H6PD Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

H6PD Antibody Overview

H6PD is an ER-localized enzyme involved in generating NADPH for redox homeostasis and biosynthetic processes. The anti-H6PD antibody [EPR12338(B)] (ab170895) from Abcam is a recombinant monoclonal antibody validated for use in Western blot (WB), immunohistochemistry (IHC-P), and other applications . Key features include:

ParameterDetails
TargetH6PD/GDH (Hexose-6-Phosphate Dehydrogenase/Glucose 1-Dehydrogenase)
HostRabbit
ClonalityMonoclonal
ReactivityHuman, Mouse, Rat
ApplicationsWB, IHC-P
SupplierAbcam
FunctionSupports NADPH production in the ER, critical for steroid metabolism .

FITC Conjugation in Antibodies

FITC is a fluorescent dye used to tag antibodies for visualization in techniques like immunofluorescence (IF) and flow cytometry. While the provided sources lack direct data on FITC-conjugated H6PD antibodies, insights from G6PD FITC conjugates illustrate typical properties :

Example: FITC-Linked G6PD Antibody (LAA716Ra81)

ParameterDetails
TargetG6PD (Glucose-6-Phosphate Dehydrogenase)
HostGoat
ClonalityPolyclonal
ConjugateFITC
ReactivityRat
ApplicationsICC, IF, IHC, WB
Concentration500 µg/mL
SupplierCloud Clone

Research Findings on H6PD

  • Role in Cancer: H6PD sustains NADPH levels in the ER, influencing cancer cell survival under oxidative stress. Knockdown studies show disrupted redox balance and increased susceptibility to apoptosis .

  • Interaction with Steroid Metabolism: H6PD-derived NADPH supports corticosteroid 11-beta-dehydrogenase (HSD11B1), linking it to glucocorticoid activation .

Comparative Analysis: H6PD vs. G6PD Antibodies

FeatureH6PD AntibodyG6PD Antibody (FITC Conjugated)
LocalizationEndoplasmic reticulum Cytosol
FunctionER-specific NADPH production Cytosolic NADPH synthesis
FITC AvailabilityNot explicitly listed in sourcesAvailable (e.g., LAA716Ra81 , sc-373886 )

Key Considerations for H6PD Antibody Use

  • Specificity: Ensure validation via knockdown/knockout controls, as cross-reactivity with G6PD is possible due to structural similarities.

  • Applications: Optimize protocols for ER-targeted studies, such as IF using ER markers (e.g., Calnexin) .

Limitations and Gaps

No FITC-conjugated H6PD antibodies are explicitly detailed in the provided sources. Current data rely on unconjugated H6PD antibodies and FITC-linked G6PD analogs . For FITC-based H6PD detection, custom conjugation or alternative fluorescent tags (e.g., CoraLite® Plus 488 ) may be required.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders within 1-3 business days of receiving them. Delivery times may vary depending on the purchase method and location. Please consult your local distributor for specific delivery timeframes.
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 residing within the lumen of the endoplasmic reticulum. It catalyzes the initial two steps in the oxidative branch of the pentose phosphate pathway (also known as the shunt), an alternative to glycolysis and a critical source of reducing power and metabolic intermediates for biosynthesis. 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 functions as a 6-phosphogluconolactonase, catalyzing the second step of the pentose phosphate pathway. It may also possess dehydrogenase activity for alternative substrates such as glucosamine 6-phosphate and glucose 6-sulfate. The primary role of H6PD is to supply reducing equivalents, notably NADPH, to maintain adequate levels of reductive cofactors within the oxidizing environment of the endoplasmic reticulum. By producing NADPH, which is required by reductases within the endoplasmic reticulum lumen, like corticosteroid 11-beta-dehydrogenase isozyme 1 (HSD11B1), H6PD indirectly regulates their activity.
Gene References Into Functions
  1. Research indicates 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 elevating luminal NADPH, thereby 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 might exhibit a potential genetic protective effect against the development of polycystic ovary syndrome (PCOS), particularly in overweight women. PCOS patients with the AG genotype of rs6688832 might be associated with an increased risk of hyperandrogenemia associated with 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 the 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 is highly polymorphic, and missense variants are common. Coding variants are rare (<1.5%) and are responsible for hyperandrogenemic polycystic ovary syndrome. PMID: 21050867
  13. This research identified a mitochondrial matrix-associated G6PDH and also provides 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 thiobarbituric acid reactive substances (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 new chemiluminescent method was applied to the determination of 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 complicated by liver disease. PMID: 16079116
  21. The observed constant expression of H6PD suggests that HSD11B1 acts as a reductase throughout the adipogenesis process in human adipose-derived mesenchymal stem cells (ADMSCs) and murine 3T3-L1 cells. PMID: 18586838
  22. Hepatic 11beta-HSD1 and H6PDH are closely interlinked. 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 increased 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. Using co-immunoprecipitation experiments with purified H6PDH and 11beta-HSD1, and with cell lysates expressing H6PDH and 11beta-HSD1, we observe 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 how does it differ functionally from G6PD?

H6PD (Hexose-6-phosphate dehydrogenase) is an endoplasmic reticulum (ER) bifunctional enzyme that oxidizes glucose-6-phosphate and other hexose-6-phosphates. Unlike cytosolic G6PD (Glucose-6-phosphate dehydrogenase), H6PD is localized to the ER lumen. H6PD contains both glucose-6-phosphate dehydrogenase (EC 1.1.1.47) activity and 6-phosphogluconolactonase (6PGL) activity (EC 3.1.1.31), making it a bifunctional enzyme . G6PD is primarily involved in the pentose phosphate pathway and plays a crucial role in cellular defense against oxidative damage through NADPH production, while H6PD contributes to ER-specific glucose metabolism and influences cortisol metabolism through 11β-hydroxysteroid dehydrogenase type 1 regulation.

What are the optimal sample preparation methods for H6PD detection?

For optimal H6PD detection using FITC-conjugated antibodies:

  • Fixation: Use 4% paraformaldehyde for 15-20 minutes at room temperature for cells or tissue sections

  • Permeabilization: For intracellular targets like H6PD, use 0.1-0.5% Triton X-100 for 5-10 minutes

  • Blocking: Block with 5% normal serum (matching secondary antibody host) with 1% BSA in PBS for 30-60 minutes

  • Antibody dilution: Prepare antibody in blocking buffer at recommended concentrations (typically 5-20μg/mL for immunofluorescence applications)

  • Wash buffer optimization: PBS with 0.05-0.1% Tween-20 works well for most applications

  • Counterstaining: For nuclear visualization, DAPI (1μg/mL) is recommended with H6PD staining

What controls should be included when using H6PD Antibody, FITC conjugated?

A robust experimental design should include:

Control TypePurposeImplementation
Positive ControlVerify antibody functionalityInclude sample known to express H6PD (e.g., liver tissue)
Negative ControlAssess background/non-specific bindingOmit primary antibody while maintaining all other steps
Isotype ControlEvaluate non-specific bindingUse matched isotype IgG at same concentration as primary antibody
Blocking PeptideConfirm specificityPre-incubate antibody with immunogen peptide to block specific binding
Genetic ControlValidate specificityUse H6PD knockout/knockdown samples when available

What are the recommended working dilutions for H6PD Antibody, FITC conjugated across different applications?

Based on experimental validation with similar FITC-conjugated antibodies targeting metabolic enzymes, optimal working dilutions vary by application technique :

ApplicationRecommended DilutionConcentration RangeIncubation Conditions
Western Blotting1:250-25000.2-2μg/mL1-2 hours at RT or overnight at 4°C
Immunohistochemistry1:25-1005-20μg/mL1-2 hours at RT or overnight at 4°C
Immunocytochemistry1:25-1005-20μg/mL1-2 hours at RT or overnight at 4°C
Flow Cytometry1:50-2002.5-10μg/mL30-60 minutes at RT

Note: Optimal working dilutions must be determined experimentally by each researcher due to variations in sample types, preparation methods, and detection systems.

How can I minimize photobleaching when imaging FITC-conjugated H6PD antibodies?

FITC is susceptible to photobleaching. Implement these strategies to preserve signal integrity:

  • Anti-fade mounting media: Use specialized mounting media containing anti-fade reagents

  • Reduced exposure: Minimize exposure time and light intensity during imaging

  • Image acquisition strategy: Locate regions of interest using lower magnification before capturing high-resolution images

  • Sequential scanning: When using confocal microscopy, employ sequential scanning rather than simultaneous detection

  • Sample storage: Store slides at 4°C in the dark and image within 1-2 weeks for optimal results

  • Alternative considerations: For long-term experiments or repeated imaging, consider antibodies with more photostable fluorophores like Alexa Fluor dyes

What is the stability profile of H6PD Antibody, FITC conjugated and what storage conditions are optimal?

For maximum stability and performance:

  • Short-term storage (up to 1 month): 2-8°C in the dark

  • Long-term storage: -20°C to -80°C in small aliquots to avoid repeated freeze-thaw cycles

  • Buffer composition: FITC-conjugated antibodies typically perform best in PBS (pH 7.4) with 50% glycerol and 0.02-0.03% preservative

  • Stability indicators: Monitor for changes in solution color, precipitation, or decreased signal intensity

  • Aliquoting: Upon receipt, prepare small working aliquots to avoid repeated freeze-thaw cycles which significantly reduce antibody performance

  • Light protection: Always store and handle in amber tubes or wrapped in foil to protect the FITC fluorophore

How can H6PD Antibody, FITC conjugated be applied in cancer research studies?

H6PD antibodies can provide valuable insights in cancer research:

  • Metabolic reprogramming analysis: Study alterations in glucose metabolism within the ER of cancer cells

  • Correlation with treatment response: Assess relationship between H6PD expression and immunotherapy efficacy, similar to findings with G6PD

  • Tumor microenvironment characterization: Examine H6PD expression in stromal cells using multiplexed immunofluorescence

  • Patient stratification: Analyze H6PD expression patterns in tumor biopsies to identify potential biomarkers, as low G6PD expression has been associated with better immunotherapy outcomes in melanoma and lung cancer patients

  • Therapeutic target exploration: Investigate H6PD as a potential therapeutic target given the emerging role of metabolic enzymes in cancer progression

What methodological approaches can be used to study the relationship between H6PD and ER stress responses?

To investigate H6PD's role in ER stress:

  • Colocalization studies: Combine H6PD-FITC antibody with ER stress markers (e.g., BiP/GRP78, CHOP, ATF6) using different fluorophores

  • ER stress induction: Compare H6PD localization and expression before and after treatment with ER stress inducers (tunicamycin, thapsigargin, DTT)

  • Proximity labeling: Employ proximity ligation assays to detect interactions between H6PD and ER stress-related proteins

  • Functional assays: Correlate H6PD activity with ER redox status using redox-sensitive probes

  • Genetic manipulation: Analyze changes in ER stress responses following H6PD knockdown/overexpression

  • Ex vivo tissue analysis: Examine H6PD expression in tissue samples from conditions associated with ER stress (obesity, diabetes, neurodegenerative diseases)

How can researchers distinguish between H6PD and G6PD when studying glucose metabolism?

Differentiating these similar enzymes requires specific methodological approaches:

  • Subcellular fractionation: Separate ER (H6PD) from cytosolic (G6PD) fractions before western blotting

  • Confocal microscopy: Use H6PD antibody alongside ER markers (e.g., calnexin, KDEL) and G6PD antibody with cytosolic markers

  • Enzyme-specific inhibitors: Apply selective inhibitors for functional discrimination in activity assays

  • Genetic tools: Utilize siRNA knockdown of each enzyme separately to determine specific contributions

  • Mass spectrometry: Employ targeted proteomics to distinguish between the two proteins based on unique peptide signatures

  • Activity assays: Develop selective biochemical assays exploiting the bifunctional nature of H6PD versus G6PD

What are common causes of weak or no signal when using H6PD Antibody, FITC conjugated?

Common issues and solutions include:

ProblemPotential CausesSolutions
No signalInsufficient antibody concentrationIncrease antibody concentration or incubation time
Target protein denaturationOptimize fixation protocol; consider alternative fixatives
Photobleached FITCMinimize light exposure; use anti-fade mounting media
Incorrect filter setEnsure microscope has appropriate filter for FITC (Ex: 490nm, Em: 525nm)
Weak signalSuboptimal permeabilizationAdjust detergent concentration or permeabilization time
Low target protein expressionIncrease exposure time; consider signal amplification methods
Inefficient antibody penetrationExtend incubation time; optimize tissue section thickness
Buffer incompatibilityCheck pH and ionic strength of buffers; avoid sodium azide >0.1%
High backgroundInsufficient blockingIncrease blocking time or blocking agent concentration
Non-specific bindingAdd 0.1-0.3% Triton X-100 to antibody diluent; try different blocking sera
AutofluorescenceUse autofluorescence quenching reagents; include background subtraction in analysis

How should quantitative image analysis be performed for H6PD localization studies?

For rigorous quantification of H6PD localization and expression:

  • Image acquisition standardization:

    • Maintain consistent exposure settings across all samples

    • Capture multiple fields per sample (minimum 5-10)

    • Include z-stacks for 3D analysis when appropriate

  • Analysis workflow:

    • Use specialized software (ImageJ/FIJI, CellProfiler, Imaris)

    • Perform background subtraction using negative control samples

    • Apply appropriate thresholding to segment positive signals

    • Create regions of interest (ROIs) for subcellular compartments using ER markers

  • Quantification parameters:

    • Mean fluorescence intensity (MFI) within defined regions

    • Colocalization coefficients (Pearson's, Manders') for H6PD with ER markers

    • Percentage of cells showing specific staining patterns

    • Signal-to-noise ratio calculations

  • Statistical considerations:

    • Analyze at least 50-100 cells per condition

    • Apply appropriate statistical tests based on data distribution

    • Present data with proper visualization (box plots, violin plots)

What approaches can address data inconsistencies when examining H6PD in different experimental conditions?

To resolve experimental inconsistencies:

  • Standardization protocols:

    • Implement strict sampling time controls (H6PD might follow circadian regulation)

    • Standardize cell culture conditions (passage number, confluence, serum batch)

    • Use identical lot numbers of antibodies when possible

  • Validation strategies:

    • Confirm findings with alternative detection methods (western blot, qPCR)

    • Employ multiple antibody clones targeting different epitopes

    • Include both positive and negative genetic controls

  • Normalization techniques:

    • Utilize multiple housekeeping proteins/genes for normalization

    • Apply global normalization methods for high-throughput analyses

    • Consider cell type-specific reference markers

  • Advanced troubleshooting:

    • Perform spike-in controls with recombinant proteins

    • Evaluate post-translational modifications affecting epitope recognition

    • Consider sex differences in H6PD expression and regulation when using animal or human samples

How can H6PD Antibody be integrated into single-cell analysis techniques?

Emerging strategies for single-cell H6PD analysis include:

  • Single-cell imaging:

    • Combine H6PD-FITC antibody with additional markers for multiplexed imaging

    • Apply imaging mass cytometry for highly multiplexed single-cell protein profiling

    • Utilize live-cell imaging with cell-permeable H6PD activity probes

  • Flow cytometry applications:

    • Develop intracellular staining protocols optimized for flow cytometric analysis

    • Implement index sorting to correlate H6PD expression with functional readouts

    • Combine with cell cycle markers to assess cell cycle-dependent regulation

  • Integration with omics:

    • Correlate H6PD protein levels with single-cell transcriptomics data

    • Apply spatial transcriptomics to map H6PD expression in tissue contexts

    • Combine with metabolomic profiling to link enzyme expression to metabolic states

What methodological considerations should be made when studying H6PD in relation to immunogenic cell death?

Recent findings linking G6PD inhibition to immunogenic cell death (ICD) suggest H6PD may have similar roles . When investigating this:

  • Experimental design considerations:

    • Compare H6PD and G6PD inhibition effects on ICD markers

    • Examine H6PD expression in relation to calreticulin exposure on plasma membrane

    • Assess correlation between H6PD levels and response to immunotherapy

  • Technical approaches:

    • Multiplex staining for H6PD with ICD markers (calreticulin, HMGB1, ATP release)

    • Establish in vitro assays measuring dendritic cell activation following H6PD manipulation

    • Develop tumor models with H6PD knockdown/overexpression to test immunotherapy response

  • Translational considerations:

    • Analyze patient samples for correlations between H6PD expression and immunotherapy outcomes

    • Stratify cancer patient cohorts based on H6PD expression levels

    • Investigate combination approaches targeting H6PD alongside immune checkpoint inhibitors

How might advanced microscopy techniques enhance H6PD research using FITC-conjugated antibodies?

Next-generation microscopy approaches offer new possibilities:

  • Super-resolution microscopy:

    • Apply STED, STORM, or PALM imaging to visualize H6PD distribution within ER subdomains

    • Achieve 20-50nm resolution to examine H6PD clustering and microdomains

    • Combine with proximity labeling to identify novel interaction partners

  • Intravital microscopy applications:

    • Develop protocols for in vivo H6PD visualization in animal models

    • Monitor dynamic changes in H6PD expression during disease progression

    • Track H6PD in response to therapeutic interventions in real-time

  • Correlative light and electron microscopy (CLEM):

    • Precisely localize H6PD at the ultrastructural level within the ER

    • Examine H6PD distribution in relation to ER morphological changes

    • Investigate H6PD localization during ER stress responses

What methodological advances could improve the study of H6PD in metabolic disorders?

Innovative approaches for metabolic disease research include:

  • Tissue-specific analysis:

    • Develop protocols for H6PD detection in difficult tissues (adipose, pancreatic islets)

    • Apply laser capture microdissection with subsequent H6PD quantification

    • Establish organoid models for examining H6PD regulation in 3D culture systems

  • Functional readouts:

    • Combine H6PD detection with real-time metabolic measurements

    • Develop biosensors for monitoring H6PD activity in living cells

    • Correlate H6PD localization with local NADPH production

  • Translational approaches:

    • Establish standardized protocols for H6PD assessment in clinical biopsies

    • Develop image analysis pipelines for automated H6PD quantification

    • Create reference datasets for H6PD expression across human tissues and disease states

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.