Research questions focusing on PTGDS interactions with specific partners (e.g., MYH9) may require antibodies that do not interfere with the relevant binding domains . Similarly, studies on glycosylated versus non-glycosylated forms should employ antibodies capable of distinguishing these variants or use complementary approaches to assess glycosylation status.

How can researchers address common challenges in PTGDS detection?

Several challenges commonly arise when working with PTGDS antibodies:

ChallengePotential CausesRecommended Solutions
Multiple bands in Western blotGlycosylation variants, proteolytic fragmentsUse deglycosylation enzymes to confirm identity; include protease inhibitors during extraction
Variable staining intensity in IHCTissue fixation differences, antigen maskingOptimize antigen retrieval; standardize fixation protocols; use positive controls
Discrepancies between different detection methodsMethod-specific artifacts, different epitope accessibilityValidate findings with multiple antibodies and techniques; correlate with functional assays
Inconsistent results in co-IP experimentsBuffer incompatibility, epitope maskingTest multiple antibodies for pull-down; adjust buffer conditions to preserve interactions
Limited detection of secreted PTGDSLow concentration in supernatantsConcentrate samples; use sandwich ELISA for increased sensitivity

For discrepancies in PTGDS expression across different cancer types, researchers should consider context-specific regulators of PTGDS that may vary between experimental systems . Additionally, comparing results with public databases (e.g., The Cancer Genome Atlas, Gene Expression Omnibus) can help identify systematic variations versus technical artifacts.

What controls are essential for interpreting PTGDS antibody results?

Robust experimental design requires appropriate controls:

  • Technical Controls:

    • Primary antibody omission: To assess background staining

    • Isotype controls: To evaluate non-specific binding

    • Loading controls: For quantitative comparisons (e.g., β-actin for total protein, lamin for nuclear fractions)

  • Biological Controls:

    • PTGDS knockdown/knockout samples: Created via siRNA, shRNA, or CRISPR-Cas9 methods

    • Recombinant PTGDS protein: As positive control and for standard curves

    • Tissues/cells with known PTGDS expression levels: As reference standards

  • Treatment Controls:

    • For functional studies: Vehicle controls for treatments affecting PTGDS (e.g., AT56)

    • For interaction studies: Conditions known to disrupt specific interactions

    • For glycosylation studies: Enzymatically deglycosylated samples

  • Correlation Controls:

    • Parallel analysis of PTGDS mRNA levels: To confirm expression changes

    • Functional readouts: To connect PTGDS levels with biological effects

In studies examining the relationship between PTGDS and clinical outcomes, multivariate analyses controlling for relevant clinical factors (age, stage, treatment) are essential for meaningful interpretation .

How can contradictory findings about PTGDS function be reconciled?

The contrasting roles of PTGDS reported across different cancer types present a scientific puzzle requiring systematic investigation:

  • Context-Specific Analysis:

    • Examine cell type-specific signaling networks in different tissues

    • Analyze the expression of PTGDS interaction partners (e.g., MYH9) across different systems

    • Consider the impact of the tumor microenvironment on PTGDS function

  • Methodological Reconciliation:

    • Compare antibody epitopes used in contradictory studies

    • Assess whether studies distinguished between different PTGDS isoforms or glycoforms

    • Evaluate differences in experimental conditions (in vitro vs. in vivo, 2D vs. 3D culture)

  • Integrative Approaches:

    • Combine genetic manipulation (overexpression/knockdown) with pharmacological approaches

    • Use multi-omics analyses to place PTGDS in broader signaling networks

    • Develop computational models integrating contradictory findings

When designing experiments to resolve contradictions, researchers should consider that PTGDS may function differently based on:

  • Its enzymatic activity (producing PGD2) versus its lipid-binding properties

  • Its localization (intracellular versus secreted)

  • The specific signaling pathways active in different cellular contexts

What novel approaches are emerging for PTGDS-targeted research?

Recent technological advances have opened new avenues for PTGDS research:

  • Advanced Imaging Technologies:

    • Super-resolution microscopy to visualize subcellular PTGDS localization

    • Live-cell imaging with tagged PTGDS to track trafficking and secretion

    • Multiplexed imaging to simultaneously detect PTGDS and interaction partners

  • Proteomic Approaches:

    • Proximity labeling techniques (BioID, APEX) to identify novel PTGDS interaction partners

    • Mass spectrometry-based glycoproteomics to characterize PTGDS glycoforms

    • Antibody-based proteomics for large-scale expression profiling

  • Therapeutic Targeting:

    • Development of small molecule inhibitors like AT56

    • Neutralizing antibodies against secreted PTGDS

    • Gene therapy approaches to modulate PTGDS expression

  • Clinical Applications:

    • Liquid biopsy methods to detect circulating PTGDS as a biomarker

    • Combination therapies targeting PTGDS alongside standard treatments

    • Patient stratification based on PTGDS expression patterns

The integration of high-throughput antibody-based screening with functional genomics will likely accelerate the discovery of context-specific PTGDS functions and therapeutic applications.

How might PTGDS antibodies contribute to translational research?

PTGDS antibodies have significant potential for translational applications:

  • Diagnostic Development:

    • IHC-based classification of tumor subtypes based on PTGDS expression

    • Prognostic panels incorporating PTGDS alongside other markers

    • Companion diagnostics for PTGDS-targeted therapies

  • Therapeutic Monitoring:

    • Assessing treatment efficacy through PTGDS expression changes

    • Monitoring resistance mechanisms involving PTGDS pathways

    • Evaluating on-target effects of PTGDS inhibitors

  • Precision Medicine Applications:

    • Patient stratification based on PTGDS expression and glycosylation

    • Tailoring treatment approaches based on PTGDS-associated pathways

    • Developing combinatorial strategies targeting PTGDS and interacting proteins

The finding that PTGDS inhibition enhances sensitivity to chemotherapeutic agents like adriamycin and bendamustine suggests potential for PTGDS-targeted approaches to overcome treatment resistance in certain cancers.

What additional research is needed to fully understand PTGDS biology?

Despite significant progress, several knowledge gaps remain:

  • Structural Biology:

    • Determination of crystal structures for different PTGDS glycoforms

    • Structural basis of PTGDS interactions with partners like MYH9

    • Conformational changes associated with lipid binding versus enzymatic activity

  • Regulatory Mechanisms:

    • Comprehensive understanding of transcriptional and post-transcriptional PTGDS regulation

    • Factors controlling PTGDS glycosylation and their impact on function

    • Signaling pathways regulating PTGDS secretion versus nuclear translocation

  • Systems Biology:

    • Network-level understanding of PTGDS in health and disease

    • Feedback mechanisms between PTGDS enzymatic products and expression

    • Integration of PTGDS function with broader prostaglandin signaling networks

  • Translational Gaps:

    • Development of standardized PTGDS detection methods for clinical applications

    • Establishment of clinically relevant PTGDS expression thresholds

    • Validation of PTGDS as a therapeutic target across different cancer types

Future research employing cutting-edge antibody-based techniques alongside complementary approaches will be essential to address these questions and realize the full potential of PTGDS-targeted therapies.

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.