PLGF2 Human, CHO

Placental Growth Factor-2 Human Recombinant, CHO
Shipped with Ice Packs
In Stock

Description

Table 1: Comparison of PLGF Isoforms

PropertyPLGF1PLGF2
Amino Acids131152
Heparin BindingNoYes
Neuropilin BindingNoYes (NRP1/2)
SolubilityFully solubleMatrix-associated
Primary SourcePlacenta, endothelial cellsPlacenta, tumor microenvironments

Production and Purification

PLGF2 is typically produced in recombinant systems:

  • E. coli: Non-glycosylated, 34.6 kDa homodimer purified via chromatographic techniques .

  • Insect Cells (Sf9): Glycosylated, 44 kDa form with enhanced stability .

  • Mammalian Systems: While CHO-cell production is not detailed here, mammalian systems (e.g., HEK293) are preferred for post-translational modifications .

Table 2: Recombinant PLGF2 Production Methods

Expression SystemGlycosylationMolecular WeightPurity
E. coliNo34.6 kDa>95% (SDS-PAGE)
Insect CellsYes44 kDa>90% (HPLC)

Biological Functions

PLGF2 drives pathological angiogenesis and inflammation via:

  • Endothelial Cell Activation: Stimulates proliferation and migration through VEGFR-1 and neuropilin signaling .

  • Anti-Apoptotic Effects: Upregulates Bcl2 and pErk while suppressing Bax in rheumatoid arthritis fibroblasts (RA-FLSs), promoting cell survival .

  • Heterodimerization: Forms VEGF-A/PLGF2 heterodimers that retain angiogenic activity, compensating for reduced VEGF-A homodimers in tumors .

In Oncology

  • Tumor Growth: PLGF2 overexpression in ovarian carcinoma (A2780 cells) increased monocyte-macrophage infiltration but did not alter tumor growth, unlike PLGF2-DE (a dominant-negative variant), which suppressed angiogenesis .

  • Dominant-Negative PLGF2-DE: Reduced VEGF-A homodimer production, inhibiting xenograft tumor growth by 60% compared to controls .

In Pregnancy Disorders

  • Preeclampsia: Serum PLGF2 levels (median 139.1 pg/mL) were 3.7-fold higher than PLGF1, serving as a biomarker for early detection .

Therapeutic Implications

  • Anti-Angiogenic Therapy: PLGF2-DE variant inhibits VEGF-A-driven angiogenesis, showing promise in ovarian cancer models .

  • Inflammatory Diseases: Targeting PLGF2 in rheumatoid arthritis reduces synovial hyperplasia and pannus formation .

Product Specs

Introduction
Placental growth factor (PLGF) is a growth factor involved in angiogenesis and endothelial cell growth. It promotes the proliferation and migration of these cells. PLGF binds to the receptor VEGFR-1/Flt1. Notably, PLGF-2 interacts with neuropilin-1 and neuropilin-2 in a manner dependent on heparin.
Description
Recombinant human PLGF2 (amino acids 19-170) is produced in CHO cells. It is a disulfide-linked homodimeric glycoprotein with a molecular weight of 33 kDa. The protein consists of 152 amino acids. Purification of recombinant human PLGF2 is achieved using proprietary chromatographic methods.
Physical Appearance
The product is a sterile, white, lyophilized powder.
Formulation
PLGF-2 was lyophilized from a 0.2 µm filtered solution containing HCl.
Solubility
To reconstitute lyophilized PLGF2, it is recommended to dissolve it at a concentration of 200 µg/ml in PBS. This solution can then be diluted further into other aqueous solutions.
Stability
Lyophilized PLGF-2 remains stable at room temperature for up to 3 weeks; however, it is recommended to store it desiccated below -18°C. Once reconstituted, PLGF2 should be stored at 4°C for 2-7 days. For long-term storage, it is advisable to add a carrier protein (0.1% HSA or BSA) and store it below -18°C. Avoid repeated freeze-thaw cycles.
Purity
The purity is determined to be greater than 95.0% using SDS-PAGE analysis.
Biological Activity
The biological activity, measured as the ED50 in a cell proliferation assay using MDA-MB-231 cells, is less than 10 µg/ml.
Synonyms
PIGF, PGF, PlGF-2, PLGF-2.
Source
Chinese Hamster Ovary cells (CHO).
Amino Acid Sequence
LPAVPPQQWA LSAGNGSSEV EVVPFQEVWG RSYCRALERL VDVVSEYPSE VEHMFSPSCV SLLRCTGCCG DENLHCVPVE TANVTMQLLK IRSGDRPSYV ELTFSQHVRC ECRPLREKMK PERRRPKGRG KRRREKQRPT DCHLCGDAVP RR.

Q&A

What distinguishes PLGF2 from other PLGF isoforms and why is this significant for experimental design?

PLGF2 is one of at least three human PLGF isoforms resulting from alternative splicing. The key distinguishing feature of PLGF2 is its highly basic heparin-binding 21 amino acid insert at the C-terminus, which is absent in PLGF-1 (131 aa) and differs from PLGF-3 (203 aa) .

This structural difference has significant functional implications:

IsoformAmino AcidsKey FeaturesBinding Properties
PLGF-1131 aaBasic isoformVEGFR-1/Flt1 only
PLGF2152 aaContains 21 aa heparin-binding domainVEGFR-1/Flt1 and neuropilin-1,2 (heparin-dependent)
PLGF-3203 aaExtended structureVEGFR-1/Flt1 only

When designing experiments, researchers should consider that PLGF2's ability to bind neuropilins in a heparin-dependent manner introduces additional signaling pathways compared to other isoforms, potentially affecting experimental outcomes . This becomes particularly relevant when comparing results across different model systems, as mice only express one PLGF form equivalent to human PLGF2 .

How does CHO-produced PLGF2 differ from E. coli-produced PLGF2, and when should each be used?

The expression system significantly impacts PLGF2's properties and experimental utility:

CharacteristicCHO-produced PLGF2E. coli-produced PLGF2
StructureDisulfide-linked homodimeric, glycosylatedDisulfide-linked homodimeric, non-glycosylated
Molecular Mass~33 kDa~25 kDa
Post-translational modificationsContains glycosylationLacks glycosylation
ReconstitutionPBS compatibleRequires 4 mM HCl with carrier protein
ApplicationsPreferred for in vivo studies and complex cellular assaysSuitable for binding studies and basic assays

PLGF2 Human Recombinant produced in CHO cells is a disulfide-linked homodimeric, glycosylated polypeptide chain containing 152 amino acids with a molecular mass of 33kDa . This glycosylated form more closely resembles native human PLGF2, making it preferable for studies requiring physiological relevance.

E. coli-derived human PLGF (Ala21-Arg149) lacks glycosylation, affecting stability and potentially immunogenicity in vivo models . For binding assays, E. coli-produced PLGF2 demonstrates 50% optimal binding response at approximately 1-6 ng/mL .

Researchers should select CHO-produced PLGF2 for experiments modeling complex physiological processes, while E. coli-produced protein may be sufficient for basic receptor-binding studies and assays where glycosylation is not critical.

What are the optimal reconstitution and storage conditions for maintaining PLGF2 bioactivity?

Proper handling of recombinant PLGF2 is critical for maintaining biological activity:

For CHO-produced PLGF2:

  • Reconstitute lyophilized protein at 200μg/ml in PBS

  • For short-term storage (2-7 days), keep at 4°C

  • For long-term storage, keep desiccated below -18°C

  • Add carrier protein (0.1% HSA or BSA) to enhance stability

  • Avoid repeated freeze-thaw cycles which significantly reduce bioactivity

For E. coli-produced PLGF2:

  • Reconstitute at 100 μg/mL in sterile 4 mM HCl containing at least 0.1% human or bovine serum albumin

  • Use manual defrost freezer for storage and avoid repeated freeze-thaw cycles

To verify bioactivity after reconstitution, a cell proliferation assay using MDA-MB-231 cells can be employed, where active PLGF2 typically shows biological activity (ED50) at concentrations less than 10μg/ml .

How can researchers validate neutralizing antibodies against PLGF2 for functional blocking experiments?

When designing blocking experiments with anti-PLGF neutralizing antibodies, researchers should implement the following methodology:

  • Antibody Selection: Use validated clones such as Clone #37203, which has demonstrated neutralizing activity against human PLGF .

  • Functional Validation Assay: Tube formation assays using Endothelial Colony Forming Cells (ECFCs) provide a robust method to validate neutralizing activity. These cells should be:

    • Stained with calcein

    • Grown on Matrigel®

    • Treated with conditioned medium containing PLGF2

    • Exposed to either anti-PLGF neutralizing antibody or isotype control IgG

  • Quantification Method: Total tube area (μm²) should be quantified using imaging software (e.g., NIS Elements) after 48h incubation .

  • Controls: Include parallel experiments with:

    • Vehicle + isotype control IgG (negative control)

    • DMOG (dimethyloxalylglycine) + isotype control IgG (positive control)

    • Vehicle + anti-PLGF neutralizing antibody (antibody control)

    • DMOG + anti-PLGF neutralizing antibody (test condition)

Successful neutralization is confirmed when anti-PLGF antibody blocks DMOG-induced inhibition of ECFC tube formation, with statistical significance (p < 0.01 or p < 0.001) when comparing treatment groups .

How do PLGF2 levels correlate with hypertensive disorders in pregnancy, and what methodological approaches provide consistent results?

PLGF levels exhibit specific patterns during normal and complicated pregnancies:

In normal pregnancy:

  • Circulating PLGF increases during pregnancy

  • Peak levels occur at mid-gestation

  • Gradually decrease toward term

In preeclampsia and hypertensive disorders:

  • The normal increase in PLGF is attenuated

  • Significantly lower serum levels of PLGF-2 (by approximately 22%) have been observed in obese hypertensive pregnant rat models compared to wild-type controls (p=0.006)

  • These reductions occur even when sFlt-1 (soluble fms-like tyrosine kinase-1) levels remain unchanged

Methodological recommendations for consistent PLGF measurement:

  • Sample Timing: Collect samples at standardized gestational timepoints, particularly around mid-gestation (GD 19 in rodent models)

  • Assay Selection: Use assays that specifically quantitate free bioavailable levels of PLGF-2 rather than total PLGF

  • Complementary Measurements: Always measure sFlt-1 levels and calculate the sFlt-1:PLGF ratio, which provides greater diagnostic value than either marker alone

  • Reference Ranges: Establish model-specific reference ranges, as PLGF2 levels vary significantly between species and strains

What is the therapeutic potential of recombinant human PLGF administration in hypertensive pregnancy disorders?

Administration of recombinant human PLGF (rhPLGF) shows promising results in experimental models of hypertensive pregnancy disorders:

Dosage and Administration Protocol:

  • In rat models of obese hypertensive pregnancy, rhPLGF infusion at 180 μg/kg per day from gestational day (GD) 13 to 19 has demonstrated efficacy

  • Administration via osmotic minipumps provides consistent delivery

Experimental Considerations:

  • The administration of rhPLGF did not significantly affect serum levels of sFlt-1 between treated groups (WT+rhPLGF: 621±332 pg/mL vs. MC4R-def+rhPLGF: 229±101 pg/mL, p=0.7)

  • The sFlt-1:PLGF-2 ratio remained statistically unchanged (WT+rhPLGF: 4.3±1.8 vs. MC4R-def+rhPLGF: 2.8±1.4, p=0.2)

Researchers investigating therapeutic applications should carefully monitor both exogenous (administered) and endogenous PLGF levels, as well as related angiogenic factors, to fully characterize treatment effects.

How does PLGF2 influence macrophage polarization in tumor microenvironments, and what experimental approaches best elucidate these effects?

PLGF2 plays a significant role in regulating macrophage polarization within tumor microenvironments:

Macrophage Polarization States in Tumors:

  • M1-like macrophages: Associated with non-progressing/regressing tumors; characterized by proinflammatory activity, antigen presentation, and tumor lysis

  • M2-like macrophages: Present in malignant tumors; promote angiogenesis, tumor cell intra/extravasation and growth; suppress antitumor immunity

PLGF2's Role:

  • Down-regulation of PLGF has been associated with skewing tumor-associated macrophages (TAMs) away from M2- toward M1-like phenotype

  • This polarization shift promotes antitumor immune response and vessel normalization, effects known to decrease tumor growth and metastasis

Recommended Experimental Approaches:

  • Macrophage Phenotyping:

    • Flow cytometry analysis of surface markers (CD80, CD86, MHC II for M1; CD163, CD206 for M2)

    • Quantitative PCR for polarization-associated genes (iNOS, TNF-α, IL-12 for M1; Arginase-1, IL-10, TGF-β for M2)

    • Cytokine profiling of macrophage secretome

  • PLGF2 Manipulation Strategies:

    • Neutralizing antibody administration in tumor models

    • Genetic approaches (siRNA, CRISPR/Cas9) to modulate PLGF expression

    • Recombinant PLGF2 administration at varying concentrations

  • Functional Assessment:

    • T-cell proliferation assays to measure immune activation

    • Vessel normalization analysis (pericyte coverage, permeability)

    • Tumor growth and metastasis quantification

  • Controls and Validation:

    • Include histidine-rich glycoprotein treatments as positive control for M1 polarization

    • Use multiple tumor models to confirm consistency of findings

When designing such experiments, researchers should account for potential compensatory mechanisms and temporal dynamics of macrophage polarization in response to PLGF2 modulation.

What are the methodological considerations for studying PLGF2 in knockout mouse models compared to neutralizing antibody approaches?

Both genetic knockout models and neutralizing antibody approaches offer valuable but distinct insights into PLGF2 function:

PLGF Knockout Mouse Models:

  • plgf knockout mice are born at Mendelian frequency, are healthy and fertile, suggesting PLGF is dispensable for normal embryonic development

  • These models demonstrate that PLGF is redundant for physiological angiogenesis but is critical during pathological conditions

  • In adult knockout mice, angiogenesis and arteriogenesis are impaired during pathological conditions including tumor growth, heart, limb, and ocular ischemia

Methodological Advantages of Knockout Models:

  • Complete absence of the target protein throughout development

  • No concerns about antibody specificity or incomplete neutralization

  • Stable phenotype allowing long-term studies

  • Ability to generate tissue-specific knockouts using Cre-lox systems

Limitations of Knockout Models:

  • Potential developmental compensation mechanisms

  • Cannot study temporal aspects of PLGF inhibition

  • May not translate directly to pharmacological inhibition approaches

  • Limited to rodent biology (species differences in PLGF isoforms)

Neutralizing Antibody Approaches:

  • Allow temporal control of PLGF inhibition

  • Can be applied to various animal models and potentially human tissues

  • Validated antibodies like Clone #37203 have demonstrated effective neutralization of PLGF activity in functional assays

Experimental Design Recommendations:

  • For mechanistic studies: Use knockout models to establish baseline understanding of PLGF2 function

  • For therapeutic potential: Use neutralizing antibodies to better approximate clinical intervention

  • For comprehensive analysis: When possible, compare both approaches within the same study to distinguish between developmental and acute effects of PLGF2 inhibition

  • Controls: Include isotype-matched antibodies as controls for neutralizing antibody experiments

Quantification endpoints should include both molecular readouts (changes in downstream signaling) and functional outcomes (angiogenesis, inflammation, etc.) for comprehensive assessment of PLGF2 function.

How can researchers address inconsistent results when measuring PLGF2 levels across different experimental platforms?

Measuring PLGF2 levels consistently across different platforms requires attention to several technical aspects:

Common Sources of Variability:

  • Assay specificity: Some immunoassays may detect total PLGF2 while others detect only free (unbound) PLGF2

  • Cross-reactivity with other PLGF isoforms: Assays may have varying specificity for PLGF-1, PLGF-2, and PLGF-3

  • Species differences: Human PLGF-1 shares only 56% and 55% amino acid identity with mouse and rat PLGF respectively

  • Sample handling: Pre-analytical variables (collection, processing, storage) significantly affect measured levels

Standardization Recommendations:

  • Assay Selection and Validation:

    • Use assays that clearly specify whether they measure free or total PLGF2

    • Validate each assay with recombinant standards of known concentration

    • When comparing CHO-produced and E. coli-produced PLGF2, account for glycosylation differences

  • Sample Preparation Protocol:

    • Standardize collection tubes (EDTA, citrate, or serum)

    • Implement consistent processing times (immediate processing vs. delayed)

    • Use standardized centrifugation protocols (speed, temperature, duration)

    • Aliquot samples to avoid freeze-thaw cycles

  • Data Normalization Strategies:

    • Consider reporting sFlt-1:PLGF ratio rather than absolute PLGF2 values

    • When appropriate, normalize to total protein content

    • For pregnancy studies, standardize reporting by gestational age

  • Cross-Platform Calibration:

    • Include common reference samples across platforms

    • Develop conversion factors between different assay systems

    • Consider measuring the same samples on multiple platforms for critical studies

By implementing these standardization practices, researchers can minimize technical variability and focus on true biological differences in PLGF2 levels across experimental conditions.

What are the critical quality control parameters for verifying the integrity of PLGF2 in experimental preparations?

To ensure experimental validity, researchers should verify PLGF2 integrity using these quality control parameters:

Physical and Chemical Parameters:

  • Purity Assessment:

    • SDS-PAGE analysis should confirm >95% purity

    • Both reducing and non-reducing conditions should be tested to verify dimer formation

  • Structural Integrity:

    • Verify disulfide-linked homodimeric structure (approximately 33kDa for CHO-produced PLGF2)

    • Confirm glycosylation status using appropriate glycoprotein staining methods for CHO-produced PLGF2

  • Stability Monitoring:

    • Implement stability-indicating analytical methods

    • Monitor for degradation products or aggregation

    • Protein concentration verification using validated methods (BCA, Bradford, or amino acid analysis)

Functional Parameters:

  • Binding Activity:

    • Verify binding to VEGFR-1/Flt1 in a functional ELISA

    • For PLGF2, confirm heparin-dependent binding to neuropilin-1 and neuropilin-2

  • Biological Activity:

    • Perform cell proliferation assay using MDA-MB-231 cells

    • Verify ED50 < 10μg/ml (typical for active PLGF2)

    • Consider tube formation assays with endothelial cells as additional functional verification

  • Receptor Specificity Controls:

    • Include receptor-blocking antibodies to confirm specificity

    • Use cells expressing individual receptors (VEGFR-1, NRP1, NRP2) to confirm binding specificity

Documentation and Reporting:

  • Record lot-specific activity data

  • Document storage conditions and reconstitution procedures

  • Maintain detailed records of freeze-thaw cycles

  • Consider implementing expiration dates based on stability data

These quality control measures ensure that experimental outcomes reflect true biological responses to PLGF2 rather than artifacts from compromised protein preparations.

What emerging research areas show promise for novel PLGF2 applications beyond current angiogenesis and preeclampsia models?

Several emerging research directions indicate expanded roles for PLGF2 beyond traditional applications:

Immunomodulation and Cancer Immunotherapy:

  • PLGF's influence on macrophage polarization suggests potential applications in cancer immunotherapy

  • The ability of PLGF inhibition to skew tumor-associated macrophages from M2 to M1 phenotype could be exploited to enhance immunotherapy efficacy

  • Research combining PLGF2 modulation with immune checkpoint inhibitors represents a promising frontier

Metabolic Disorders and Obesity:

  • The established link between PLGF2 levels and outcomes in obese hypertensive pregnancy models suggests broader applications in metabolic disorders

  • Investigating PLGF2's role in adipose tissue vascularization and inflammation could yield insights into obesity pathophysiology

  • The MC4R-deficient rat model demonstrates connections between melanocortin signaling, obesity, and PLGF2 levels that warrant further exploration

Regenerative Medicine Applications:

  • PLGF's role in pathological angiogenesis but redundancy in normal development suggests potential for targeted tissue regeneration

  • Tissue-specific delivery of CHO-produced PLGF2 could promote vascularization in ischemic tissues without systemic effects

  • Biomaterial scaffolds incorporating PLGF2 represent an untapped approach for guided tissue regeneration

Neurological Disorders:

  • PLGF2's ability to bind neuropilins suggests potential roles in neuronal development and function

  • Investigation of PLGF2 in neurodegenerative diseases and stroke models could reveal new therapeutic approaches

  • The heparin-binding domain unique to PLGF2 may enable specific targeting of neuronal populations

Researchers exploring these emerging areas should consider developing new methodological approaches, including:

  • Tissue-specific conditional knockout models

  • Advanced protein engineering to create PLGF2 variants with enhanced receptor specificity

  • Combination approaches targeting multiple angiogenic pathways simultaneously

  • Novel delivery systems for targeted PLGF2 therapy

Product Science Overview

Introduction

Placental Growth Factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family, which plays a crucial role in angiogenesis and vasculogenesis. PlGF has four isoforms (PlGF-1 to PlGF-4), with PlGF-2 being one of the most biologically active forms. Recombinant human PlGF-2 (rhPlGF-2) is produced using Chinese Hamster Ovary (CHO) cells, which are commonly used in biotechnology for the production of therapeutic proteins.

Biological Role

PlGF-2 is primarily involved in the formation of new blood vessels (angiogenesis) and the growth of existing ones (arteriogenesis) under pathological conditions. It exerts its effects by binding to the VEGF receptor-1 (VEGFR-1), which is expressed on various cell types, including endothelial cells, monocytes, and macrophages .

Therapeutic Potential

The therapeutic potential of rhPlGF-2 has been explored in various preclinical and clinical studies. One of the key areas of interest is its application in cardiovascular diseases, particularly in the context of myocardial infarction (heart attack) and ischemic cardiomyopathy. Studies have shown that rhPlGF-2 can induce angiogenesis and improve cardiac function in animal models of myocardial infarction .

Preclinical Studies

In a preclinical study involving pigs with induced myocardial infarction, continuous intravenous infusion of rhPlGF-2 was found to be safe and associated with favorable post-infarct remodeling. However, the study did not show significant improvements in global cardiac function or neovascularization . Another study in mice with advanced atherosclerosis and chronic ischemic cardiomyopathy demonstrated that rhPlGF-2 improved contractile function and increased capillary and arteriolar density in ischemic myocardium without aggravating atherosclerosis .

Challenges and Future Directions

Despite the promising results in preclinical studies, the translation of rhPlGF-2 therapy to clinical practice faces several challenges. One of the main challenges is the need for well-designed clinical trials in representative large animal models to ensure the safety and efficacy of the therapy. Additionally, understanding the molecular mechanisms underlying the effects of rhPlGF-2 and optimizing its delivery methods are crucial for its successful clinical application .

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.