GH antibodies are produced when the immune system recognizes endogenous or exogenous GH as foreign. Key contexts include:
Therapeutic Neutralization: Antibodies generated during rhGH treatment for growth hormone deficiency (GHD) can block GH activity, reducing treatment efficacy .
Autoimmune Response: Autoantibodies targeting GH-producing pituitary cells are observed in idiopathic GHD and short stature syndromes .
Research Tools: Monoclonal antibodies (mAbs) against GH are used to study GH receptor (GHR) signaling and develop cancer therapies .
Incidence: 8.5% of rhGH-treated patients develop anti-GH antibodies, though modern synthetic GH formulations reduce this risk .
Impact on Growth: High antibody titers correlate with blunted growth responses, particularly in isolated GHD type 1A and cerebral midline defects .
Pituitary Tumor Diagnosis: Anti-GH antibodies aid in classifying GH-secreting adenomas and diagnosing acromegaly .
Novel anti-GH mAbs are being developed to inhibit GH signaling in cancers and endocrine disorders:
Mechanism: mAbs 1-46-3 and 1-8-2 block GH-dependent cell proliferation and migration in breast cancer models .
Half-Life Optimization: Humanized mAbs like Hu-13H02m incorporate Fc mutations to prolong serum stability .
Commercial anti-GH antibodies enable precise GH detection in research and clinical settings:
Assay Innovations: Polyethylene glycol (PEG)-based separation distinguishes free GH from antibody-bound GH in plasma, improving accuracy in antibody-positive patients .
Neutralization in Therapy: High-affinity antibodies (e.g., 1-46-3 with KD = 2.68 nM) risk overriding therapeutic benefits in non-oncological uses .
Autoantibody Detection: Indirect immunofluorescence assays identify GH-targeting antibodies in idiopathic short stature, predicting GHD development .
Research Gaps: Long-term effects of anti-GH antibodies in metabolic disorders remain underexplored .
Growth Hormone antibodies serve dual purposes in research and clinical contexts. In clinical settings, they represent immunoglobulins produced by patients receiving growth hormone therapy that potentially neutralize treatment effects. In research settings, they function as laboratory tools for GH detection and quantification.
Clinically, GH antibodies are monitored when treatment appears to be failing despite adequate dosing. The patient's immune system may produce antibodies against administered GH, neutralizing its therapeutic effects and resulting in treatment resistance .
In research applications, monoclonal or polyclonal antibodies against GH serve as reagents for detection and quantification of GH in various laboratory assays. These antibodies bind to GH with high specificity and can be used in techniques like Western blot, where they can detect GH at approximately 22 kDa in human tissue samples .
GH antibodies play several important roles in the management of growth hormone deficiency:
Treatment Monitoring: GH antibodies are primarily monitored to assess treatment efficacy in patients receiving growth hormone therapy . Their presence may explain treatment failure despite adequate dosing.
Diagnostic Indicators: The most common symptom of GH antibody development is persistent lack of growth despite adequate GH dosing . This clinical presentation triggers testing for GH antibodies to determine if immunological resistance is the cause.
Clinical Management Decision-making: Positive GH antibody test results (indicating antibody presence) inform healthcare providers about the need to reconsider treatment strategies, potentially adjusting dosage or exploring alternative approaches .
Complementary Biomarker Assessment: GH antibody testing is typically performed alongside insulin-like growth factor I (IGF-I) measurement, as low IGF-I levels despite GH treatment suggest treatment ineffectiveness, possibly due to neutralizing antibodies .
Laboratory research utilizes several types of GH antibodies with distinct characteristics and applications:
Monoclonal Antibodies: These highly specific antibodies derive from identical immune cells that are clones of a unique parent cell. For example, Mouse Anti-Human Growth Hormone Monoclonal Antibody (Clone #178902) is used for Western blot detection of human growth hormone . These antibodies target specific epitopes on the GH molecule with high specificity.
Neutralizing Antibodies: These antibodies bind to GH in a manner that blocks its biological activity. Research assays can measure this neutralization capacity, with the neutralization dose (ND50) typically between 2-8 μg/mL in the presence of 0.2 ng/mL recombinant human GH .
Detection Antibodies: Used primarily in analytical techniques such as Western blot, ELISA, or immunohistochemistry. These antibodies can identify specific GH bands at approximately 22 kDa in human pituitary gland tissue .
Researchers can employ several techniques for GH antibody detection in both research and clinical contexts:
Western Blot Analysis: This technique separates proteins by electrophoresis, transfers them to a membrane, and probes with anti-GH antibodies. The protocol typically employs 2 μg/mL of Mouse Anti-Human Growth Hormone Monoclonal Antibody followed by HRP-conjugated secondary antibody to detect GH at approximately 22 kDa .
Cell Proliferation Assays: GH stimulates proliferation in specific cell lines like Nb2-11 rat lymphoma cells. GH antibodies can neutralize this proliferation in a dose-dependent manner, providing a functional assay to measure antibody activity. The typical neutralization dose (ND50) ranges from 2-8 μg/mL in the presence of 0.2 ng/mL recombinant human GH .
Immunoassays for Patient Samples: Clinical settings utilize blood tests to detect antibodies developed against therapeutic GH. These typically employ enzyme-linked immunosorbent assays (ELISA) or radioimmunoassays (RIA) .
IGF-I Measurement: As a complementary approach, IGF-I levels can be measured to indirectly assess GH activity. Low IGF-I levels despite GH treatment may indicate the presence of neutralizing GH antibodies .
Optimal Western Blot protocols for GH detection require attention to several parameters:
For optimal results, researchers should optimize antibody concentration, incubation times, and washing steps for their specific experimental conditions. The protocol should be validated with appropriate positive and negative controls before experimental use .
The Nb2-11 rat lymphoma cell line stands as the primary model system for studying GH antibody neutralization effects:
Nb2-11 Rat Lymphoma Cells: This cell line proliferates in response to human GH in a dose-dependent manner, making it suitable for functional neutralization assays. Recombinant human GH stimulates proliferation, which can then be neutralized by increasing concentrations of anti-GH antibodies .
The experimental system demonstrates:
GH stimulates proliferation in a dose-dependent manner
This proliferation is neutralized by anti-GH antibodies in a concentration-dependent fashion
The neutralization dose at which 50% inhibition occurs (ND50) is typically 2-8 μg/mL of antibody in the presence of 0.2 ng/mL GH
This cellular system provides a functional readout of antibody neutralizing capacity beyond simple binding assays, offering insights into the biological significance of GH-antibody interactions.
GH antibodies can significantly affect growth hormone treatment efficacy through several mechanisms:
Neutralization of Therapeutic Effect: GH antibodies bind to and neutralize administered growth hormone, preventing it from exerting its intended therapeutic effects .
Treatment Failure Manifestation: The most common clinical manifestation is lack of growth despite adequate GH dosing. As stated in clinical resources, "The most common symptom of GH antibodies is a lack of growth even with adequate doses of GH" .
Resistance Development: Patients who develop GH antibodies may show progressive resistance to treatment, potentially requiring alternative therapeutic approaches or dosage adjustments .
Monitoring Requirements: The potential development of GH antibodies necessitates ongoing monitoring of treatment efficacy through growth measurements and complementary biomarkers like IGF-I levels .
It's noteworthy that "GH antibodies are becoming more rare thanks to the use of manmade (synthetic) growth hormone" . This indicates that modern recombinant human GH formulations have reduced—though not eliminated—the risk of antibody development.
Several factors contribute to GH antibody development in patients receiving growth hormone therapy:
Protein Structure Recognition: The immune system may recognize therapeutic GH as foreign, particularly if its structure differs from endogenous human GH in conformation or post-translational modifications.
Formulation Characteristics: The development of recombinant synthetic GH has reduced immunogenicity compared to earlier preparations. As clinical resources note, "GH antibodies are becoming more rare thanks to the use of a manmade (synthetic) growth hormone" .
Individual Immune Factors: Patient-specific factors including genetic predisposition to immune reactions, prior sensitization, or concurrent immune dysfunction may contribute to antibody development.
Distinguishing between neutralizing and non-neutralizing GH antibodies requires functional assays that assess biological impact, not merely binding:
Cell Proliferation Assays: The Nb2-11 rat lymphoma cell line proliferates in response to GH. Neutralizing antibodies inhibit this proliferation, while non-neutralizing antibodies will not, despite potentially binding to GH .
Neutralization Dose Determination: The ND50 (dose at which 50% neutralization occurs) can be calculated from dose-response curves. The typical ND50 is 2-8 μg/mL of antibody in the presence of 0.2 ng/mL GH .
Comparative Analysis: Researchers should implement both binding assays (like ELISA or Western blot) and functional assays to comprehensively characterize antibodies. While binding assays detect the presence of antibodies that interact with GH, they cannot distinguish neutralizing capacity.
In clinical settings, the distinction may be inferred from the correlation between antibody presence and treatment failure, though this represents an indirect assessment rather than a direct measurement of neutralizing activity .
Comprehensive analysis of GH receptor-antibody interactions requires multiple complementary approaches:
Binding Assays:
Surface Plasmon Resonance (SPR) to measure real-time binding kinetics
Competitive binding assays to determine if antibodies compete with GH receptors
ELISA-based methods to quantify binding interactions
Functional Assays:
Cell proliferation assays using Nb2-11 rat lymphoma cells or other GH-responsive cell lines
Signal transduction assays measuring JAK/STAT pathway activation
Reporter gene assays where GH receptor activation drives expression of measurable outputs
Structural Studies:
X-ray crystallography or cryo-electron microscopy to visualize complexes
Epitope mapping to identify specific GH regions recognized by antibodies
Mutagenesis studies to identify critical residues for antibody binding versus receptor binding
The combination of these approaches provides comprehensive understanding of how antibodies interact with GH and potentially interfere with receptor binding and signaling, which has implications for both research applications and clinical treatment resistance.
Epitope mapping provides critical insights into the specific regions of GH recognized by antibodies:
Peptide Scanning: This approach involves synthesizing overlapping peptides spanning the GH sequence (Phe27-Phe217 as identified for certain antibodies ) to identify regions recognized by antibodies.
Mutagenesis Approaches:
Alanine scanning mutagenesis: Systematically replacing amino acids in GH with alanine to identify critical binding residues
Creating chimeric proteins combining regions of GH with non-immunogenic scaffold proteins
Competition Assays: Using known GH receptor binding domains to compete with antibody binding helps determine if antibodies target receptor-binding regions of GH.
Structural Analysis:
X-ray crystallography of antibody-GH complexes
Hydrogen-deuterium exchange mass spectrometry to identify protected regions
Cross-linking coupled with mass spectrometry to identify proximity relationships
Understanding the specific epitopes recognized by GH antibodies can:
Determine if antibodies target regions critical for receptor binding (explaining neutralizing effects)
Guide engineering of GH variants with reduced immunogenicity
Enable development of better detection assays with enhanced specificity
Cross-reactivity evaluation requires systematic testing against structurally related hormones:
Cross-Reactivity ELISA Panels:
Coating plates with GH and related hormones (prolactin, placental lactogen)
Testing antibody binding across this panel of related proteins
Quantifying relative binding affinities
Competitive Binding Assays:
Using labeled GH and unlabeled related hormones as competitors
Measuring displacement of antibody binding to determine cross-reactivity
Calculating IC50 values for each potential cross-reactant
Western Blot Analysis:
Running purified samples of GH and related hormones
Probing with the GH antibody as described in research protocols
Assessing band detection patterns and intensities
Functional Neutralization Assays:
Testing if the GH antibody neutralizes biological activity of related hormones
Using cell proliferation assays with related hormones as stimulants
Comparing neutralization potencies (ND50 values)
These experimental approaches provide comprehensive characterization of antibody specificity, which is critical for both research applications and clinical diagnostic interpretation.
Multiple factors influence GH antibody detection assay performance:
Optimization of these factors is essential for developing robust and reliable GH antibody detection assays for both research and clinical applications .
Proper storage and handling are critical for maintaining GH antibody functionality:
Storage Temperature and Duration:
12 months from date of receipt, -20 to -70 °C as supplied
1 month, 2 to 8 °C under sterile conditions after reconstitution
6 months, -20 to -70 °C under sterile conditions after reconstitution
Freeze-Thaw Considerations:
These protocols indicate that:
Long-term storage requires freezer temperatures (-20 to -70 °C)
Working stocks can be maintained at refrigerator temperatures (2 to 8 °C) for up to a month
Sterile conditions are essential for reconstituted antibodies
Minimizing freeze-thaw cycles preserves antibody activity
Additional best practices include aliquoting stock solutions, maintaining proper documentation of lot numbers and expiration dates, and centrifuging antibody solutions before use to remove aggregates.
Comprehensive validation ensures reliable performance across experimental contexts:
Cross-Platform Validation:
Test antibodies in multiple applications (Western blot, ELISA, immunohistochemistry)
Positive and Negative Controls:
Use well-characterized positive controls (e.g., human pituitary gland tissue)
Include appropriate negative controls (tissues not expressing GH)
Consider knockdown or knockout systems as definitive negative controls
Sensitivity Assessment:
Determine limits of detection using serial dilutions
Compare sensitivity across different detection systems
Specificity Testing:
Test for cross-reactivity with related proteins
Confirm binding to the expected molecular weight target (22 kDa)
Lot-to-Lot Consistency:
Compare performance across different antibody lots
Maintain reference standards for long-term comparisons
Functional Validation:
Confirm biological relevance using functional assays like neutralization tests
Calculate and compare functional parameters (e.g., ND50 values of 2-8 μg/mL)
As noted in research protocols, "Optimal dilutions should be determined by each laboratory for each application," emphasizing the importance of system-specific optimization and validation .
Growth hormone (GH), also known as somatotropin, is a peptide hormone that stimulates growth, cell reproduction, and cell regeneration in humans and other animals. It is crucial for human development and is produced by the pituitary gland. The mouse anti-human growth hormone antibody is a monoclonal antibody developed in mice that specifically targets human growth hormone. This antibody is widely used in research and diagnostic applications.
Growth hormone is a 191-amino acid, single-chain polypeptide that is synthesized, stored, and secreted by somatotropic cells within the lateral wings of the anterior pituitary gland. GH plays a key role in growth, metabolism, and overall health. It stimulates the liver and other tissues to secrete insulin-like growth factor 1 (IGF-1), which in turn promotes the growth of bones and other tissues.
The secretion of growth hormone is regulated by the hypothalamus, which releases growth hormone-releasing hormone (GHRH) to stimulate GH release and somatostatin to inhibit it. Factors such as sleep, stress, exercise, and nutrition can influence GH levels. GH secretion follows a pulsatile pattern, with the highest levels occurring during sleep.
The mouse anti-human growth hormone antibody is a monoclonal antibody produced by immunizing mice with human growth hormone. This antibody is designed to specifically bind to human GH, allowing for its detection and quantification in various assays. Monoclonal antibodies are preferred in research due to their high specificity and consistency.
Mouse anti-human growth hormone antibodies are used in a variety of scientific applications, including:
These antibodies are essential tools in endocrinology research, particularly in studies related to growth disorders, pituitary function, and metabolic diseases.