H6PD Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Overview of H6PD Antibody, Biotin Conjugated

H6PD Antibody, Biotin conjugated is a rabbit recombinant monoclonal antibody designed to target the bifunctional enzyme glucose-6-phosphate dehydrogenase (G6PD)/6-phosphogluconolactonase (6PGL), also referred to as hexose-6-phosphate dehydrogenase (H6PD). This antibody is conjugated with biotin, enabling its use in immunoassays such as sELISA (single-step enzyme-linked immunosorbent assay) for detecting H6PD in biological samples.

Key Features

ParameterSpecification
TargetH6PD/GDH (Hexose-6-phosphate dehydrogenase/6-phosphogluconolactonase)
HostRabbit
ClonalityMonoclonal
ConjugateBiotin
ReactivityMouse (tested); Other species not verified
ApplicationssELISA
ImmunogenRecombinant human H6PD/GDH protein
IsotypeIgG
Storage-20°C (long-term); 4°C (short-term)

Biological Role of H6PD

H6PD/GDH is a bifunctional enzyme localized in the lumen of the endoplasmic reticulum. It catalyzes the first two steps of the oxidative branch of the pentose phosphate pathway (PPP):

  1. Hexose-6-phosphate dehydrogenase activity: Converts glucose-6-phosphate to 6-phosphogluconolactone.

  2. 6-phosphogluconolactonase activity: Hydrolyzes 6-phosphogluconolactone to 6-phosphogluconate.

This enzyme generates NADPH, a critical reducing agent for reductive biosynthesis and maintaining redox balance in the ER. Unlike the cytoplasmic G6PD, H6PD has broader substrate specificity and operates in a distinct cellular compartment .

sELISA for Quantitative Detection

The antibody is validated for sELISA, a streamlined immunoassay that combines sample incubation and detection in a single step. This method is ideal for high-throughput screening of H6PD levels in mouse models or cell lysates. A representative standard curve from a corresponding ELISA kit (e.g., Mouse GDH ELISA Kit) demonstrates its sensitivity and specificity .

Comparative Analysis of H6PD vs. G6PD Antibodies

MarkerH6PD AntibodyG6PD Antibody (Common Counterpart)
LocalizationEndoplasmic reticulum lumenCytoplasm
Primary RolePPP oxidative phase, NADPH generationPPP oxidative phase, NADPH generation
SubstrateGlucose-6-phosphate, glucosamine-6-phosphateGlucose-6-phosphate
ApplicationssELISA, ER-specific pathway studiesELISA, Western blot, IHC (e.g., cancer, hemolytic anemia)

Challenges and Considerations

  1. Limited Species Reactivity: Currently validated for mouse samples only. Cross-reactivity with human or other species requires experimental confirmation.

  2. Conjugate Specificity: Biotin conjugation enables streptavidin-based detection but may introduce non-specific binding in complex samples.

  3. ER-Specificity: H6PD's localization necessitates optimized lysis protocols to preserve ER integrity during sample preparation.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your order. The delivery time may vary depending on the purchasing method or location. For specific delivery time, please contact your local distributors.
Synonyms
GDH/6PGL endoplasmic bifunctional protein [Includes: Glucose 1-dehydrogenase (EC 1.1.1.47) (Glucose-6-phosphate dehydrogenase) (EC 1.1.1.363), 6-phosphogluconolactonase (6PGL) (EC 3.1.1.31)], H6PD, GDH
Target Names
H6PD
Uniprot No.

Target Background

Function
Hexose-6-phosphate dehydrogenase (H6PD) is a bifunctional enzyme located within the lumen of the endoplasmic reticulum. It catalyzes the initial two steps of the oxidative branch of the pentose phosphate pathway (also known as the pentose phosphate shunt), an alternative pathway to glycolysis and a primary source of reducing power and metabolic intermediates required for biosynthetic processes. H6PD exhibits hexose-6-phosphate dehydrogenase activity with broad substrate specificity compared to glucose-6-phosphate 1-dehydrogenase (G6PD). It catalyzes the first step of the pentose phosphate pathway. Additionally, H6PD acts as a 6-phosphogluconolactonase, catalyzing the second step of the pentose phosphate pathway. It might also possess dehydrogenase activity for alternative substrates, including glucosamine 6-phosphate and glucose 6-sulfate. The primary role of this enzyme is to provide reducing equivalents, such as NADPH, to maintain adequate levels of reductive cofactors within the oxidizing environment of the endoplasmic reticulum. By producing NADPH, which is required by reductases in the lumen of the endoplasmic reticulum, like corticosteroid 11-beta-dehydrogenase isozyme 1 (HSD11B1), H6PD indirectly regulates their activity.
Gene References Into Functions
  1. Research suggests that the tetramer is the most active form of glucose-6-phosphate dehydrogenase (G6PDH). PMID: 28370139
  2. Cortisol reduces glucose-6-phosphate (G6P) flux through H6PDH by increasing luminal NADPH, thus allowing more G6P for hydrolysis via G6Pase. PMID: 26860459
  3. Inhibition of glucose-6-phosphate dehydrogenase sensitizes cisplatin-resistant cells to death. PMID: 26337086
  4. The G allele of rs6688832 in H6PD may potentially provide genetic protection against the development of Polycystic Ovary Syndrome (PCOS), particularly in overweight women. PCOS patients with the AG genotype of rs6688832 might be at risk for the hyperandrogenemia phenotype of PCOS. PMID: 26452272
  5. The study aimed to investigate the expression of HSD11B1, HSD11B2, H6PDH, and glucocorticoids receptor (GR) mRNA in subcutaneous adipose tissue (SAT) from obese women with or without Polycystic Ovary Syndrome. PMID: 23979790
  6. Cytoplasmic Mg2+ regulates glucose 6-phosphate utilization by reticular H6PD. PMID: 24631573
  7. GR and p300 are involved in the induction of H6PD by cortisol in human amnion fibroblasts. PMID: 23125313
  8. Polymorphisms in the H6PD gene may not be associated with type 2 diabetes and metabolic syndrome. PMID: 21869537
  9. Antioxidant supplementation was observed to increase G6PDH in the pentose phosphate cycle and 18S rRNA in the ribosome. There were no significant changes in the gene expression levels of beta-ACT. PMID: 22285204
  10. R453Q and D151A variants of the H6PD gene are associated with PCOS and obesity, respectively, and may contribute to the PCOS phenotype by influencing obesity, insulin resistance, and hyperandrogenism. PMID: 22306327
  11. Results suggest a novel role for the H6PD gene in atherosclerosis susceptibility. PMID: 21858044
  12. The H6PD gene exhibits significant polymorphism, and missense variants are prevalent. Coding variants (less than 1.5%) are rarely responsible for hyperandrogenemic polycystic ovary syndrome. PMID: 21050867
  13. Researchers identified a mitochondrial matrix-associated G6PDH and provided evidence that metabolic state/glucose availability modulates enzymatic sources of NADPH. PMID: 20228249
  14. This study corroborates the association of one locus determined by genome-wide association study and points to H6PD as a new candidate gene for multiple sclerosis. PMID: 19935835
  15. Findings indicate that the decrease in G6PDH activity that occurs after exposure of lung epithelial cells to chrysotile results from the carbonylation of G6PDH by TBARS. PMID: 20211231
  16. Mutations in the genes encoding 11beta-hydroxysteroid dehydrogenase type 1 and hexose-6-phosphate dehydrogenase interact to cause cortisone reductase deficiency. PMID: 12858176
  17. H6PDH directly determines the reaction direction of 11beta-Hydroxysteroid dehydrogenase1 in intact cells as an oxoreductase. PMID: 15280030
  18. A novel chemiluminescent method was applied to determine G6PDH in healthy and enzyme-deficient individuals. PMID: 15767211
  19. Two new missense mutations in the glucose-6-phosphate dehydrogenase gene were associated with chronic hemolytic anemia. PMID: 16079115
  20. Mutations were found in beta thalassemia patients experiencing complications related to liver disease. PMID: 16079116
  21. The observed constant expression of H6PD suggests that HSD11B1 acts as a reductase throughout the adipogenesis process in human ADMSCs and murine 3T3-L1 cells. PMID: 18586838
  22. Hepatic 11beta-HSD1 and H6PDH are closely interconnected. 11beta-HSD1 gene expression does not appear to be involved in the pathogenesis of fatty liver or nonalcoholic steatohepatitis. PMID: 18665910
  23. Data show that adipose tissue 11-beta-Hydroxysteroid Dehydrogenase Type 1 and Hexose-6-Phosphate Dehydrogenase gene expressions are elevated in patients with type 2 diabetes mellitus. PMID: 18963204
  24. The impact of H6PDH on the modulation of 11beta-HSD1-dependent interconversion of cortisone and cortisol by inhibitors and alternative substrates was investigated. PMID: 19010388
  25. Through co-immunoprecipitation experiments with purified H6PDH and 11beta-HSD1, and with cell lysates expressing H6PDH and 11beta-HSD1, researchers observed direct physical interaction between the two enzymes. PMID: 19121282

Show More

Hide All

Database Links

HGNC: 4795

OMIM: 138090

KEGG: hsa:9563

STRING: 9606.ENSP00000366620

UniGene: Hs.463511

Involvement In Disease
Cortisone reductase deficiency 1 (CORTRD1)
Protein Families
Glucose-6-phosphate dehydrogenase family; Glucosamine/galactosamine-6-phosphate isomerase family, 6-phosphogluconolactonase subfamily
Subcellular Location
Endoplasmic reticulum lumen.
Tissue Specificity
Present in most tissues examined, strongest in liver.

Q&A

What is H6PD and why is it significant in research studies?

H6PD (Hexose-6-Phosphate Dehydrogenase) is an endoplasmic reticulum enzyme also known as GDH/6PGL endoplasmic bifunctional protein. It plays a crucial role in the pentose phosphate pathway within the endoplasmic reticulum lumen, catalyzing the first two steps of this pathway. The enzyme converts glucose-6-phosphate to 6-phosphogluconate while generating NADPH, which is essential for various redox reactions within the cell. H6PD is particularly important in research due to its involvement in redox balance maintenance, cortisol metabolism, and its potential role in metabolic diseases.

The significance of H6PD in research extends to its potential implications in oxidative stress responses, steroid metabolism, and glucose homeostasis. Recent studies have shown that alterations in H6PD activity may contribute to various pathological conditions, making it an important target for immunodetection in both basic research and disease studies. H6PD is distinct from the cytosolic glucose-6-phosphate dehydrogenase (G6PD), though they catalyze similar reactions in different cellular compartments .

What are the key differences between biotin-conjugated and unconjugated H6PD antibodies?

Biotin-conjugated H6PD antibodies offer several distinct advantages over unconjugated versions in experimental applications. The primary difference lies in the detection system: biotin-conjugated antibodies have biotin molecules covalently attached to the antibody, allowing for signal amplification through the high-affinity biotin-streptavidin interaction. This enhances detection sensitivity, which is particularly valuable when working with low-abundance proteins like H6PD.

The biotin conjugation enables versatile detection options, as researchers can use various streptavidin-conjugated reporter molecules (HRP, fluorophores, gold particles) without needing multiple species-specific secondary antibodies. For example, in ELISA applications, biotin-conjugated H6PD antibodies can be detected using streptavidin-HRP, providing enhanced signal amplification compared to conventional detection methods .

What are the recommended protocols for H6PD Antibody, Biotin conjugated in ELISA applications?

ELISA applications using H6PD Antibody, Biotin conjugated require careful optimization to ensure specificity and sensitivity. Based on established protocols, the following methodological approach is recommended:

Sample Preparation:

  • Cell/tissue lysates should be prepared in compatible buffers (PBS-based with protease inhibitors)

  • Protein concentration should be determined and standardized (typically 1-10 μg/mL for coating)

Protocol Steps:

  • Coat ELISA plates with capture antibody or antigen depending on the assay format

  • Block with 3-5% BSA or 5% non-fat milk in PBS for 1-2 hours at room temperature

  • Add samples and standards in duplicate or triplicate

  • Use H6PD Antibody, Biotin conjugated at a dilution range of 1:1000 to 1:5000 (optimal dilution should be determined empirically)

  • Detect using streptavidin-HRP (typically at 1:2000 to 1:10000 dilution)

  • Develop with appropriate substrate (TMB recommended)

  • Stop reaction and read at 450nm with 570nm reference

Critical Parameters:

  • Temperature and timing consistency between wells is essential

  • Streptavidin detection systems require thorough washing to remove unbound biotin

  • Background control wells should be included to account for any endogenous biotin

  • Validation with positive and negative controls is necessary

What are the optimal storage conditions for H6PD Antibody, Biotin conjugated?

H6PD Antibody, Biotin conjugated requires specific storage conditions to maintain its functionality and stability. The optimal storage conditions include:

Temperature: Store at -20°C or -80°C for long-term storage. Avoid repeated freeze-thaw cycles which can compromise antibody activity and binding specificity. If frequent use is anticipated, prepare working aliquots to minimize freeze-thaw events .

Buffer Composition: The antibody is typically provided in a storage buffer containing 50% glycerol, 0.01M PBS at pH 7.4, with 0.03% Proclin 300 as a preservative. This formulation enhances stability during freeze-thaw cycles and prevents microbial contamination .

Working Solution Handling: Once thawed for use, keep on ice or at 4°C for short-term use (1-2 days). Return to -20°C promptly after use. For diluted working solutions, prepare fresh on the day of experiment as biotin conjugates may show reduced stability in diluted form.

Light Protection: Biotin-conjugated antibodies should be protected from direct light exposure to prevent potential photodegradation of the biotin moiety, which could reduce detection efficiency.

Shipping Considerations: If the antibody must be transported, use dry ice for shipping and monitor temperature to ensure it remains frozen throughout transit .

How can I optimize Western blotting protocols when using H6PD Antibody, Biotin conjugated?

Western blotting with H6PD Antibody, Biotin conjugated requires specific optimization strategies to achieve clear, specific detection. While the search results indicate that some H6PD antibodies are validated for Western blotting applications, the biotin-conjugated variant is primarily tested for ELISA . Nevertheless, the following protocol adaptations are recommended when using biotin-conjugated antibodies for Western blotting:

Sample Preparation:

  • Ensure complete denaturation of H6PD (89 kDa) by heating samples at 95°C for 5 minutes in reducing sample buffer

  • Load sufficient protein (30-50 μg total protein) as H6PD expression varies across tissues

Electrophoresis and Transfer:

  • Use 6-8% SDS-PAGE gels to properly resolve H6PD protein

  • Transfer at lower voltage (25V) for longer duration (overnight at 4°C) to ensure complete transfer of larger proteins

Blocking and Antibody Incubation:

  • Block with 5% BSA in TBST rather than milk (which contains endogenous biotin)

  • Dilute H6PD Antibody, Biotin conjugated at 1:500 to 1:2000 in blocking buffer

  • Extend primary antibody incubation to overnight at 4°C for improved sensitivity

  • Use streptavidin-HRP (1:2000 to 1:5000) as the detection reagent

Signal Development:

  • Employ enhanced chemiluminescence with extended exposure times (30-60 seconds)

  • Consider signal enhancers specifically designed for biotin-streptavidin systems

Controls and Validation:

  • Include mouse liver tissue lysate as a positive control, which shows high H6PD expression

  • Prepare a negative control by pre-adsorbing the antibody with immunizing peptide when available

Based on published validation data for similar antibodies, a well-optimized protocol should yield a distinct band at approximately 89 kDa, with potential secondary bands representing different isoforms or post-translational modifications .

What controls should be included when using H6PD Antibody, Biotin conjugated in experimental designs?

When designing experiments using H6PD Antibody, Biotin conjugated, incorporating appropriate controls is essential for result validation and troubleshooting. The following control strategy ensures experimental rigor:

Positive Controls:

  • Mouse liver tissue lysate shows reliable H6PD expression and can serve as a positive tissue control

  • Human prostate cancer tissue sections have been validated for H6PD immunodetection

  • Recombinant H6PD protein (specifically amino acids 422-712) can serve as a defined positive control

Negative Controls:

  • Omission of primary antibody while maintaining all other reagents detects non-specific binding of detection system

  • Non-expressing cell lines or tissues (must be empirically determined)

  • Blocking peptide competition assay using the immunizing peptide sequence to confirm specificity

Technical Controls:

  • Endogenous biotin blocking step using streptavidin/biotin blocking kits is essential in biotin-rich tissues (liver, kidney)

  • Secondary-only controls (streptavidin-reporter only) detect endogenous biotin

  • Loading controls such as β-actin antibodies confirm equal protein loading and transfer efficiency

Validation Controls:

  • Parallel analysis with alternative H6PD antibody clones (non-biotin conjugated)

  • siRNA or shRNA knockdown of H6PD in cell line models to demonstrate specificity

  • Gradient dilution series to establish the linear detection range

A robust control strategy should include at minimum a positive tissue control, a negative control (primary antibody omission), and appropriate technical controls for the specific application being used .

How does H6PD expression vary across different tissue types and what are the implications for antibody selection?

H6PD expression demonstrates significant tissue-specific patterns, which has important implications for antibody selection and experimental design. Understanding this variation is critical for proper control selection and interpretation of results:

Tissue Expression Profile:

  • Liver: High expression levels, particularly in endoplasmic reticulum-rich hepatocytes

  • Prostate: Moderate to high expression, with altered patterns in cancerous tissue

  • Adipose tissue: Moderate expression, with variations between white and brown adipose

  • Muscle: Low to moderate expression, primarily in sarcoplasmic reticulum

  • Brain: Variable expression, with region-specific patterns

  • Blood cells: Generally low expression levels

Cellular Localization:
H6PD is predominantly localized to the endoplasmic reticulum lumen, with expression patterns that follow the distribution of smooth ER in different cell types. This subcellular localization is important for selecting fixation and permeabilization protocols that preserve ER structure while allowing antibody access .

Implications for Antibody Selection:

  • For tissues with high expression (liver), lower antibody concentrations (1:400 to 1:1000) are typically sufficient

  • For tissues with low expression, higher antibody concentrations (1:20 to 1:100) and signal amplification systems may be necessary

  • When examining tissues with high endogenous biotin (liver, kidney), non-biotin detection systems or thorough biotin blocking steps are essential

  • For multiplex applications, careful selection of antibodies raised in different host species is required

  • When examining cancerous tissues, antibodies targeting preserved epitopes should be selected as expression patterns may be altered

The implications of this varied expression include the need for tissue-specific protocol optimization and careful selection of dilution factors based on the expected abundance in the target tissue.

How can H6PD Antibody, Biotin conjugated be used to investigate the role of this enzyme in oxidative stress responses?

H6PD Antibody, Biotin conjugated provides a valuable tool for investigating the complex relationship between H6PD activity and cellular oxidative stress responses. H6PD generates NADPH within the endoplasmic reticulum lumen, which serves as a critical electron donor for various redox reactions, particularly those involved in detoxification pathways and antioxidant defense.

Experimental Approaches:

  • Co-localization Studies:

    • Double immunofluorescence staining of H6PD with oxidative stress markers (8-OHdG, 4-HNE)

    • Counterstaining with ER markers (calnexin, PDI) to confirm localization

    • Protocol: Use H6PD Antibody, Biotin conjugated (1:100) with streptavidin-fluorophore and co-stain with antibodies against oxidative stress markers

  • Functional Analysis in Oxidative Stress Models:

    • Treatment of cells with H₂O₂ or other oxidative stress inducers

    • Analysis of H6PD expression and localization changes

    • Measurement of NADPH/NADP+ ratios in subcellular fractions

  • Knockdown/Inhibition Studies:

    • siRNA knockdown of H6PD followed by oxidative challenge

    • Measurement of ROS levels, lipid peroxidation, and cell viability

    • Similar approaches have been validated with G6PD inhibition, showing decreased tolerance to H₂O₂ in a concentration-dependent manner

Data from Related Studies:
Research on G6PD (related to H6PD) demonstrated that melanoma cells with genetically blocked G6PD showed significantly reduced expression and decreased tolerance to H₂O₂, indicating the importance of this pathway in oxidative stress protection. COLO679 melanoma cells with inhibited G6PD showed decreased tolerance to H₂O₂ in a concentration-dependent manner in XTT assay, suggesting a similar protective role may exist for H6PD .

Methodological Considerations:

  • When using biotin-conjugated antibodies in oxidative stress studies, ensure that oxidative conditions do not affect biotin-streptavidin interactions

  • Include appropriate controls for each oxidative treatment condition

  • Consider using complementary approaches (enzyme activity assays, gene expression analysis) alongside immunodetection

What are the technical challenges in detecting H6PD in different subcellular compartments and how can they be overcome?

Detecting H6PD in its native subcellular compartment (endoplasmic reticulum lumen) presents several technical challenges that require specific methodological approaches for accurate localization and quantification. The following challenges and solutions are particularly relevant when using H6PD Antibody, Biotin conjugated:

Challenge 1: Preserving ER Structure During Fixation

  • Solution: Use mild fixation with 2-4% paraformaldehyde for 10-15 minutes at room temperature rather than harsh fixatives

  • Evidence: Studies have shown that over-fixation can mask ER luminal epitopes

  • Implementation: For immunocytochemistry of cultured cells, 4% paraformaldehyde for 10 minutes preserves H6PD antigenicity while maintaining ER structure

Challenge 2: Achieving Adequate Permeabilization of ER Membrane

  • Solution: Sequential permeabilization with digitonin (plasma membrane) followed by saponin (ER membrane)

  • Protocol: Treat fixed cells with 25μg/ml digitonin for 5 minutes, then 0.1% saponin for 10 minutes

  • Validation: This approach has been successful for other ER luminal proteins

Challenge 3: Distinguishing H6PD from Cytosolic G6PD

  • Solution: Use subcellular fractionation to separate ER and cytosolic compartments before immunodetection

  • Protocol: Differential centrifugation followed by sucrose gradient separation to isolate pure ER fractions

  • Controls: Co-stain with established markers for ER (calnexin) and cytosol (GAPDH) to confirm fraction purity

Challenge 4: Signal Amplification for Low-Abundance Detection

  • Solution: Utilize the biotin-streptavidin system with tyramide signal amplification (TSA)

  • Implementation: Following primary antibody incubation and streptavidin-HRP binding, employ TSA to deposit additional biotin or fluorophore molecules at the binding site

  • Quantification: Use appropriate imaging software with background subtraction for accurate quantification

Challenge 5: Endogenous Biotin Interference

  • Solution: Pre-block endogenous biotin using commercially available biotin blocking kits

  • Protocol: Apply avidin solution (15 minutes), wash, then apply biotin solution (15 minutes) before antibody incubation

  • Validation: Include controls without biotin blocking to assess endogenous biotin levels

How does H6PD interact with 11β-HSD1 in regulating glucocorticoid metabolism and what detection methods are most appropriate?

H6PD plays a crucial role in glucocorticoid metabolism through its functional interaction with 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) in the endoplasmic reticulum lumen. This interaction represents an important metabolic regulatory mechanism with implications for various metabolic disorders.

Mechanism of Interaction:
H6PD generates NADPH within the ER lumen, which serves as the essential cofactor for 11β-HSD1's reductase activity. This activity converts inactive cortisone to active cortisol (in humans) or 11-dehydrocorticosterone to corticosterone (in rodents). This physical and functional coupling creates a "cortisol amplification" system within target tissues.

Optimal Detection Methods:

  • Co-immunoprecipitation (Co-IP):

    • Protocol: Use anti-H6PD antibody for immunoprecipitation followed by Western blotting for 11β-HSD1

    • Optimization: When using biotin-conjugated H6PD antibody, employ magnetic streptavidin beads for pull-down

    • Controls: Include reverse Co-IP (anti-11β-HSD1 for IP, anti-H6PD for detection)

  • Proximity Ligation Assay (PLA):

    • Methodology: Apply H6PD Antibody, Biotin conjugated alongside a non-biotin 11β-HSD1 antibody from a different host species

    • Detection: Use species-specific PLA probes and rolling circle amplification

    • Advantage: Provides visual confirmation of protein proximity (<40nm) in intact cells

  • FRET/BRET Analysis:

    • Approach: Express fluorescently-tagged H6PD and 11β-HSD1 in cell models

    • Validation: Use antibody detection to confirm proper localization

    • Application: Allows real-time monitoring of protein interactions

  • Functional Coupling Assays:

    • Method: Measure 11β-HSD1 activity (cortisone to cortisol conversion) in microsomes with or without H6PD inhibition

    • Detection: Use LC-MS/MS to quantify steroid metabolites

    • Controls: Include positive control with exogenous NADPH addition

Tissue-Specific Considerations:

Different tissues exhibit varying levels of H6PD and 11β-HSD1 expression, requiring adapted detection strategies:

Tissue TypeH6PD Expression11β-HSD1 ExpressionRecommended Detection Approach
LiverHighHighStandard Western blot (1:500 dilution)
AdiposeModerateHighIHC with signal amplification (1:100 dilution)
MuscleLowVariablePLA or FRET for co-localization
BrainRegion-dependentRegion-dependentIn situ hybridization with immunofluorescence

When using H6PD Antibody, Biotin conjugated in these contexts, the biotin-streptavidin interaction provides enhanced sensitivity for detecting the protein in tissues with lower expression levels, though careful blocking of endogenous biotin is essential .

What methodologies are most effective for validating H6PD Antibody, Biotin conjugated specificity in complex experimental systems?

Validating the specificity of H6PD Antibody, Biotin conjugated in complex experimental systems is crucial for generating reliable research data. The following comprehensive validation strategy incorporates multiple complementary approaches:

1. Genetic Validation Approaches:

  • siRNA/shRNA Knockdown:

    • Methodology: Transfect cells with H6PD-specific siRNA or shRNA and compare antibody signal in knockdown vs. control cells

    • Analysis: Quantify signal reduction corresponding to knockdown efficiency

    • Controls: Include non-targeting siRNA controls and validate knockdown by qRT-PCR

    • Expected Results: Proportional reduction in signal intensity correlating with mRNA reduction

  • CRISPR/Cas9 Knockout:

    • Approach: Generate H6PD knockout cell lines using CRISPR/Cas9 genome editing

    • Validation: Complete absence of antibody signal in knockout cells

    • Controls: Include wild-type and heterozygous cells as positive controls

    • Application: Provides definitive negative control for antibody specificity

2. Biochemical Validation:

  • Immunoprecipitation-Mass Spectrometry:

    • Methodology: Use H6PD Antibody, Biotin conjugated for immunoprecipitation followed by mass spectrometry

    • Analysis: Confirm that the predominant protein identified is H6PD

    • Criteria: >70% of peptides matching H6PD sequence indicates high specificity

  • Western Blot with Recombinant Proteins:

    • Approach: Test antibody against recombinant H6PD and related proteins (G6PD)

    • Analysis: Antibody should detect recombinant H6PD but not related proteins

    • Quantification: Calculate cross-reactivity percentage if any signal appears in related proteins

3. Immunological Validation:

  • Peptide Competition Assay:

    • Methodology: Pre-incubate antibody with immunizing peptide (human H6PD protein, amino acids 422-712) before application

    • Expected Result: Significant reduction or elimination of specific signal

    • Controls: Include non-specific peptide control that should not reduce signal

    • Quantification: >90% signal reduction indicates high specificity

  • Multiple Antibody Comparison:

    • Approach: Compare staining patterns using alternative H6PD antibodies targeting different epitopes

    • Analysis: Consistent localization patterns across antibodies supports specificity

    • Implementation: Use at least three independent antibodies recognizing different regions

4. Signal Validation in Complex Samples:

  • Tissue Panel Analysis:

    • Methodology: Apply antibody to tissues with known differential H6PD expression

    • Analysis: Signal intensity should correlate with known expression patterns

    • Controls: Include tissues with established high expression (liver) and low expression

  • Subcellular Fractionation:

    • Approach: Separate cellular compartments and confirm signal primarily in ER fractions

    • Validation: Co-localization with ER markers but not with markers of other organelles

    • Quantification: >80% of signal should localize to ER fraction for H6PD

These validation approaches provide a comprehensive assessment of antibody specificity and should be applied according to the complexity and requirements of the experimental system.

How can H6PD Antibody, Biotin conjugated be used in immunohistochemistry applications?

While the H6PD Antibody, Biotin conjugated (CSB-PA010111LD01HU) is primarily tested for ELISA applications according to the product information , related H6PD antibodies have been successfully employed in immunohistochemistry (IHC) . The following protocol provides a methodological framework for adapting biotin-conjugated H6PD antibodies for IHC applications:

Tissue Preparation and Fixation:

  • Fix tissues in 10% neutral buffered formalin for 24-48 hours

  • Process through graded alcohols and embed in paraffin

  • Section tissues at 4-5 μm thickness and mount on charged slides

  • For frozen sections, fix in cold acetone for 10 minutes

Antigen Retrieval Methods:

  • Heat-induced epitope retrieval in citrate buffer (pH 6.0) is recommended

  • Microwave for 15-20 minutes at 95°C or pressure cooker for 3-5 minutes

  • Allow slides to cool in buffer for 20 minutes before proceeding

Endogenous Biotin Blocking (Critical for Biotin-Conjugated Antibodies):

  • Apply avidin solution for 15 minutes

  • Rinse thoroughly

  • Apply biotin solution for 15 minutes

  • Rinse thoroughly

Antibody Application Protocol:

  • Block endogenous peroxidase with 3% H₂O₂ in methanol, 10 minutes

  • Block non-specific binding with 5% normal serum in PBS, 30 minutes

  • Apply H6PD Antibody, Biotin conjugated (recommended starting dilution 1:20 to 1:50)

  • Incubate overnight at 4°C in humidity chamber

  • Wash thoroughly with PBS-T (3 × 5 minutes)

  • Apply streptavidin-HRP (1:100 to 1:500 depending on system), 30 minutes

  • Wash thoroughly with PBS-T (3 × 5 minutes)

  • Develop with DAB chromogen, 5-10 minutes (monitor microscopically)

  • Counterstain with hematoxylin, dehydrate, clear, and mount

Controls and Validation:

  • Human prostate cancer tissue has been validated as a positive control for H6PD detection

  • Original magnification of 200× is recommended for initial evaluation

  • For frozen sections, a modified fixation protocol with acetone is recommended

Based on published data with related antibodies, successful H6PD immunohistochemistry should reveal cytoplasmic/reticular staining patterns consistent with ER localization. The biotin-conjugated format may offer enhanced sensitivity through signal amplification, particularly valuable in tissues with lower H6PD expression .

What are the considerations for using H6PD Antibody, Biotin conjugated in multi-color flow cytometry?

Using H6PD Antibody, Biotin conjugated in multi-color flow cytometry requires specific technical considerations to achieve successful detection while avoiding common pitfalls. While the specific H6PD antibody (CSB-PA010111LD01HU) is not explicitly validated for flow cytometry in the search results , related H6PD antibodies have been used for FACS applications . The following methodological approach outlines key considerations for adapting biotin-conjugated antibodies for flow cytometry:

Sample Preparation Considerations:

  • Cell fixation with 2% paraformaldehyde preserves H6PD antigenicity

  • Permeabilization is essential as H6PD is an intracellular (ER luminal) protein

  • Recommended permeabilization: 0.1% saponin or commercial permeabilization buffers designed for ER proteins

  • Maintain permeabilizer in all buffers throughout the procedure to prevent resealing of membranes

Staining Protocol Optimization:

  • Fix 1×10⁶ cells in 2% paraformaldehyde for 10 minutes at room temperature

  • Permeabilize with 0.1% saponin in PBS for 15 minutes

  • Block with 2% BSA, 5% normal serum in permeabilization buffer for 30 minutes

  • Apply H6PD Antibody, Biotin conjugated (1:100 to 1:500 dilution)

  • Incubate for 45-60 minutes at room temperature or overnight at 4°C

  • Wash 3× with permeabilization buffer

  • Apply streptavidin-fluorophore conjugate at manufacturer-recommended dilution

  • Incubate for 30 minutes at room temperature protected from light

  • Wash 3× with permeabilization buffer

  • Resuspend in appropriate buffer for flow cytometric analysis

Fluorophore Selection and Panel Design:

  • Streptavidin conjugates are available with various fluorophores (PE, APC, BV421, etc.)

  • Choose fluorophores based on cytometer configuration and other markers in panel

  • Avoid fluorophores with spectral overlap with FITC if measuring GFP or other green reporters

  • PE-streptavidin offers high signal-to-noise ratio for lower abundance proteins like H6PD

Critical Controls for Biotin-Conjugated Antibodies:

  • Unstained cells to establish autofluorescence baseline

  • Streptavidin-fluorophore only (no primary antibody) to detect endogenous biotin

  • Isotype control-biotin conjugate followed by same streptavidin-fluorophore

  • Blocking control: pre-incubation with unconjugated anti-H6PD before adding biotin-conjugated version

  • Biological negative control: cell type with minimal H6PD expression

  • Biological positive control: cells with confirmed high H6PD expression

Panel Design Considerations:

  • H6PD detection is compatible with common lineage markers when using appropriate compensation

  • When combining with other biotin-conjugated antibodies, sequential staining with intermediate blocking is required

  • For live/dead discrimination, use amine-reactive dyes before fixation and permeabilization

By following these methodological considerations, researchers can successfully incorporate H6PD Antibody, Biotin conjugated into multi-color flow cytometry panels for investigating H6PD expression in various cell populations .

How can H6PD Antibody, Biotin conjugated be used to investigate the relationship between H6PD and immunogenic cell death?

Recent research has highlighted important connections between metabolic enzymes and immunogenic cell death (ICD), a form of cell death that elicits an immune response. While the search results specifically discuss G6PD's role in ICD , the methodological approaches can be adapted to investigate potential roles of H6PD using biotin-conjugated antibodies.

Experimental Design Approaches:

  • H6PD Expression Analysis During ICD Induction:

    • Methodology: Treat cells with established ICD inducers (anthracyclines, oxaliplatin, radiation)

    • Detection: Use H6PD Antibody, Biotin conjugated (1:1000) with streptavidin-HRP for Western blot or streptavidin-fluorophore for microscopy

    • Analysis: Quantify changes in H6PD expression level and subcellular localization

    • Controls: Include non-ICD inducing cell death stimuli (staurosporine) for comparison

  • Calreticulin and H6PD Co-localization During ER Stress:

    • Rationale: Calreticulin exposure is a hallmark of ICD and originates from the ER

    • Protocol: Double immunofluorescence with anti-calreticulin (1:75, ab2907, Abcam) and H6PD Antibody, Biotin conjugated

    • Detection: Anti-rabbit secondary for calreticulin and streptavidin-fluorophore for H6PD

    • Analysis: Confocal microscopy to assess co-localization patterns and translocation during ICD

    • Control: Include β-actin staining as reference

  • HMGB1 Release Assay Following H6PD Modulation:

    • Background: HMGB1 release is another critical marker of ICD

    • Methodology: Modulate H6PD activity through inhibitors or siRNA knockdown

    • Assessment: Measure HMGB1 release using ELISA (HMGB1 ELISA Kit)

    • Controls: Similar to those used in G6PD studies, include control siRNA plasmid

    • Data Analysis: Quantify HMGB1 release relative to control conditions

  • H6PD Role in Oxidative Stress During ICD:

    • Approach: Compare H₂O₂ sensitivity in cells with normal versus reduced H6PD

    • Methodology: XTT viability assay following oxidative challenge

    • Protocol: Treat cells with H₂O₂ after H6PD knockdown or inhibition

    • Detection: Use H6PD Antibody, Biotin conjugated to confirm knockdown efficiency

    • Analysis: Generate dose-response curves similar to those established for G6PD

Experimental Data from Related Studies:

Studies with G6PD have shown that its inhibition decreased tolerance to oxidative damage in a concentration-dependent manner and influenced immunogenic cell death pathways. For example, COLO679 melanoma cells with inhibited G6PD showed significantly reduced tolerance to H₂O₂. Similarly, A375 melanoma cells genetically blocked for G6PD with shRNA showed significantly reduced G6PD expression and altered responses to oxidative stress .

These methodological approaches can be adapted to investigate whether H6PD plays parallel or distinct roles in ICD compared to its cytosolic counterpart G6PD, potentially revealing new therapeutic approaches for enhancing immunogenicity of cancer cell death.

What are common troubleshooting strategies for weak or non-specific signals when using H6PD Antibody, Biotin conjugated?

When working with H6PD Antibody, Biotin conjugated, researchers may encounter various signal issues that require systematic troubleshooting. The following methodological approach addresses common problems and their solutions:

Problem 1: Weak or Absent Signal

Potential CauseDiagnostic ApproachSolution Strategy
Insufficient antigenCompare with positive control tissueIncrease protein loading (Western) or use thicker sections (IHC)
Inadequate permeabilizationTest with known ER markersOptimize permeabilization with saponin or Triton X-100
Epitope masking by fixationTry different fixativesReduce fixation time or try alternative fixatives
Insufficient primary antibodyPerform dilution seriesIncrease concentration (start with 1:100 instead of 1:500)
Inefficient biotin-streptavidin detectionTest detection system with control biotin-antibodyUse fresher streptavidin reagent or try signal amplification systems
Antibody degradationTest new lot of antibodyAliquot antibody upon arrival and minimize freeze-thaw cycles

Problem 2: High Background or Non-specific Signal

Potential CauseDiagnostic ApproachSolution Strategy
Endogenous biotin interferenceInclude streptavidin-only controlImplement thorough avidin-biotin blocking steps
Insufficient blockingCompare different blocking agentsExtend blocking time (2 hours) or use different blocking agent (5% BSA)
Overly sensitive detectionPerform dilution series of detection reagentDilute streptavidin-conjugate further (1:1000 instead of 1:500)
Non-specific antibody bindingTest with isotype controlIncrease salt concentration in wash buffer (150mM to 300mM NaCl)
Cross-reactivity with related proteinsPeptide competition assayEnsure using proper negative controls and validation steps
Sample over-fixationCompare different fixation timesReduce fixation time or enhance antigen retrieval

Problem 3: Inconsistent Results Between Experiments

Potential CauseDiagnostic ApproachSolution Strategy
Variable sample preparationStandardize extraction protocolsEstablish consistent protocol for sample preparation
Antibody lot variationTest different antibody lots side-by-sideWhen possible, reserve single lot for complete study
Degradation of biotin conjugateTest storage conditionsStore in small aliquots, avoid repeated freeze-thaw cycles
Protocol inconsistenciesDocument detailed protocolsStandardize all steps with precise timing and temperature control
Variable expression levelsInclude internal reference standardsNormalize to housekeeping proteins or total protein stain

Implementation Strategy:
When troubleshooting, adopt a systematic approach by changing one variable at a time and maintaining detailed records of all protocol modifications. Include appropriate controls with each experiment, particularly when testing new conditions. For critical experiments, consider running optimization experiments with both positive and negative controls to establish ideal conditions before analyzing experimental samples .

How can the specificity and sensitivity of H6PD Antibody, Biotin conjugated be optimized for challenging samples?

Optimizing H6PD Antibody, Biotin conjugated for challenging samples requires specialized approaches to enhance both specificity and sensitivity. The following methodological strategies address common challenges encountered with difficult sample types:

For Samples with Low H6PD Expression:

  • Signal Amplification Methods:

    • Tyramide Signal Amplification (TSA): Apply streptavidin-HRP followed by biotin-tyramide substrate

    • Protocol: After primary antibody binding, use streptavidin-HRP (1:1000), then apply biotin-tyramide (1:50) for 10 minutes

    • Expected Outcome: 10-50 fold signal enhancement while maintaining specificity

    • Validation: Include non-amplified controls to assess background increase

  • Optimized Antibody Incubation:

    • Extended incubation time: 48-72 hours at 4°C with gentle agitation

    • Concentration optimization: Test 1:50 to 1:200 dilution range

    • Buffer enhancement: Add 0.1% Triton X-100 and 1% BSA to improve penetration

    • Temperature cycling: Alternate between 4°C (16 hours) and room temperature (2 hours)

For Samples with High Background:

  • Enhanced Blocking Strategies:

    • Sequential blocking: 10% normal serum (2 hours), followed by 1% BSA with 0.3M glycine

    • Commercial background reducers containing proprietary protein mixtures

    • For tissues with high endogenous biotin: Apply avidin (30 minutes), wash, then biotin (30 minutes) before antibody incubation

    • Implementation: Critical for highly metabolic tissues like liver and kidney

  • Alternative Detection Systems:

    • Two-step polymer detection systems with minimal cross-reactivity

    • Directly labeled primary antibodies to eliminate secondary detection steps

    • Biotin-free detection systems for tissues with high endogenous biotin

For Samples with Limited Quantities or Degraded Quality:

  • Sample Preparation Optimization:

    • Antigen retrieval optimization: Test multiple buffers (citrate pH 6, EDTA pH 8, Tris-EDTA pH 9)

    • Mild extraction buffers to preserve epitope integrity

    • Ultracentrifugation to concentrate proteins from dilute samples

    • Protocol: For FFPE tissue sections, extend antigen retrieval to 30 minutes

  • Microfluidic Immunoassay Systems:

    • Automated microfluidic platforms that require minimal sample volume

    • Continuous flow of reagents to enhance binding kinetics

    • Implementation: Especially valuable for needle biopsies or rare cell populations

For Samples with Complex Matrices or Contamination:

  • Pre-clearing Strategies:

    • Pre-adsorption of antibody with tissue powder from non-target species

    • Protein A/G pre-clearing step to remove interfering immunoglobulins

    • Implementation: Incubate diluted antibody with 5% non-target tissue homogenate for 2 hours before use

  • Sequential Extraction Methods:

    • Differential detergent fractionation to isolate ER-enriched fractions

    • Density gradient centrifugation to separate cellular compartments

    • Protocol: Extract first with digitonin (plasma membrane), then with stronger detergents

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.