GH1 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

Introduction

The GH1 Antibody, FITC conjugated (Catalog # NBP2-54361F) is a high-specificity monoclonal antibody developed for research applications targeting human Growth Hormone (GH). Conjugated with Fluorescein Isothiocyanate (FITC), this reagent enables fluorescence-based detection in immunohistochemistry (IHC), protein arrays, and related assays. Its utility lies in studying pituitary biology, tumor diagnostics, and endocrine disorders such as acromegaly .

Key Features

ParameterDetails
TypeMonoclonal Mouse IgG2b Kappa
ReactivityHuman
ImmunogenRecombinant fragment (aa58-187) of human GH
ConjugateFITC (Excitation: 495 nm, Emission: 519 nm)
ApplicationsIHC, IHC-Paraffin, Protein Array
PurificationProtein A/G affinity chromatography
Storage4°C in the dark (PBS, 0.05% sodium azide)

Applications

Immunohistochemistry (IHC)

  • Used to localize GH in pituitary tissues, including somatotropic cells and adenomas .

  • Requires antigen retrieval (e.g., TE buffer pH 9.0 or citrate buffer pH 6.0) .

Protein Array

  • Detects GH in multiplex assays for hormone profiling .

Research Relevance

  • Serves as a diagnostic marker for pituitary tumors and acromegaly .

  • Complements studies on GH signaling pathways (e.g., IGF-1 regulation) .

Research Insights

  • Pituitary Tumor Classification: The antibody aids in distinguishing GH-secreting adenomas from non-functional tumors .

  • Acromegaly Pathogenesis: Studies using this antibody have linked GH overexpression to increased cell proliferation and metabolic dysregulation .

  • Fluorophore Properties: FITC’s emission at 519 nm minimizes spectral overlap in multicolor assays .

Comparison with Other GH Antibodies

Catalog NumberHost/TypeConjugateApplications
NBP2-54361F Mouse/IgG2bFITCIHC, Protein Array
55243-1-AP Rabbit/IgGUnconjugatedWB, IHC, IF
RP1023

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receiving them. Delivery times may vary depending on your location and preferred shipping method. For specific delivery estimates, please consult your local distributor.
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
Growth hormone (GH) plays a pivotal role in regulating growth and development. Its primary function is to stimulate the liver and other tissues to release insulin-like growth factor 1 (IGF-1), which is crucial for promoting cell growth and proliferation. GH also directly stimulates the differentiation and proliferation of myoblasts, the precursor cells of muscle fibers. Additionally, GH enhances amino acid uptake and protein synthesis in muscle and other tissues.
Gene References Into Functions
  1. Studies have revealed significant correlations between GH concentration and impairments in various aspects of eating disorders, including drive for thinness, body dissatisfaction, interoceptive awareness, sense of ineffectiveness, interpersonal distrust, and maturity fear. This finding suggests a potential hormonal influence on specific aspects of eating disorders. PMID: 29179911
  2. A substantial proportion of patients with mild traumatic brain injury, even years after the event, exhibit isolated GH deficiency. PMID: 27878771
  3. Genetic variations within the GH1 gene have been associated with isolated growth hormone deficiency. PMID: 28910730
  4. In newborns, serum prolactin (PRL) and hGH levels demonstrate a positive correlation, potentially explained by shared regulatory mechanisms or a drift phenomenon. Furthermore, a higher gestational week is associated with a higher PRL/hGH ratio. PMID: 28700563
  5. A comprehensive review highlights the endocrine profile of centenarians, particularly focusing on the GH/IGF-I/insulin system and its role in modulating aging and longevity. PMID: 27932301
  6. Research suggests that patients with non-islet cell tumor hypoglycemia (NICTH) exhibit reduced serum growth hormone levels during hypoglycemic episodes. [Retrospective Study & REVIEW of Case Reports] PMID: 28529277
  7. The absence of both copies of the GH1 gene is linked to growth hormone deficiency. PMID: 28525353
  8. Human Growth Hormone has been shown to suppress CLAUDIN-1 expression by activating Signal Transducer and Activator of Transcription 3 (STAT3). PMID: 28617312
  9. Serum GH levels were found to be independent of type 2 diabetes, fasting blood glucose, or HbA1c levels. PMID: 27060213
  10. Genetic screening of GH1 and GHRHR genes in patients with isolated growth hormone deficiency revealed several variations, including four novel deleterious mutations, one novel non-pathogenic variant, and six previously reported changes. PMID: 27114065
  11. Evidence suggests that GH directly regulates energy metabolism in myocytes, with uncoupling protein 2 (UCP2) playing a role in the signaling pathway downstream of the GHR/JAK/STAT system. PMID: 27150070
  12. Research findings indicate that TIMP3, a tissue inhibitor of metalloproteinases, acts as a modulator of growth hormone receptor (GHR) abundance on the cell surface, influencing the ability of GH to stimulate cellular signaling. PMID: 27075707
  13. Patients with neurofibromatosis type 1 (NF1) who exhibit GH excess have been effectively treated with lanreotide. This study emphasizes that GH excess should be considered a relatively frequent endocrine manifestation in NF1 patients, comparable to central precocious puberty. PMID: 28631895
  14. Growth hormone-releasing hormone (GHRH) analogs from the Miami series effectively suppress tumor growth but demonstrate only weak endocrine GH inhibitory activity. This tumor growth suppression could be partially attributed to the downregulation of GHRH receptor levels. PMID: 28130121
  15. While both 22K-GH and 20K-GH variants activate signaling pathways, the levels and kinetics of phosphorylation mediated by key signaling proteins differ slightly. PMID: 28427901
  16. Androgens and their receptor (AR) negatively regulate locally produced GH in prostate cancer cells following treatment with AR agonists (R1881) and antagonists (enzalutamide, bicalutamide). PMID: 28444169
  17. The inherent amyloidogenicity of growth hormone, in the presence of contaminating prion protein (and potentially prolactin and amyloid-beta) in certain cadaver pituitaries, might contribute to the observed co-occurrence of Creutzfeldt-Jakob disease and Alzheimer's disease. PMID: 27214308
  18. GH potentially exerts a negative regulatory effect on the maturation and accumulation of lipids in adipocytes. PMID: 27802441
  19. The c.-223C>T homozygous point mutation in the GH1 promoter represents the first identified case of short stature due to idiopathic growth hormone deficiency. PMID: 27252485
  20. Research has demonstrated that the recombinant protein produced by a plasmid-free E. coli strain was purified and characterized as human growth hormone (hGH). PMID: 27542624
  21. Findings suggest that the 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 is associated with dynamic interactions between the circadian machinery and a specific component of the hGH1 locus chromosomal structure, which is essential for efficient gene expression. PMID: 27151213
  23. Genetic polymorphisms in leptin (rs7799039) and leptin receptor (rs1137101) have been linked to an increased susceptibility to growth hormone deficiency. PMID: 26915772
  24. Hybrid training on a cycle ergometer (CE) proved more effective in stimulating acute increases in GH, lactate, and IL-6 compared to CE at the same workload. PMID: 26522057
  25. The Growth Hormone 1 T1663A polymorphism was associated with a decreased risk of breast cancer. PMID: 26225688
  26. A family exhibiting short stature due to autosomal dominant growth hormone deficiency type II has been reported, highlighting a severe impact on intracellular GH folding, stability, and secretion. PMID: 26485222
  27. GH and IGF-1 suppression persists for up to 25 months during pasireotide LAR treatment for acromegaly. PMID: 25103549
  28. In women with normal somatotroph function, GH levels remain unchanged during the first trimester of pregnancy. PMID: 25179796
  29. A large study of pregnancy outcomes in women with hypopituitarism revealed no association between GH replacement therapy regimens and pregnancy outcomes. PMID: 26256649
  30. The influence of oral glucose administration on rebound growth hormone release in normal and obese women, considering the roles of adiposity, insulin sensitivity, and ghrelin, has been investigated. PMID: 25782001
  31. Research using MIP-FoxM1-hGH mice suggests that the phenotype observed is primarily due to hGH activity, with FoxM1 protein remaining largely inactive. PMID: 26202070
  32. A case report describes Klinefelter syndrome with short stature attributed to growth hormone deficiency. PMID: 25241616
  33. Findings suggest that growth hormone deficiency might be a prevalent feature among patients with vernal keratoconjunctivitis. PMID: 25079463
  34. Human Growth Hormone stimulates the microRNA 96-182-183 cluster, promoting epithelial-mesenchymal transition and invasion in breast cancer. PMID: 25873390
  35. Research demonstrates that activation of noncoding transcription represents an autonomous activity of the human growth hormone long-range enhancer, independent of interactions with linked gene promoters and occurring synchronously with the initiation of GH expression in the embryonic pituitary. PMID: 25662214
  36. An analysis of GH1 in a Brazilian patient cohort revealed that the autosomal recessive form of isolated growth hormone deficiency (IGHD) is more prevalent than the dominant form, with both found only in severe IGHD cases. PMID: 25116472
  37. Recipients of living donor liver transplantation (LDLT) experience rapid recovery of the GH/IGF1 hormonal axis and liver function, while donors demonstrate altered GH signaling and delayed regeneration in the early post-donation period. PMID: 24889799
  38. JAK2 is activated by growth hormone and other cytokines. (Review) PMID: 25656053
  39. Genotyping has proven valuable in diagnosing children with suspected growth hormone insensitivity and short stature. PMID: 25411237
  40. hGH production is highly sensitive to increased caloric intake. PMID: 25295535
  41. Following liver transplantation (LT), GH levels correlate with the extent of cytolysis, while IGF-1 serves as an indicator of liver synthetic function recovery. IGF-1 levels above 90 mug/L (day 15-30) are indicative of short-term survival. PMID: 24804205
  42. This review highlights the evidence for extrapituitary synthesis of GH in humans. [review] PMID: 24642386
  43. This review summarizes the impact of growth hormone on in utero and neonatal cellular and metabolic profiles related to bone and adipose tissue. PMID: 25015810
  44. The study presents experimental data on 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 might be relatively common. PMID: 23789946
  46. In one family, a novel splice site mutation in GH1 (c.172-1G>C, IVS2-1G>C) was identified. Two other families presented with a previously reported splice site mutation (c.291+1G>A, IVS3+1G>A). 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 might 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, 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 GH1 Antibody, FITC conjugated and what are its fundamental characteristics?

GH1 Antibody, FITC conjugated is an immunological reagent consisting of antibodies against human Growth Hormone 1 (GH1/Somatotropin) that have been labeled with Fluorescein Isothiocyanate (FITC), a fluorescent dye with excitation at 495 nm and emission at 519 nm . These antibodies are available in different formats including rabbit polyclonal and mouse monoclonal variants . They specifically target human Growth Hormone, which plays a crucial role in stimulating and controlling growth, metabolism, and differentiation of many mammalian cell types by modulating the synthesis of multiple mRNA species . The primary applications include immunofluorescence techniques, allowing researchers to visualize GH1 protein localization within cellular compartments without requiring secondary antibody detection steps.

The antibodies demonstrate cytoplasmic localization when binding to GH1, which is primarily synthesized by acidophilic or somatotropic cells of the anterior pituitary gland . The conjugation process typically follows established protocols involving crosslinking of the primary antibody with the FITC fluorophore . These antibodies serve as valuable markers in the classification of pituitary tumors and the study of pituitary diseases such as acromegaly .

What are the critical storage and handling recommendations for maintaining GH1 Antibody, FITC conjugated integrity?

Proper storage and handling of GH1 Antibody, FITC conjugated is essential for maintaining reagent performance and extending its useful lifespan. Based on manufacturer recommendations, researchers should adhere to the following protocols:

For lyophilized formulations:

  • Store at -20°C for up to one year from the date of receipt

  • After reconstitution, store at 4°C for one month

  • For longer storage after reconstitution, aliquot and store at -20°C for up to six months

For liquid formulations:

  • Store at 2-8°C in the dark, as FITC is light-sensitive

  • Some formulations may be stored at -20°C or -80°C depending on manufacturer specifications

Critical handling considerations include:

  • Avoid repeated freeze-thaw cycles, as they significantly decrease antibody activity and binding efficiency

  • Protect from prolonged exposure to light due to the photosensitivity of the FITC fluorophore

  • When working with antibodies containing sodium azide preservative (0.01-0.05%), be aware of its toxicity and potential chemical incompatibilities

Following these storage and handling protocols will minimize degradation of both the antibody and the fluorescent conjugate, ensuring consistent experimental results over time.

What are the validated applications for GH1 Antibody, FITC conjugated in research settings?

GH1 Antibody, FITC conjugated has been validated for multiple research applications across different experimental platforms. The validated applications include:

ApplicationValidation StatusTechnical Considerations
Immunofluorescence (IF)ValidatedDirect visualization of target localization
Immunocytochemistry (ICC)ValidatedCellular-level detection
Immunohistochemistry (IHC)ValidatedTissue section analysis
Immunohistochemistry-Paraffin (IHC-P)ValidatedFixed tissue section analysis
Flow Cytometry (FACS)ValidatedQuantitative cell population analysis
ELISAValidated for some productsQuantitative protein detection
Western Blot (WB)Validated for some productsProtein size verification
Protein ArrayValidated for some productsHigh-throughput screening

Researchers should note that optimal dilutions for each application should be experimentally determined, as the working concentration may vary depending on the specific experimental conditions and the manufacturer's formulation . When used for Western blot applications, the expected band size for GH1 is approximately 22-25 kD . For immunohistochemistry applications, human placenta tissue has been successfully used as a positive control .

The direct FITC conjugation eliminates the need for secondary antibody incubation steps, which can reduce background signal and simplify experimental workflows, particularly in multi-color immunofluorescence studies.

What are the key experimental design considerations when implementing GH1 Antibody, FITC conjugated in multi-parameter flow cytometry?

When designing multi-parameter flow cytometry experiments using GH1 Antibody, FITC conjugated, researchers should consider several critical factors to ensure reliable and interpretable results:

Spectral Overlap Management:
FITC has an emission spectrum (519 nm) that can overlap with other commonly used fluorophores like PE and GFP . Implementation of proper compensation controls is essential, which should include:

  • Single-color controls for each fluorophore used

  • Fluorescence minus one (FMO) controls to establish gating boundaries

  • Isotype controls conjugated with FITC to assess non-specific binding

Sample Preparation Optimization:

  • Fixation protocols must be optimized as overfixation can mask GH1 epitopes

  • Permeabilization is required for detecting intracellular GH1

  • Cell concentration should be maintained at 1 × 10^6 cells/ml for optimal antibody-to-cell ratio

Antibody Titration:
Researchers should perform titration experiments to determine the optimal antibody concentration that provides maximum specific signal with minimal background. Starting with the manufacturer's recommended concentration (typically 2 μg/mL) , serial dilutions should be tested to identify the optimal signal-to-noise ratio for each specific experimental system.

Data Analysis Considerations:

  • Implement hierarchical gating strategies beginning with forward/side scatter to identify viable cells

  • Use appropriate statistical methods for comparing GH1 expression levels between experimental groups

  • Consider using geometric mean fluorescence intensity (gMFI) rather than mean for more accurate quantification of fluorescence intensity

This systematic approach to experimental design will help minimize artifacts and ensure the generation of high-quality, reproducible data when using GH1 Antibody, FITC conjugated in flow cytometry applications.

What troubleshooting strategies should be employed when facing sensitivity or specificity issues with GH1 Antibody, FITC conjugated?

When researchers encounter challenges with GH1 Antibody, FITC conjugated experiments, a systematic troubleshooting approach is essential. The following methodological strategies address common issues:

For Weak or Absent Signal:

  • Antibody Concentration Adjustment: Increase the antibody concentration incrementally while monitoring background signal. The recommended starting concentration of 2 μg/mL may require optimization for specific applications .

  • Antigen Retrieval Enhancement: For fixed tissues, optimize antigen retrieval methods (heat-induced vs. enzymatic) to improve epitope accessibility.

  • Signal Amplification Systems: Consider implementing tyramide signal amplification (TSA) to enhance FITC signal while maintaining specificity.

  • Storage Condition Verification: Confirm that the antibody has been stored according to manufacturer recommendations to prevent degradation of either the antibody or FITC moiety .

For High Background or Non-specific Binding:

  • Blocking Protocol Optimization: Increase blocking agent concentration (BSA, serum, or commercial blocking solutions) and duration.

  • Dilution Buffer Modification: Add 0.1-0.3% Triton X-100 to reduce hydrophobic interactions while maintaining specific binding.

  • Autofluorescence Reduction: Implement specialized treatments such as Sudan Black B (0.1-0.3%) to quench tissue autofluorescence.

  • Washing Stringency Increase: Extend washing steps with PBS-T (0.05-0.1% Tween-20) to remove unbound antibody more effectively.

For Cross-Reactivity Issues:

  • Validation with Multiple Techniques: Compare results across different detection methods (e.g., IF vs. WB) to confirm specificity .

  • Antibody Pre-absorption: Perform pre-absorption tests with recombinant GH1 protein to confirm binding specificity.

  • Control Implementation: Include appropriate negative controls (non-GH1 expressing tissues) and positive controls (human placenta tissue has been validated) .

For Degraded Signal Over Time:

  • Photobleaching Prevention: Minimize exposure to light during all experimental steps and consider using anti-fade mounting media containing DABCO or NPG.

  • Aliquoting Strategy: Aliquot antibody solutions into single-use volumes to prevent protein degradation from repeated freeze-thaw cycles .

By systematically applying these troubleshooting approaches, researchers can resolve most technical issues encountered with GH1 Antibody, FITC conjugated experiments.

How can GH1 Antibody, FITC conjugated be effectively implemented in dual or multi-color immunofluorescence studies?

Implementing GH1 Antibody, FITC conjugated in dual or multi-color immunofluorescence studies requires careful methodological planning to achieve optimal signal separation and minimize cross-interference. The following protocol outlines a comprehensive approach:

Fluorophore Selection and Spectral Considerations:

  • When pairing with FITC (Ex: 495nm, Em: 519nm) , select secondary fluorophores with minimal spectral overlap, such as:

    • Cy3 (Ex: 550nm, Em: 570nm)

    • Alexa Fluor 647 (Ex: 650nm, Em: 668nm)

    • Texas Red (Ex: 596nm, Em: 615nm)

  • Avoid fluorophores with significant FITC spectral overlap such as GFP, BODIPY-FL, or Alexa Fluor 488.

Sequential Staining Protocol:

  • Primary Antibody Cocktail Preparation:

    • For same-species antibodies: Apply GH1 Antibody, FITC conjugated first, followed by complete washing steps, then apply other unconjugated primary antibodies.

    • For different-species antibodies: Co-incubation may be possible after careful validation.

  • Blocking Strategy:

    • Implement double blocking approach using both protein blocking (5% normal serum) and Fab fragment blocking when using multiple antibodies from the same species.

    • Consider using specialized multi-color blocking reagents containing both protein blockers and Fc receptor blockers.

  • Cross-Reactivity Elimination:
    When using multiple rabbit antibodies, employ the following sequence:

    • Apply first primary antibody (unconjugated)

    • Detect with anti-rabbit secondary antibody

    • Block with anti-rabbit IgG Fab fragments

    • Apply GH1 Antibody, FITC conjugated

Image Acquisition Optimization:

  • Capture single-channel controls first to establish proper exposure settings

  • Utilize sequential scanning rather than simultaneous acquisition to prevent bleed-through

  • Implement channel unmixing algorithms for fluorophores with partial spectral overlap

Validation Controls:

  • Single-stained controls for each antibody

  • Secondary-only controls to assess non-specific binding

  • Absorption controls with recombinant GH1 protein

Following this methodological framework will enable researchers to generate reliable multi-color immunofluorescence data with GH1 Antibody, FITC conjugated while minimizing artifacts and false co-localization signals.

What experimental validation approaches should be employed to confirm GH1 Antibody, FITC conjugated specificity in novel research models?

When implementing GH1 Antibody, FITC conjugated in novel research models, comprehensive validation is essential to ensure experimental reliability. The following systematic validation approach is recommended:

Multi-technique Concordance Testing:

  • Compare staining patterns across multiple detection methods:

    • Immunofluorescence (IF) for cellular localization

    • Western blot (WB) to confirm appropriate molecular weight (expected 22-25kD)

    • Flow cytometry to quantify population-specific expression

  • Verify consistent results across different antibody clones or lots, particularly comparing monoclonal and polyclonal antibodies when possible .

Genetic Validation Approaches:

  • Knockout/Knockdown Controls:

    • Test antibody in GH1-knockdown models (siRNA or shRNA)

    • If available, use CRISPR-Cas9 generated GH1-knockout cells as negative controls

    • Compare staining intensity between wild-type and genetically modified samples using quantitative image analysis

  • Overexpression Systems:

    • Test in recombinant GH1-overexpressing cells

    • Use dual-detection of epitope-tagged GH1 (e.g., with anti-His or anti-myc antibodies)

Physiological Validation:

  • Tissue-specific Expression:

    • Confirm strong positive staining in tissues known to express GH1 (anterior pituitary)

    • Verify minimal staining in tissues with low/no GH1 expression

    • Compare staining pattern with established reference data from human placenta tissue

  • Absorption/Competition Testing:

    • Pre-incubate antibody with purified recombinant GH1 protein

    • Compare staining between pre-absorbed and non-absorbed antibody

    • Gradual signal reduction with increasing concentrations of blocking peptide confirms specificity

Technical Validation:

  • Dilution Series:

    • Perform titration experiments (0.5-10 μg/mL) to identify optimal concentration

    • Verify signal reduction with decreased antibody concentration follows expected pattern

  • Isotype Controls:

    • Include appropriate FITC-conjugated isotype controls (IgG, IgG2b, etc.)

    • Match concentration exactly to experimental antibody

This comprehensive validation approach generates multiple independent lines of evidence for antibody specificity, establishing confidence in experimental results even in novel research models.

What quantitative analysis methodologies are most appropriate for GH1 expression studies using FITC-conjugated antibodies?

Quantitative analysis of GH1 expression using FITC-conjugated antibodies requires robust methodological approaches to ensure accurate, reproducible, and statistically valid results. The following analytical frameworks are recommended based on different experimental platforms:

Flow Cytometry-Based Quantification:

  • Population Analysis:

    • Report percentage of GH1-positive cells using consistently applied gating strategies

    • Utilize fluorescence minus one (FMO) controls to establish accurate positive/negative boundaries

    • Apply biexponential transformation for proper visualization of negative populations

  • Expression Level Quantification:

    • Use geometric mean fluorescence intensity (gMFI) rather than arithmetic mean

    • Calculate Staining Index: (MFI positive - MFI negative)/2 × SD of negative population

    • For absolute quantification, implement Quantum FITC beads to convert fluorescence to Molecules of Equivalent Soluble Fluorochrome (MESF)

Image-Based Quantification:

  • Intensity Measurement Protocols:

    • Capture images with identical acquisition parameters (exposure time, gain, offset)

    • Perform background subtraction using rolling ball algorithm

    • Measure integrated density (area × mean intensity) for whole cell or compartment-specific analysis

  • Colocalization Analysis:

    • Calculate Pearson's or Manders' coefficients when assessing GH1 colocalization with other markers

    • Implement automated threshold determination using Costes method

    • Report spatial statistics including nearest neighbor distances

Western Blot Quantification (for validation):

  • Normalize GH1 band intensity (22-25kD) to loading controls

  • Utilize housekeeping proteins appropriate for the experimental context

  • Generate standard curves with recombinant GH1 protein for absolute quantification

Statistical Considerations:

  • Technical Reproducibility:

    • Perform minimum of 3 independent experiments

    • Report both technical and biological variability

    • Utilize appropriate statistical tests based on data distribution (parametric vs. non-parametric)

  • Normalization Strategies:

    • For tissue samples, normalize to cell number or tissue area

    • For cell culture, normalize to total protein concentration

    • Consider cell cycle effects on GH1 expression when interpreting results

  • Reporting Standards:

    • Document all image processing steps

    • Report antibody concentration, lot number, and incubation conditions

    • Include representative images alongside quantitative data

This comprehensive quantitative framework ensures reliable measurement of GH1 expression patterns while minimizing technical artifacts and analytical biases.

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.