GH Human 20kDa

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

Description

Growth Hormone Receptor (GHR) Activation

20kDa hGH binds to GHR but induces receptor dimerization differently than 22kDa hGH:

  • Sequential binding: 20kDa hGH binds the first GHR subunit with lower affinity (Kd1 ≈ 1.6 × 10⁻⁸ M) but exhibits higher affinity for the second subunit (Kd2 ≈ 2.5 × 10⁻¹⁰ M) .

  • Agonistic activity: Despite forming a 1:2 GHR complex, 20kDa hGH activates downstream signaling (e.g., JAK2-STAT5/3 pathways) comparably to 22kDa hGH .

Prolactin Receptor (PRLR) Engagement

20kDa hGH exhibits weaker activity on PRLR compared to 22kDa hGH:

  • Lower potency: EC₅₀ for 20kDa hGH in PRLR-mediated cell proliferation is ~10× higher than 22kDa hGH (15 nM vs. 1.5 nM) .

  • Zinc dependency: Zinc ions enhance 20kDa hGH activity more than 22kDa hGH, but even at physiological Zn²⁺ levels (0.25–1 µM), 20kDa hGH remains less potent .

Osteogenic Differentiation in MSCs

Bone marrow-derived mesenchymal stem cells (BM-MSCs) respond differently to 20kDa and 22kDa hGH:

  • Signaling divergence:

    • 22kDa hGH: Strongly activates STAT5/3 and JAK2 phosphorylation at lower concentrations (5–40 nM) .

    • 20kDa hGH: Requires higher concentrations (≥100 nM) and shows reduced STAT5 activation .

  • Internalization: 20kDa hGH localizes to the cytoplasm, while 22kDa hGH enters the nucleus, suggesting distinct intracellular trafficking .

Parameter20kDa hGH22kDa hGH
Nuclear uptakeAbsent Present
GHR internalizationCytoplasmic Nuclear
Osteogenic differentiationReduced vs. 22kDa hGH Enhanced

Pharmacokinetics and Secretion

  • Half-life: 20kDa hGH has a longer half-life (18.7 ± 0.8 min) compared to 22kDa hGH (14.7 ± 0.8 min) .

  • Cosecretion: 20kDa hGH is secreted in a pulsatile manner coinciding with 22kDa hGH, maintaining a constant 20:22 ratio under physiological conditions .

  • Exogenous suppression: Administration of recombinant 22kDa hGH rapidly reduces endogenous 20kDa hGH levels, indicating feedback regulation .

Potential Applications

  • Reduced PRLR-mediated side effects: 20kDa hGH’s weaker PRLR activation may mitigate risks like breast cancer in therapeutic settings .

  • IGF-I stimulation: 20kDa hGH enhances IGF-I mRNA expression at lower concentrations than 22kDa hGH in GHR-expressing cells .

Challenges and Research Gaps

  • Limited physiological role: The exact biological significance of 20kDa hGH remains unclear, though it may modulate stem cell behavior and tissue repair .

  • Diagnostic utility: A 20:22 ratio is proposed as a biomarker for exogenous 22kDa hGH administration .

Product Specs

Introduction
Growth hormone (GH) is part of the somatotropin/prolactin hormone family, essential for growth regulation. Located on chromosome 17, the GH gene clusters with four related genes at the growth hormone locus. This arrangement, with all genes in the same transcriptional orientation, likely arose from gene duplications. These five genes share significant sequence similarity. Alternative splicing generates diverse isoforms of each growth hormone, expanding their functions. This specific family member is expressed in the pituitary gland, unlike the other four genes in the locus, which are also found in placental tissue. Mutations or deletions in the GH gene result in growth hormone deficiency and short stature.
Description
Recombinant Human Growth Hormone 20KDa, produced in E. coli, is a single, non-glycosylated polypeptide chain. It comprises 177 amino acids, with a molecular weight of 20,322 Daltons. HGH-20kDa is purified using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
HGH-20K was lyophilized from a concentrated solution (1mg/ml) containing 0.0045mM NaHCO3, adjusted to a pH of 10.
Solubility
Reconstitute the lyophilized Pituitary Growth Hormone in 0.4% NaHCO3 solution, adjusted to pH 10. The initial concentration should be at least 100 µg/ml. This solution can be further diluted in other aqueous solutions.
Stability
Lyophilized Pituitary Growth Hormone-20K remains stable at room temperature for up to 3 weeks. However, it should be stored desiccated below -18°C. After reconstitution and filter sterilization, GH-20K can be stored at 4°C for a maximum of 4 weeks. For long-term storage or when using more diluted solutions, adding 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.
Protein Content
HGH-20K quantification was performed using two independent methods: 1. UV spectroscopy at 280 nm. An absorbance value of 0.735 was used as the extinction coefficient for a 0.1% (1mg/ml) solution. This value is calculated by the PC GENE computer analysis program for protein sequences (IntelliGenetics). 2. Reverse Phase-High Performance Liquid Chromatography (RP-HPLC) analysis. A calibrated solution of GH-20K serves as a reference standard.
Synonyms
GH1, GH, GHN, GH-N, hGH-N,Pituitary growth hormone, Growth hormone 1, Somatotropin.
Source
Escherichia Coli.
Amino Acid Sequence
The sequence of the first five N-terminal amino acids was determined and was found to be Ala-Phe-Pro-Thr-Ile.

Q&A

What is 20kDa human growth hormone and how does it differ structurally from the standard 22kDa variant?

The 20kDa human growth hormone (20K hGH) is a naturally occurring variant protein in the human pituitary, representing approximately 5-10% of pituitary GH, while the 22kDa form (22K hGH) constitutes about 75% of pituitary GH. The 20K hGH has an amino acid sequence identical to the 22K form except for a 15 amino acid internal deletion of residues 32-46, which results from an alternative splicing event of the human GH gene . This structural difference alters the protein's three-dimensional conformation and affects its interaction with various receptors, despite both variants being products of the same gene.

What methodologies can be used to detect and quantify 20K hGH in biological samples?

Detection and quantification of 20K hGH in biological samples requires specialized techniques due to its structural similarity to 22K hGH. Effective methodological approaches include:

  • Two-site immunoassays using monoclonal antibodies specifically recognizing the unique epitopes of 20K hGH

  • Mass spectrometry-based proteomics, which can distinguish between the two variants based on their molecular weight differences

  • RT-PCR detection of alternatively spliced mRNA transcripts

  • Bioassays based on the differential effects of 20K and 22K hGH on cell proliferation in receptor-expressing cell lines
    For instance, researchers have developed novel bioassays based on hGH receptor-mediated cell proliferation that can specifically measure 20K hGH and its modified forms . When implementing these techniques, it's crucial to validate assay specificity using recombinant standards and to establish appropriate detection thresholds relevant to physiological concentrations.

How do the receptor binding properties of 20K hGH differ from 22K hGH?

The 20K hGH exhibits distinct binding properties compared to 22K hGH across different receptor types:

Receptor20K hGH Binding22K hGH BindingFunctional Implications
GH Receptor (GHR)Full agonist with unique dimerization mechanismFull agonist20K hGH stimulates IGF-I gene expression more potently at lower concentrations
Prolactin Receptor (PRLR)Minimal to no bindingStrong binding20K hGH has significantly reduced lactogenic activity
Zinc DependenceGreater enhancement by Zn²⁺Less enhancement by Zn²⁺Zn²⁺ supplementation equalizes activities of both forms
Research has demonstrated that the binding between stem regions of the GH receptor compensates for the weaker site 1 binding of 20K hGH compared to 22K hGH . This compensation mechanism enables 20K hGH to function as a full agonist for GHR despite having different binding characteristics. The EC₅₀ for 20K hGH in PRLR activation is approximately 10-fold higher than that of 22K hGH (15 nM vs. 1.5 nM), indicating substantially lower potency at the prolactin receptor .

How does 20K hGH activate differential signaling pathways compared to 22K hGH?

Both 20K and 22K hGH activate similar but not identical signaling cascades through their interactions with receptors. Key differences in signaling pathways include:

  • GHR-mediated signaling: Both variants activate the JAK2/STAT5 pathway through GHR, but 20K hGH induces stronger IGF-I gene expression at lower concentrations in Ba/F3-hGHR cells . This suggests that 20K hGH may have unique interactions with the downstream effectors of GHR signaling.

  • PRLR-mediated signaling: 20K hGH demonstrates significantly reduced ability to activate STAT5 tyrosine phosphorylation in human breast cancer T-47D cells compared to 22K hGH . This reduced activation extends to the Spi2.1 gene promoter in cells expressing hPRLR, where 20K hGH shows only modest effects .

  • Metabolic signaling: The differential signaling profile of 20K hGH likely contributes to its reduced diabetogenic activity. The underlying mechanisms may involve altered effects on insulin receptor sensitivity, hepatic glucose production, and peripheral glucose uptake pathways .
    When designing experiments to investigate these pathways, researchers should implement phospho-specific Western blotting, reporter gene assays, and RNA-seq analyses to comprehensively characterize the differential signaling profiles.

What experimental systems are most suitable for studying 20K hGH receptor interactions?

Several experimental systems have proven valuable for investigating 20K hGH receptor interactions:

  • Ba/F3-hGHR cells: This pro-B cell line stably transfected with human GH receptor has been effectively used to study IGF-I gene expression in response to 20K hGH . These cells lack endogenous GH or prolactin receptors, making them ideal for studying specific receptor-mediated effects.

  • Ba/F3-hPRLR cells: These cells express human prolactin receptor and have been employed to compare the effects of 20K and 22K hGH on prolactin receptor signaling and cell proliferation .

  • Surface plasmon resonance (SPR): This technique allows real-time measurement of binding kinetics between 20K hGH and its receptors, providing association and dissociation rates.

  • Fluorescence resonance energy transfer (FRET): Particularly useful for studying receptor dimerization upon hormone binding, which is crucial for understanding the unique dimerization mechanism of 20K hGH.

  • CHO-hPRLR cells: Chinese hamster ovary cells expressing human prolactin receptor have been utilized to study gene promoter activation and JAK2/STAT5 phosphorylation .
    When implementing these systems, researchers should carefully control for receptor expression levels, as these can significantly impact the observed binding properties and downstream signaling events.

How can researchers experimentally demonstrate the differential effects of 20K hGH on IGF-I gene expression?

To experimentally demonstrate the differential effects of 20K hGH on IGF-I gene expression compared to 22K hGH, researchers can employ several methodological approaches:

  • Dose-response studies: Ba/F3-hGHR cells can be incubated with various concentrations (10 pM to 10 nM) of 20K or 22K hGH for defined periods (e.g., 2 hours), followed by RT-PCR analysis of IGF-I mRNA expression . This approach has demonstrated that lower concentrations of 20K hGH more strongly induce IGF-I gene expression than 22K hGH.

  • Time-course experiments: Analyzing IGF-I mRNA expression at different time points after exposure to equimolar concentrations of 20K and 22K hGH to determine differences in the kinetics of gene induction.

  • Signaling pathway analysis: Using specific inhibitors of JAK2, STAT5, or other signaling molecules to determine which pathways are differentially engaged by 20K vs. 22K hGH.

  • ChIP assays: Chromatin immunoprecipitation to examine differential recruitment of transcription factors to the IGF-I promoter in response to 20K vs. 22K hGH.

  • Reporter gene assays: Constructing IGF-I promoter-reporter constructs to quantitatively measure promoter activation by both hormone variants.
    Control experiments should include treatment with non-GH hormones (such as PRL or IL-3) and testing in parental cells lacking GH receptors to confirm specificity, as these treatments do not induce IGF-I gene expression in Ba/F3-hGHR cells .

Why does 20K hGH lack the diabetogenic activity observed with 22K hGH?

The 20K hGH variant lacks the diabetogenic activity characteristic of 22K hGH, representing a significant physiological difference with important therapeutic implications. Multiple experimental models have confirmed this property:

  • In GH-deficient mice, treatment with 20K hGH did not induce the hyperinsulinemia observed in mice treated with 22K hGH .

  • Mice receiving 20K hGH demonstrated significantly greater insulin sensitivity compared to those treated with 22K hGH .

  • In high-fat-fed rats, 20K hGH treatment reduced insulin and C-peptide levels compared to rats receiving 22K hGH treatment .
    The molecular basis for this difference likely stems from several factors:

  • Altered receptor binding and downstream signaling patterns that differently affect insulin receptor sensitivity

  • Differential effects on hepatic glucose production and peripheral glucose uptake

  • Lack of prolactin receptor activation, as prolactin signaling has been implicated in insulin resistance
    When designing experiments to investigate this phenomenon, researchers should ensure that measurements of insulin sensitivity are performed within an appropriate time window (1-5 hours after GH administration), as the diabetogenic effects of GH are transient . The absence of this diabetogenic activity makes 20K hGH a promising candidate for therapeutic applications in individuals at risk for insulin resistance or metabolic disease.

How does 20K hGH affect prolactin receptor signaling and what are the implications for cancer research?

Unlike 22K hGH, which binds and activates both the GH receptor (GHR) and prolactin receptor (PRLR), 20K hGH exhibits minimal binding to PRLR and consequently minimal activation of PRLR-mediated signaling pathways . Research has confirmed that 20K hGH has a significantly reduced ability to stimulate STAT5 tyrosine phosphorylation in human breast cancer T-47D cells via PRLR compared to 22K hGH .
This limited PRLR activation has significant implications for cancer research, particularly regarding PRLR-positive cancers:

  • Experimental evidence shows that three distinct PRLR-positive human cancer cell lines (two breast and one colon cancer cell line) displayed significantly reduced proliferation rates when treated with 20K hGH compared to 22K hGH .

  • PRLR signaling has been implicated in the progression of multiple cancer types:

    • Breast cancer

    • Prostate cancer

    • Ovarian cancer

    • Colon cancer

  • The reduced ability of 20K hGH to stimulate these cancer cells suggests that it may represent a safer alternative to 22K hGH for GH replacement therapy in individuals with elevated risk of PRLR-positive cancers.
    Methodologically, when studying these effects, researchers should incorporate specific PRLR antagonists as controls and examine downstream signaling events beyond proliferation, including cell migration, invasion, and apoptosis resistance.

What are the growth-promoting effects of 20K hGH compared to 22K hGH in animal models?

Studies in animal models have demonstrated that 20K hGH exhibits growth-promoting effects comparable to 22K hGH, despite differences in receptor binding and signaling profiles. Key experimental findings include:

Growth ParameterEffect of 20K hGHComparison to 22K hGHExperimental Model
Circulating IGF-1Significant increaseSimilar increaseGH-deficient mice
Body LengthSignificant increaseSimilar increaseGH-deficient mice
Femur LengthSignificant increaseSimilar increaseGH-deficient mice
Lean Body MassSignificant increaseSimilar increaseGH-deficient mice
Fat MassSignificant decreaseSimilar decreaseGH-deficient mice
When designing animal studies to investigate these effects, researchers should consider:
  • Using appropriate GH-deficient models to eliminate interference from endogenous GH

  • Implementing dose-response studies to account for potential differences in potency

  • Conducting long-term studies to assess sustained effects on growth parameters

  • Including comprehensive body composition analysis using techniques like DEXA or MRI

  • Measuring both circulating IGF-1 and local tissue IGF-1 expression to understand systemic versus local effects
    These findings suggest that 20K hGH retains the full growth-promoting capabilities of 22K hGH while potentially offering improved metabolic and safety profiles .

How should researchers reconcile conflicting data on the effects of 20K hGH on insulin sensitivity?

Interpreting conflicting data on the effects of 20K hGH on insulin sensitivity requires a systematic methodological approach considering several factors:

What experimental approaches can be used to study the therapeutic potential of 20K hGH in PRLR-positive cancers?

To investigate the therapeutic potential of 20K hGH in PRLR-positive cancers, researchers can employ several experimental approaches:

  • In vitro cancer cell studies:

    • Comparative proliferation assays using multiple PRLR-positive cancer cell lines (breast, prostate, ovarian, colon) treated with 20K vs. 22K hGH

    • Analysis of cell cycle progression, apoptosis, migration, and invasion in response to both GH variants

    • Molecular studies examining differential activation of JAK2/STAT5 and other oncogenic signaling pathways

  • Mechanistic investigations:

    • siRNA knockdown or CRISPR/Cas9 deletion of PRLR to confirm its role in differential responses

    • Phospho-proteomic analysis to identify signaling differences between 20K and 22K hGH treatment

    • Co-immunoprecipitation studies to examine differential protein-protein interactions

  • In vivo cancer models:

    • Xenograft models using PRLR-positive human cancer cells in immunodeficient mice

    • Patient-derived xenografts to better represent tumor heterogeneity

    • Transgenic mouse models prone to developing PRLR-dependent tumors

  • Therapeutic intervention studies:

    • Comparison of 20K hGH vs. 22K hGH effects on established tumors

    • Combination approaches with conventional cancer therapies

    • Measurement of tumor growth, metastasis, angiogenesis, and survival endpoints

  • Translational models:

    • Ex vivo culture of primary human cancer samples with 20K vs. 22K hGH

    • Analysis of patient samples for markers that predict differential response to GH variants
      This comprehensive experimental approach would provide robust evidence regarding the potential advantages of 20K hGH over conventional GH therapy in patients with or at risk for PRLR-positive cancers .

What are the key challenges in translating 20K hGH research to clinical applications?

Translating 20K hGH research from preclinical models to human clinical applications faces several significant challenges that researchers must address:

  • Production and characterization challenges:

    • Developing cost-effective methods for large-scale production of pharmaceutical-grade 20K hGH

    • Ensuring proper folding, stability, and bioactivity of the recombinant protein

    • Establishing reliable analytical methods for quality control and pharmacokinetic studies

  • Regulatory considerations:

    • Extensive preclinical safety data requirements for novel biologics

    • Comparative trials with established 22K hGH therapies

    • Demonstration of meaningful advantages over existing treatments to justify approval

  • Clinical trial design:

    • Patient selection (children vs. adults with GH deficiency)

    • Determination of optimal dosing regimens, which may differ from conventional GH

    • Selection of appropriate endpoints (growth velocity, metabolic parameters, quality of life)

    • Statistical power to detect differences from standard GH therapy

  • Safety monitoring:

    • Long-term safety surveillance for unforeseen adverse effects

    • Immunogenicity assessment

    • Cancer surveillance, especially in adult populations

  • Patient-specific factors:

    • Identifying patient populations most likely to benefit from 20K hGH's unique properties

    • Developing personalized dosing strategies based on individual response markers

    • Addressing potential differences in efficacy across different etiologies of GH deficiency

  • Clinical implementation:

    • Education of clinicians about the unique properties of 20K hGH

    • Development of specific monitoring protocols

    • Integration into existing treatment guidelines
      The most promising initial clinical applications may be in adult GH deficiency patients with metabolic comorbidities or cancer history, where the reduced diabetogenic activity and lack of PRLR activation could provide meaningful benefits compared to conventional GH therapy .

What novel therapeutic applications might exploit the unique properties of 20K hGH?

The distinct properties of 20K hGH open possibilities for novel therapeutic applications beyond conventional growth hormone replacement therapy:

  • Metabolic disease applications:

    • Treatment of sarcopenia and frailty in elderly patients, leveraging the anabolic effects without diabetogenic consequences

    • Body composition improvement in obesity without exacerbating insulin resistance

    • Potential adjunctive therapy in type 2 diabetes where growth hormone is needed but its diabetogenic effects are contraindicated

  • Cancer-related applications:

    • Safer GH replacement in cancer survivors, particularly those with histories of PRLR-positive cancers

    • Cachexia treatment in cancer patients, where anabolic effects without cancer stimulation would be valuable

    • Combination with conventional cancer therapies where PRLR signaling contributes to treatment resistance

  • Tissue-specific regenerative applications:

    • Promotion of bone healing in osteoporotic fractures

    • Acceleration of wound healing in diabetic patients

    • Muscle regeneration following injury or surgery

  • Novel delivery approaches:

    • Development of long-acting formulations with reduced frequency of administration

    • Tissue-targeted delivery systems to enhance desired effects while minimizing systemic exposure

    • Combination with complementary therapies (e.g., insulin sensitizers) for synergistic effects

  • Personalized medicine applications:

    • Development of biomarker panels to identify patients most likely to benefit from 20K hGH

    • Genetic profiling to predict response based on receptor variants or metabolic characteristics
      Each of these applications would require rigorous preclinical validation followed by carefully designed clinical trials with appropriate endpoints and safety monitoring. The greatest near-term potential likely lies in populations where the metabolic and cancer-related benefits of 20K hGH would provide clinically meaningful advantages over conventional GH therapy .

How might researchers address the knowledge gaps in understanding 20K hGH biological activity?

Several significant knowledge gaps remain in understanding 20K hGH biological activity, which researchers can address through the following methodological approaches:

  • Receptor biology and signaling:

    • Cryo-electron microscopy studies to elucidate the structural basis for differential receptor binding and activation

    • Systems biology approaches to map the complete signaling networks activated by 20K vs. 22K hGH

    • CRISPR/Cas9 screening to identify novel molecular components involved in 20K hGH signaling

  • Tissue-specific effects:

    • Single-cell transcriptomics to characterize cell type-specific responses to 20K hGH

    • Tissue-specific receptor knockout models to delineate the relative contribution of different tissues

    • Metabolomics analysis to identify unique metabolic signatures of 20K hGH action

  • Developmental biology:

    • Effects of 20K hGH during different developmental stages

    • Longitudinal studies examining sustained versus transient effects

    • Epigenetic profiling to identify lasting changes induced by 20K hGH exposure

  • Comparative physiology:

    • Expanded studies across multiple species to better understand evolutionary conservation

    • Comprehensive comparison of pituitary 20K GH-N versus placental 20K GH-V effects

    • Examination of sex-specific differences in response to 20K hGH

  • Clinical translation:

    • Development of specific biomarkers of 20K hGH activity in humans

    • Non-invasive imaging methods to assess tissue-specific responses

    • Physiological studies in healthy volunteers and patients with GH deficiency
      These research approaches would significantly advance our understanding of 20K hGH biology and facilitate its potential therapeutic applications. Importantly, researchers should employ multiple complementary techniques rather than relying on single methodological approaches to address these complex questions .

What technological advances would facilitate better investigation of 20K hGH mechanisms and applications?

Several technological advances would significantly enhance the investigation of 20K hGH mechanisms and applications:

  • Protein engineering and production technologies:

    • CRISPR-engineered cell lines producing higher yields of properly folded 20K hGH

    • Cell-free protein synthesis systems for rapid production of variant forms for structure-function studies

    • Novel purification strategies for large-scale, high-purity production

  • Advanced structural biology approaches:

    • AlphaFold and other AI-based protein structure prediction to model 20K hGH-receptor interactions

    • Time-resolved crystallography to capture dynamic aspects of receptor activation

    • Hydrogen-deuterium exchange mass spectrometry to map conformational changes during signaling

  • Gene expression and regulation technologies:

    • CRISPR activation/interference systems to modulate GH receptor expression in target tissues

    • Single-molecule RNA FISH to visualize GH-induced gene expression at the cellular level

    • Spatial transcriptomics to map tissue responses with high anatomical resolution

  • In vivo imaging and analysis:

    • Reporter mice expressing fluorescent proteins under GH-responsive promoters

    • Intravital microscopy to observe real-time cellular responses to 20K hGH

    • PET ligands specific for GH receptors to assess receptor occupancy non-invasively

  • Delivery and formulation technologies:

    • Biodegradable microparticles for sustained release with optimized pharmacokinetics

    • Tissue-targeted nanoparticles to enhance delivery to specific organs

    • Novel excipients to improve stability and reduce immunogenicity

  • Clinical assessment tools:

    • Digital biomarkers and wearable sensors to continuously monitor physiological responses

    • Machine learning algorithms to identify patterns in complex datasets and predict individual responses

    • Point-of-care testing for 20K hGH levels and biomarkers of activity
      These technological advances would accelerate research progress by enabling more precise, comprehensive, and translational studies of 20K hGH biology, ultimately facilitating clinical development of this promising therapeutic candidate .

Product Science Overview

Structure and Characteristics

The 20kDa variant of human growth hormone is a single, non-glycosylated polypeptide chain consisting of 177 amino acids, with a molecular mass of approximately 20,322 Daltons . It is produced in Escherichia coli (E. coli) and purified using proprietary chromatographic techniques .

Genetic Background

Human growth hormone belongs to the somatotropin/prolactin family of hormones. The gene encoding this hormone is located on chromosome 17, along with four other related genes. These genes share a high degree of sequence identity and are thought to have evolved through gene duplications . The 20kDa variant is expressed in the pituitary gland but not in placental tissue, unlike some other members of this gene family .

Production and Purification

The recombinant 20kDa human growth hormone is produced in E. coli using a modified signal peptide from Staphylococcus aureus to facilitate secretion into the extracellular medium . This method ensures the production of an active form of the hormone, free from endotoxins . The hormone is then purified to a high degree of purity (>98%) using techniques such as size-exclusion chromatography (SEC-HPLC) and SDS-PAGE .

Stability and Storage

The lyophilized form of the 20kDa growth hormone is stable at room temperature for up to three weeks but should be stored desiccated below -18°C for long-term storage . Upon reconstitution and filter sterilization, it can be stored at 4°C for up to four weeks. For more diluted solutions, adding a carrier protein such as human serum albumin (HSA) or bovine serum albumin (BSA) is recommended to prevent freeze-thaw cycles .

Clinical Importance

Human growth hormone is essential for normal growth and development. Deficiencies in this hormone can lead to growth disorders and short stature. Recombinant human growth hormone, including the 20kDa variant, is used in the treatment of growth hormone deficiencies and other related conditions . The production of recombinant hGH in suitable hosts like E. coli provides a cost-effective and efficient method for obtaining this therapeutic protein .

Metabolic Effects

Studies have shown that the 20kDa variant of human growth hormone has significant metabolic effects in patients with growth hormone deficiency. It has been administered in various doses to evaluate its efficacy and safety . These studies help in understanding the therapeutic potential of this recombinant hormone in clinical settings.

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.