PAEP Human

Progesterone-Associated Endometrial Protein Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Isoforms

PAEP is a 180-amino-acid glycoprotein with a molecular weight of ~21 kDa . Key structural features include:

  • Disulfide bonds: Four conserved cysteinyl residues (positions 66, 106, 119, 160) stabilize its tertiary structure .

  • N-terminal sequence: M D I P Q T K Q D L E L P K L A G T W H S M, sharing homology with β-lactoglobulins .

  • Isoforms: Three glycosylation variants (GdA, GdF, GdS) exist in amniotic fluid, follicular fluid, and seminal plasma, respectively, with distinct biological roles .

Table 2: PAEP Levels in Pregnancy

Tissue/FluidConcentration RangeTiming
Maternal serum15–40 µg/L (non-pregnant)Luteal phase
Amniotic fluid232 mg/L (peak)12–20 weeks
Early decidua41–160 mg/g proteinFirst trimester

Clinical Significance in Oncology

PAEP is overexpressed in multiple cancers and correlates with immune evasion and poor prognosis:

Melanoma

  • Function: Suppresses CD4+ T-cell IL-2/IFN-γ secretion and induces lymphocyte apoptosis .

  • Prognostic marker: High expression in metastatic melanoma linked to reduced survival (HR = 2.29, P = 0.017) .

Non-Small Cell Lung Cancer (NSCLC)

  • Expression: 8,000-fold upregulation in adenocarcinomas vs. normal tissue .

  • Survival: High PAEP levels reduce female OS (P = 0.014), independent of histology .

Bladder Cancer (BLCA)

  • Association: Correlates with advanced T stage (P = 0.009) and poor OS (HR = 1.79, P = 0.042) .

  • Immune pathways: Enriched in JAK-STAT, PD-1 blockade, and NK cell cytotoxicity pathways .

Table 3: PAEP in Cancer Prognosis

Diagnostic and Therapeutic Potential

  • Biomarker utility:

    • Predicts recurrent pregnancy loss via aberrant splicing in endometrial glands .

    • Serum levels monitor NSCLC treatment response .

  • Therapeutic target: siRNA knockdown reduces melanoma proliferation and restores T-cell cytotoxicity .

Research Frontiers

  • Alternative splicing: Novel PAEP transcripts (e.g., PAEP-206, PAEP-202) identified in recurrent pregnancy loss .

  • Immune checkpoint correlation: PAEP expression positively associates with PD-L1 and CTLA-4 in BLCA (P < 0.001) .

Product Specs

Introduction
Progesterone-Associated Endometrial Protein (PAEP), a member of the kernel lipocalin superfamily, exhibits low sequence similarity but high conservation in exon/intron structure and three-dimensional protein folding among its members. Located on chromosome 9's long arm, PAEP has been previously designated as pregnancy-associated endometrial alpha-2-globulin, placental protein 14, and glycodelin.
Description
Recombinant human PAEP, produced in E. coli, is a non-glycosylated polypeptide chain consisting of 185 amino acids (19-180a.a) with a molecular weight of 21 kDa. The protein includes a 23 amino acid His-tag at the N-terminus and undergoes purification using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The PAEP protein solution (0.5 mg/ml) contains 20 mM Tris-HCl buffer (pH 8.0), 0.15 M NaCl, 30% glycerol, and 1 mM DTT.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 90% as determined by SDS-PAGE analysis.
Synonyms
GD, GdA, GdF, GdS, PAEG, PEP, PP14, Glycodelin, Placental protein 14, Pregnancy-associated endometrial alpha-2 globulin, PEG, Progestagen-associated endometrial protein.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMDIPQTK QDLELPKLAG TWHSMAMATN NISLMATLKA PLRVHITSLL PTPEDNLEIV LHRWENNSCV EKKVLGEKTE NPKKFKINYT VANEATLLDT DYDNFLFLCL QDTTTPIQSM MCQYLARVLV EDDEIMQGFI RAFRPLPRHL WYLLDLKQME EPCRF.

Q&A

What is PAEP and what are its primary biological functions?

PAEP (progestogen-associated endometrial protein), also known as glycodelin, is a glycoprotein that plays crucial roles in fertility and maintenance of pregnancy. The protein undergoes biochemical interaction changes that affect its biological activity in various reproductive processes . As a secreted glycoprotein, PAEP has immunomodulatory properties that influence implantation, placentation, and maternal-fetal tolerance. Its expression is regulated by hormones, particularly progesterone, and it exhibits tissue-specific glycosylation patterns that determine its functional properties in different reproductive tissues. Understanding these functions is essential for interpreting experimental results in reproductive biology and immunology research.

How does PAEP expression vary across different tissue types?

PAEP expression demonstrates significant tissue specificity, with primary expression in reproductive tissues. While most strongly expressed in the endometrium during the secretory phase and in early pregnancy, PAEP can also be detected in other tissues under specific conditions. Research has shown aberrant expression in certain pathological states, including bladder cancer where PAEP expression is significantly associated with tumor immune cells and immune checkpoints . When designing experiments involving PAEP, researchers should consider this tissue variability and include appropriate positive and negative controls. Expression analysis should employ tissue-specific normalization methods to account for baseline differences across sample types.

What are the established reference ranges for PAEP concentration in human biological samples?

Standard reference ranges for PAEP vary by sample type and physiological state. In commercial ELISA kits, detection ranges typically span from 7.81 pg/ml to 500 pg/ml for serum samples , while other assays report ranges from 1.563-100 ng/ml with a sensitivity of 0.938 ng/ml . These reference ranges should be used as guidelines, as PAEP levels fluctuate naturally during the menstrual cycle and pregnancy. When establishing normal values for research purposes, it is advisable to collect samples from healthy controls matched for age, sex, and reproductive status to the experimental group. Researchers should also note that sample processing can affect measurements, as PAEP is sensitive to freeze-thaw cycles.

What is the optimal method for quantitative measurement of PAEP in research settings?

Sandwich ELISA represents the gold standard for quantitative PAEP measurement in research settings. Contemporary ELISA kits employ a one-step assay approach where capture antibodies conjugated to affinity tags bind specifically to PAEP, forming an antibody-analyte sandwich complex . This methodology offers high sensitivity (as low as 3 pg/ml) and specificity for human PAEP with limited cross-reactivity to other proteins .

For optimal results, researchers should:

  • Select kits with appropriate detection ranges for their expected concentrations

  • Follow manufacturer protocols precisely regarding sample dilution

  • Include both technical and biological replicates

  • Run standard curves on each plate to account for inter-assay variability

When comparing results across studies, be aware that different kits may use different antibodies targeting various epitopes of PAEP, potentially affecting measured values.

How do the various commercially available detection systems for PAEP compare in terms of sensitivity and specificity?

Commercial PAEP detection systems demonstrate varying performance metrics that researchers should consider when selecting an assay:

Kit ParameterELISA Kit A ELISA Kit B
Detection Range7.81-500 pg/ml1.563-100 ng/ml
Sensitivity3 pg/ml0.938 ng/ml
Assay Duration90 minutes120 minutes
Sample TypesSerumSerum, plasma, cell culture supernatant, cell/tissue lysate
Intra-Assay CV5.2%Not specified
Inter-Assay CV7.98%Not specified
Recovery Rate107% (89-116%)Not specified

The selection of an appropriate detection system should be guided by research requirements including required sensitivity, available sample types, and time constraints. For studies requiring ultra-sensitive detection, the system with 3 pg/ml sensitivity might be preferable , while investigations using diverse sample types might benefit from platforms accepting multiple sample matrices . Researchers should validate assay performance in their specific sample types prior to conducting full-scale experiments.

What technical challenges are commonly encountered in PAEP measurement and how can they be addressed?

Several technical challenges can affect PAEP measurement accuracy:

  • Sample matrix effects: Components in biological samples can interfere with antibody binding. Solution: Optimize sample dilution and use appropriate blocking buffers.

  • Hook effect: At extremely high analyte concentrations, sandwich ELISA signal may paradoxically decrease. Solution: Test multiple sample dilutions when high concentrations are suspected.

  • Cross-reactivity: Some antibodies may recognize similar proteins. Solution: Verify antibody specificity through western blot validation or competitive inhibition tests.

  • Pre-analytical variables: Sample collection, processing, and storage conditions affect PAEP stability. Solution: Standardize protocols for sample handling, including consistent collection tubes, processing times, and storage temperatures.

  • Inter-assay variability: Differences between kit lots or assay runs can introduce systematic error. Solution: Include internal quality controls on each plate and normalize to standards.

For robust PAEP quantification, researchers should employ methodological triangulation when possible, comparing ELISA results with orthogonal techniques such as mass spectrometry or immunohistochemistry in a subset of samples.

How is PAEP utilized as a biomarker in cancer research?

Methodologically, researchers utilize PAEP expression for:

  • Prognostic stratification of patients

  • Correlation with tumor immune infiltrate characteristics

  • Identification of patients who might benefit from immunotherapy approaches

When employing PAEP as a biomarker, researchers should establish appropriate expression cutoffs based on their specific cohorts, as differences in detection methods and patient populations can affect optimal thresholds. Integration with other biomarkers often provides superior predictive value compared to PAEP assessment alone.

What is the relationship between PAEP expression and immune system function in pathological contexts?

PAEP demonstrates significant correlations with immune function parameters in pathological contexts. In bladder cancer, PAEP expression is associated with immune-related pathways and cell populations. Gene set enrichment analysis (GSEA) reveals that high PAEP expression correlates with enrichment in crucial immune-related pathways including:

  • Cytokine-Cytokine Receptor Interaction

  • Antigen Processing and Presentation

  • JAK-STAT Signaling Pathway

  • Natural Killer Cell Mediated Cytotoxicity

  • T Cell Receptor Signaling Pathway

  • Cancer Immunotherapy by PD1 Blockade

GO functional enrichment analysis further confirms PAEP's association with 15 immune-related functions . When designing studies investigating PAEP's immunological roles, researchers should consider concurrent assessment of these pathways through transcriptomic or proteomic approaches. For functional validation, ex vivo immune cell assays using PAEP-treated cells can elucidate direct immunomodulatory mechanisms.

How do clinical parameters correlate with PAEP expression in cancer patients?

PAEP expression demonstrates significant correlations with multiple clinicopathological characteristics in cancer patients. In bladder cancer specifically, PAEP expression correlates with:

What are best practices for sample collection and preparation when studying PAEP?

Optimal sample collection and preparation for PAEP analysis requires careful attention to pre-analytical variables:

  • Sample timing: For reproductive studies, document menstrual cycle day or gestational age as PAEP levels fluctuate hormonally.

  • Collection containers: Use standardized tubes (preferably without additives for serum, or EDTA for plasma) to minimize interference.

  • Processing window: Process samples within 2 hours of collection to prevent degradation.

  • Centrifugation protocol: Employ consistent centrifugation parameters (typically 2000g for 10 minutes) to ensure comparable sample preparation.

  • Aliquoting strategy: Prepare multiple small aliquots to avoid freeze-thaw cycles.

  • Storage conditions: Store at -80°C for long-term preservation of protein integrity.

For tissue samples, immediate flash freezing or preservation in appropriate fixatives is essential for maintaining PAEP antigenicity. When working with cell culture supernatants, standardize collection timing relative to media changes to account for secretion kinetics.

How should researchers design controlled experiments for investigating PAEP function?

Robust experimental design for PAEP functional studies should incorporate:

  • Multiple experimental models: Employ complementary in vitro, ex vivo, and when possible, in vivo models to establish functional relevance across systems.

  • Dose-response assessments: Test PAEP at physiologically relevant concentrations (typically 1-100 ng/ml based on biological fluid measurements) and include concentration gradients to identify threshold effects.

  • Temporal dynamics: Assess both acute and prolonged PAEP exposure to distinguish between immediate signaling events and adaptive responses.

  • Genetic manipulation approaches: Utilize CRISPR-Cas9 or siRNA for gene knockout/knockdown experiments alongside recombinant protein addition to differentiate between endogenous and exogenous PAEP effects.

  • Isoform specificity: Account for glycosylation variants of PAEP that may exhibit differential activities by using appropriate glycoform-specific antibodies or recombinant proteins.

  • Comprehensive controls: Include vehicle controls, isotype-matched control antibodies, and when applicable, heat-inactivated PAEP to confirm specificity of observed effects.

Statistical power calculations should guide sample sizes, with consideration for potentially high biological variability in PAEP responses.

What qualitative research methodologies are applicable to studying PAEP in clinical settings?

While quantitative methods dominate PAEP research, integrating qualitative methodologies can provide contextual depth, particularly in clinical applications. Drawing from qualitative healthcare research frameworks, researchers might consider:

  • Phenomenological research: Exploring lived experiences of patients with conditions characterized by PAEP dysregulation, such as infertility or endometriosis .

  • Qualitative case studies: Detailed examination of unusual PAEP expression patterns in specific patients to generate hypotheses about novel functions .

  • Grounded theory: Developing theoretical frameworks about PAEP's role in disease progression based on patient and clinician observations .

  • Content analysis: Systematically analyzing clinical reports or patient narratives for patterns related to PAEP-associated conditions .

For validation of qualitative PAEP research, researchers should employ triangulation of data sources, member checking with participants, and peer debriefing with expert clinicians. Integration of qualitative findings with quantitative PAEP measurements can yield more holistic understanding of its clinical significance.

What statistical approaches are recommended for analyzing PAEP expression data in cancer studies?

Statistical analysis of PAEP expression in cancer studies should be carefully tailored to the research questions and data characteristics:

  • Expression comparisons: For comparing PAEP levels between groups, non-parametric tests (Mann-Whitney U or Kruskal-Wallis) are often appropriate given the typically non-normal distribution of expression data.

  • Survival analysis: Kaplan-Meier curves with log-rank tests should be used to assess survival differences between high and low PAEP expression groups, with cutpoints determined through methodologically sound approaches such as receiver operating characteristic (ROC) curve analysis or minimum p-value approaches .

  • Multivariable modeling: Cox proportional hazards models should adjust for established prognostic factors, as demonstrated in bladder cancer studies where PAEP remained independently significant (P = 0.045) alongside age, pathologic stage, and other clinical parameters .

  • Correlation analyses: Spearman's rank correlation is preferable for assessing relationships between PAEP and continuous variables (immune markers, other biomarkers) due to robustness against outliers and non-linearity.

  • Multiple testing correction: When assessing PAEP correlations with multiple features, appropriate methods such as Benjamini-Hochberg false discovery rate should be applied.

The analysis should be complemented with proper visualization techniques, including forest plots for multivariate analyses and heatmaps for correlation patterns.

How can researchers effectively interpret discrepancies in PAEP measurements across different studies?

When confronting discrepancies in PAEP measurements across studies, researchers should systematically evaluate potential sources of variation:

  • Detection methodology differences: ELISA kits vary in antibody specificity, detection ranges (7.81-500 pg/ml vs. 1.563-100 ng/ml), and sensitivity (3 pg/ml vs. 0.938 ng/ml) .

  • Sample processing variations: Inter-study differences in collection tubes, processing times, and storage conditions affect PAEP stability and measurement.

  • Population heterogeneity: Demographic factors, disease stage distribution, treatment history, and genetic background influence baseline PAEP levels and associations.

  • Reporting conventions: Some studies report raw concentrations while others use normalized values (z-scores, percentiles, etc.).

  • Statistical approach differences: Varying cutpoints for "high" versus "low" expression can dramatically affect reported associations.

To reconcile these discrepancies, researchers should:

  • Conduct meta-analyses with random-effects models to account for between-study heterogeneity

  • Perform sensitivity analyses excluding outlier studies

  • When possible, reanalyze raw data using standardized methods across studies

  • Report results with multiple statistical approaches to demonstrate robustness

What approaches should be used to integrate PAEP expression data with other -omics datasets?

Integration of PAEP expression with other -omics data requires sophisticated computational approaches:

  • Multi-omics correlation analysis: Beyond simple pairwise correlations, researchers should employ methods like similarity network fusion or canonical correlation analysis to identify patterns across data types.

  • Pathway enrichment integration: GSEA has revealed PAEP's association with immune-related pathways including JAK-STAT signaling, T cell receptor signaling, and PD1 blockade response . These findings should guide focused integration with corresponding phosphoproteomic or transcriptomic signatures.

  • Causal network inference: Bayesian network approaches can help establish potential causal relationships between PAEP expression and downstream molecular changes.

  • Machine learning frameworks: Supervised learning methods (random forests, support vector machines) can identify multi-omics signatures that, in combination with PAEP, enhance predictive power for outcomes of interest.

  • Data visualization techniques: Dimension reduction methods (t-SNE, UMAP) can visualize complex relationships between PAEP and high-dimensional -omics data.

When interpreting integrated analyses, researchers should consider the temporal relationships between measurements and biological processes, as some molecular changes may precede or follow PAEP expression changes.

How can researchers effectively study the glycosylation patterns of PAEP and their functional significance?

PAEP's glycosylation patterns critically influence its biological activities. To study these patterns:

  • Glycoform separation: Employ lectin affinity chromatography or isoelectric focusing to isolate different PAEP glycoforms before functional testing.

  • Glycoproteomics analysis: Use mass spectrometry with electron transfer dissociation to characterize site-specific glycosylation patterns, identifying both N-linked and O-linked modifications.

  • Glycosylation inhibitors: Apply tunicamycin (N-glycosylation inhibitor) or benzyl-α-GalNAc (O-glycosylation inhibitor) in expression systems to produce PAEP with altered glycosylation for functional comparison.

  • Glycosidase treatments: Systematically remove specific sugar moieties using endoglycosidases and exoglycosidases to map structure-function relationships.

  • Glycoengineering: Express PAEP in different cell types (CHO, HEK293, insect cells) that produce distinct glycosylation patterns to assess functional consequences.

  • Mutational analysis: Generate site-directed mutants at predicted glycosylation sites to study contribution of specific glycans to PAEP function.

These approaches should be combined with functional assays (immune cell modulation, receptor binding, etc.) to establish glycosylation-function relationships.

What are the current hypotheses regarding PAEP's role in modulating immune checkpoint pathways in cancer?

Current research indicates significant connections between PAEP and immune checkpoint pathways in cancer, generating several mechanistic hypotheses:

  • Direct checkpoint regulator: PAEP may directly interact with checkpoint molecules (PD-1/PD-L1), modulating their expression or signaling. GSEA analysis of bladder cancer samples has shown enrichment of "Cancer Immunotherapy By PD1 Blockade" pathway in PAEP-high tumors .

  • Immune microenvironment modifier: PAEP might alter the tumor immune microenvironment, affecting infiltration and activation status of T cells, NK cells, and myeloid populations. This is supported by enrichment of multiple immune-related pathways in PAEP-high tumors .

  • Cytokine network influencer: PAEP may modify cytokine production profiles, indirectly affecting checkpoint expression. The association with "Cytokine-Cytokine Receptor Interaction" pathway supports this hypothesis .

  • Antigen presentation modulator: PAEP's correlation with "Antigen Processing And Presentation" pathways suggests it might affect the visibility of tumor cells to the immune system .

  • Signal transduction effector: Enrichment in JAK-STAT and MAPK signaling in PAEP-high tumors indicates potential roles in transducing immunomodulatory signals .

Testing these hypotheses requires integrated approaches combining in vitro immune co-culture systems, in vivo models, and comprehensive analysis of patient samples, particularly those undergoing immunotherapy.

What emerging technologies might enhance future PAEP research?

Several cutting-edge technologies promise to advance PAEP research:

  • Single-cell multi-omics: Integrated single-cell RNA/protein profiling can reveal cell-specific PAEP production and response patterns, particularly in heterogeneous tissues like tumors or the endometrium.

  • Spatial transcriptomics/proteomics: Technologies like Visium, CODEX, or imaging mass cytometry can map PAEP expression and effects within tissue microenvironments, providing crucial spatial context.

  • Organoid models: Patient-derived organoids from reproductive tissues or tumors offer physiologically relevant systems for studying PAEP function in controlled environments.

  • CRISPR screening: Genome-wide or targeted CRISPR screens can identify genes that modulate PAEP expression or mediate its biological effects.

  • Protein interaction proteomics: BioID or APEX2 proximity labeling coupled with mass spectrometry can map PAEP's protein interaction networks in living cells.

  • AI-driven predictive modeling: Machine learning approaches can integrate PAEP data with clinical parameters to enhance predictive power for treatment responses or disease outcomes.

  • Digital pathology with AI analysis: Automated quantification of PAEP in tissue samples using deep learning algorithms can standardize assessment and reveal subtle patterns invisible to human observers.

Researchers should consider forming interdisciplinary collaborations to leverage these technologies effectively, as they often require specialized expertise beyond traditional biomedical research domains.

Product Science Overview

Gene and Protein Structure

The PAEP gene is located on the long arm of chromosome 9 and encodes a protein that is secreted in various forms, each with distinct glycosylation profiles. These forms are found in amniotic fluid, follicular fluid, and seminal plasma . The protein has been referred to by several names, including pregnancy-associated endometrial alpha-2-globulin, placental protein 14, and glycodelin .

Function and Expression

PAEP is primarily expressed in the female reproductive system, particularly in the endometrium during the secretory phase of the menstrual cycle. Its expression is regulated by progesterone, a hormone produced by the corpus luteum after ovulation . The protein plays a vital role in preparing the endometrium for implantation of the blastocyst (fertilized egg) and in regulating the uterine environment to support pregnancy .

Recombinant Production

Recombinant human PAEP is produced using genetic engineering techniques. The gene encoding PAEP is inserted into a suitable expression system, such as bacteria or mammalian cells, which then produce the protein. The recombinant protein is purified and used for various research and clinical applications .

Clinical Significance

PAEP has immunomodulatory effects and is involved in critical steps during fertilization. It has been associated with several reproductive disorders, including ectopic pregnancy and Kleefstra Syndrome 1 . The protein’s ability to modulate the immune response makes it a potential target for therapeutic interventions in reproductive health.

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.