GH1 Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Applications in Research

The biotin-conjugated GH1 antibody is optimized for high-sensitivity assays due to its strong binding affinity (~25 kDa target) . Key applications include:

  • ELISA: Quantitative detection of GH1 in serum or tissue lysates .

  • Western Blot (WB): Detection of GH1 in human placenta (1:500–1:2000 dilution) .

  • Immunohistochemistry (IHC): Staining of pituitary and adenoma tissues (1:50–1:500) .

Myogenic Differentiation

A 2015 study showed that a GHRP-6-biotin conjugate (not the antibody) stimulated myogenic differentiation by upregulating IGF-1 and collagen synthesis . While this compound differs from the antibody, its findings highlight biotin’s role in enhancing bioactivity.

Antibody Validation

Proteintech’s GH1 antibody (55243-1-AP) demonstrated specificity in WB and IHC, with no cross-reactivity to CSH1 . Its use in publications includes:

  • PMID: 31666193: Elongator pathway regulation in melanocortin signaling .

  • PMID: 31666193: Pregnancy-related growth hormone disruption .

Biotin’s Role

Biotin conjugation enhances antibody stability and assay sensitivity. For example, Assaypro’s biotin-conjugated GH1 antibody (11711-05121) is validated for EIA/RIA, with a purity of >90% affinity-purified IgG .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We are typically able to ship your orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please consult your local distributor for specific delivery timelines.
Synonyms
gH antibody; GH-N antibody; GH1 antibody; GHB5 antibody; GHN antibody; Growth hormone 1 antibody; Growth hormone antibody; Growth hormone B5 antibody; Growth hormone; normal antibody; Growth hormone; pituitary antibody; HG1 antibody; hGH-N antibody; IGHD1B antibody; Pituitary growth hormone antibody; RNGHGP antibody; SOMA_HUMAN antibody; Somatotropin antibody
Target Names
GH1
Uniprot No.

Target Background

Function
GH1 antibody plays a crucial role in regulating growth. Its primary function in stimulating body growth is to induce the liver and other tissues to secrete IGF-1. It also stimulates both the differentiation and proliferation of myoblasts, ultimately promoting amino acid uptake and protein synthesis in muscle and other tissues.
Gene References Into Functions
  1. Studies have found significant correlations between GH concentration and impairments in several EDI-2 subscales (drive for thinness, body dissatisfaction, interoceptive awareness, sense of ineffectiveness, interpersonal distrust, maturity fear) and SCL-90 subitems (depression, hostility, obsessivity compulsivity, anxiety). This suggests a potential hormonal modulatory effect on specific aspects of eating disorders. PMID: 29179911
  2. A high occurrence of isolated GH deficiency has been observed in patients examined one year or several years after experiencing a complicated mild traumatic brain injury. PMID: 27878771
  3. A single nucleotide variant in the GH1 gene has been associated with isolated growth hormone deficiency. PMID: 28910730
  4. In newborns, serum PRL and hGH levels exhibit a positive correlation, potentially explained by shared regulatory factors or a drift phenomenon. Higher gestational weeks are associated with a higher PRL/hGH ratio. PMID: 28700563
  5. This review examines the endocrine profile of centenarians concerning the GH/IGF-I/insulin system, highlighting the pathway's relevance in modulating aging and longevity. PMID: 27932301
  6. Research suggests that individuals with NICTH (non-islet cell tumor hypoglycemia) demonstrate low serum growth hormone levels during hypoglycemic episodes. This conclusion is based on a retrospective study and a review of case reports. PMID: 28529277
  7. Homozygous GH1 deletion has been linked to growth hormone deficiency. PMID: 28525353
  8. Human Growth Hormone inhibits CLAUDIN-1 expression by activating Signal Transducer and Activator of Transcription 3 (STAT3). PMID: 28617312
  9. Serum GH levels were not found to be related to type 2 diabetes, fasting blood glucose, or HbA1c levels. PMID: 27060213
  10. GH1 and GHRHR screening identified eleven variations in 24 (21%) patients with isolated growth hormone deficiency. Of these variations, four were novel deleterious, one was a novel non-pathogenic change, and six were previously reported changes. PMID: 27114065
  11. Research suggests that GH directly regulates energy metabolism in myocytes, and that UCP2 plays a role in the signal transduction pathway downstream of the GHR/JAK/STAT. PMID: 27150070
  12. Evidence indicates that TIMP3 acts as a modulator of cell surface GHR abundance and influences the ability of GH to promote cellular signaling. PMID: 27075707
  13. Children with GH excess have undergone medical treatment with lanreotide, with a minimum clinical/biochemical follow-up of 2 years. This study demonstrates that GH excess should be considered as a relatively frequent endocrine manifestation in NF1 patients, similar to central precocious puberty. PMID: 28631895
  14. GHRH analogs of the Miami series have been shown to effectively suppress tumor growth while exhibiting only weak endocrine GH inhibitory activity. The suppression of tumor growth may be partially attributed to the downregulation of GHRH receptor levels. PMID: 28130121
  15. Studies have found that the levels and kinetics of phosphorylation mediated by the main signaling proteins triggered by 22K-GH or 20K-GH are not identical. PMID: 28427901
  16. Research indicates a negative regulation of locally produced GH by androgens/AR in prostate cancer cells following treatment with AR agonists (R1881) and antagonists (enzalutamide, bicalutamide). PMID: 28444169
  17. The intrinsic amyloidogenicity of growth hormone, in the presence of contaminating prion protein (and perhaps prolactin as well) and amyloid-beta found in some cadavers' pituitaries, may have contributed to the observed co-occurrence of Creutzfeldt-Jakob disease and Alzheimer's disease. PMID: 27214308
  18. GH potentially negatively modulates the maturation and accumulation of lipid in adipocytes. PMID: 27802441
  19. To our knowledge, c.-223C>T is the first homozygous point mutation in the GH1 promoter identified to cause short stature due to idiopathic growth hormone deficiency. PMID: 27252485
  20. Data demonstrate that the recombinant protein produced by the plasmid-free E coli strain was purified and characterized as human growth hormone (hGH). PMID: 27542624
  21. Research suggests that the known protective effect of GH signaling deficiency on neoplastic tissue growth is at least partially mediated by regulating p53 expression. PMID: 27226307
  22. Evidence indicates that hGH synthesis follows a diurnal rhythm, and there are dynamic associations between the circadian machinery and a component of the chromosomal structure of the hGH1 locus that is essential for efficient expression. PMID: 27151213
  23. Gene polymorphism of leptin (loci rs7799039) and leptin receptor (loci rs1137101) have been correlated with susceptibility to Growth hormone deficiency. PMID: 26915772
  24. Results showed that a hybrid training system on a cycle ergometer (CE) was more efficient in stimulating acute increases in GH, lactate, and IL-6 compared to CE at the same workload. PMID: 26522057
  25. Individuals with Growth Hormone 1 T1663A Polymorphism demonstrated a reduced risk of breast cancer. PMID: 26225688
  26. This is the first reported case of a family affected by short stature caused by an autosomal dominant form of GH deficiency II, which significantly impacts intracellular GH folding and stability as well as secretion. PMID: 26485222
  27. GH and IGF-1 suppression has been maintained for up to 25 months during pasireotide LAR treatment of acromegaly. PMID: 25103549
  28. In women with normal somatotroph function, GH levels do not change in the first trimester of pregnancy. PMID: 25179796
  29. Data from a large group of women with hypopituitarism revealed no association between GH replacement therapy regimens and pregnancy outcomes. PMID: 26256649
  30. This study investigates the effect of oral glucose administration on rebound growth hormone release in normal and obese women, exploring the role of adiposity, insulin sensitivity, and ghrelin. PMID: 25782001
  31. Research suggests that the phenotype of MIP-FoxM1-hGH mice is primarily attributed to hGH activity, with the FoxM1 protein largely remaining inactive. PMID: 26202070
  32. Case Report: of Klinefelter syndrome with short stature due to growth hormone deficiency. PMID: 25241616
  33. Research suggests that growth hormone deficiency may be a common feature in vernal keratoconjuntivitis patients. PMID: 25079463
  34. Human Growth Hormone stimulates the microRNA 96-182-183 cluster, which promotes epithelial-mesenchymal transition and invasion in breast cancer. PMID: 25873390
  35. Results demonstrate that activation of noncoding transcription reflects an autonomous activity of the human growth hormone long-range enhancer, fully independent of interactions with linked gene promoters and occurring in spatial and temporal synchrony with the initiation of GH expression in the embryonic pituitary. PMID: 25662214
  36. Analysis of GH1 in a cohort of Brazilian patients revealed that the autosomal recessive form of isolated growth hormone deficiency (IGHD) was more prevalent than the dominant form. Both forms were found only in severe IGHD. PMID: 25116472
  37. Recipients showed a rapid recovery of the GH/IGF1 hormonal axis and liver function following LDLT, while donors exhibited altered GH signaling and delayed regeneration in the early days after living donation. PMID: 24889799
  38. JAK2 is activated by growth hormone and other cytokines. (Review) PMID: 25656053
  39. Genotyping has contributed to the diagnosis of children suspected of having growth hormone insensitivity and short stature. PMID: 25411237
  40. hGH production is highly sensitive to increased caloric intake. PMID: 25295535
  41. Following LT, GH levels correlate with the extent of cytolysis, while IGF-1 serves as an indicator of liver synthetic function recovery. IGF-1 levels exceeding 90 mug/L (day 15-30) appear to be a predictor of short-term survival. PMID: 24804205
  42. This review highlights the evidence of extrapituitary synthesis of GH in humans. [review] PMID: 24642386
  43. This review summarizes findings on growth hormone's influence on in utero and neonatal cellular and metabolic profiles related to bone and adipose tissue. PMID: 25015810
  44. The study presents experimental data for the mechanism of thiol-disulfide exchange in tryptic peptides derived from human growth hormone in aqueous solution. PMID: 24549831
  45. Screening for mutations in GH1 and GHRHR genes in a large cohort of Argentinian patients with IGHD suggests that the p.Arg183His mutation associated with the type II dominant form of IGHD may be relatively common. PMID: 23789946
  46. In the first family, a novel splice site mutation in GH1 was identified (c.172-1G>C, IVS2-1G>C). In two other families, a previously reported splice site mutation (c.291+1G>A, IVS3+1G>A) was found. PMID: 24280736
  47. Bilateral involvement of a pituitary adenoma and severely decreased immediate postoperative serum GH levels at 72 hours after transsphenoidal adenomectomy may be independent risk factors for accelerated GH deficiency in acromegalic patients. PMID: 24972779
  48. The activity of autocrine GH may differ from that of endocrine GH in prostate cancer cells. PMID: 23238889
  49. Robust GH-stimulated hepatic Igf1 gene transcription utilizes tissue-specific mechanisms of epigenetic regulation that are established independently of GH signaling. PMID: 24109593
  50. A meta-analysis indicates that the GH1 T1663A polymorphism may contribute to the risk of colorectal cancer, particularly among Asian populations. PMID: 24464925

Show More

Hide All

Database Links

HGNC: 4261

OMIM: 139250

KEGG: hsa:2688

STRING: 9606.ENSP00000312673

UniGene: Hs.655229

Involvement In Disease
Growth hormone deficiency, isolated, 1A (IGHD1A); Growth hormone deficiency, isolated, 1B (IGHD1B); Kowarski syndrome (KWKS); Growth hormone deficiency, isolated, 2 (IGHD2)
Protein Families
Somatotropin/prolactin family
Subcellular Location
Secreted.

Q&A

What is a GH1 antibody with biotin conjugation and what are its primary applications?

A GH1 antibody with biotin conjugation is an immunoglobulin that specifically recognizes human growth hormone 1 (GH1) and has been chemically linked to biotin molecules. The biotin conjugation provides a means for secondary detection through strong binding with streptavidin or avidin reagents, which are commonly labeled with enzymes, fluorophores, or other detection molecules. The primary applications of biotin-conjugated GH1 antibodies include enzyme immunoassays (EIA), radioimmunoassays (RIA), immunohistochemistry, and immunocytochemistry/immunofluorescence techniques .

Specifically, GH1 antibodies with biotin conjugation are valuable tools in experimental designs where signal amplification is needed, such as in detecting low abundance GH1 in biological samples. The biotin-streptavidin interaction is one of the strongest non-covalent biological interactions known, making it highly advantageous for detection purposes. Most commercially available biotin-conjugated GH1 antibodies have been validated for specificity and sensitivity, ensuring reliable detection of human growth hormone in research applications .

The choice between monoclonal and polyclonal biotin-conjugated GH1 antibodies depends on the specific research objectives. Monoclonal antibodies like GH-45 offer high specificity with an affinity constant of approximately 3.8 x 10^10 l/mol, without cross-reactivity to human prolactin or other pituitary hormones . Polyclonal antibodies may provide broader epitope recognition but should be carefully validated for cross-reactivity with other somatotropin/prolactin family members.

How should researchers store and handle biotin-conjugated GH1 antibodies to maintain optimal activity?

Proper storage and handling of biotin-conjugated GH1 antibodies are critical for maintaining their activity and specificity. Most biotin-conjugated GH1 antibodies should be stored at -20°C or below as indicated by manufacturers' guidelines . Some preparations contain glycerol (typically 50%) to prevent freeze-thaw damage, allowing for longer-term storage stability .

The storage buffer composition is specifically formulated to maintain antibody stability and functionality. For example, the Assaypro GH1 antibody biotin conjugate is provided in PBS pH 7.4 with 50% glycerol, 0.25% BSA, and 0.02% sodium azide . Similarly, Proteintech's product uses PBS with 0.02% sodium azide and 50% glycerol at pH 7.3 . These buffer components serve specific purposes: glycerol prevents freeze-thaw damage, BSA reduces non-specific binding, and sodium azide prevents microbial growth.

To minimize activity loss, researchers should aliquot the antibody upon receipt to avoid repeated freeze-thaw cycles. Each thaw cycle can potentially reduce antibody activity. For antibodies stored in small volumes (e.g., 20 μL), the presence of carrier proteins like BSA (0.1%) helps maintain stability . When working with the antibody, it should be kept on ice when in use and returned to -20°C storage promptly after use.

Prior to each experimental application, centrifuge the antibody vial briefly to collect the solution at the bottom of the tube. When diluting the antibody for experimental use, use appropriate buffers as recommended in the specific application protocols to ensure optimal performance in various assay formats .

What factors should be considered when selecting between polyclonal and monoclonal biotin-conjugated GH1 antibodies?

Monoclonal antibodies, like the mouse-derived GH-45 clone, recognize a single epitope with exceptional specificity and consistent lot-to-lot reproducibility . The GH-45 antibody has a documented affinity constant of 3.8 x 10^10 l/mol for human growth hormone and demonstrates no binding to human prolactin or other pituitary hormones, making it ideal for studies requiring absolute specificity .

The experimental application significantly influences this selection decision. For Western blotting and ELISA where high specificity is required to avoid cross-reactivity with related hormones, monoclonal antibodies often perform better. For immunohistochemistry applications, particularly in fixed tissues where some epitopes may be masked, polyclonal antibodies might provide superior detection by recognizing multiple sites .

Host species considerations are also important, especially when performing multi-color immunofluorescence studies or working with tissue samples containing endogenous immunoglobulins. For example, when studying mouse tissues, a rabbit-derived polyclonal antibody might be preferred to avoid detection of endogenous mouse antibodies .

Finally, researchers should evaluate whether isoform-specific detection is required. Some antibodies are specifically designed to discriminate between GH1 variants (such as the 22 kDa and 20 kDa isoforms), which may be critical for certain research applications .

How can biotin-conjugated GH1 antibodies be optimized for use in ELISA assays?

Optimizing biotin-conjugated GH1 antibodies for ELISA assays requires careful consideration of multiple parameters to achieve maximum sensitivity and specificity. The optimization process should begin with antibody titration experiments to determine the optimal concentration range. While manufacturers suggest starting dilutions (e.g., 1:500-1:1000 for ELISA applications), these should be experimentally verified for each specific assay setup and sample type .

The blocking buffer composition significantly impacts assay performance with biotin-conjugated antibodies. Due to the presence of the biotin moiety, blocking solutions containing milk proteins may be suboptimal as they contain endogenous biotin that could interfere with detection. Instead, researchers should consider BSA-based blockers (typically 1-3%) that have been confirmed to be biotin-free or specially formulated commercial blockers designed for biotin-based detection systems .

For detection systems, the choice between streptavidin or avidin conjugates affects assay sensitivity. Streptavidin typically provides lower background and higher signal-to-noise ratios compared to avidin, particularly in serum samples. The detection conjugate (e.g., streptavidin-HRP, streptavidin-AP) should be titrated independently of the primary antibody to optimize signal development without increasing background .

Sandwich ELISA configurations require careful selection of capture and detection antibodies to ensure they recognize distinct, non-overlapping epitopes on the GH1 molecule. When using a biotin-conjugated detection antibody, researchers must verify that the capture antibody's epitope does not overlap with the binding site of the detection antibody. In some cases, using antibodies raised in different host species (e.g., mouse monoclonal for capture and rabbit polyclonal for detection) may enhance assay performance .

Biotin interference is a critical consideration when analyzing clinical samples. High levels of endogenous biotin in patient samples (particularly those taking biotin supplements) can significantly affect assay results. Validation experiments show that biotin concentrations up to 1000 ng/mL do not affect some GH1 immunocapture assays , but this threshold should be verified for each specific assay configuration.

What methodologies ensure optimal performance of biotin-conjugated GH1 antibodies in immunohistochemistry?

Successful immunohistochemistry (IHC) with biotin-conjugated GH1 antibodies requires methodological optimization across several critical steps. Antigen retrieval methods significantly impact epitope accessibility and antibody binding efficiency. For formalin-fixed, paraffin-embedded tissues, heat-induced epitope retrieval using TE buffer at pH 9.0 is recommended for GH1 detection, although citrate buffer at pH 6.0 may serve as an alternative . The optimal retrieval conditions should be determined empirically for each tissue type and fixation protocol.

The working dilution range for biotin-conjugated GH1 antibodies in IHC applications varies considerably between products. While manufacturers recommend starting dilutions (e.g., 1:50-1:500 for Proteintech's antibody ), optimization through a dilution series is essential for each specific tissue type and detection system. Researchers should establish the minimum concentration that provides specific staining with minimal background.

When using biotin-conjugated primary antibodies in IHC, researchers must be aware of potential endogenous biotin interference, particularly in biotin-rich tissues such as liver, kidney, and some neuroendocrine tissues. Pre-blocking endogenous biotin activity using an avidin-biotin blocking kit before antibody incubation is strongly recommended to enhance signal specificity. This approach reduces false-positive staining that may result from detection reagents binding to endogenous biotin rather than the antibody-linked biotin .

The detection system used with biotin-conjugated antibodies requires careful consideration. Standard streptavidin-HRP systems work effectively, but for tissues with high background or weak GH1 expression, amplification systems such as tyramide signal amplification may provide enhanced sensitivity. When performing dual or multi-label immunofluorescence, spectral considerations are critical to avoid fluorophore crosstalk when biotin-streptavidin detection is combined with other detection methods .

Validation of staining specificity should include appropriate controls, including: (1) positive controls using human pituitary adenoma or placenta tissues that are known to express GH1 ; (2) negative controls by omitting the primary antibody; and (3) pre-absorption controls where available, using the immunizing peptide or recombinant GH1 protein to demonstrate staining specificity.

How can researchers perform effective immunocapture of GH1 using biotinylated antibodies?

Immunocapture of GH1 using biotinylated antibodies represents a powerful approach for isolating and quantifying human growth hormone from complex biological matrices. An effective immunocapture protocol for GH1 begins with optimizing the biotinylation procedure for the capture antibody. As demonstrated in published methodologies, the molar ratio of biotin-7-NHS to antibody significantly impacts capture efficiency. For example, a protocol using 3 mM biotin-7-NHS in DMSO with a 120-minute conjugation reaction at room temperature has been validated for GH1 capture antibodies .

After biotinylation, purification of the conjugated antibody is essential to remove unreacted biotin molecules. Sephadex G-25 gel filtration columns effectively separate the biotinylated antibody from free biotin, and spectrophotometric analysis at 280 nm can confirm the final concentration of the purified conjugate. For long-term stability, dividing the biotinylated antibody solution into small aliquots (e.g., 0.1 mL) in low-protein-binding tubes and storing at -80°C prevents repeated freeze-thaw cycles .

The immunocapture procedure typically employs streptavidin-coated magnetic beads or plates as the solid phase for antibody immobilization. For optimal capture efficiency from biological samples like serum or plasma, researchers should determine the minimum effective concentration of biotinylated antibody needed for maximum GH1 recovery. The binding of biotinylated antibodies to streptavidin-coated surfaces should be conducted under controlled conditions (e.g., 4°C overnight) to maintain antibody functionality .

Sample preparation prior to immunocapture significantly affects recovery rates. Dilution of samples in appropriate buffers (often PBS with detergents and carrier proteins) can reduce matrix effects. Optimization of incubation parameters (time, temperature, and agitation) is essential for maximizing analyte recovery while minimizing non-specific binding. For example, validation studies have demonstrated that optimized immunocapture methods can achieve recoveries between 94-102% across calibration ranges .

After immunocapture, researchers can directly analyze the captured GH1 or proceed with additional processing, such as tryptic digestion for LC-MS/MS analysis. These advanced workflows allow for distinction between GH1 isoforms through detection of signature peptides that are specific to particular variants, such as the 22 kDa isoform .

How can researchers distinguish between different GH1 isoforms using biotin-conjugated antibodies?

Distinguishing between different GH1 isoforms presents a significant challenge in growth hormone research, and biotin-conjugated antibodies can be valuable tools when employed within sophisticated analytical frameworks. The major isoforms of interest include the predominant 22 kDa form and the less abundant 20 kDa variant produced by alternative splicing. These isoforms differ structurally in that the 20 kDa variant lacks amino acids 32-46, which affects not only molecular weight but potentially biological activity and immunoreactivity .

Advanced immunoanalytical approaches combine biotin-conjugated antibodies with mass spectrometry to achieve isoform-specific detection. In this methodology, biotinylated anti-GH antibodies are used for initial immunocapture of all GH isoforms from biological samples. Following capture, enzymatic digestion (typically using trypsin) generates isoform-specific signature peptides. For instance, the amino acid sequence deleted in the 20 kDa isoform produces unique tryptic peptides in the 22 kDa variant that can serve as specific markers. LC-MS/MS analysis then quantifies these signature peptides, allowing researchers to distinguish and quantify specific isoforms .

The specificity of this approach depends on careful selection of signature peptides unique to each isoform. As shown in Figures 1 and 2 from the research literature, the sequence underlined in the 191-amino-acid sequence of GH1 is deleted in the 20 kDa isoform, creating distinct tryptic digestion patterns. Biotinylated antibodies that recognize common epitopes present in all isoforms ensure comprehensive capture, while the mass spectrometric detection provides the isoform specificity .

For researchers without access to LC-MS/MS capabilities, alternative approaches involve using pairs of biotin-conjugated antibodies with different epitope specificities. For example, an antibody targeting an epitope in the region absent in the 20 kDa isoform (amino acids 32-46) would selectively bind only the 22 kDa variant. When used in parallel with an antibody recognizing a common epitope, researchers can develop sandwich immunoassays with differential responsiveness to the isoforms .

Validation of isoform specificity should include analysis of samples containing known concentrations of recombinant 22 kDa and 20 kDa GH1 variants, as well as evaluation using the WHO International Standard for Somatropin. Correlation between nominal and measured concentrations of these standards confirms the analytical accuracy of the isoform-specific detection method .

What strategies help overcome biotin interference in GH1 detection assays?

Biotin interference represents a significant analytical challenge in immunoassays utilizing biotin-conjugated antibodies or biotin-streptavidin detection systems. This interference has gained particular attention in clinical diagnostics as patients taking biotin supplements (often at high doses for conditions like multiple sclerosis) can have serum biotin levels sufficient to disrupt assay performance. Several methodological strategies have been developed to mitigate this issue in GH1 detection assays.

Alternative detection chemistries that avoid the biotin-streptavidin interaction entirely represent another solution. While biotin-conjugated GH1 antibodies are widely available, many manufacturers also offer the same antibody clones with alternative conjugations such as HRP, fluorophores, or other haptens. For example, the same monoclonal and polyclonal antibodies against GH1 are often available with different conjugates, allowing researchers to select non-biotin detection systems when biotin interference is a concern .

For mass spectrometry-based GH1 quantification methods, the impact of biotin interference is primarily limited to the immunocapture step. By incorporating isotope-labeled internal standards early in the sample preparation workflow, researchers can correct for any variations in capture efficiency caused by biotin interference. The inclusion of stable-isotope-labeled forms of signature peptides as internal standards enables accurate quantification even if immunocapture efficiency is reduced by sample biotin .

Statistical correction approaches have also been developed for situations where biotin interference cannot be completely eliminated. By establishing a dose-response relationship between biotin concentration and assay bias, researchers can apply mathematical corrections to results from samples with known or measured biotin levels. This approach requires thorough validation across the relevant concentration ranges for both biotin and GH1.

How can researchers validate the specificity of biotin-conjugated GH1 antibodies in their experimental systems?

Validating the specificity of biotin-conjugated GH1 antibodies is crucial for ensuring experimental reliability and accurate interpretation of results. A comprehensive validation strategy employs multiple complementary approaches to confirm that the antibody specifically detects GH1 without cross-reactivity to related proteins or interference from the biotin conjugation.

Positive and negative control tissues or cell lines represent the first line of validation. Human pituitary adenoma and placenta tissues are excellent positive controls for GH1 expression as confirmed by IHC validation data . Negative controls should include tissues known not to express GH1 or samples from GH1 knockout models where available. When using cell lines, those with documented GH1 expression (e.g., GH-secreting pituitary adenoma cell lines) versus non-expressing lines provide important specificity controls.

Cross-reactivity testing with structurally related proteins is essential, particularly with other members of the somatotropin/prolactin family. The GH-45 monoclonal antibody, for example, has been specifically validated not to bind human prolactin or other pituitary hormones despite their structural similarities to GH1 . For polyclonal antibodies, comprehensive cross-reactivity testing should include prolactin, placental lactogen, and other growth hormone variants.

Molecular weight verification through Western blotting provides confirmation that the detected protein matches the expected size of GH1. The observed molecular weight for GH1 is approximately 22 kDa , and detection of a single band at this size supports antibody specificity. Multiple bands may indicate detection of different isoforms, proteolytic fragments, or non-specific binding.

Competition assays using the immunizing antigen offer perhaps the most definitive specificity validation. Pre-incubation of the biotin-conjugated antibody with excess recombinant GH1 or the specific immunizing peptide should abolish or significantly reduce signal in all detection formats. This approach directly demonstrates that the observed signal results from specific epitope recognition rather than non-specific binding .

For biotin-conjugated antibodies specifically, additional validation should address potential artifacts related to the biotin moiety. This includes testing with streptavidin-only controls (omitting the primary antibody) to check for direct binding to endogenous biotin, and comparison of staining patterns between biotin-conjugated and unconjugated versions of the same antibody clone where available .

What are common challenges when using biotin-conjugated GH1 antibodies and how can they be resolved?

Researchers working with biotin-conjugated GH1 antibodies frequently encounter several technical challenges that can compromise experimental outcomes. By understanding these common issues and implementing targeted solutions, researchers can significantly improve assay performance and data reliability.

High background signal is perhaps the most frequently reported problem with biotin-conjugated antibodies. This issue often stems from endogenous biotin in biological samples or from excessive biotin conjugation ratios on the antibody itself. To address this, researchers should implement avidin/streptavidin blocking steps before antibody incubation when working with biotin-rich tissues. Additionally, optimizing the antibody dilution is critical—starting with higher dilutions (e.g., 1:1000 for ELISA) and titrating to find the optimal signal-to-noise ratio can substantially reduce background .

Signal variability across experiments represents another significant challenge. This variability may result from antibody degradation during storage or inconsistent sample preparation. Researchers should aliquot antibodies upon receipt to minimize freeze-thaw cycles and strictly adhere to storage recommendations (-20°C or -80°C depending on the product) . For immunohistochemistry applications, standardizing fixation conditions, antigen retrieval protocols, and incubation times will improve reproducibility. When working with clinical samples, pre-analytical variables such as sample collection, processing time, and storage conditions should be carefully controlled .

Interference from binding proteins in biological samples can mask epitope recognition and reduce assay sensitivity. The presence of growth hormone binding protein (GHBP) in serum or plasma samples can particularly affect GH1 detection. Validation studies have shown that properly designed immunocapture methods remain functional even in the presence of a 100-fold molar excess of GHBP . For samples with potentially high GHBP levels, sample pre-treatment with acidification followed by neutralization can dissociate GH1-GHBP complexes and improve detection efficiency.

Cross-reactivity with other growth hormone isoforms may complicate interpretation when studying specific variants. This challenge is particularly relevant when distinguishing between the 22 kDa and 20 kDa GH1 isoforms. Researchers should carefully select antibodies with documented epitope specificity or combine antibody detection with mass spectrometry approaches that can distinguish isoform-specific peptides following enzymatic digestion .

For multicolor immunofluorescence applications, spectral overlap between fluorophores conjugated to streptavidin and other detection reagents can lead to false co-localization signals. This issue requires careful selection of fluorophore combinations with minimal spectral overlap and implementation of appropriate controls, including single-stained samples for compensation settings when using confocal or flow cytometry analysis methods .

How should researchers optimize sample preparation for maximum detection sensitivity of GH1?

Optimizing sample preparation is fundamental for achieving maximum detection sensitivity when working with biotin-conjugated GH1 antibodies. Different biological sample types require specific preparation strategies to preserve GH1 integrity while minimizing interfering substances.

Sample storage conditions significantly affect GH1 stability. Analytical validation has confirmed GH1 stability during standard sample storage conditions, including room temperature (for limited periods), refrigeration at 4°C, and long-term storage at -20°C or -80°C . For prolonged storage, samples should be kept at -80°C to minimize degradation. Repeated freeze-thaw cycles should be avoided, as they can reduce immunoreactivity; accordingly, samples should be aliquoted before freezing.

Pre-analytical sample treatment can enhance detection sensitivity for complex matrices. For samples containing potential interfering substances, dilution in assay buffer containing detergents (e.g., Tween-20) and carrier proteins (e.g., BSA) can reduce matrix effects and improve signal-to-noise ratios. When working with tissue samples for immunohistochemistry, optimization of fixation protocols is critical—overfixation can mask epitopes, while underfixation may compromise tissue morphology and antigen retention.

For mass spectrometry-based approaches following immunocapture with biotinylated antibodies, sample preparation includes critical enzymatic digestion steps. Optimized tryptic digestion conditions (enzyme-to-protein ratio, digestion time, temperature, and buffer composition) ensure complete and reproducible generation of signature peptides for GH1 quantification. The inclusion of stable-isotope-labeled internal standards early in the sample preparation workflow provides control for variations in recovery throughout the entire analytical process .

When analyzing samples with potentially high concentrations of interfering substances like biotin or growth hormone binding protein (GHBP), additional pretreatment steps may be necessary. For biotin interference, specialized scavenging reagents or solid-phase extraction methods can reduce biotin levels before immunoassay analysis. For GHBP interference, acidification techniques can dissociate GH-GHBP complexes, making epitopes more accessible for antibody binding .

What considerations are important when designing multiplex assays that include biotin-conjugated GH1 antibodies?

Designing multiplex assays that incorporate biotin-conjugated GH1 antibodies requires careful consideration of several key factors to ensure accurate and reliable detection of multiple analytes simultaneously. Multiplexing introduces additional complexity compared to single-analyte detection, particularly when biotin-streptavidin chemistry is involved in one or more detection channels.

The selection of complementary detection chemistries is paramount when including biotin-conjugated GH1 antibodies in multiplex panels. Since the biotin-streptavidin system occupies one detection channel, other analytes must utilize non-biotin detection methods to avoid cross-interference. For example, directly conjugated fluorophores, enzyme conjugates (HRP, AP), or alternative hapten systems (DNP, digoxigenin) can be employed for other targets. When working with fluorescence-based multiplex systems, careful selection of fluorophores with minimal spectral overlap is essential to avoid bleed-through between detection channels .

Antibody cross-reactivity testing becomes even more critical in multiplex settings. Beyond confirming that the GH1 antibody does not recognize non-target proteins, researchers must verify that all antibodies in the multiplex panel do not exhibit cross-reactivity with each other or with non-target analytes. This validation should include tests where each antibody is omitted in turn to confirm signal specificity in each detection channel. The GH-45 monoclonal antibody, for example, has been validated not to bind human prolactin or other pituitary hormones, making it suitable for inclusion in multiplex panels targeting multiple pituitary hormones .

Order of addition and incubation parameters require optimization when incorporating biotin-conjugated antibodies into multiplex workflows. If multiple detection steps are required, the biotin-streptavidin detection should typically be performed last to prevent blocking of epitopes needed for other detection systems. Incubation times may need adjustment compared to single-analyte assays, as binding kinetics can be affected by the presence of multiple antibodies competing for sample access .

For multiplex immunohistochemistry or immunofluorescence applications, the antigen retrieval method becomes a critical consideration. Different targets may require different optimal retrieval conditions (e.g., citrate buffer pH 6.0 versus TE buffer pH 9.0) . Researchers must identify a compromise retrieval condition that adequately exposes epitopes for all targets or implement sequential staining protocols with multiple retrieval steps.

Signal normalization and quality control are particularly important in multiplex assays. Including internal standards for each analyte helps control for variability in detection efficiency across the different channels. When designing calibration curves for quantitative multiplex assays, matrix-matched calibrators containing all analytes at varying concentrations should be used to account for potential interactions or competition effects .

What methods should researchers use to validate lot-to-lot consistency of biotin-conjugated GH1 antibodies?

Validating lot-to-lot consistency of biotin-conjugated GH1 antibodies is essential for ensuring reproducible experimental results over time, particularly for long-term studies or clinical applications. Implementing a comprehensive validation protocol helps researchers identify and address potential variability between antibody lots.

Spectrophotometric analysis provides the first level of quality assessment. Measuring protein concentration (A280) and the degree of biotinylation (typically using HABA assay or other biotin quantification methods) allows comparison of basic physicochemical properties between lots. The biotin-to-protein ratio is particularly important, as over-biotinylation can compromise antibody function while under-biotinylation may reduce detection sensitivity. Consistent biotinylation ratios between lots indicate reproducible conjugation processes .

Functional performance validation using standardized positive controls is the cornerstone of lot comparison. Researchers should maintain a reference sample set (e.g., recombinant GH1 standards at multiple concentrations, reference tissue sections, or cell lysates with known GH1 expression) that is tested with each new antibody lot. Quantitative assays like ELISA can generate dose-response curves for different lots, allowing comparison of parameters such as EC50 values, maximum signal intensity, and detection limits. Acceptance criteria should be established a priori, for example, requiring that new lots demonstrate EC50 values within ±20% of the reference lot .

Epitope specificity assessment confirms consistent target recognition between lots. Competition assays using the immunizing antigen or peptide should demonstrate comparable inhibition profiles across different antibody lots. For antibodies claiming isoform specificity, testing with recombinant standards of different GH variants (e.g., 22 kDa vs. 20 kDa) should show consistent selectivity patterns .

Cross-reactivity profiling against structurally related proteins (such as prolactin, placental lactogen, or growth hormone 2) should be performed for each lot to ensure maintenance of specificity. The GH-45 monoclonal antibody, for example, should consistently demonstrate its documented lack of binding to human prolactin or other pituitary hormones across different production lots .

Side-by-side testing in the specific application of interest provides the most relevant validation. For immunohistochemistry applications, this involves staining serial sections of reference tissues with different antibody lots and comparing staining intensity, pattern, and background. For immunocapture-MS applications, comparing recovery rates and precision metrics between lots using standardized samples provides direct functional comparisons .

How can researchers determine the optimal dilution and concentration of biotin-conjugated GH1 antibodies for different applications?

Determining the optimal dilution and concentration of biotin-conjugated GH1 antibodies is a critical step in assay development that significantly impacts sensitivity, specificity, and cost-effectiveness. A systematic optimization approach involves titration experiments designed specifically for each application platform.

For ELISA applications, researchers should perform checkerboard titrations where both the biotin-conjugated GH1 antibody and the detection reagent (typically streptavidin-HRP or streptavidin-AP) are varied across a range of concentrations. Starting with the manufacturer's recommended dilution range (e.g., 1:500-1:1000) , serial dilutions should be tested against a concentration gradient of GH1 standard. The optimal antibody dilution will provide the steepest dose-response curve with acceptable background at zero analyte concentration. Signal-to-noise ratios should be calculated for each condition, with ratios >10 typically indicating good assay performance. Additional considerations include the linear range of detection and the lower limit of quantification, which are particularly important for clinical applications.

For immunohistochemistry and immunofluorescence applications, the optimization strategy should include testing multiple antibody dilutions on positive control tissues with known GH1 expression patterns, such as human pituitary adenoma or placenta tissues . The manufacturer's recommended dilution range (e.g., 1:50-1:500 for IHC) provides a starting point , but the optimal dilution may vary based on tissue fixation methods, antigen retrieval protocols, and detection systems. The ideal dilution should produce strong specific staining with minimal background and should be determined for each tissue type and preparation method.

For immunocapture applications preceding mass spectrometry analysis, optimization involves balancing maximum analyte recovery with antibody consumption. Titration experiments should measure recovery rates of spiked GH1 standards across different antibody concentrations immobilized on capture surfaces (e.g., streptavidin-coated magnetic beads or plates). The optimal concentration is the lowest amount that achieves maximum or near-maximum recovery (e.g., >90%) of the target analyte. Published methods have demonstrated that properly optimized immunocapture protocols can achieve recoveries between 94% and 102% across calibration ranges .

Finally, validation across the intended sample types is essential, as matrix effects may necessitate further adjustment of antibody concentrations. Spike-recovery experiments using the optimized antibody dilution in different matrices (e.g., serum, plasma, tissue extracts) can identify potential interference that might require matrix-specific protocol modifications .

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 2025 TheBiotek. All Rights Reserved.