PDAP1 Human

PDGFA Associated Protein 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

PDAP1 Recombinant Human produced in E.Coli is a single, non-glycosylated polypeptide chain containing 189 amino acids (1-181 a.a.) and having a molecular mass of 21.7 kDa.
The PDAP1 is fused to an 8 amino acid His-Tag at C-terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
PDAP1 enhances PDGFA-stimulated cell growth in fibroblasts but inhibits the growth-promoting effects of PDGFB. TNF-alpha induces PDAP1 expression, and cells with high PDAP1 levels exhibit increased resistance to TNF-alpha-induced apoptosis.
Description
Recombinant Human PDAP1, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 189 amino acids (1-181 a.a.) with a molecular weight of 21.7 kDa. This protein is engineered with an 8 amino acid His-Tag at the C-terminus and purified using proprietary chromatographic methods.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The PDAP1 solution is provided at a concentration of 1mg/ml in a buffer consisting of 20mM Tris-HCl (pH 8), 2mM DTT, 0.1M NaCl, 0.1mM PMSF, and 20% glycerol.
Stability
For short-term storage (up to 2-4 weeks), keep at 4°C. For long-term storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 90.0% as determined by SDS-PAGE analysis.
Synonyms
PAP1, HASPP28.
Source
Escherichia Coli.
Amino Acid Sequence
MPKGGRKGGH KGRARQYTSP EEIDAQLQAE KQKAREEEEQ KEGGDGAAGD PKKEKKSLDS DESEDEEDDY QQKRKGVEGL IDIENPNRVA
QTTKKVTQLD LDGPKELSRR EREEIEKQKA KERYMKMHLA GKTEQAKADL ARLAIIRKQR EEAARKKEEE RKAKDDATLS GKRMQSLSLN
KLEHHHHHH.

Q&A

What is PDAP1 and what is its basic function in human cells?

PDAP1 (PDGFA-associated protein 1), also known as the 28 kDa heat- and acid-stable phosphoprotein, is highly conserved across vertebrate taxa. It was originally characterized as a novel casein kinase II substrate in the rat brain and was associated with platelet-derived growth factor (PDGF)-A with low affinity in a rat neural retina cell line . Recent studies have identified PDAP1 as an essential regulator of cellular homeostasis in mature B cells, protecting them from stress-induced cell death and promoting antibody gene diversification . PDAP1 functions as an RNA-binding protein according to several RNA-protein interactome studies . The protein plays a critical role in countering chronic activation of the integrated stress response (ISR), which otherwise would lead to cell death through proapoptotic pathways .

How is PDAP1 expression regulated in normal and pathological conditions?

In colorectal cancer cells, c-Myc has been identified as a direct transcriptional regulator of PDAP1. Mechanistic studies have demonstrated that c-Myc directly transactivates PDAP1, which contributes to the high PDAP1 expression observed in CRC cells . This represents a key regulatory mechanism in pathological conditions. Single-cell genomic analysis has revealed differential expression patterns between normal and cancerous tissues, with PDAP1 being significantly overexpressed in CRC compared to paired paracancerous tissues . In normal B cells, PDAP1 expression appears necessary for maintaining cellular homeostasis, though the specific transcriptional regulators in this context haven't been fully characterized in the available data .

What is the evolutionary conservation pattern of PDAP1 across species?

PDAP1 demonstrates high evolutionary conservation across vertebrate taxa, suggesting fundamental biological importance . This conservation pattern indicates that PDAP1 likely serves essential cellular functions that have been preserved throughout vertebrate evolution. The protein structure and functional domains appear to be maintained across species, allowing for translational research using mouse models with potential relevance to human biology. The high degree of conservation further supports PDAP1's role in basic cellular processes like stress response regulation and potentially explains its involvement in both normal physiology and disease states .

What evidence supports PDAP1's role in colorectal cancer development?

Multi-omics data analysis has identified PDAP1 as a potential key player in CRC development. Immunohistochemistry studies on two independent cohorts of CRC biopsies revealed significantly higher PDAP1 expression in cancer tissues compared to adjacent normal tissues . The table below summarizes these findings:

CohortPDAP1 Positive Rate in CRCPDAP1 Positive Rate in Adjacent Normal Tissuep-value
Cohort 1 (n=280)83.57% (234/280)23.93% (67/280)<0.01
Cohort 2 (n=75)96.00% (72/75)25.33% (19/75)<0.01

Survival analysis demonstrated that patients with high PDAP1 expression had worse outcomes than those with low expression . Functional studies with PDAP1 knockdown showed decreased cell proliferation, migration, invasion, and colony formation in CRC cell lines, while overexpression enhanced these tumorigenic properties . In vivo xenograft assays confirmed reduced tumor progression and metastatic potential with PDAP1 knockdown .

What molecular mechanisms underlie PDAP1's contribution to cancer progression?

PDAP1 contributes to cancer progression through several molecular mechanisms:

  • EGFR-MAPK Pathway Activation: PDAP1 interacts with the juxtamembrane domain of epidermal growth factor receptor (EGFR) and facilitates EGFR-mitogen-activated protein kinase (MAPK) signaling activation .

  • FRA-1 Expression Regulation: The activated EGFR-MAPK signaling results in FOS-related antigen 1 (FRA-1) expression, thereby facilitating CRC progression .

  • Cell Cycle Regulation: PDAP1 knockdown arrests the cell division cycle during the S/G2 phase transition and delays the completion of DNA replication, as evidenced by weak and prolonged PCNA-cb foci compared to control cells .

  • c-Myc Target Activation: PDAP1 is directly transactivated by c-Myc, which is frequently dysregulated in cancers, creating a potential oncogenic feedback loop .

These mechanisms collectively contribute to enhanced proliferation, migration, invasion, and metastasis of CRC cells.

How can researchers design experiments to evaluate PDAP1 as a therapeutic target?

Researchers can design comprehensive experiments to evaluate PDAP1 as a therapeutic target through the following approaches:

  • Patient-Derived Xenograft (PDX) Models: Establish PDX models as demonstrated in previous studies where in vivo-optimized small interfering RNAs (siRNAs) targeting PDAP1 were administered to mice with implanted patient tumors . Monitor tumor growth after PDAP1 silencing through multiple treatment rounds.

  • Conditional Knockout Mouse Models: Use Pdap1 conditional knockout mice (Pdap1^fl/fl) to establish CRC mouse models and evaluate tumor development and progression in the absence of PDAP1 .

  • Combination Therapy Assessment: Test PDAP1 inhibition in combination with established CRC therapies to identify potential synergistic effects.

  • Biomarker Development: Correlate PDAP1 expression levels with treatment response to identify patient subgroups most likely to benefit from PDAP1-targeted therapies.

  • Target Validation Methods: Employ RNA sequencing, phosphoprotein antibody arrays, and interactome analysis to identify downstream effects of PDAP1 inhibition and potential resistance mechanisms .

  • Pharmacological Approaches: Develop small molecule inhibitors targeting PDAP1 or its interaction with EGFR, and validate their efficacy in preclinical models.

How does PDAP1 regulate B cell function and antibody diversification?

PDAP1 serves as an essential regulator of mature B cell physiology through several key mechanisms:

  • Protection from Chronic Stress: PDAP1 counters chronic activation of the integrated stress response (ISR), preventing sustained expression of activating transcription factor 4 (Atf4) and subsequent cell death .

  • Class Switch Recombination (CSR) Support: CRISPR-Cas9–mediated deletion of PDAP1 in CH12 cells resulted in considerable reduction of CSR efficiency. Both indel knockout (Pdap1^−/−) and in-frame deletion mutant (Pdap1^mut) clones showed impaired CSR .

  • Antibody Isotype Switching: Primary B cells isolated from Pdap1^F/F^Cd19^Cre/+^ mice displayed reduced levels of switching to multiple isotypes (IgG1, IgG3, IgG2b, and IgA) under various stimulation conditions .

  • AID Expression Regulation: PDAP1 is essential for efficient induction of activation-induced cytidine deaminase (AID) expression, which is required for both CSR and somatic hypermutation (SHM) .

  • Germinal Center Formation: Loss of Pdap1 reduces germinal center B cell formation, which is critical for proper humoral immune responses .

The combined effect of these mechanisms makes PDAP1 crucial for establishing protective humoral immunity.

What methodological approaches can researchers use to study PDAP1's role in B cell stress response?

Researchers can employ several methodological approaches to study PDAP1's role in B cell stress response:

  • Conditional Gene Targeting: Generate B cell-specific Pdap1 knockout models by breeding Pdap1^F/F^ mice with Cd19^Cre/+^ mice to specifically ablate Pdap1 expression at early stages of B cell differentiation .

  • In Vitro B Cell Stimulation Assays: Isolate resting splenocytes from Pdap1^F/F^Cd19^Cre/+^ mice and stimulate them under various conditions to assess their capability to undergo class switch recombination to different isotypes .

  • Integrated Stress Response (ISR) Monitoring: Measure Atf4 expression levels and activation of the ISR transcriptional program using RNA sequencing and protein expression analysis in Pdap1-deficient versus control B cells .

  • Cell Death Assays: Quantify cell death rates in Pdap1-deficient B cells exposed to various stressors to determine the protective role of PDAP1 .

  • AID Expression Analysis: Assess AID levels using techniques like western blotting and qPCR to understand how PDAP1 regulates this key enzyme for antibody diversification .

  • Germinal Center B Cell Analysis: Use flow cytometry to quantify germinal center B cell formation in Pdap1-deficient mice following immunization .

What multi-omics approaches are most suitable for comprehensive PDAP1 function analysis?

For comprehensive analysis of PDAP1 function, researchers should consider integrating these multi-omics approaches:

  • Single-Cell RNA Sequencing: Particularly valuable for understanding PDAP1's role in heterogeneous tumor ecosystems, as demonstrated in studies identifying PDAP1 dysregulation in CRC . This technique can reveal cell type-specific functions and effects.

  • Proteomics Analysis: Mass spectrometry-based proteomics can identify PDAP1 interactors and post-translational modifications. Previous studies used an LTQ-Orbitrap hybrid mass spectrometer with Proteome Discoverer software to analyze proteins eluted from His pull-down assays with His6-tagged PDAP1 as bait .

  • Phosphoproteomics: Since PDAP1 was originally characterized as a casein kinase II substrate, phosphoprotein antibody arrays can help identify downstream signaling events affected by PDAP1 modulation .

  • RNA-Protein Interaction Profiling: Given PDAP1's identification as an RNA-binding protein, techniques like CLIP-seq (Cross-linking immunoprecipitation followed by sequencing) can identify its RNA targets and binding motifs .

  • Integrated Bioinformatics Analysis: Combining datasets from The Cancer Genome Atlas (TCGA) with experimental data can provide comprehensive insights into PDAP1's role in cancer and normal physiology .

How can researchers effectively model PDAP1-related phenotypes in animal systems?

Researchers can effectively model PDAP1-related phenotypes through several sophisticated animal approaches:

  • Conditional Knockout Strategies: Generate tissue-specific Pdap1 knockout mice, as demonstrated with the Pdap1^fl/fl^ system where loxP fragments were inserted into introns 2 and 4, allowing conditional removal of exons 3 and 4 using Cre recombinase . This approach allows investigation of PDAP1 function in specific tissues while avoiding potential embryonic lethality.

  • Inducible Systems: Implement temporally controlled Pdap1 deletion using inducible Cre systems to study PDAP1's role at different developmental stages or disease progression points.

  • Disease-Specific Models:

    • Colitis and CRC Models: Utilize dextran sulfate sodium salt-induced colitis and colitis-associated cancer models in Pdap1 conditional knockout mice to study its role in inflammation and cancer .

    • Immunological Challenge Models: Immunize Pdap1-deficient mice to examine germinal center formation and antibody responses .

  • Humanized Models: Develop patient-derived xenograft (PDX) models in immunodeficient mice to test PDAP1-targeting therapies, as demonstrated in the research where in vivo-optimized siRNAs were used to silence PDAP1 .

  • Metastasis Models: Employ tail vein injection assays with PDAP1-modulated cells to investigate its influence on metastatic potential .

What are the current contradictions or knowledge gaps in PDAP1 research?

Several significant knowledge gaps and potential contradictions exist in current PDAP1 research:

  • Dual Role in Different Cell Types: PDAP1 promotes cancer cell survival and proliferation but also protects normal B cells from stress-induced death . This apparent contradiction requires further investigation to understand context-dependent functions.

  • Regulatory Mechanisms: While c-Myc has been identified as a direct transcriptional regulator of PDAP1 in CRC cells , regulatory mechanisms in normal tissues and other cell types remain poorly understood.

  • RNA-Binding Function: Despite being identified as an RNA-binding protein in several studies , the specific RNA targets and functional significance of this activity remain undefined.

  • Therapeutic Targeting Challenges: Inhibiting PDAP1 for cancer therapy might impair normal immune function, given its role in B cells . This potential trade-off needs thorough evaluation.

  • Upstream Activators: The signaling events that activate or repress PDAP1 function beyond transcriptional regulation are largely unknown.

  • Post-Translational Modifications: Despite being identified as a phosphoprotein, the specific kinases (beyond casein kinase II) and phosphatases regulating PDAP1 activity through post-translational modifications are not well characterized.

How might PDAP1's diverse functions be integrated into a unified molecular model?

A unified molecular model of PDAP1 function could integrate its diverse roles through the following framework:

  • Stress Response Hub: PDAP1 appears to function centrally as a regulator of cellular stress responses across different cell types. In both cancer cells and B cells, it protects against chronic stress activation but through potentially different downstream mechanisms .

  • Cell Type-Specific Interactome: PDAP1 may interact with different partner proteins depending on cellular context:

    • In cancer cells: Interaction with EGFR promotes MAPK signaling and FRA-1 expression

    • In B cells: Interactions that regulate the integrated stress response and AID expression

  • Transcriptional Regulation Circuits: Different transcriptional programs may control PDAP1 expression:

    • c-Myc driven in cancer cells

    • Potentially different regulators in immune cells and normal tissues

  • RNA-Binding Functionality: PDAP1's RNA-binding capacity may contribute to post-transcriptional regulation of different target mRNAs in various cellular contexts .

  • Signal Integration Model: PDAP1 might function as a signal integrator that senses cellular stress levels and modulates survival/proliferation pathways accordingly.

This unified model would need experimental validation through comparative interactome studies across cell types and contexts.

What innovative approaches might accelerate therapeutic applications targeting PDAP1?

Several innovative approaches could accelerate therapeutic applications targeting PDAP1:

  • Context-Specific Inhibition Strategies: Develop inhibitors that selectively target PDAP1 in cancer cells while sparing its function in normal cells, possibly by targeting cancer-specific post-translational modifications or protein-protein interactions .

  • Domain-Specific Targeting: Identify and target specific functional domains of PDAP1 that are critical for its oncogenic functions but dispensable for its normal physiological roles.

  • RNA-Based Therapeutics: Design RNA-based therapeutics that modulate PDAP1 expression, such as the in vivo-optimized siRNAs that have shown efficacy in PDX models .

  • Combination Therapy Approaches: Develop rational combination strategies pairing PDAP1 inhibition with existing therapies, particularly those targeting the EGFR-MAPK pathway that PDAP1 has been shown to facilitate .

  • Biomarker-Driven Patient Selection: Establish PDAP1 expression or activation signatures as biomarkers to identify patients most likely to benefit from PDAP1-targeted therapies.

  • Proteolysis-Targeting Chimeras (PROTACs): Design PROTACs to selectively degrade PDAP1 in cancer cells by taking advantage of cancer-specific E3 ligases.

  • Allosteric Modulators: Develop compounds that bind to allosteric sites on PDAP1 to modulate its function rather than completely inhibiting it, potentially offering more nuanced control of its activity.

Product Science Overview

Gene and Protein Structure

The PDAP1 gene is located on chromosome 7 and encodes a protein that is approximately 28 kDa in size . The protein is characterized by its heat and acid stability, which makes it resilient under various physiological conditions. PDAP1 is a phosphoprotein, meaning it undergoes phosphorylation, a process that can alter its activity and function within the cell .

Function and Mechanism

PDAP1 is known to enhance the growth of fibroblasts stimulated by Platelet-Derived Growth Factor A (PDGFA) . Fibroblasts are cells that play a crucial role in wound healing and tissue repair. PDAP1 achieves this by binding to PDGFA with low affinity, thereby promoting cell proliferation . Interestingly, PDAP1 also has an inhibitory effect on the mitogenic activity of Platelet-Derived Growth Factor B (PDGFB), which suggests a regulatory role in balancing the effects of different growth factors .

Expression and Localization

PDAP1 is predominantly localized in the cytosol of cells, with additional presence at the plasma membrane . Its expression is observed in various tissues, indicating its widespread role in cellular functions. The protein’s localization to the cytosol and plasma membrane suggests its involvement in intracellular signaling pathways and interactions with membrane-bound receptors .

Clinical Significance

The regulatory functions of PDAP1 in cell growth and proliferation make it a protein of interest in the study of cancer and other proliferative disorders. Abnormal expression or mutations in the PDAP1 gene could potentially lead to dysregulated cell growth, contributing to the development of tumors . Additionally, PDAP1’s role in fibroblast proliferation highlights its potential importance in wound healing and tissue regeneration research .

Research and Applications

Recombinant PDAP1 is used in various research applications to study its function and interactions with other proteins. By using human recombinant PDAP1, researchers can investigate its role in cellular processes under controlled conditions, providing insights into its potential therapeutic applications .

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.