PF4 Human, His

Platelet Factor-4 Human Recombinant (CXCL4), His Tag
Shipped with Ice Packs
In Stock

Description

Heparin-Induced Thrombocytopenia (HIT)

PF4 Human, His is used to study HIT pathogenesis, where anti-PF4/heparin IgG antibodies trigger platelet activation and thrombosis. Key findings:

  • PF4/heparin complexes bind to platelet FcγRIIA receptors, inducing prothrombotic microvesicle release and calpain-mediated platelet death .

  • His-tagged PF4 maintains antigenicity for antibody detection assays, critical for HIT diagnosis .

Bacterial Defense Mechanisms

PF4 binds to bacterial cell walls (e.g., Staphylococcus aureus), promoting opsonization and phagocytosis. Anti-PF4 antibodies cross-react with PF4-coated bacteria, suggesting an evolutionary role in innate immunity .

Neuroinflammation and Cognitive Enhancement

Recent studies highlight PF4’s role in reducing neuroinflammation and improving cognition in aged mice. His-tagged variants enable mechanistic studies of PF4’s blood-brain barrier penetration and hippocampal effects .

Comparative Analysis: Native vs. Recombinant PF4

FeatureNative PF4PF4 Human, His
SourceHuman platelet α-granules Recombinant (E. coli)
Post-Translational ModificationsGlycosylated Non-glycosylated
ApplicationsCoagulation studies, HIT diagnostics Structural biology, antibody production
StabilitySensitive to freeze-thaw cycles Enhanced stability with glycerol/DTT

Research Limitations and Considerations

  • Immunogenicity: The His-tag may interfere with antibody binding in some epitope-specific assays .

  • Functional Assays: While tetrameric assembly is preserved, heparin-binding affinity may vary compared to native PF4 .

  • Storage: Long-term stability requires avoidance of repeated freeze-thaw cycles .

Product Specs

Introduction
Platelet factor-4 (PF4) is a protein released from activated platelets that plays a role in blood clotting, inflammation, and wound healing. It binds to heparin and neutralizes its anti-clotting effects. PF4 also attracts immune cells like neutrophils and fibroblasts to injury sites. Recombinant human PF4 is used in research to study heparin-induced thrombocytopenia, angiogenesis inhibition, and other related processes.
Description
Recombinant human PF4 is a non-glycosylated protein with a molecular weight of 10 kDa. It consists of a single polypeptide chain of 91 amino acids (residues 32-101) and includes a 21 amino acid His-tag at the N-terminus for purification. The protein is produced in E. coli and purified using chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The PF4 protein solution is provided at a concentration of 0.25 mg/ml in a buffer containing 20mM Tris-HCl (pH 8.0), 0.2M NaCl, 50% glycerol, and 2mM DTT.
Stability
For short-term storage (up to 4 weeks), store the PF4 protein at 4°C. For long-term storage, freeze the protein at -20°C. Adding a carrier protein such as 0.1% HSA or BSA is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity of the PF4 protein is greater than 85% as determined by SDS-PAGE analysis.
Synonyms
CXCL4, PF-4, PF4, Iroplact, Oncostatin-A, SCYB4, MGC138298.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MEAEEDGDLQ CLCVKTTSQV RPRHITSLEV IKAGPHCPTA QLIATLKNGR KICLDLQAPL YKKIIKKLLE S.

Q&A

What is the molecular structure of human PF4 and how does this relate to its function?

Human PF4 (CXCL4) is a specific protein synthesized from platelet α-granules with a complex quaternary structure. Full-length human PF4 comprises 101 amino acids, including a hydrophobic signal-like sequence involved in transmembrane transport and a mature monomeric peptide with a molecular weight of 7.8 kDa containing 70 amino acids . PF4 naturally functions as a tetramer (four identical subunits that assemble to form a globular protein) . This tetrameric structure is critical for its biological activities, as demonstrated by studies showing that disruption of tetramerization significantly impacts function. Nuclear magnetic resonance spectroscopy studies have shown that pH and ionic strength play key roles in regulating PF4's structural state . The genes encoding human PF4 are located at q13.1 in the global run-on (GRO) region of chromosome 4, containing 3'-untranslated and 5' regions alongside the amino acid coding sequence .

How does human PF4 interact with various binding partners to exert its effects?

PF4 has a high affinity for polyanions due to its strong positive charge. It forms complexes with various negatively charged molecules including surface mucopolysaccharides, platelet polyphosphates, and DNA from endothelial cells . Most notably, PF4 binds heparin to form antigenic complexes that can trigger immune responses in heparin-induced thrombocytopenia (HIT) . Beyond heparin, PF4 can interact with other polyanions from vaccines, bacteria, and other sources to form immune complexes involved in thrombosis . A particularly interesting interaction occurs between PF4 and von Willebrand factor (VWF), forming PF4-VWF complexes that may induce and promote immune-associated thrombosis . Unlike typical chemokines with clear receptor binding patterns, PF4's receptor interactions remain less certain, although CXCR3 and LDLR have been identified as receptors transmitting PF4 signals in hematopoietic stem cells .

What are the known physiological and pathological roles of human PF4?

PF4 serves diverse physiological functions while being implicated in several pathological conditions. In inflammation, PF4 triggers chemotaxis of human polymorphonuclear leukocytes and monocytes, attracting inflammatory cells to injury sites, promoting neutrophil degranulation, and stimulating cytokine production in monocytes . It exerts antithetic effects on T cell subsets, inhibiting CD4+CD25- T cells while inducing expansion of CD4+CD25+ T regulatory cells . PF4 plays a crucial role in hematopoietic stem cell regulation, with deficiency leading to phenotypes resembling accelerated stem cell aging, including lymphopenia, increased myeloid output, and DNA damage .

Pathologically, PF4 is central to heparin-induced thrombocytopenia (HIT) when PF4-heparin complexes trigger antibody production . Similar mechanisms occur in vaccine-induced immune thrombotic thrombocytopenia (VITT) related to COVID-19 vaccines . PF4 is also involved in chronic obstructive pulmonary disease, pancreatic cancer, periodontitis, polycystic ovary syndrome, and thyroiditis .

What expression systems are optimal for producing recombinant human PF4 with a His-tag?

The choice of expression system for His-tagged human PF4 depends on research requirements for yield, purity, proper folding, and post-translational modifications. Based on published research, several systems demonstrate effectiveness:

Expression SystemAdvantagesLimitationsNotable Considerations
Drosophila S2 cellsProper eukaryotic folding, good secretionSlower growth than bacteriacDNA encoding human PF4 can be cloned into pMT/BiP/V5-His A vector
E. coliHigh yield, cost-effectiveMay require refolding, lacks eukaryotic modificationsOptimal for structural studies requiring large quantities
Mammalian cells (HEK293, CHO)Native folding, complete post-translational modificationsHigher cost, potentially lower yieldBest for functional studies requiring authentic structure

Regardless of system choice, researchers should verify tetramerization after purification as this quaternary structure is essential for many biological functions. The search results indicate successful use of Drosophila Expression System with human PF4 cloned into the plasmid pMT/BiP/V5-His A .

What are effective methods for detecting PF4-polyanion complexes and antibody interactions?

Detection of PF4-polyanion complexes and antibody interactions requires carefully designed assays:

  • Enzyme-Linked Immunosorbent Assay (ELISA): Ultra-large complexes (ULCs) can be detected by:

    • Coating wells with KKO (a murine monoclonal antibody that mimics human HIT antibodies)

    • Incubating with PF4-polyanion complexes

    • Detecting bound complexes using horseradish peroxidase-conjugated sheep polyclonal anti-human PF4 antibody

    • Developing with 3,3',5,5'-tetramethylbenzidine substrate and measuring absorbance at 450nm

  • Complex Formation Dynamics: For studying complex dissociation, PF4 can be incubated with heparin for 30 minutes before adding potential dissociating agents, followed by overnight incubation at 37°C before measuring binding .

  • Functional Cellular Assays: FcγRIIA-dependent activation can be measured using:

    • DT40 cells expressing FcγRIIA receptors

    • Luciferase reporter systems to measure cellular activation

    • Controls including monoclonal antibody IV.3 as positive control for FcγRIIA signaling

  • Platelet Activation Assays: Serotonin release assays can assess platelet activation in response to PF4-heparin complexes and anti-PF4/heparin antibodies .

These methods provide complementary information about complex formation, antibody binding, and functional consequences of these interactions.

How can researchers study the tetramerization properties of PF4 and identify tetramerization inhibitors?

Studying PF4 tetramerization and identifying inhibitors involves several complementary approaches:

  • Size Exclusion Chromatography (SEC): This technique differentiates between monomeric, dimeric, and tetrameric forms of PF4. Research has shown that mutations like K50E result in dimers that fail to tetramerize , demonstrating SEC's utility in characterizing oligomerization states.

  • Computational Chemistry: Researchers have identified binding sites on the PF4 dimer near Glu28 and Lys50 that are critical for dimer-dimer interaction . In silico screening of compound libraries targeting this interface can identify potential tetramerization inhibitors, as demonstrated by the identification of compounds scoring greater than 10 standard deviations below the mean in docking studies .

  • Experimental Validation: Candidate inhibitors can be tested for:

    • Direct inhibition of tetramerization using SEC

    • Prevention of ultra-large complex (ULC) formation with heparin

    • Ability to disrupt preformed PF4-heparin ULCs

    • Inhibition of cellular activation in functional assays

  • Structure-Activity Relationship Analysis: Systematic analysis of inhibitor structural features can guide optimization of lead compounds for improved potency and specificity.

This combined computational and experimental approach has successfully identified PF4 antagonists (PF4As) that inhibit tetramerization at micromolar concentrations .

What assays can evaluate PF4's effects on hematopoietic stem cells?

Several experimental approaches can assess PF4's impacts on hematopoietic stem cells (HSCs):

  • DNA Damage Assessment: Measuring γH2AX positivity in HSCs after PF4 treatment. Studies show short-term recombinant PF4 treatment of old HSCs in culture decreased γH2AX positive cells by approximately 20% .

  • Proliferation and Cell Cycle Analysis: After 4-day culture with PF4, researchers can quantify:

    • Absolute cell counts to measure proliferation inhibition

    • Cell cycle distribution using flow cytometry

    • Effects on specific HSC subpopulations (e.g., myeloid-biased CD41+ HSCs)

  • In Vivo Reconstitution Assays: Testing HSCs treated with or without PF4 for their ability to reconstitute the hematopoietic system in irradiated recipients. PF4 treatment has been shown to enhance in vivo reconstitution capacity of aged HSCs .

  • Lineage Output Analysis: Evaluating the differentiation potential by analyzing:

    • Myeloid output (typically increased in aged HSCs)

    • Lymphoid output (B and T cells, typically decreased in aged HSCs)

    • Balance between different lineages

  • Cell Polarity Measurements: Assessing intracellular distribution of polarity markers, as PF4 improves cell polarity in aged HSCs .

These complementary approaches provide comprehensive assessment of PF4's rejuvenating effects on aged HSCs.

How can site-directed mutagenesis be used to investigate critical amino acids in human PF4?

Site-directed mutagenesis provides powerful insights into PF4's structure-function relationships:

  • Targeting Critical Residues: Research has identified several key amino acids for focused investigation:

    • Glu28 and Lys50 at the dimer-dimer interface are critical for tetramerization

    • The K50E mutation creates dimers incapable of tetramerization or forming ultra-large complexes

    • Species-specific differences between human, rat, and bovine PF4 (74% homologous) affect T cell modulation

  • Mutagenesis Strategies:

    • Charge reversal mutations to disrupt electrostatic interactions

    • Alanine scanning of residues at protein interfaces

    • Species-specific residue swapping between human and rat/bovine PF4

    • Systematic mutation of residues at the dimer-dimer interface

  • Functional Readouts:

    • Oligomerization state analysis

    • Complex formation with polyanions

    • T cell proliferation modulation

    • Receptor binding studies

This approach has revealed that despite high homology between species variants, rat and bovine PF4 cannot induce proliferation of CD4+CD25+ Tr cells like human PF4 does, indicating that relatively few amino acid differences determine this critical function .

What computational strategies can identify potential PF4 antagonists for research applications?

Computational identification of PF4 antagonists involves several sophisticated steps:

  • Target Site Identification: Research has identified a critical binding site on the PF4 dimer surface near residues Lys50 and Glu28, which are essential for dimer-dimer interactions and tetramerization .

  • Library Selection and Preparation: Researchers can screen "lead-like compounds" (those serving as potential drug nuclei requiring further optimization) from libraries containing millions of structures .

  • Docking Methodology: The search results describe using DOCK software to screen approximately 1.1 million compounds (processing about 1 compound/second) over a 10-day period .

  • Scoring and Selection: Compounds are scored empirically, with lower values representing higher predicted affinity. The distribution of scores approximates a Gaussian curve with mean value of -26.6 and standard deviation of 3.3 . Selecting compounds scoring greater than 10 standard deviations below the mean (-60 or less) provides high-confidence candidates for experimental validation .

  • Validation: Candidate compounds require experimental testing for:

    • Tetramerization inhibition

    • ULC formation prevention

    • Disruption of preformed complexes

    • Inhibition of cellular activation

This approach successfully identified four compounds (designated PF4As) that inhibit tetramerization at micromolar concentrations, with three able to promote breakdown of preformed ULCs .

How can PF4's immunomodulatory effects on T cell populations be characterized and exploited?

PF4 exerts complex immunomodulatory effects on T cell populations that can be characterized through:

  • Differential Proliferation Assays: PF4 has antithetic effects on T cell subsets:

    • Inhibits proliferation of CD4+CD25- conventional T cells

    • Induces expansion of CD4+CD25+ T regulatory cells when stimulated with anti-CD3 or anti-CD3/CD28 antibodies

  • Suppression Assays: PF4-induced CD4+CD25+ T regulatory cells lose their potent suppressor function in vitro , an effect that can be measured through co-culture experiments with responder T cells.

  • Specificity Controls: These effects are specific to PF4 and not mimicked by:

    • Protamine (another positively charged heparin-binding protein)

    • Heparin alone

  • Species-Specific Effects: Rat and bovine PF4, despite 74% homology to human PF4, cannot induce proliferation of CD4+CD25+ T regulatory cells, suggesting the few amino acid differences between species are critical for this function .

  • Receptor Identification: While the exact mechanism remains unclear, researchers can investigate potential receptors and signaling pathways mediating these effects.

These immunomodulatory properties could potentially be exploited for treating autoimmune disorders, transplant rejection, or other conditions where T cell regulation is central to pathology.

What are the mechanisms underlying PF4's role in hematopoietic stem cell aging and rejuvenation?

PF4 regulates hematopoietic stem cell (HSC) aging through several interconnected mechanisms:

  • Receptor-Mediated Signaling: LDLR and CXCR3 receptors on HSCs transmit PF4 signals, as demonstrated by double knockout mice showing exacerbated HSC aging phenotypes similar to PF4-deficient mice .

  • DNA Damage Control: PF4 treatment reduces DNA damage in aged HSCs, measured by decreased γH2AX positivity , suggesting a role in maintaining genomic integrity.

  • Cell Cycle Regulation: PF4 inhibits proliferation of old HSCs, including myeloid-biased CD41+ HSCs, as demonstrated by reduced cell counts and altered cell cycle profiles after treatment .

  • Niche Interaction: Age-related attrition of the megakaryocytic niche and associated PF4 downregulation represent central mechanisms in HSC aging . This is supported by findings that MK progenitor cell function is enhanced upon aging, and aged HSCs are redistributed away from old MK niches .

  • Lineage Balance Restoration: PF4 deficiency leads to increased myeloid and decreased lymphoid output, mimicking an aged immune system. Conversely, PF4 administration restores balanced lineage output in aged HSCs .

  • Translational Relevance: Human HSCs across various age groups respond to PF4 signaling, highlighting its potential as a rejuvenating factor for aged hematopoietic systems .

These mechanisms collectively demonstrate PF4's potential as a therapeutic target for age-related hematopoietic disorders.

How might PF4-VWF interactions contribute to immune-associated thrombosis?

The interaction between PF4 and von Willebrand factor (VWF) represents an emerging area of investigation in immune-associated thrombosis:

This research direction represents an important frontier in understanding immune-associated thrombosis beyond the classical PF4-heparin interaction.

What novel therapeutic applications of recombinant PF4 or PF4 antagonists are being explored?

Several innovative therapeutic applications of PF4 and its antagonists are under investigation:

While no drugs targeting PF4 have been successfully marketed to date , these diverse approaches highlight PF4's therapeutic potential across multiple disease areas.

How can multi-omics approaches advance our understanding of PF4 biology?

Multi-omics strategies can substantially advance PF4 research through integrated analysis of:

  • Genomics: Studying genetic variations in:

    • PF4 gene and regulatory regions

    • Receptors like LDLR and CXCR3 identified as transmitting PF4 signals

    • Components of downstream signaling pathways
      This could reveal population differences in PF4 responses and disease susceptibility.

  • Transcriptomics: RNA sequencing of cells treated with PF4 can identify:

    • Gene expression changes in different cell types (HSCs, T cells, platelets)

    • Altered signaling pathways and biological processes

    • Temporal dynamics of PF4-induced transcriptional programs

  • Proteomics: Mass spectrometry-based approaches can elucidate:

    • PF4 interactome (including VWF and other binding partners)

    • Post-translational modifications affecting PF4 function

    • Signaling pathway activation through phosphoproteomics

  • Metabolomics: Analysis of metabolic changes induced by PF4 treatment might reveal:

    • Altered cellular energetics in responding cells

    • Metabolic signatures associated with aging/rejuvenation of HSCs

    • Novel metabolic pathways affected by PF4 signaling

  • Single-Cell Analysis: Applying multi-omics at single-cell resolution can:

    • Identify cell subpopulations with differential PF4 responses

    • Map heterogeneity in aging HSCs and their response to PF4 treatment

    • Characterize rare cell populations in complex tissues

Integration of these multi-omics datasets through computational methods would provide unprecedented insights into PF4 biology and potentially identify novel therapeutic targets.

What are the translational challenges in developing PF4-based interventions for clinical applications?

Developing PF4-based interventions faces several translational challenges:

  • Stability and Delivery:

    • PF4's tetrameric structure may present stability challenges in vivo

    • Targeted delivery to specific tissues (e.g., bone marrow for HSC effects )

    • Achieving appropriate pharmacokinetics for sustained therapeutic effects

  • Specificity and Off-Target Effects:

    • PF4's multiple biological functions across different cell types

    • Potential for unintended consequences in platelet function or immunity

    • Individual variation in PF4 responses across patient populations

  • Antagonist Optimization:

    • Current PF4 antagonists work at micromolar concentrations

    • Need for improved potency, selectivity, and drug-like properties

    • Structure-based optimization to enhance specific activities

  • Diagnostic Integration:

    • Limited availability of PF4-targeted diagnostics (only one HIT diagnostic kit available)

    • Need for companion diagnostics to identify suitable patients

    • Biomarkers to monitor treatment response

  • Targeted Applications:

    • Identifying optimal disease targets (HIT, VITT, age-related hematopoietic disorders)

    • Patient stratification strategies

    • Combination approaches with existing therapies

  • Regulatory Pathway:

    • Novel mechanism of action requiring comprehensive safety data

    • Appropriate clinical endpoints for age-related hematopoietic interventions

    • Need for biomarkers correlating with clinical outcomes

Addressing these challenges requires coordinated basic and translational research efforts to fully realize the therapeutic potential of PF4-based interventions.

Product Science Overview

Structure and Composition

Platelet Factor-4 is a 70-amino acid protein with a molecular weight of approximately 7.8 kDa . It contains four highly conserved residues present in CXC chemokines . The His-tag, a sequence of histidine residues, is added to facilitate purification using metal affinity chromatography.

Physiological Role

The primary physiological role of PF-4 is to neutralize heparin-like molecules on the endothelial surface of blood vessels . This action inhibits local antithrombin III activity, thereby promoting coagulation . PF-4 is also a strong chemoattractant for neutrophils and fibroblasts, suggesting a role in inflammation and wound repair .

Biological Activity

Recombinant human PF-4 exhibits several biological activities:

  • Antiproliferative and Antiangiogenic: PF-4 can interfere with FGF2 and VEGF heparin binding, inhibiting their signaling pathways .
  • Proinflammatory and Proatherogenic: PF-4 affects monocytes, macrophages, and endothelial cells, contributing to inflammation and atherosclerosis .
Applications in Research

Recombinant PF-4 is used in various research applications, including:

  • Chemoattraction Assays: To study the migration of fibroblasts and neutrophils .
  • Coagulation Studies: To investigate the role of PF-4 in blood coagulation and its interaction with heparin .
  • Inflammation and Wound Healing: To explore the involvement of PF-4 in inflammatory responses and tissue repair .
Storage and Handling

Recombinant PF-4 is typically lyophilized and should be stored at -20°C for long-term stability . Upon reconstitution, it remains stable for up to one week at 4°C or up to three months at -20°C . It is recommended to avoid repeated freeze-thaw cycles to maintain protein integrity .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.