GH Human, HEK

Growth Hormone Human Recombinant, HEK
Shipped with Ice Packs
In Stock

Description

Introduction to GH Human, HEK

GH Human, HEK refers to recombinant human growth hormone (GH) produced in HEK293 (Human Embryonic Kidney 293) cells. This system leverages the HEK293 cell line’s ability to express complex proteins with native post-translational modifications (PTMs), making it ideal for research and therapeutic applications. GH Human, HEK is a non-glycosylated protein with a molecular weight of 20–22 kDa, depending on the isoform .

Production in HEK293 Cells

HEK293 cells are widely used for recombinant protein production due to their human origin, which ensures compatibility with human therapeutic proteins. Key features include:

  • Transient Transfection: High-yield expression via mammalian expression vectors .

  • Stable Cell Lines: Engineered lines (e.g., HEK293hGHR) enable constitutive GH secretion studies .

  • Non-Glycosylated Output: HEK-produced GH lacks carbohydrate modifications, which is critical for certain research applications .

Table 1: GH Human, HEK Production Platforms

FeatureHEK293 SystemE. coli System Plant System
Post-Translational ModificationsNone (non-glycosylated)NonePlant-specific glycosylation
Molecular Weight (kDa)20–22 (isoform-dependent)20.3 (20kDa variant)22.9 (with 6-His-tag)
Purity>98% (SDS-PAGE, SEC-HPLC)>98%>97%
Biological ActivityED50 < 0.1 ng/mL (Nb2-11 cell assay) ED50 < 0.1 ng/mL ED50 < 0.1 ng/mL

Biological Activity and Signaling Pathways

GH Human, HEK exhibits high potency in cell-based assays, with EC50 values ≤0.5 ng/mL . Studies in HEK293-derived models reveal:

  • GHR Activation: Recombinant GH induces STAT5 phosphorylation and SOCS2 upregulation via growth hormone receptor (GHR) .

  • Cross-Talk with FGF21: GH and FGF21 interact in human growth plates, modulating insulin sensitivity and skeletal growth .

  • GPR101-Mediated Secretion: Constitutive activation of Gαs and Gq/11 pathways in HEK293 cells drives GH hypersecretion, mimicking pituitary tumor phenotypes .

Applications in Research

HEK293-produced GH is pivotal in studying:

  1. GH Resistance: Mechanisms underlying reduced GH signaling in metabolic disorders .

  2. Pituitary Pathophysiology: GHRH/Gα15 stable cell lines model GH secretion dynamics .

  3. Therapeutic Development: Functional assays validate recombinant GH efficacy .

Comparative Analysis with Other Systems

AspectHEK293E. coli Plant
Protein YieldModerate (mg/L scale)High (grams/L scale)Low (μg/g biomass)
Host OriginHuman (native PTMs)Bacterial (no PTMs)Plant (non-human PTMs)
Cost-EffectivenessHigh (complex media)LowModerate
Research RelevanceHigh (humanized models)Moderate (structural studies)Low

Stability and Handling

  • Lyophilization: Sterile-filtered powder stored at -18°C to avoid degradation .

  • Reconstitution: Avoid freeze-thaw cycles; use 0.1% HSA/BSA for long-term storage .

Research Findings and Implications

  • GPR101 Signaling: Constitutive GH secretion in HEK293 models highlights pathways linking GPR101 to gigantism .

  • FGF21/GH Interplay: HEK293hGHR cells demonstrate reduced insulin sensitivity when exposed to FGF21, suggesting therapeutic targets for metabolic diseases .

Product Specs

Introduction
Growth hormone (GH) is part of the somatotropin/prolactin hormone family, crucial for growth regulation. This gene, along with four related ones, resides on chromosome 17's growth hormone locus, arranged in the same transcriptional direction. This arrangement likely arose from gene duplications. These five genes share significant sequence similarity. Alternative splicing further diversifies them, creating additional isoforms for potential specialization. While expressed in the pituitary gland, this specific family member isn't found in placental tissue, unlike the other four genes in the growth hormone locus. Gene mutations or deletions result in growth hormone deficiency and restricted growth.
Description
Recombinant Human Growth Hormone, produced in HEK cells, is a non-glycosylated monomer with a molecular weight of 22kDa. It undergoes purification using proprietary chromatographic methods.
Physical Appearance
White, sterile-filtered powder, lyophilized (freeze-dried).
Formulation
The Growth Hormone was lyophilized from a 1.13mg/ml solution in 1xPBS.
Solubility
Reconstitute the lyophilized Growth Hormone in sterile 1xPBS containing 0.1% endotoxin-free recombinant HSA.
Stability
Lyophilized Growth Hormone remains stable at room temperature for 3 weeks but should be stored desiccated below -18°C. After reconstitution, store Growth Hormone at 4°C for 2-7 days. For long-term storage, keep it below -18°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 95% based on SDS-PAGE analysis.
Biological Activity
Activity is determined by the dose-dependent stimulation of rat lymphoma cell line Nb2-11 (prolactin indicator cell line) proliferation. The ED50 is 0.1ng/ml.
Synonyms
GH1, GH, GHN, GH-N, hGH-N,Pituitary growth hormone, Growth hormone 1, Somatotropin.
Source
HEK.

Q&A

What is the molecular mechanism of Growth Hormone receptor signaling in human cells?

Human Growth Hormone (GH) functions by binding to a dimer of its high-affinity receptor (GHR) on target cells, initiating a complex signaling cascade. Upon binding, phosphorylation of associated JAK2 tyrosine kinases occurs, along with phosphorylation of the receptor itself. This activation triggers multiple intracellular signaling pathways that culminate in GH's biological effects: changes in gene expression, enhanced proliferation, blocking of apoptosis, cellular differentiation, and metabolic activity regulation .

The biological consequences of GH signaling are directly proportional to the number and functional status of GHRs in target tissues. Individuals with low GHR levels or dysfunctional receptors demonstrate abnormalities including decreased bone mineral density and increased adiposity, while those with enhanced GH response may exhibit excessive growth and metabolic abnormalities associated with various pathologies .

Why are HEK293 cells preferred as a model system for investigating GH signaling?

HEK293 cells serve as a primary model system for GH signaling studies due to several advantageous characteristics:

  • Transfection efficiency: HEK293 cells demonstrate exceptional transfection capabilities, allowing for reliable expression of recombinant proteins and reporter constructs .

  • Signaling machinery: These cells possess the complete molecular machinery necessary for GH and GHR-mediated signaling cascades, including JAK-STAT pathways .

  • Experimental versatility: HEK293 cells are compatible with various molecular techniques including luciferase reporter assays and bioluminescent resonance energy transfer (BRET) analyses .

  • Physiological relevance: Despite being immortalized cells, HEK293 maintain many characteristics of human cells, providing greater translational value than non-human or non-mammalian systems .

  • Reproducibility: HEK293 cells yield consistent results across experiments, facilitating reliable data generation and interpretation .

How can researchers effectively utilize BRET analysis to study GH receptor-G protein coupling specificity?

Bioluminescent Resonance Energy Transfer (BRET) analysis offers a powerful approach for investigating specific receptor-G protein coupling in GH signaling pathways. The methodology employs energy transfer between a bioluminescent donor (Rluc) inserted within the Gα subunit sequence and an acceptor (GFP10) fused to the N-terminus of Gγ2 subunit .

To implement this technique:

  • Co-transfect HEK293 cells with:

    • GHR or related receptor constructs

    • Gα-RLuc (options include Gαi1, i2, i3, q, oA, oB)

    • Gγ2-GFP10

    • Untagged Gβ1

  • After transfection (typically 48 hours), measure BRET signals following agonist stimulation.

  • Calculate the ligand-promoted BRET change (ΔBRET) as the difference between agonist-stimulated and basal BRET values .

This technique has revealed critical insights about receptor-specific G protein coupling. For example, wild-type somatostatin receptor type 5 (SST5), which influences GH secretion, activates Gαi1–3 and GαoA/B, while a specific mutant (R240W SST5) fails to activate the GαoA splicing variant despite maintaining other G protein interactions .

What methodologies are optimal for studying miRNA regulation of GHR expression?

The investigation of miRNA-mediated regulation of GHR expression requires a multi-faceted experimental approach:

  • Bioinformatic prediction and validation:

    • Use algorithms to identify potential miRNA binding sites in the GHR 3′-UTR

    • Create 3′-UTR luciferase reporter constructs containing wild-type and mutated miRNA binding sites

  • Functional analysis in cell models:

    • Transfect HEK293 cells with the 3′-UTR luciferase reporter vector and miRNA binding site mutants

    • Co-transfect with miRNA mimics (50nM concentration using appropriate transfection reagents like Dharmafect-1)

    • Measure reporter activity 24-48 hours post-transfection

  • Evaluation of endogenous effects:

    • Transfect cells with miRNA mimics

    • Extract RNA after 24 hours for qRT-PCR analysis

    • Extract proteins after 48 hours for Western blot analysis

    • Quantify changes in GHR mRNA and protein levels

This approach has identified miR-129–5p, miR-142–3p, miR-202, and miR-16 as potent inhibitors of human GHR expression in both normal (HEK293) and cancer (MCF7 and LNCaP) cells, suggesting potential therapeutic applications in GH/GHR-related pathophysiologies .

How should researchers design experiments to investigate G protein specificity in GH-related signaling pathways?

To effectively investigate G protein specificity in GH-related signaling, researchers should implement a systematic experimental strategy:

  • Pertussis toxin (PTX) sensitivity studies:

    • Pretreat cells with PTX to inactivate Gαi/o proteins

    • Generate PTX-resistant G protein constructs by introducing specific mutations

    • Rescue experiments with PTX-resistant G proteins can identify specific G protein involvement

  • G protein silencing approaches:

    • Use siRNA to selectively silence specific G protein subunits

    • Evaluate effects on downstream signaling pathways

    • Confirm silencing efficiency via Western blot

  • Pathway-specific readouts:

    • cAMP measurements for Gαi-mediated inhibition of adenylyl cyclase

    • ERK1/2 phosphorylation assays for MAPK pathway activation

    • Hormone secretion assays (e.g., GH release from somatotrophs)

G ProteinPTX SensitivityKey Downstream PathwaysAppropriate Readouts
Gαi1-3SensitivecAMP inhibitioncAMP assays
GαoASensitiveERK1/2, hormone secretionphospho-ERK1/2 immunoblot, hormone ELISA
GαoBSensitiveVariable (cell-type dependent)Context-specific assays

Using this approach, researchers have identified critical roles for specific G proteins. For example, GαoA has been shown to mediate ERK1/2 inhibition and GH secretion inhibition in somatotrophs, while cAMP inhibition involves multiple G proteins .

What experimental design approaches can optimize recombinant human GH production in mammalian cells?

The Taguchi method provides a powerful experimental design approach for optimizing recombinant human GH (rhGH) production. This orthogonal array design allows researchers to investigate multiple parameters simultaneously while minimizing experimental runs and maintaining statistical power .

Implementation involves:

  • Parameter selection: Identify key culture components affecting protein production (e.g., DMSO, glycerol, ZnSO₄, sodium butyrate)

  • Orthogonal array design: Establish an M16 experimental matrix with defined parameter combinations

  • Production assessment: Evaluate rhGH levels using:

    • Dot blotting

    • Western blotting

    • ELISA assays

  • Data analysis: Determine optimal conditions through statistical analysis of production levels across conditions

The application of this method has identified optimal conditions for rhGH production in CHO cells: 1% DMSO, 1% glycerol, 25 μM ZnSO₄ and 0 mM sodium butyrate .

How can researchers effectively assess the biological activity of recombinant human GH?

Evaluating the biological activity of recombinant human GH requires functional assays that reflect physiological responses. The LHRE-TK-Luciferase reporter gene system in HEK-293 cells provides a sensitive and reproducible bioassay for this purpose .

The methodology involves:

  • Reporter system: Transfect HEK293 cells with the LHRE-TK-Luciferase construct, which contains Growth Hormone Response Elements linked to a luciferase reporter

  • Stimulation: Treat transfected cells with different concentrations of the purified rhGH

  • Activity measurement: Assess luciferase activity, which directly correlates with the biological activity of the GH sample

  • Reference comparison: Compare results to a standardized GH preparation (e.g., prokaryotic rhGH)

This approach allows for quantitative assessment of GH bioactivity and has demonstrated that mammalian cell-produced rhGH typically exhibits higher bioactivity than prokaryotic versions at equivalent concentrations, likely due to proper folding and post-translational modifications .

How do mutations in signaling proteins affect GH-mediated cellular responses?

Mutations in signaling proteins can significantly alter GH-mediated cellular responses through selective disruption of specific signaling pathways. A methodological approach to investigating such effects includes:

  • Identification of critical residues: Through structural analysis and sequence alignment, identify conserved regions likely important for protein-protein interactions

  • Mutant generation: Create expression constructs containing specific mutations (e.g., the R240W mutation in SST5)

  • Functional characterization:

    • Cell surface expression (using immunofluorescence or flow cytometry)

    • Ligand binding profiles

    • G-protein coupling (using BRET technology)

    • Downstream signaling effects (e.g., cAMP levels, ERK1/2 phosphorylation)

  • Pathway-specific effects: Determine which pathways remain intact and which are disrupted

Research using this approach has revealed that mutations can cause selective uncoupling from specific G proteins. For instance, the R240W mutation in SST5 disrupts coupling to GαoA while preserving coupling to other G proteins (Gαi1–3 and GαoB). This selective uncoupling prevents SST5-mediated inhibition of GH release and cell proliferation despite maintaining the ability to inhibit cAMP production .

What is the relationship between GHR expression levels and cancer development?

The relationship between GHR expression and cancer development represents an important area of research with therapeutic implications. Methodological approaches to investigating this relationship include:

  • Comparative expression analysis:

    • Quantify GHR mRNA and protein levels in cancer versus normal tissues

    • Use RT-qPCR for mRNA quantification

    • Use Western blotting or immunohistochemistry for protein quantification

  • Functional studies:

    • Manipulate GHR levels through overexpression or knockdown

    • Assess effects on proliferation, apoptosis, and invasion

    • Evaluate changes in oncogenic signaling pathways

  • miRNA regulation studies:

    • Investigate how miRNAs regulate GHR expression in normal versus cancer cells

    • Determine if cancer-specific alterations in miRNA expression contribute to GHR upregulation

Research has demonstrated that GHR mRNA and protein are increased 2- to 5-fold in various cancers compared to corresponding control tissues, including:

  • Breast carcinomas

  • Prostatic carcinomas

  • Colorectal adenomas and adenocarcinomas

  • Gastric adenocarcinomas

  • Hepatic carcinomas

These findings suggest that the GH/GHR axis plays a significant role in cancer progression and may represent a therapeutic target.

What bioinformatic approaches can help identify regulatory elements affecting GH signaling?

Bioinformatic approaches provide powerful tools for identifying regulatory elements in GH signaling pathways:

  • 3'-UTR analysis for miRNA binding sites:

    • Employ algorithms (e.g., TargetScan, miRanda, PicTar) to predict miRNA binding sites

    • Validate predictions through reporter assays and functional studies in HEK293 cells

  • Promoter analysis for transcription factor binding sites:

    • Use tools like JASPAR and TRANSFAC to identify potential regulatory elements

    • Validate through chromatin immunoprecipitation (ChIP) and reporter assays

  • Protein-protein interaction networks:

    • Analyze potential interactions between GH signaling proteins and other cellular factors

    • Employ BRET technology to validate predicted interactions experimentally

  • Structural modeling for mutational impact assessment:

    • Use computational modeling to predict how mutations might affect protein structure and function

    • Generate hypotheses for experimental validation

These approaches can reveal unexpected regulatory mechanisms, such as the identification of miR-129–5p, miR-142–3p, miR-202, and miR-16 as regulators of human GHR expression .

How can researchers effectively troubleshoot contradictory results in GH signaling experiments?

When facing contradictory results in GH signaling experiments, researchers should implement a systematic troubleshooting approach:

  • Cell-type considerations:

    • Recognize that signaling pathways may be cell-type specific

    • For example, while GαoA inhibits ERK1/2 in somatotrophs, it activates ERK1/2 in CHO cells via PKC-dependent mechanisms

  • G-protein coupling specificity:

    • Use BRET analysis to verify specific receptor-G protein coupling

    • Consider that mutations may cause selective G-protein uncoupling

  • Experimental design validation:

    • Verify experimental conditions using positive and negative controls

    • Consider transfection efficiency and protein expression levels

  • Signal pathway crosstalk:

    • Investigate potential crosstalk between signaling pathways

    • Use pathway-specific inhibitors to dissect complex signaling networks

  • Alternative splicing and isoform variation:

    • Check for expression of different protein isoforms (e.g., GαoA vs. GαoB)

    • Validate using isoform-specific tools (antibodies, primers, etc.)

By systematically addressing these factors, researchers can resolve seemingly contradictory results and gain deeper insights into the complexity of GH signaling mechanisms.

Product Science Overview

Introduction

Human Growth Hormone (hGH), also known as somatotropin, is a glycoprotein secreted by the pituitary gland. It plays a crucial role in human growth and development by stimulating the production of insulin-like growth factor I (IGF-I), which in turn promotes bone density, muscle mass, and overall growth .

Historical Context

The history of hGH dates back to the 1920s when scientists first identified its role in growth. Initially, hGH was extracted from the pituitary glands of cadavers, a process that continued for over two decades. However, this method posed significant risks, including contamination with Creutzfeldt-Jakob disease (CJD), a fatal brain disorder . In 1985, the U.S. Food and Drug Administration halted the distribution of cadaver-derived hGH due to these risks .

Recombinant DNA Technology

The advent of recombinant DNA technology revolutionized the production of hGH. Biotechnology companies like Genentech and Eli Lilly developed methods to produce hGH using genetically modified bacteria, eliminating the need for cadaver-derived hormone . This recombinant hGH is produced by inserting the human growth hormone gene into bacterial or mammalian cells, which then express the hormone.

Human Embryonic Kidney (HEK) Cells

Human recombinant growth hormone can also be produced using Human Embryonic Kidney (HEK) cells, specifically the HEK293 cell line. These cells were created by transforming human embryonic kidney cells with adenovirus type 5 DNA, which immortalized them . HEK293 cells are widely used in biotechnology for their ability to produce high yields of recombinant proteins, including hGH .

Applications and Benefits

Recombinant hGH has numerous clinical applications. It is used to treat growth hormone deficiencies in children and adults, Turner syndrome, chronic kidney disease, and muscle wasting diseases . Additionally, it has been used off-label for its anabolic effects, although this practice is controversial and regulated.

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.