PF 4 Human

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

Description

Molecular Structure and Characteristics

Human PF4 is a homotetrameric protein composed of identical 70-amino acid subunits, each with a molecular weight of 7,756 Da . The amino acid sequence is:

E A E E D G D L Q C L C V K T T S Q V R P R H I T S L E V I K A G P H C P T A Q L I A T L K N G R K I C L D L Q A P L Y K K I I K K L L E S .

Key structural features include:

  • Disulfide bonds: Cys10–Cys36 and Cys12–Cys52 stabilize the tertiary structure .

  • Heparin-binding domain: Positively charged residues in the C-terminal region enable high-affinity interactions with heparin and other glycosaminoglycans .

  • Tetrameric form: Enhances heparin neutralization and chemotactic activity .

Biological Functions

PF4 exhibits diverse roles in physiology and pathology:

FunctionMechanismReferences
Coagulation regulationNeutralizes heparin by forming stable complexes, promoting clot formation.
Immune modulationBinds CXCR3 receptors on T-cells and CCR1 on monocytes, inducing migration.
Hematopoiesis inhibitionMaintains quiescence of hematopoietic stem cells via TGF-β signaling.
Anti-angiogenesisInhibits endothelial cell proliferation and vascular remodeling.
NeuroprotectionReduces age-related brain inflammation and enhances cognitive function.

Clinical Significance

PF4 is implicated in several thrombotic and immune disorders:

Heparin-Induced Thrombocytopenia (HIT)

  • Mechanism: Anti-PF4/heparin IgG antibodies activate platelets, causing thrombocytopenia and thrombosis .

  • Diagnostic assays:

    • PF4-dependent P-selectin expression assay (PEA): Higher sensitivity (92% accuracy) than serotonin release assay (SRA) .

    • Serum ELISA: Detects anti-PF4 antibodies but lacks specificity for clinical HIT .

Vaccine-Induced Immune Thrombotic Thrombocytopenia (VITT)

  • Association: Rare cases linked to adenoviral COVID-19 vaccines (e.g., Janssen, AstraZeneca) involve anti-PF4 antibodies .

  • Pathogenesis: Autoantibodies trigger platelet activation independently of heparin .

Systemic Sclerosis and Long COVID

  • Elevated PF4 levels correlate with interstitial lung disease in scleroderma .

  • PF4 expression changes are linked to persistent inflammation in long COVID .

PF4 in Aging and Neuroinflammation

  • PF4 administration in aged mice reduced neuroinflammation and restored cognitive function to levels equivalent to younger adults .

  • Mechanism: Downregulates pro-aging immune factors (e.g., CCL11) and enhances synaptic plasticity .

Recombinant PF4 Applications

  • Recombinant human PF4 (produced in E. coli) is used to study thrombotic mechanisms and immune cell recruitment .

Table 2: Clinical Conditions Associated with PF4 Dysregulation

ConditionKey FeaturesDiagnostic Markers
HITThrombocytopenia, thrombosis post-heparinAnti-PF4/heparin IgG antibodies
VITTThrombosis after adenoviral COVID-19 vaccinesAnti-PF4 antibodies (heparin-independent)
Systemic sclerosisInterstitial lung disease, fibrosisElevated serum PF4

Product Specs

Introduction
Platelet factor-4 (PF4), also known as CXCL4, is a small protein released by activated platelets. Its primary function is to regulate blood clotting by neutralizing molecules on blood vessel walls that prevent clotting. PF4 also acts as a chemoattractant, drawing neutrophils and fibroblasts to sites of injury, suggesting a role in inflammation and tissue repair. Oncostatin-A, while belonging to the same chemokine family as PF4, has distinct functions and is notably involved in inhibiting blood vessel formation (angiogenesis) in tumors.
Description
Recombinant human CXCL4, produced in E. coli bacteria, is a single chain protein consisting of 70 amino acids. It lacks glycosylation and has a molecular weight of 7.8 kDa.
Physical Appearance
Sterile white powder, free of visible particles, obtained by filtration and freeze-drying.
Formulation
CXCL4 protein was filtered through a 0.2 µm filter and subsequently freeze-dried. The solution prior to freeze-drying contained 20mM PB (phosphate buffer) and 1.5M NaCl (sodium chloride) at a pH of 7.4.
Solubility
To reconstitute lyophilized CXCL4, it is recommended to dissolve it in sterile, ultrapure water (18 megaohm-cm resistivity) to a minimum concentration of 100 µg/ml. This solution can be further diluted using other aqueous solutions as needed.
Stability
Lyophilized CXCL4 remains stable for 3 weeks at room temperature but should ideally be stored in a dry environment below -18°C. After reconstitution, CXCL4 should be kept at 4°C and used within 2-7 days. For long-term storage, freezing at -18°C is recommended, and the addition of a carrier protein like HSA or BSA (0.1%) is advisable. Avoid repeated freezing and thawing.
Purity
The purity of CXCL4 is greater than 97%, as confirmed by: (a) High-performance liquid chromatography (RP-HPLC) analysis and (b) Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis.
Biological Activity
The biological activity of CXCL4 is evaluated through a chemotaxis assay, which measures the movement of human fibroblasts towards a chemical attractant. The effective concentration range for CXCL4 in this assay is 1.0-10 ng/ml.
Synonyms
CXCL4, PF-4, PF4, Iroplact, Oncostatin-A, SCYB4, MGC138298.
Source
Escherichia Coli.
Amino Acid Sequence

EAEEDGDLQC LCVKTTSQVR PRHITSLEVI KAGPHCPTAQ LIATLKNGRK ICLDLQAPLY

Q&A

What is Human Platelet Factor 4 and what are its primary structural characteristics?

Human Platelet Factor 4 (PF4, also known as CXCL4) is a 70 amino acid heparin-binding protein with a molecular weight of approximately 7.8 kDa that is released from the alpha-granules of activated platelets . It belongs to a multigene family involved in critical biological processes including chemotaxis, coagulation, inflammation, and cell growth regulation . Structurally, PF4 is a strongly cationic chemokine that can undergo conformational changes upon binding to negatively-charged molecules, which is significant for both its physiological functions and pathological roles .

The protein's cationic nature is particularly important for its ability to bind to negatively-charged prokaryotic microorganisms and other biological surfaces, which can trigger structural alterations that serve as danger signals recognized by the immune system .

What is known about the genetic structure and expression of human PF4?

The human PF4 gene contains three exons and spans approximately 1,000 base pairs (bp) . The 5'-untranslated region of PF4 is 73 bp long with a TATA box present 30 bp upstream of the transcription start site . A notable feature is a 90 bp stretch of pyrimidines (including 53 consecutive thymidine residues) that begins at -227 bp, which is similar to a 30-residue region found in the rodent PF4 gene .

Northern blot analysis using gene-specific oligonucleotides has shown that PF4 mRNA in platelets is approximately 800 nucleotides in length . Importantly, steady-state platelet PF4 mRNA levels are approximately one order of magnitude greater than those of its homolog PF4alt, indicating differential regulation of these related genes .

How does PF4 differ from its homolog PF4alt?

PF4 and PF4alt are non-allelic genes with significant structural and expression differences as outlined in Table 1 below . While the human PF4 gene is encoded on a 10 kilobase pair (kb) EcoRI fragment, PF4alt is encoded in a polymorphic 3 or 5 kb EcoRI fragment . Despite their differences, DNA homology exists between the two human genes in both the 5'- and 3'-flanking regions and extends for over 3.6 kb .

Table 1: Comparison of PF4 and PF4alt Characteristics

FeaturePF4PF4altDivergence
Genomic location10 kb EcoRI fragment3 or 5 kb EcoRI fragment-
DNA sequence in signal peptideReference sequenceVariant14%
Amino acid sequence in signal peptideReference sequenceVariant38%
DNA sequence in mature protein coding regionReference sequenceVariant2.6%
Amino acid sequence in mature proteinReference sequenceVariant4.3%
Key amino acid substitutionsReference sequenceP58→L, K66→E, L67→H-
Pyrimidine-rich region at -227 bpPresent (90 bp)Absent-
mRNA size800 nucleotides900 nucleotides-
Relative mRNA expression in plateletsHigherLower~10-fold difference

What are the optimal methods for isolating and purifying human PF4 for experimental use?

For isolating human PF4, researchers should consider a multi-step approach beginning with platelet isolation from fresh human blood samples using differential centrifugation. After platelet activation with thrombin or calcium ionophore, alpha-granule contents including PF4 are released and can be collected in the supernatant .

Purification typically involves heparin-affinity chromatography, exploiting PF4's strong binding to heparin, followed by ion-exchange chromatography and size-exclusion chromatography for higher purity . When working with recombinant PF4, bacterial or mammalian expression systems can be utilized, but researchers must be cautious about potential differences in post-translational modifications compared to native platelet-derived PF4.

For storage, purified human PF4 should be maintained desiccated below -18°C, despite its reported stability at 25°C for up to one week . It is critical to minimize freeze-thaw cycles as they can compromise protein integrity and biological activity .

How can researchers effectively detect and measure PF4 in experimental and clinical samples?

Detection and quantification of PF4 in research and clinical samples can be accomplished through several complementary approaches:

  • Enzyme-Linked Immunosorbent Assays (ELISA): Commercial PF4 ELISA kits offer high sensitivity and specificity for quantifying PF4 in plasma, serum, or tissue culture supernatants . These assays typically have detection ranges in the picogram to nanogram per milliliter range.

  • Western Blotting: For qualitative detection and molecular weight confirmation, western blotting with specific anti-PF4 antibodies provides information about protein integrity and potential degradation products.

  • Mass Spectrometry: For detailed characterization of PF4 variants or modifications, liquid chromatography coupled with mass spectrometry (LC-MS/MS) offers unparalleled resolution and can identify post-translational modifications.

  • Flow Cytometry: For cellular studies, intracellular staining of PF4 in megakaryocytes or platelets can be performed using fluorescently-labeled antibodies.

When measuring PF4 levels, researchers should be mindful of sample collection conditions as improper handling can cause platelet activation and artificial elevation of PF4 concentrations.

What methods are most effective for studying PF4's role in thromboinflammatory conditions?

Investigating PF4's role in thromboinflammation requires a comprehensive methodological approach combining in vitro, ex vivo, and in vivo techniques:

  • Platelet Activation Assays: Functional assays using washed platelets can detect antibody-mediated platelet activation. For heparin-induced thrombocytopenia (HIT) antibody detection, adding heparin to these assays is recommended, while for vaccine-induced immune thrombotic thrombocytopenia (VITT) antibody detection, adding PF4 alone is more appropriate .

  • Immunoassays: Solid-phase PF4-dependent immunoassays using microtiter plates are sensitive for both HIT and VITT antibodies, while rapid immunoassays with PF4/heparin antigen coated on beads are sensitive and specific for HIT but not for VITT antibodies .

  • Biophysical Tools: Techniques such as circular dichroism spectroscopy, X-ray crystallography, and molecular dynamics simulations can characterize structural alterations in PF4 during complex formation with heparin or other binding partners .

  • Animal Models: Transgenic mouse models expressing human PF4 and FcγRIIA can recapitulate key aspects of HIT and provide valuable insights into the pathogenesis of PF4-related disorders.

How can researchers differentiate between HIT and VITT in experimental and diagnostic settings?

Differentiating between HIT and VITT requires careful consideration of their distinct but related pathophysiological mechanisms as outlined in Table 2 :

Table 2: Distinguishing Features and Diagnostic Approaches for HIT and VITT

FeatureHeparin-Induced Thrombocytopenia (HIT)Vaccine-Induced Immune Thrombotic Thrombocytopenia (VITT)
Triggering agentHeparin exposureAdenoviral vector vaccines
Antibody targetPF4/heparin complexesPF4 alone
Optimal functional assayPlatelet activation with heparin addedPlatelet activation with PF4 added
Recommended immunoassaySolid-phase PF4-dependent assays or rapid immunoassays with PF4/heparinSolid-phase PF4-dependent assays
Typical clinical presentationThrombosis in venous and arterial circulationPredominance of cerebral venous sinus thrombosis and splanchnic thrombosis
Temporal relationship5-14 days after heparin exposure (typical)4-28 days after vaccination
Response to therapeutic heparinUsually worsens symptomsMay not exacerbate symptoms

For accurate differentiation, laboratories should employ a combination of clinical history, platelet count monitoring, and specialized assays. Functional assays using washed platelets with appropriate additives (heparin for HIT, PF4 for VITT) provide the highest specificity, while solid-phase immunoassays offer high sensitivity for initial screening .

What approaches can best elucidate the conformational dynamics of PF4 during interactions with binding partners?

Understanding PF4's conformational changes requires sophisticated biophysical and computational approaches:

  • Circular Dichroism (CD) Spectroscopy: This technique allows researchers to monitor changes in PF4's secondary structure upon interaction with heparin, DNA, or other binding partners.

  • Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS): This method can map the regions of PF4 involved in interactions with binding partners at peptide-level resolution.

  • Nuclear Magnetic Resonance (NMR) Spectroscopy: For more detailed structural information, solution-state NMR can provide atomic-level insights into PF4's conformational dynamics.

  • X-ray Crystallography: While challenging due to PF4's tendency to oligomerize, crystallographic studies can reveal detailed structural information about PF4 complexes.

  • Molecular Dynamics Simulations: Computational approaches can model PF4's structural alterations in complex with various binding partners and predict regions susceptible to conformational changes.

These methods collectively have revealed that PF4 undergoes significant structural rearrangements upon binding to negatively-charged molecules, exposing neoepitopes that can be recognized by antibodies in conditions like HIT and VITT .

What gene expression and regulatory mechanisms govern PF4 production during megakaryopoiesis?

PF4 expression is primarily restricted to megakaryocytes and platelets, with tight transcriptional regulation during megakaryopoiesis. Researchers investigating these mechanisms should consider:

  • Chromatin Immunoprecipitation (ChIP): To identify transcription factors binding to the PF4 promoter region, including the TATA box located 30 bp upstream of the transcription start site .

  • Reporter Gene Assays: Constructs containing the PF4 promoter region can help delineate key regulatory elements controlling gene expression.

  • CRISPR/Cas9 Genome Editing: For functional studies, targeted modification of regulatory regions can provide insights into their importance for PF4 expression.

  • Single-Cell RNA Sequencing: This approach can track PF4 expression dynamics throughout megakaryocyte maturation and platelet production.

  • DNA Methylation Analysis: Epigenetic studies may reveal regulatory mechanisms underlying the differential expression of PF4 versus PF4alt .

Research has shown that the unique pyrimidine-rich region (including 53 consecutive thymidine residues) beginning at -227 bp in the PF4 gene may play a role in its regulation, as this feature is absent in the PF4alt gene despite high homology in other regions .

What are the most promising approaches for targeting PF4 therapeutically in thromboinflammatory disorders?

Emerging therapeutic strategies targeting PF4-mediated pathologies include:

  • Engineered Decoy Molecules: Designing synthetic compounds that can bind to PF4 and prevent formation of immunogenic complexes without triggering antibody recognition.

  • Monoclonal Antibodies: Developing antibodies that bind to PF4 at sites distinct from pathogenic epitopes to block formation of antigenic complexes.

  • Small Molecule Inhibitors: Compounds that disrupt PF4 oligomerization or binding to cell surfaces may prevent downstream immune activation.

  • Non-Heparin Anticoagulants: For patients with HIT, alternative anticoagulants that don't interact with PF4 can prevent formation of immunogenic complexes while maintaining necessary anticoagulation.

  • Immunomodulatory Approaches: Strategies targeting FcγRIIA receptors or downstream signaling pathways could block effector functions of anti-PF4 antibodies without interfering with PF4 itself.

Research in this area should focus on developing in vitro screening assays and relevant animal models to evaluate efficacy and safety of these approaches.

How can researchers better understand the evolutionary significance of PF4 and its homologs across species?

Understanding the evolutionary aspects of PF4 requires comparative genomic and functional approaches:

  • Phylogenetic Analysis: Comparing PF4 sequences across species can identify conserved regions that may be critical for fundamental functions.

  • Functional Studies with Orthologs: Testing PF4 proteins from different species in identical assay systems can reveal differences in activity or binding properties.

  • Genomic Organization Analysis: Studying the arrangement of PF4 and related genes across species can provide insights into gene duplication events and evolutionary pressures.

  • Molecular Clock Analysis: Estimating the timing of divergence between PF4 and its homologs, including PF4alt, can correlate with evolutionary milestones.

  • Structural Biology Approaches: Comparing three-dimensional structures of PF4 proteins across species can reveal conserved functional domains and species-specific adaptations.

These approaches can help determine whether PF4's roles in hemostasis and antimicrobial defense represent its original evolutionary functions, and how its involvement in pathological conditions like HIT and VITT may have emerged .

Product Science Overview

Gene and Protein Structure

The gene for human PF4 is located on chromosome 4 . The recombinant human PF4 is a 7.8 kDa protein containing 70 amino acid residues, including the four highly conserved residues present in CXC chemokines . The protein is expressed in megakaryocytes and stored in the α-granules of platelets .

Biological Functions

PF4 has several biological functions:

  • Anticoagulant Neutralization: PF4 binds with high affinity to heparin, neutralizing its anticoagulant effect and promoting coagulation .
  • Chemoattraction: It acts as a chemoattractant for neutrophils, fibroblasts, and monocytes .
  • Inflammation and Wound Repair: PF4 is involved in wound repair and inflammation by moderating the effects of heparin-like molecules .
  • Antiproliferative and Antiangiogenic Effects: PF4 can interfere with FGF2 and VEGF heparin binding, inhibiting their signaling and thus exhibiting antiproliferative and antiangiogenic properties .
Clinical Significance

PF4 is clinically significant in several contexts:

  • Heparin-Induced Thrombocytopenia (HIT): The heparin:PF4 complex is the antigen in HIT, an autoimmune reaction to the administration of heparin .
  • Vaccine-Induced Immune Thrombotic Thrombocytopenia (VITT): Antibodies against PF4 have been implicated in cases of thrombosis and thrombocytopenia following vaccination with the Oxford–AstraZeneca or Janssen COVID-19 vaccines .
  • Long COVID and Systemic Sclerosis: Changes in PF4 expression have been associated with symptoms of long COVID and increased levels in patients with systemic sclerosis and interstitial lung disease .
Recombinant PF4

Recombinant human PF4 is produced using expression systems such as HEK 293 cells. It is lyophilized after extensive dialysis against PBS and can be reconstituted in ddH₂O or PBS . The recombinant protein is used for laboratory research and is not intended for direct human use .

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.