pGH 20kDa Human

Growth Hormone Placental 20kDa Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to Placental Growth Hormone 20kDa (pGH 20kDa Human)

Placental growth hormone 20kDa (pGH 20kDa), a variant of human growth hormone (GH), is a 20-kilodalton isoform produced during pregnancy. Unlike the pituitary-derived 22-kilodalton GH (22kDa-hGH), which is the predominant form in circulation, pGH 20kDa is synthesized by the placenta and lacks lactogenic activity mediated by prolactin receptors . This isoform is critical in supporting maternal-fetal interactions during gestation.

Production and Purification

pGH 20kDa is manufactured via recombinant DNA technology in E. coli and purified through proprietary chromatographic methods . Key steps include:

  1. Expression: Recombinant DNA encoding the 20kDa GH isoform is expressed in bacterial systems.

  2. Purification: Chromatographic techniques (e.g., size-exclusion or reverse-phase HPLC) ensure >98% purity .

  3. Quality Control:

    • SDS-PAGE: Confirms uniform protein size.

    • RP-HPLC: Validates peptide integrity .

Biological Activities and Receptor Interactions

pGH 20kDa binds to the growth hormone receptor (GHR) but exhibits distinct signaling profiles compared to the pituitary 20kDa-hGH:

  • Lack of Lactogenic Activity: Unlike pituitary 20kDa-hGH, pGH 20kDa does not activate prolactin receptors , reducing potential side effects such as breast cancer risk .

  • GHR Signaling: Activates JAK2-STAT pathways, albeit with weaker phosphorylation kinetics compared to 22kDa-hGH .

PropertypGH 20kDaPituitary 20kDa-hGH
Lactogenic ActivityAbsent Present (reduced vs. 22kDa)
Receptor Binding AffinitySpecific to GHRBinds GHR and hGH-BP
Insulin-like EffectsNot reportedDetectable (20% of 22kDa)

Comparative Analysis with Pituitary 20kDa-hGH

While both isoforms share structural similarities (e.g., amino acid sequence except for a 15-residue deletion), their biological roles differ significantly:

AspectpGH 20kDaPituitary 20kDa-hGH
Primary FunctionPregnancy supportGrowth regulation
Complex FormationNot reportedForms 1:2 complexes with hGH-BP
Metabolic EffectsLimited dataComparable to 22kDa-hGH

Therapeutic Potential

  • Reduced Side Effects: The absence of lactogenic activity may mitigate risks associated with prolactin receptor activation .

  • Bone Metabolism: Pituitary 20kDa-hGH increases osteocalcin and lean mass , suggesting potential applications in osteoporosis, though placental-derived variants are less studied.

Challenges and Future Directions

  • Mechanistic Gaps: The precise role of pGH 20kDa in placental development and maternal physiology requires deeper investigation.

  • Comparative Studies: Further research is needed to contrast pGH 20kDa with pituitary isoforms in receptor internalization and nuclear transport .

Product Specs

Introduction
Growth Hormone (GH) belongs to the somatotropin/prolactin family of hormones, which are essential for regulating growth. This gene, along with four similar genes, is found on chromosome 17 at the growth hormone locus. These genes are arranged in the same transcriptional orientation, suggesting they evolved through gene duplication. The five genes exhibit a high degree of sequence similarity. Alternative splicing further amplifies the diversity of these hormones by producing additional isoforms of each, potentially leading to functional specialization. While expressed in the pituitary gland, this particular family member is not found in placental tissue, unlike the other four genes in the growth hormone locus. Mutations or deletions within this gene can lead to growth hormone deficiency and short stature.
Description
Recombinant Human Growth Hormone Placental 20kDa, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 177 amino acids. With a molecular mass of 20498 Daltons, it has a predicted isoelectric point (pI) of 8.20. Unlike pituitary GHs, GH 20K placental lacks lactogenic activity (mediated by the prolactin receptor). The purification of GH 20K placental is achieved using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
pGH 20kDa was lyophilized from a concentrated (1mg/ml) solution with 0.0045mM NaHCO3, pH adjusted to 11 prior to lyophilization.
Solubility
Reconstitute the lyophilized Placental Growth Hormone in 0.4% NaHCO3 or water adjusted to pH 11, at a concentration not less than 100 µg/ml. Further dilutions can be made using other aqueous solutions.
Stability
Lyophilized Growth Hormone 20K Placental remains stable at room temperature for up to 3 weeks. However, for long-term storage, it is recommended to store it desiccated below -18°C. After reconstitution and filter sterilization, GH 20K pl can be stored at 4°C for up to 4 weeks. For extended storage or more diluted solutions, the addition of a carrier protein (0.1% HSA or BSA) is recommended. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 98.0% as determined by: (a) Size Exclusion Chromatography-High Performance Liquid Chromatography (SEC-HPLC) analysis. (b) Sodium Dodecyl Sulphate-Polyacrylamide Gel Electrophoresis (SDS-PAGE) analysis.
Synonyms
GHL, GHV, GH-V, hGH-V, PGH.
Source
Escherichia Coli.
Amino Acid Sequence

AFPTI PLSRLFDNAM LRARRLYQLA YDTYQEFNPQ TSLCFSESIP TPSNRVKTQQ KSNLELLRIS LLLIQSWLEP VQLLRSVFAN SLVYGASDSN VYRHLKDLEE GIQTLMWRLE DGSPRTGQIF NQSYSKFDTK SHNDDALLKN YGLLYCFRKD MDKVETFLRI VQCRSVEGSC GF

Q&A

What is 20kDa human placental growth hormone and how does it differ from 22kDa GH?

20kDa human placental growth hormone (20kDa GH-V) is a naturally occurring variant of growth hormone produced by the placenta during pregnancy. It differs from the more abundant 22kDa GH variant primarily through the absence of amino acids 32-46 in its protein structure. While 22kDa GH (also called GH-N) is predominant in blood circulation (90-95%), 20kDa GH represents approximately 5-10% of circulating GH . The structural difference leads to significant functional variations, particularly in metabolic activity, receptor binding, and signaling kinetics. Both hormones bind to the same growth hormone receptor (GHR), but 20kDa GH-V notably lacks the diabetogenic activity of the 22kDa variant and does not bind to prolactin receptors (PRLR), which has important implications for cancer research and therapeutic development .

What experimental models are most appropriate for studying 20kDa human placental GH?

Research on 20kDa GH-V has utilized several experimental models, each with specific advantages. GH-deficient mouse models have proven valuable for evaluating growth-promoting effects, with studies demonstrating that 20kDa GH-V administration produces significant increases in circulating IGF-1, femur length, and body length compared to saline controls . For cellular signaling studies, both 3T3-F442A cells and Chinese hamster ovary (CHO) cells transfected with growth hormone receptor (CHO-GHR) have been successfully employed to analyze signaling cascades through Western blot analyses . Bone marrow-derived mesenchymal stem cells (BM-MSCs) provide an excellent model for studying differential effects on osteogenic differentiation . When selecting a model, researchers should consider whether they are investigating metabolic effects (mouse models), signaling properties (transfected cell lines), or specific tissue responses (primary cell cultures) to align with research objectives.

How should researchers approach the purification of recombinant 20kDa human placental GH for experimental use?

Purification of recombinant 20kDa human placental GH requires careful consideration of expression systems and purification strategies. Based on recombinant protein purification principles, researchers should consider: (1) Selection of an appropriate expression system such as Origami B DE3 or Rosettagami B DE3 bacterial strains, which provide enhanced protein folding environments for disulfide bond formation ; (2) Utilization of fusion protein approaches, with SUMO fusion protein expression systems showing particular promise for enhancing solubility and proper folding ; (3) Implementation of multi-parameter cleavage experimental designs to optimize protein recovery while maintaining biological activity; (4) Verification of biological activity through functional assays such as cell migration or phosphorylation studies. Researchers should validate their purified protein through both structural analysis (SDS-PAGE, mass spectrometry) and functional assays before application in experimental settings to ensure biological relevance of findings.

How does receptor internalization differ between 20kDa and 22kDa GH, and what methodologies best capture these differences?

Growth hormone receptor (GHR) internalization patterns differ significantly between 20kDa and 22kDa GH stimulation. Research indicates that while both variants induce GHR internalization in a time-dependent manner, 20kDa GH uniquely facilitates GHR transport into cell nuclei of bone marrow mesenchymal stem cells (BM-MSCs) . To effectively capture these differences, researchers should employ multiple complementary methodologies: (1) Fluorescence microscopy with GHR-GFP fusion proteins to visualize trafficking patterns in real-time; (2) Subcellular fractionation followed by Western blotting to quantify receptor distribution across cellular compartments; (3) Flow cytometry to measure surface receptor depletion rates; (4) Co-immunoprecipitation studies to identify differential interaction partners during internalization. Additionally, pulse-chase experimental designs using biotinylated or radio-labeled GH variants can provide valuable kinetic data on receptor recycling versus degradation pathways. When interpreting results, researchers should control for cell type-specific effects, as internalization patterns may vary significantly between different tissues and experimental models.

What experimental approaches are most effective for investigating the lack of diabetogenic activity in 20kDa human placental GH?

Investigating the lack of diabetogenic activity in 20kDa human placental GH requires a multi-faceted experimental approach spanning in vivo and in vitro methodologies. In vivo approaches should include: (1) Glucose tolerance tests in GH-deficient mice treated with either 20kDa GH-V or 22kDa GH-N, measuring blood glucose clearance rates at specific intervals (typically 15, 30, 60, and 120 minutes) post-glucose challenge; (2) Insulin tolerance tests to directly assess insulin sensitivity; (3) Hyperinsulinemic-euglycemic clamp studies for precise quantification of insulin-stimulated glucose disposal; (4) Analysis of pancreatic beta cell function through measurement of insulin and C-peptide levels, as 20kDa GH-V has been shown to reduce these parameters compared to 22kDa GH . In vitro approaches should focus on molecular pathways in insulin-responsive tissues: (5) Primary hepatocyte cultures to measure GH effects on gluconeogenesis and glycogenolysis; (6) Adipocyte models to quantify insulin receptor signaling and glucose transporter translocation; (7) Skeletal muscle cells to assess changes in insulin receptor substrate (IRS) phosphorylation and PI3K/Akt pathway activation. Timing of measurements is critical—diabetogenic effects are transient, requiring assessment within 1-5 hours after GH administration .

How does 20kDa human placental GH influence osteogenic differentiation in mesenchymal stem cells compared to 22kDa GH?

Research has demonstrated that 20kDa and 22kDa GH variants exert differential effects on osteogenic differentiation of bone marrow mesenchymal stem cells (BM-MSCs) . To properly investigate these differences, researchers should implement a comprehensive experimental design including: (1) Assessment of early osteogenic markers (RUNX2, ALP) through qPCR and enzymatic activity assays; (2) Evaluation of matrix mineralization via Alizarin Red staining at multiple time points (typically days 7, 14, and 21); (3) Quantification of later-stage markers such as osteocalcin and osteopontin through ELISA; (4) Analysis of signaling pathway activation patterns, particularly MAPK and STAT pathways, which may be differentially affected by the two GH variants. Western blot analysis reveals that both variants activate the JAK2-STAT5/3 signaling pathway in BM-MSCs in a dose-dependent manner, though with different kinetics and intensities . These differential signaling profiles likely contribute to observed variations in osteogenic capacity. Researchers should carefully consider dose-response relationships, as optimal concentrations for osteogenic induction may differ between the variants, with 20-40nM shown to be effective for 22kDa GH activation of critical pathways .

What methods are most appropriate for studying the differential effects of 20kDa GH on PRLR-positive cancer proliferation?

The lack of prolactin receptor (PRLR) binding by 20kDa GH-V (in contrast to 22kDa GH-N) has significant implications for PRLR-positive cancer research. When designing experiments to study these differential effects, researchers should employ multiple complementary approaches: (1) Cell proliferation assays using PRLR-positive cancer cell lines (breast, prostate, ovarian, and colon cancers have demonstrated PRLR expression) treated with equivalent concentrations of either 20kDa or 22kDa GH variants; (2) Receptor binding studies using competitive displacement assays to confirm differential PRLR binding; (3) PRLR signaling pathway analysis focusing on JAK2-STAT5 activation, which is typically induced by PRLR activation; (4) 3D tumor spheroid models to better recapitulate the tumor microenvironment; (5) In vivo xenograft models with PRLR-positive tumors to assess tumor growth kinetics in response to 20kDa versus 22kDa GH treatment. Previous studies have demonstrated that three distinct PRLR-positive human cancer cell lines (two breast and one colon cancer) showed significantly reduced proliferation rates when treated with 20kDa GH-V compared to 22kDa GH-N . These findings suggest potential therapeutic applications for 20kDa GH in contexts where PRLR signaling promotes cancer progression.

What are the key considerations when developing a functional assay system for testing bioactivity of recombinant and synthetic 20kDa human placental GH?

Developing robust functional assay systems for 20kDa GH-V requires careful attention to several methodological aspects. First, researchers must consider cell model selection: primary cells provide physiological relevance but exhibit donor variability, while stably transfected cell lines offer consistency but may lack complete physiological signaling networks . Chemotaxis assays have proven particularly effective, with studies demonstrating that regioselectively cyclized synthetic rhesus CCL20 (a chemokine) shows high bioactivity in migration assays . When establishing such systems, researchers should: (1) Validate receptor expression through flow cytometry or Western blotting; (2) Determine optimal hormone concentration ranges through dose-response studies (typically 5-100nM); (3) Include appropriate positive and negative controls, including commercially validated 22kDa GH; (4) Implement multiple readout methodologies (proliferation, signaling activation, gene expression) to comprehensively assess bioactivity. For stable cell line development, pre-treatment protocols may be necessary to ensure consistent receptor expression, as demonstrated by studies showing that only pre-treated stable CCR6-expressing L1.2 cells exhibited migration toward synthetic macaque CCL20 . Rigorous validation of synthetic or recombinant 20kDa GH preparations is essential before proceeding to more complex experimental applications.

What methodological approaches best address the challenge of studying species-specific differences in 20kDa GH activity?

Investigating species-specific differences in 20kDa GH activity presents significant methodological challenges, particularly given the limited availability of species-specific reagents for non-human primates and other experimental models . To address these challenges, researchers should implement: (1) Recombinant DNA technology optimized for expression of species-specific GH variants—systems utilizing Origami B DE3 or Rosettagami B DE3 bacterial strains have shown promise for non-human primate chemokine expression ; (2) Comparative sequence analysis coupled with structural modeling to identify functionally significant interspecies differences; (3) Cross-species receptor binding and activation studies using cells transfected with species-specific GH receptors; (4) Development of species-specific antibodies and ELISAs for accurate quantification; (5) Careful selection of heterologous systems when homologous systems are unavailable, with appropriate controls to account for interspecies variations. Given that most commercially available reagents have human specificity, developing efficient protocols for expression and purification of non-human primate growth hormones is essential . Researchers should also consider using regioselective cyclization peptide synthesis as an alternative to recombinant protein expression, as this approach has successfully generated bioactive non-human primate chemokines .

What evidence supports the therapeutic potential of 20kDa human placental GH for patients with growth hormone deficiency?

Several lines of evidence support the therapeutic potential of 20kDa human placental GH (GH-V) for growth hormone deficiency (GHD), particularly in populations at risk for metabolic complications. Studies in GH-deficient mouse models demonstrate that 20kDa GH-V treatment produces significant increases in circulating IGF-1, femur length, and body length compared to saline-treated controls . Importantly, these growth-promoting effects were comparable to those observed with standard 22kDa GH-N (hGH) treatment, indicating that 20kDa GH-V retains full capacity to stimulate IGF-1 production and longitudinal bone growth . Additionally, 20kDa GH-V significantly increases lean body mass while decreasing fat mass in GH-deficient mice, mirroring the body composition effects of standard GH therapy . The critical therapeutic advantage lies in 20kDa GH-V's metabolic profile—it lacks the diabetogenic activity observed with standard GH treatment, failing to cause hyperinsulinemia and demonstrating significantly improved insulin sensitivity compared to 22kDa GH-N treated mice . This metabolic benefit makes 20kDa GH-V particularly promising for adult GHD patients, who face increased risk of metabolic syndrome and diabetes with advancing age. Though clinical translation requires additional research, these findings from multiple independent laboratories suggest 20kDa GH-V may represent a significant improvement over current GH therapies for specific patient populations.

What experimental approaches should be prioritized to advance understanding of 20kDa human placental GH's potential in cancer-related applications?

Given 20kDa GH-V's unique inability to bind prolactin receptors (PRLR), prioritizing research into its cancer-related applications requires focused experimental approaches. Researchers should implement: (1) Comprehensive comparison studies examining proliferation rates across multiple PRLR-positive cancer cell lines (breast, prostate, ovarian, colon) treated with equivalent concentrations of 20kDa GH-V versus 22kDa GH-N; (2) Mechanistic investigations into downstream signaling pathways specific to PRLR activation that may contribute to cancer progression; (3) Xenograft models using PRLR-positive human cancers to assess in vivo tumor growth kinetics in response to different GH variants; (4) Combination therapy experiments evaluating whether 20kDa GH-V can provide growth-promoting benefits while mitigating cancer risk when combined with standard oncology treatments. Previous studies have already established that three distinct PRLR-positive human cancer cell lines showed significantly reduced proliferation rates when treated with 20kDa GH-V compared to standard hGH . These findings align with the theory that GH stimulation of PRLR partially drives proliferation of PRLR-positive cancers. For clinical translation, additional studies should address pharmacokinetics, optimal dosing strategies, and potential development of antibodies specific to 20kDa GH-V to enable precise monitoring during therapy.

How should researchers design comparative studies between 20kDa and 22kDa GH to identify novel therapeutic applications?

Designing effective comparative studies between 20kDa and 22kDa GH variants requires careful consideration of experimental parameters to identify novel therapeutic applications. Researchers should implement: (1) Parallel dose-response studies across multiple cell types and tissues to identify differential sensitivity thresholds—Western blot analyses have shown that activation of JAK2-STAT signaling occurs at different concentrations for each variant, with 5nM sufficient for 22kDa GH activation but differential activation patterns for 20kDa GH ; (2) Temporal analysis of signaling cascade activation, as the kinetics of phosphorylation differ significantly between variants ; (3) Transcriptomic and proteomic profiling to identify variant-specific gene expression patterns that may reveal unique therapeutic targets; (4) Tissue-specific response evaluation, particularly in metabolically active tissues (liver, muscle, adipose) and those with high GH receptor expression; (5) Age-dependent and sex-dependent comparative studies, as hormone responsiveness may vary with these parameters. Experimental design should account for the distinct lack of PRLR binding by 20kDa GH-V and its reduced diabetogenic potential . Given that 20kDa GH-V induces GHR translocation to the nucleus in BM-MSCs , nuclear signaling mechanisms represent an intriguing area for investigation that might reveal novel therapeutic applications beyond traditional GH replacement.

Product Science Overview

Introduction

Growth Hormone Placental 20kDa (Human Recombinant), often abbreviated as GH-V, is a variant of human growth hormone (hGH) produced in the placenta. Unlike the pituitary-derived human growth hormone (hGH-N), which is a 22-kilodalton (kDa) protein, GH-V is a 20-kDa protein. This variant is produced through recombinant DNA technology, which allows for the synthesis of proteins identical to those naturally occurring in the human body.

Structure and Expression

GH-V is a single, non-glycosylated polypeptide chain containing 177 amino acids and has a molecular mass of approximately 20,498 Daltons . It is expressed in the syncytiotrophoblast and extravillous cytotrophoblast layers of the human placenta. The levels of GH-V increase throughout pregnancy and eventually replace hGH-N in the maternal circulation .

Biological Functions

GH-V plays a crucial role in regulating in utero growth and development. It has similar somatogenic effects to hGH-N but with reduced lactogenic activity . This means that while GH-V promotes growth, it does not stimulate the production of milk as effectively as hGH-N. Additionally, GH-V has been shown to have a beneficial reduction in diabetogenic activity, making it a potentially safer alternative for therapeutic use .

Preparation Methods

The recombinant form of GH-V is produced using Escherichia coli (E. coli) as the host organism. The gene encoding GH-V is inserted into the bacterial DNA, allowing the bacteria to produce the protein. The protein is then purified through various biochemical techniques to ensure its purity and functionality .

Chemical Reactions and Analysis

Studies have shown that GH-V can stimulate insulin-like growth factor-1 (IGF-1) and subsequent longitudinal body growth in GH-deficient models, similar to hGH-N . However, unlike hGH-N, GH-V does not inhibit insulin action or promote the growth of prolactin receptor-positive cancers . This makes GH-V a promising candidate for growth hormone therapies, especially for individuals at risk for metabolic syndrome or certain types of cancer.

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.