PIP Human

Prolactin-Induced Protein Human
Shipped with Ice Packs
In Stock

Description

Introduction to Prolactin-Induced Protein (PIP)

Prolactin-Induced Protein (PIP), also known as Gross Cystic Disease Fluid Protein-15 (GCDFP-15), is a 17 kDa glycoprotein encoded by the PIP gene in humans. It is primarily expressed in tissues responsive to hormonal signals, including apocrine glands, lacrimal glands, and breast epithelium. PIP has garnered attention for its dual roles in physiological processes (e.g., immune modulation) and pathological contexts, particularly in breast cancer progression and therapy resistance .

Table 1: Key Molecular Properties of PIP

PropertyValue/DescriptionSource
Molecular Weight17 kDa
Isoelectric Point (pI)4.7
Binding PartnersCD4, HLA-DR, Fibronectin
Expression SitesBreast, Salivary Glands, Prostate

Biological Functions and Mechanisms

PIP exhibits multifunctional roles across physiological and pathological systems:

Immune Regulation

  • Adaptive Immunity: Binds to CD4⁺ T-cell receptors and MHC-II molecules, modulating antigen presentation and T-cell activation .

  • Cytokine Interactions: Expression is upregulated by IL-4 and IL-13, linking it to Th2-mediated immune responses .

Table 2: PIP-Mediated Pro-Apoptotic Gene Regulation in BC

GeneFunctionFold Change (PIP⁺ vs. PIP⁻)
CRADDCaspase recruitment domain adaptor3.2×
DAPK1Serine/threonine kinase2.8×
FASLGDeath receptor ligand4.1×
Data derived from in vitro models of MDA-MB-231 BC cells .

Diagnostic and Prognostic Utility

  • Biomarker Potential: PIP expression inversely correlates with tumor grade and triple-negative BC status, serving as a favorable prognostic marker .

  • Therapeutic Target: Soluble PIP activates extrinsic apoptosis pathways via surface receptors, suggesting utility in overcoming chemoresistance .

Challenges and Controversies

  • Dual Roles in Cancer: While PIP sensitizes BC cells to chemotherapy, it also promotes adhesion to fibronectin, potentially facilitating metastasis in advanced stages .

Research Advancements and Future Directions

Recent studies highlight PIP’s involvement in novel pathways:

  • Immune Checkpoint Modulation: PIP-CD4 interactions may suppress T-cell hyperactivation, offering avenues for autoimmune disease therapy .

  • Nanoparticle Delivery: Encapsulation of recombinant PIP improves blood-brain barrier penetration in preclinical neurological models, expanding its therapeutic scope .

Table 3: Emerging PIP-Associated Pathways

PathwayMechanismDisease Relevance
PI3K/AKT SuppressionDownregulates PI(3,4,5)P₃ signalingBreast Cancer
CD4-MHC-II InteractionInhibits antigen recognitionAutoimmunity

Product Specs

Introduction
Prolactin inducible protein (PIP), a 17kDa glycoprotein found in human seminal plasma, is synthesized as a 146 amino acid polypeptide. PIP shares a high degree of sequence similarity with mouse submaxillary gland and possesses a single glycosylation site. While its exact biological functions remain unclear, PIP is thought to play various roles due to its high concentration in biological fluids. It exhibits binding affinity for proteins like fibrinogen, actin, keratin, myosin, and tropomyosin. PIP expression is also observed in pathological conditions of the mammary gland and certain exocrine tissues, including lacrimal, salivary, and sweat glands. Its association with secretory cell differentiation has led to its use in diagnosing breast, salivary gland, and skin tumors.
Description
Derived from Human Seminal Plasma, the produced Prolactin-Induced Protein has a molecular mass of 13.52kDa (excluding glycosylation) and comprises 118 amino acid residues.
Physical Appearance
White, lyophilized powder after filtration.
Formulation
The PIP protein undergoes filtration (0.4µm) and lyophilization in a solution containing 0.5mg/ml protein, 0.05M phosphate buffer, and 0.075M NaCl at a pH of 8.0.
Solubility
To prepare a working stock solution, add deionized water to the lyophilized pellet to achieve a concentration of approximately 0.5 mg/ml and allow for complete dissolution. Note: PIP is not sterile. Prior to use in cell culture, filter the product through an appropriate sterile filter.
Stability
Store the lyophilized protein at -20°C. After reconstitution, aliquot the product to prevent repeated freeze-thaw cycles. The reconstituted protein remains stable at 4°C for a limited period; no changes are observed after two weeks at 4°C.
Purity
SDS-PAGE analysis indicates a purity greater than 95%.
Human Virus Test
Donor samples have been tested and confirmed negative for HBsAg, HIV1+2, HCV, syphilis, aHBc, and RRR.
Synonyms
Prolactin-inducible protein, Gross cystic disease fluid protein 15, GCDFP-15, Prolactin-induced protein, Secretory actin-binding protein, SABP, gp17, PIP, GCDFP15, GPIP4.
Source
Human Seminal Plasma.
Amino Acid Sequence
QDNTRKIIIK NFDIPKSVRP NDEVTAVLAV QTELKECMVV KTYLISSIPL QGAFNYKYTA CLCDDNPKTF YWDFYTNRTV QIAAVVDVIR ELGICPDDAA VIPIKNNRFY TIEILKVE.

Q&A

What is the PIP compound and how does it interact with human DNA?

Pyrrole-imidazole polyamides (PIPs) are small molecules specifically designed to bind to minor grooves in the DNA helix. These compounds can recognize and bind to specific DNA nucleic acid sequences, functioning similarly to transcription factors that regulate gene expression. PIPs have generated significant interest for their potential to turn genes on and off, making them promising candidates for developing new treatments for cancers and hereditary diseases .

The binding mechanism involves:

  • Sequence-specific recognition of DNA base pairs

  • Formation of hydrogen bonds with DNA bases

  • Insertion into the minor groove of the DNA double helix

  • Potential disruption or enhancement of transcription factor binding

What are the fundamental mechanisms of PIP compounds in human applications?

PIPs operate through several key mechanisms in human cellular systems:

  • DNA Binding: PIPs bind to minor grooves found in the DNA helix, recognizing specific nucleic acid sequences .

  • Transcription Factor Mimicry: They can mimic and potentially disrupt transcription factor pairs from binding to DNA, resulting in various biological effects .

  • Epigenetic Regulation: When combined with epigenetic regulators (forming "ePIP-HoGu" systems), they can mark specific DNA sequences for epigenetic modification .

  • Gene Expression Modulation: Depending on their design and target sequence, PIPs can either inhibit or activate gene expression.

The versatility of these mechanisms makes PIPs valuable tools for both research and potential therapeutic applications.

How are PIP compounds designed for human research applications?

The design of PIP compounds for human research involves sophisticated molecular engineering approaches:

  • Structural Optimization: Researchers select molecules that strongly bind to DNA while maintaining favorable pharmacological properties (non-toxicity, cell-permeability, water-solubility, and chemical stability) .

  • Sequence Targeting: Scientists fine-tune the molecules to target specific DNA nucleic acid sequences with flexible gap spacings .

  • Host-Guest Assembly: Some advanced PIPs are combined with a 'host-guest assembly' (HoGu) that can strongly bind to DNA and act similarly to transcription factors .

  • Functional Conjugation: For epigenetic applications, PIPs are attached to epigenetic regulator molecules to form "ePIP-HoGu" systems that can mark specific sequences for modification .

The rational design process allows researchers to create increasingly specific and effective DNA-targeting molecules for human applications.

How do PIP-HoGu systems enable precise epigenetic regulation in human cells?

PIP-HoGu systems represent an advancement in epigenetic regulation technology through a multi-component approach:

  • Component Integration: The system combines a DNA-binding PIP with a host-guest assembly (HoGu) and an epigenetic regulator molecule .

  • Sequential Functionality:

    • The PIP component provides sequence-specific DNA targeting

    • The HoGu component enhances binding strength and stability

    • The epigenetic regulator component modifies the chromatin state at the target site

  • Enhanced Specificity: Studies have shown that ePIP-HoGu systems more specifically bind to targeted nucleic acid sequences and efficiently mark them for epigenetic modification compared to conventional approaches .

  • Precision Control: This system allows researchers to target specific genomic locations for epigenetic modification, potentially enabling more precise control over gene expression patterns .

This methodological approach opens new possibilities for studying and potentially treating conditions with epigenetic dysregulation.

What challenges exist in translating PIP research from in vitro to in vivo human applications?

Translating PIP research from laboratory settings to human applications involves several significant challenges:

Challenge CategorySpecific IssuesMethodological Approaches
DeliveryCell membrane penetration, tissue-specific targeting, nuclear localizationConjugation with cell-penetrating peptides, nanoparticle formulations, targeted delivery systems
StabilityMetabolic degradation, serum protein binding, excretion kineticsChemical modifications, protective formulations, pharmacokinetic optimization
SpecificityOff-target binding, competition with endogenous DNA-binding proteinsIterative design refinement, genome-wide binding analysis, selectivity screening
EfficacyAchieving sufficient target occupation, overcoming chromatin accessibility barriersDose optimization, combination with chromatin-opening agents, cell-specific targeting
SafetyPotential toxicity, immunogenicity, long-term effectsRigorous toxicology studies, biodistribution analysis, long-term follow-up studies

Addressing these challenges requires interdisciplinary approaches combining chemistry, molecular biology, pharmacology, and clinical medicine.

How can contradictory data on PIP efficacy in different human cell types be reconciled?

Contradictory findings regarding PIP efficacy across different human cell types can be methodologically reconciled through:

  • Chromatin Accessibility Analysis: Different cell types present unique chromatin landscapes affecting DNA accessibility. Systematic comparison of chromatin states using techniques like ATAC-seq can reveal why PIPs may function effectively in some cells but not others.

  • Cellular Uptake Quantification: Variation in nuclear transport mechanisms between cell types can be assessed through quantitative imaging of labeled PIPs to identify cell-specific uptake differences.

  • Transcription Factor Competition Mapping: Cell-type-specific transcription factors may compete with PIPs for identical DNA binding sites. ChIP-seq analysis can identify potential competitive binding factors.

  • Epigenetic Landscape Integration: Different epigenetic modifications may affect PIP binding efficacy across cell types. Correlation analysis between epigenetic marks and PIP activity can reveal patterns explaining variable efficacy.

  • Systematic PIP Derivative Testing: Developing panels of structurally distinct PIP variants for testing across multiple cell types can identify specific features conferring cell-type specificity.

This multi-dimensional analytical approach transforms contradictory observations into valuable insights about cell-specific factors influencing PIP function.

What methodological approaches best evaluate PIP-mediated gene expression changes in human cells?

Evaluating PIP-mediated gene expression changes requires comprehensive methodological approaches:

  • Transcriptomic Analysis:

    • RNA-seq for genome-wide expression changes

    • Quantitative PCR for targeted gene expression measurement

    • Single-cell RNA-seq to assess cellular heterogeneity in response

  • Chromatin Interaction Assessment:

    • ChIP-seq to map PIP binding sites across the genome

    • CUT&RUN or CUT&TAG for higher resolution binding site identification

    • HiC or chromatin conformation capture to identify long-range interactions

  • Functional Validation:

    • Reporter gene assays to quantify promoter activity changes

    • CRISPR interference/activation to compare with PIP effects

    • Phenotypic assays to link gene expression changes to cellular functions

  • Temporal Analysis:

    • Time-course experiments to track the dynamics of gene expression changes

    • Nuclear run-on assays to measure nascent transcription rates

    • Protein half-life studies to distinguish transcriptional from post-transcriptional effects

Integration of these complementary approaches provides comprehensive understanding of how PIPs influence gene expression regulation in human cells.

What are optimal experimental controls for PIP compound studies in human cell models?

Robust experimental design for PIP studies requires comprehensive controls:

Control TypePurposeImplementation
Negative ControlsEstablish baseline cellular responsesUntreated cells, vehicle-only treatment, non-binding PIP analogs
Positive ControlsValidate experimental systemKnown transcription factor modulators, CRISPR-based gene regulators
Sequence Specificity ControlsConfirm target selectivityPIPs targeting mutated binding sites, scrambled sequence PIPs
Concentration ControlsEstablish dose-response relationshipMultiple PIP concentrations ranging from sub-effective to saturating
Temporal ControlsDetermine kinetics of PIP effectsTime-course sampling from immediate to extended time points
Cell Type ControlsAssess cell-specific responsesMultiple relevant cell lines, primary cells, isogenic cell lines with specific mutations
Technical ControlsEnsure methodological validityTechnical replicates, spike-in standards, batch effect controls

Implementation of this comprehensive control framework ensures reliable interpretation of PIP effects in human cell systems.

How should researchers approach PIP physical interaction prediction studies?

Physical interaction prediction studies involving PIPs require sophisticated methodological approaches:

  • Mental Simulation with Span Selection: Recent innovations like Physical Interaction Prediction via Mental Simulation with Span Selection (PIP) utilize deep generative models to simulate physical interactions before employing selective temporal attention for outcome prediction .

  • Attention-Based Mechanisms: The PIP model employs span selection as a temporal attention mechanism to focus on key physical interaction moments, providing both accuracy and interpretability advantages .

  • Experimental Design Considerations:

    • Use of multiscale simulations to capture interactions across different time and length scales

    • Implementation of selective attention to focus computational resources on physically relevant moments

    • Integration of both generative modeling and predictive analytics

  • Validation Approaches:

    • Comparison with human performance on identical tasks

    • Evaluation against baseline models and related intuitive physics approaches

    • Assessment using diverse interaction scenarios and object types

These approaches have shown promising results, with the PIP model outperforming human predictions, baseline models, and related intuitive physics models that utilize mental simulation .

What methodologies can identify PIP binding sites with highest precision in the human genome?

Identifying precise PIP binding sites in the human genome requires integrating multiple cutting-edge methodologies:

  • In Vitro Binding Assays:

    • Systematic Evolution of Ligands by Exponential Enrichment (SELEX)

    • High-throughput sequencing of PIP-bound DNA fragments

    • Competitive binding assays with known transcription factors

  • Genomic Mapping Techniques:

    • ChIP-seq adapted for small molecules using photo-crosslinking

    • Chem-seq for direct detection of small molecule binding sites

    • CUT&RUN or CUT&TAG for higher resolution and lower background

  • Computational Prediction:

    • Position Weight Matrix (PWM) models based on binding rules

    • Machine learning approaches trained on experimental binding data

    • Molecular dynamics simulations of PIP-DNA interactions

  • Validation Methods:

    • CRISPR-based genomic modifications of predicted binding sites

    • Functional assays to confirm biological relevance of binding

    • Cross-validation using orthogonal detection methods

Integration of these complementary approaches yields a comprehensive and high-confidence map of PIP binding sites throughout the human genome.

How can researchers analyze contradictory data in PIP-DNA binding studies?

When faced with contradictory data in PIP-DNA binding studies, researchers should implement a systematic analytical framework:

  • Methodological Reconciliation:

    • Compare experimental conditions across studies (buffer composition, temperature, pH)

    • Evaluate differences in PIP concentrations and purification methods

    • Assess detection method sensitivities and potential artifacts

  • Contextual Analysis:

    • Examine chromatin state differences between experimental systems

    • Consider competing DNA-binding factors present in different systems

    • Analyze DNA structural variations that might affect binding

  • Quantitative Assessment:

    • Perform meta-analysis when sufficient quantitative data is available

    • Develop mathematical models that can account for observed variations

    • Use Bayesian approaches to integrate prior knowledge with new data

  • Resolution Strategies:

    • Design bridging experiments that directly address discrepancies

    • Implement side-by-side comparisons under standardized conditions

    • Utilize orthogonal methods to validate controversial findings

This systematic approach transforms seemingly contradictory results into deeper insights about context-dependent PIP-DNA interactions.

What statistical approaches best evaluate PIP efficacy across different human tissue types?

Evaluating PIP efficacy across diverse human tissue types requires sophisticated statistical methodologies:

  • Hierarchical Mixed-Effects Models:

    • Account for tissue-specific, donor-specific, and experimental variation

    • Incorporate nested dependencies in experimental design

    • Enable identification of tissue-specific responses while controlling for confounding factors

  • Multivariate Analysis Techniques:

    • Principal Component Analysis (PCA) to identify patterns across tissues

    • Canonical Correlation Analysis to relate PIP properties to tissue responses

    • Cluster analysis to group tissues by response profiles

  • Meta-Analytical Approaches:

    • Random-effects models to integrate data across independent studies

    • Forest plots to visualize tissue-specific effect sizes

    • Publication bias assessment using funnel plots and Egger's test

  • Machine Learning Integration:

    • Random forest models to identify predictive features of tissue response

    • Support vector machines to classify responder/non-responder tissues

    • Neural networks to model complex tissue-specific response patterns

How should researchers interpret epigenetic changes induced by PIP-HoGu systems?

Interpretation of epigenetic changes induced by PIP-HoGu systems requires a comprehensive analytical framework:

  • Multi-Omics Integration:

    • Correlation of epigenetic modifications with transcriptional changes

    • Integration with chromatin accessibility data to assess functional impact

    • Protein-level validation of expression changes for key targets

  • Temporal Dynamics Analysis:

    • Time-course studies to distinguish primary from secondary effects

    • Persistence evaluation to determine stability of induced modifications

    • Reversal experiments to assess epigenetic memory

  • Specificity Assessment:

    • Genome-wide mapping of induced epigenetic changes

    • Comparison with predicted binding sites to identify off-target effects

    • Evaluation of effects on related gene families or pathways

  • Causal Relationship Establishment:

    • Directed epigenetic editing using orthogonal systems for validation

    • Genetic knockout/rescue experiments of affected pathways

    • Dose-response studies to establish quantitative relationships

This interpretive framework allows researchers to move beyond correlative observations to establish mechanistic understanding of PIP-HoGu-induced epigenetic changes.

What are the most promising applications of PIP compounds in human disease research?

PIP compounds show significant promise in several human disease research applications:

Disease CategoryPotential ApplicationsCurrent Research Status
CancerTargeted silencing of oncogenes, Activation of tumor suppressors, Disruption of fusion oncoproteinsPreclinical studies showing efficacy in multiple cancer types, Early transition to targeted delivery systems
Genetic DisordersAllele-specific targeting of dominant mutations, Activation of compensatory genes, Correction of splicing defectsProof-of-concept studies in cell models of several monogenic disorders, Animal model validation underway
Viral InfectionsTargeting of viral genomes, Blocking viral integration sites, Modulation of host restriction factorsDemonstrated in vitro efficacy against several DNA viruses, Studies on delivery to infected cells ongoing
Inflammatory ConditionsTargeted regulation of inflammatory gene expression, Modulation of immune cell differentiation pathwaysEarly-stage research showing potential in autoimmune disease models, Development of tissue-specific delivery approaches
Neurodegenerative DiseasesSilencing of toxic repeat expansions, Activation of neuroprotective factors, Modulation of neuroinflammationChallenges in BBB penetration being addressed, Promising results in cellular models

The translation of these applications from laboratory research to clinical development represents a major frontier in PIP research.

What methodological innovations could advance PIP compound research in human systems?

Several methodological innovations hold particular promise for advancing PIP research:

  • Advanced Delivery Systems:

    • Tissue-specific targeting using aptamer-conjugated PIPs

    • Stimuli-responsive nanoparticles for controlled release

    • Exosome-based delivery systems for enhanced cellular uptake

  • Multiplexed PIP Technologies:

    • Combinatorial PIP libraries for simultaneous targeting of multiple genes

    • Orthogonal PIP systems for independent regulation of separate pathways

    • Logic-gated PIP designs that respond to specific cellular conditions

  • Temporal Control Mechanisms:

    • Photoswitchable PIPs for light-controlled gene regulation

    • Chemically inducible systems for dose-dependent activation

    • Self-limiting PIPs with programmed degradation pathways

  • Integration with Other Technologies:

    • CRISPR-PIP hybrid systems combining targeting advantages

    • PIP-directed epigenetic editors for precise chromatin modification

    • Antibody-PIP conjugates for enhanced specificity

  • Advanced Analytical Approaches:

    • Single-molecule tracking of PIP interactions in living cells

    • Spatial transcriptomics to map PIP effects across tissue architecture

    • AI-driven design platforms for optimized PIP development

These innovations address current limitations and could substantially expand the research and therapeutic potential of PIP compounds.

How might computational advances enhance PIP design for human applications?

Computational advances are poised to transform PIP design through several key approaches:

  • AI-Driven Design Platforms:

    • Deep learning models trained on experimental binding data

    • Generative adversarial networks for novel PIP structure creation

    • Reinforcement learning systems optimizing for multiple parameters simultaneously

  • Molecular Dynamics Simulations:

    • Quantum mechanical calculations of PIP-DNA interactions

    • Microsecond-scale simulations of binding dynamics

    • Free energy calculations for binding affinity prediction

  • Systems Biology Integration:

    • Network modeling of PIP effects on gene regulatory networks

    • Multi-scale simulations linking molecular interactions to cellular outcomes

    • Predictive models of tissue-specific responses

  • Predictive Toxicology:

    • QSAR models for rapid toxicity prediction

    • Molecular docking with off-target proteins

    • Simulation of metabolic pathways affecting PIP compounds

  • Interactive Design Tools:

    • Visual programming interfaces for non-computational scientists

    • Real-time feedback systems linking design to predicted properties

    • Collaborative platforms integrating experimental and computational insights

These computational approaches are likely to accelerate PIP development while reducing experimental iterations required to achieve optimal compounds for human applications.

Product Science Overview

Gene Location and Expression

The PIP gene is located on chromosome 7 (7q34) in humans . It is expressed in various tissues, including the apocrine glands in the axilla, vulva, eyelid, and ear canal, as well as the serous cells of the submandibular salivary gland, submucosal glands of the bronchi, and accessory lacrimal glands . Additionally, PIP is found in amniotic fluid and seminal fluid .

Physiological Functions

PIP plays a significant role in the regulation of water transport in the aforementioned glands . It has the ability to bind to immunoglobulin G (IgG), IgG-Fc, and CD4-T cell receptors, suggesting a wide range of immunological functions . PIP also binds to AZGP1 and exhibits aspartyl proteinase activity, which allows it to cleave fibronectin .

Immunological and Antimicrobial Roles

PIP is involved in various immunological processes, including the negative regulation of T cell apoptotic processes and the regulation of immune system processes . It can bind to different species of bacteria, showing the highest affinity to streptococci, thus playing a role in the non-immune defense of the body against pathogenic bacterial strains .

Clinical Significance

PIP has been observed to have a mitogenic effect on both normal and malignant breast epithelial cells . This protein is also associated with certain diseases, such as breast cysts and perivascular epithelioid cell tumors . Its expression and function in various exocrine tissues, such as the lacrimal, salivary, and sweat glands, highlight its versatile nature and importance in human reproductive and immunological systems .

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.