GUCY2D Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery time may vary depending on the shipping method or location. Please consult your local distributor for specific delivery timelines.
Synonyms
GUCY2D antibody; CORD6 antibody; GUC1A4 antibody; GUC2D antibody; RETGC antibody; RETGC1 antibody; Retinal guanylyl cyclase 1 antibody; RETGC-1 antibody; EC 4.6.1.2 antibody; CG-E antibody; Guanylate cyclase 2D antibody; retinal antibody; Rod outer segment membrane guanylate cyclase antibody; ROS-GC antibody
Target Names
GUCY2D
Uniprot No.

Target Background

Function
GUCY2D Antibody catalyzes the synthesis of cyclic GMP (cGMP) in rods and cones of photoreceptors. This enzyme plays a crucial role in phototransduction, specifically mediating cGMP replenishment. Additionally, it may contribute to the trafficking of membrane-associated proteins to the photoreceptor outer segment membrane.
Gene References Into Functions
  1. As further genotypes were identified, efforts were made to classify them based on phenotype: GUCY2D-LCA was considered a cone-rod dystrophy, while other genotypes were designated as rod-cone dystrophies PMID: 28212877
  2. Sequencing of GUCY2D revealed a novel missense mutation (c.2129C:T; p.Ala710Val) leading to the substitution of alanine with valine at position 710. This alteration modifies the conformation of the regulatory segment within the kinase styk-domain of GC1, disrupting its helical structure. This likely inhibits phosphorylation of threonine residue within this segment, which is essential for activating the catalytic domain of the protein. PMID: 27475985
  3. These findings highlight distinct effects of blindness on the visual pathway. Notably, the relatively intact postgeniculate white matter pathway in GUCY2D-LCA offers promising prospects for visual function recovery with gene augmentation therapy. PMID: 28403437
  4. Two novel mutations causing phenotypic LCA and Alstrom syndrome in Saudi patients from consanguineous families expand the genotypic spectrum of congenital retinal dystrophies. PMID: 26957854
  5. Data suggest that GCAP1 (guanylate cyclase activator 1A; Mg2+ vs. Ca2+) exhibits conformational changes in the Ca2+ switch helix that are critical for activating RetGC1. Myristoylation of GCAP1 is also essential in attaining the activator conformation. PMID: 26703466
  6. The Gc1s/Gc1s phenotype variant of DBP and the unbound fraction of plasma RBP4 may be considered as factors associated with the incidence, and potentially the risk, of IR in CHC patients. PMID: 26962819
  7. The guanylate cyclase signaling pathway is downregulated in the pathogenesis of inflammatory bowel diseases. PMID: 25979109
  8. Studies indicate that mutations in retinal guanylate cyclase-1 (GUCY2D) are linked to a leading cause of recessive Leber congenital amaurosis (LCA1). PMID: 26427419
  9. GUCY2D mutations were prevalent in Chinese families with autosomal dominant cone or cone-rod dystrophies. All mutations were identified in exon 13. PMID: 26298565
  10. Cardiac fibrosis and the endogenous natriuretic peptide system were evaluated in end-stage heart failure to assess the anti-fibrotic actions of the dual GC-A/-B activator. PMID: 25117468
  11. A deletion mutation in the GUCY2D gene is associated with Leber congenital amaurosis in a consanguineous Pakistani family. PMID: 25189253
  12. GUCY2D is a significant contributor to autosomal dominant cone and cone-rod dystrophies in Israel PMID: 25515582
  13. Neurodevelopmental delay is a potential characteristic of strictly defined LCA, documented in our series for some children with homozygous RPGRIP1 and GUCY2D mutations. PMID: 24997176
  14. Screening of the GUCY2D gene revealed the mutation p.R838H in all affected individuals with autosomal dominant cone dystrophy and was absent in asymptomatic patients. PMID: 24480840
  15. A missense mutation in the GUCY2D gene caused ADCRD in this family. Clinical follow-up of this family with a typical CRD phenotype revealed disease progression over time. PMID: 23686677
  16. Data suggest that the dimerization domain of GUCY2D functions as a calcium-sensitive regulatory module. GUCY2D requires the correct conformation of the monomer-monomer interface to interact with guanylate cyclase activating proteins (GCAP1; GCAP2). PMID: 23815670
  17. A novel missense mutation of the GUCY2D gene was identified in this study. PMID: 23734073
  18. This study proposes a relationship between the level of RetGC1 activity and the severity of cone vision abnormality. It advocates that cone function should be the primary efficacy outcome in clinical trials of gene augmentation therapy for LCA1 PMID: 23035049
  19. Expression of mutant human RETGC-1 leads to a retinal phenotype characterized by aberrant photoreceptor morphology and a reduced number of photoreceptors. PMID: 23328348
  20. This is the first report of a GUCY2D mutation causing central areolar choroidal dystrophy and enhances our understanding of genotype-phenotype correlation within this diverse group of choroidoretinal dystrophies. PMID: 22695961
  21. A recurrent heterozygous (p.Arg838His) mutation in GUCY2D is associated with autosomal dominant cone dystrophy in a Chinese family. PMID: 22194653
  22. Following subretinal delivery of a vector containing GUCY2D in Gucy2e(-/-) mice, GC1 protein was detected in the rod and cone outer segments, transducin was appropriately localized to cone outer segments, and an improvement in visual behavior was observed. PMID: 21671801
  23. Two macular dystrophy-associated disease mutations at codon 838 of the GUCY2D gene were found among the 22 unrelated Spanish families, one of which had not been previously described (p.R838P). This novel mutation exhibited phenotypic variability. PMID: 21552474
  24. This study establishes GUCY2D, a common cause for both recessive Leber's congenital amaurosis and dominant cone-rod dystrophy, as a potential candidate for autosomal recessive cone-rod dystrophy. PMID: 20517349
  25. Variations in macular microstructures were observed among LCA (Leber congenital amaurosis) patients with different genotypes. PMID: 19959640
  26. Studies show that a fold recognition-based model of the catalytic domain of ROS-GC1 was constructed, and neurocalcin delta docking simulations were performed to define the three-dimensional features of the interacting domains of the two molecules. PMID: 18500817
  27. The coexpression of ROS-GC1 and its activators in spermatozoa suggests that the Ca(2+)-modulated ROS-GC1 transduction system may be involved in the fertilization process. PMID: 19111294
  28. Clustering and frequency of mutations in patients with dominant cone-rod dystrophies PMID: 11565546
  29. Some carrier parents of patients with Leber congenital amaurosis and a GUCY2D mutation develop measurable cone and possibly rod abnormalities, consistent with mild cone-rod dysfunction. PMID: 12365911
  30. Two amino acid substitution missense mutations at R838C and R838H have been identified, along with 11 new polymorphic markers. PMID: 12552567
  31. Leber congenital amaurosis (LCA) caused by mutant GUCY2D had only light perception but retained a substantial number of cones and rods in the macula and far periphery. PMID: 12623820
  32. A heterozygous complex mutation of I915T and G917R in the GUCY2D gene caused autosomal dominant CORD (cone-rod dystrophy). PMID: 15111605
  33. LCA (Leber congenital amaurosis) is caused by the modifying effect of a heterozygous GUCY2D mutation observed against the disease background of a homozygous RPE65 mutation. PMID: 15512997
  34. AIPL1, CRB1, GUCY2D, RPE65, and RPGRIP1 mutations may play a role in juvenile retinitis pigmentosa. PMID: 16272259
  35. Microarray-based mutation detection enabled the identification of 32% of LCA sequence variants and represents an efficient first-pass screening tool. Mutations in CRB1, and to a lesser extent, in GUCY2D, underlie most LCA cases in this cohort. PMID: 16505055
  36. There is a phenotype-genotype correlation of autosomal dominant cone-rod dystrophy due to the R838C mutation of the GUCY2D gene encoding retinal guanylate cyclase-1. PMID: 17041576
  37. Testing confirms the diagnosis at the molecular level and allows for a more precise prognosis of the potential future clinical evolution. PMID: 17651254
  38. This illustrates the use of haplotype information about allele-sharing decay around a mutation as a genetic clock, utilizing data for two GUCY2D mutations in Mediterranean populations. PMID: 17684531
  39. RPE65 gene mutations represented a significant cause of LCA in the Italian population, while GUCY2D and CEP290 mutations had a lower frequency than reported elsewhere. PMID: 17724218
  40. A novel mutation, P575L, was found in exon 8 of the GUCY2D gene in 12 members of a family with autosomal dominant progressive cone degeneration. PMID: 18332321
  41. GUCY2D is a major gene responsible for progressive autosomal dominant cone degeneration. All identified mutations localize to codon 838. PMID: 18487367

Show More

Hide All

Database Links

HGNC: 4689

OMIM: 204000

KEGG: hsa:3000

STRING: 9606.ENSP00000254854

UniGene: Hs.592109

Involvement In Disease
Leber congenital amaurosis 1 (LCA1); Cone-rod dystrophy 6 (CORD6)
Protein Families
Adenylyl cyclase class-4/guanylyl cyclase family
Subcellular Location
Photoreceptor outer segment membrane; Single-pass type I membrane protein. Endoplasmic reticulum membrane; Single-pass type I membrane protein.
Tissue Specificity
Retina.

Q&A

What is GUCY2D and what is its role in retinal function?

GUCY2D encodes a retina-specific guanylate cyclase that belongs to the membrane guanylyl cyclase family. Unlike other membrane guanylyl cyclases, this enzyme is not activated by natriuretic peptides . GUCY2D plays a critical role in the phototransduction cascade, specifically in the recovery and regulation phases. It functions to restore cGMP levels in photoreceptors after light stimulation, which is essential for returning the photoreceptor to its dark-adapted state.

Mutations in GUCY2D result in severe visual impairments including Leber congenital amaurosis (LCA1) and cone-rod dystrophy-6 . These conditions typically manifest as decreased visual acuity and sensitivity in the central visual field, followed by progressive loss of peripheral vision . Interestingly, despite significant visual impairment, patients with GUCY2D-LCA1 demonstrate preserved optic chiasm volume and white matter organization of the optic radiations .

What types of GUCY2D antibodies are commercially available for research?

Several types of GUCY2D antibodies are available for research applications, with the most common being rabbit polyclonal antibodies. These include:

Antibody TypeHostReactivityApplicationsImmunogen Region
PolyclonalRabbitHuman, Mouse, RatWBRecombinant fusion protein of human GUCY2D
PolyclonalRabbitHuman, RatWB, FACS, IHCSynthetic peptide between AA 540-570

The antibodies are typically generated using either recombinant fusion proteins of human GUCY2D or synthetic peptides corresponding to specific amino acid sequences, such as the region between amino acids 540-570 from the central portion of the human GUCY2D protein . These different immunogens may result in antibodies with varying epitope recognition patterns, which can be advantageous for different experimental applications.

What applications are GUCY2D antibodies most commonly used for in research?

GUCY2D antibodies are primarily utilized in the following research applications:

  • Western Blotting (WB): All commercially available GUCY2D antibodies are validated for Western blot applications with recommended dilution ranges of 1:200-1:2000 . This technique allows for detection and quantification of GUCY2D protein in tissue lysates.

  • Immunohistochemistry (IHC): Some GUCY2D antibodies are validated for use in immunohistochemistry on paraffin-embedded sections , enabling the visualization of GUCY2D distribution within retinal tissues.

  • Flow Cytometry (FACS): Select antibodies are suitable for flow cytometry applications , which can be useful for analyzing GUCY2D expression in dissociated retinal cells.

In research contexts, these applications are particularly valuable for:

  • Characterizing GUCY2D expression patterns in normal versus diseased retinal tissues

  • Validating gene therapy approaches targeting GUCY2D

  • Investigating the molecular mechanisms of retinal degenerative diseases

  • Monitoring changes in GUCY2D expression levels in response to therapeutic interventions

How should GUCY2D antibodies be stored and handled for optimal performance?

Proper storage and handling of GUCY2D antibodies is critical for maintaining their activity and specificity. Based on manufacturer recommendations:

  • Long-term storage: Store at -20°C for up to one year . Some antibodies are formulated with 50% glycerol to prevent freeze damage.

  • Short-term storage: For frequent use, store at 4°C for up to one month .

  • Avoid freeze-thaw cycles: Repeated freezing and thawing can degrade antibody quality and should be minimized .

  • Buffer composition: GUCY2D antibodies are typically supplied in PBS with 0.02% sodium azide and 50% glycerol at pH 7.2 . This formulation helps maintain antibody stability during storage.

To maximize antibody performance:

  • Aliquot antibodies upon receipt to minimize freeze-thaw cycles

  • Thaw frozen antibodies completely before use and mix gently

  • Centrifuge briefly before opening to collect all material at the bottom of the tube

  • Handle antibodies with care to avoid contamination and degradation

What disease models are available for studying GUCY2D function?

Several mouse models have been developed to study GUCY2D function and test potential therapeutic approaches:

  • Guanylate Cyclase 1/2 Double Knockout (GCDKO) Mice: These mice carry disruptions in both the Gucy2e and Gucy2f genes, resulting in loss of rod and cone structure and function . They lack all retinal guanylate cyclase activity, making them useful for evaluating AAV-mediated guanylate cyclase enzyme activity .

  • Guanylate Cyclase 1 Knockout (GC1KO) Mice: These mice lack cone function, and their cones degenerate over time. Rod function and structure are retained due to the presence of retGC2 (encoded by Gucy2f) . This model allows for:

    • Evaluation of cone function restoration via photopic ERG

    • Assessment of cone structure via optical coherence tomography (OCT)

    • Safety monitoring through scotopic (rod) ERG and ONL thickness measurement

These models have been instrumental in preclinical studies for gene therapy approaches targeting GUCY2D-related diseases. For example, subretinal delivery of AAV5-hGRK1-GUCY2D has been shown to significantly improve both cone and rod function in the GCDKO mouse model of LCA1 .

What is the relationship between GUCY2D mutations and retinal diseases?

Mutations in the GUCY2D gene are associated with several retinal degenerative disorders:

DiseaseCharacteristicsPublications with GUCY2D
Leber Congenital Amaurosis (LCA1)Severe early-onset retinal dystrophy with profound vision lossNumerous
Cone-Rod Dystrophy 5 (CORD5)Progressive loss of cone then rod photoreceptor function>13 publications
Retinal Cone Dystrophy 2 (RCD2)Primary degeneration of cone photoreceptors>2 publications
Retinitis PigmentosaProgressive degeneration of rod and then cone photoreceptors>30 publications

These conditions result from disruptions in the phototransduction cascade, particularly in the recovery and regulation phases. GUCY2D mutations lead to decreased visual acuity and sensitivity in the central visual field, followed by progressive loss of peripheral vision .

The gene therapy approach using AAV5-hGRK1-GUCY2D is currently being evaluated in phase I/II clinical trials (ClinicalTrials.gov: NCT03920007) and is showing early signs of efficacy and safety . This treatment represents a potential first-in-class therapy for GUCY2D-LCA1.

How can Western blot protocols be optimized for detecting GUCY2D in retinal samples?

Optimizing Western blot protocols for GUCY2D detection requires careful consideration of several parameters:

Sample Preparation:

  • Use fresh retinal tissue when possible, or snap-freeze samples immediately after collection

  • Include protease inhibitors in lysis buffers to prevent GUCY2D degradation

  • Consider membrane fractionation techniques, as GUCY2D is a membrane-associated protein

Antibody Selection and Dilution:

  • GUCY2D antibodies typically work in a dilution range of 1:200-1:2000 for Western blot applications

  • Validate optimal dilution for each new lot of antibody using positive control samples

  • For stronger signals with rabbit GUCY2D antibodies, use HRP-conjugated Goat Anti-Rabbit IgG (H+L) as a secondary antibody at approximately 1:10000 dilution

Protocol Optimization:

  • Load sufficient protein (approximately 25μg per lane)

  • Use 3% nonfat dry milk in TBST as a blocking buffer

  • Consider longer exposure times, as documented examples used 90-second exposures

  • For detection, ECL Basic Kit has been successfully used in published protocols

Controls and Validation:

  • Include protein extracts from various cell lines as controls

  • Consider using retinal tissue from GUCY2D knockout models as negative controls

  • For human samples, use known GUCY2D-expressing tissues like retina as positive controls

The expected molecular weight of GUCY2D is approximately 120kDa , which should be confirmed in all Western blot experiments to verify specificity.

What considerations are important when using GUCY2D antibodies in immunohistochemistry of retinal tissues?

Successful immunohistochemical detection of GUCY2D in retinal tissues requires several key considerations:

Tissue Processing:

  • Choose appropriate fixation methods that preserve GUCY2D epitopes while maintaining tissue morphology

  • Consider brief fixation times (4-8 hours) with 4% paraformaldehyde for retinal tissues

  • For paraffin embedding, use careful dehydration protocols to minimize protein denaturation

Antigen Retrieval:

  • Heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) may be necessary to unmask GUCY2D epitopes

  • Optimize retrieval time and temperature based on tissue preparation method

Antibody Selection:

  • Choose antibodies validated for IHC applications, such as those recognizing the AA 540-570 region of GUCY2D

  • Start with recommended dilutions and optimize for your specific experimental conditions

  • Consider using fluorescent secondary antibodies for colocalization studies with other retinal markers

Controls and Interpretation:

  • Include tissue sections from GUCY2D knockout models as negative controls

  • Use adjacent sections with primary antibody omitted as technical controls

  • Consider double-labeling with established photoreceptor markers to confirm cellular localization

  • Be aware that GUCY2D expression is primarily confined to photoreceptor cells in the retina

Analysis:

  • Document imaging parameters carefully to allow for comparative analyses

  • Consider quantitative approaches to measure staining intensity or pattern changes in disease models

  • Use consistent anatomical landmarks in the retina for reproducible analyses

How can GUCY2D antibodies be used to validate gene therapy approaches?

GUCY2D antibodies play a crucial role in validating gene therapy approaches targeting GUCY2D-related retinal diseases:

Pre-clinical Validation:

  • Confirm transgene expression in appropriate cellular compartments following vector administration

  • Quantify GUCY2D protein levels in treated versus untreated retinas using Western blot

  • Verify the spatial distribution of expressed GUCY2D using immunohistochemistry

  • Compare expression patterns between AAV-delivered human GUCY2D and endogenous mouse Gucy2e in animal models

Mechanism Confirmation:

  • Use antibodies to demonstrate restoration of downstream signaling pathways

  • Correlate GUCY2D expression levels with functional outcomes measured by ERG

  • Assess the relationship between protein expression and structural preservation using OCT and immunostaining

Translational Applications:
In ongoing clinical trials of AAV5-hGRK1-GUCY2D (NCT03920007) , antibodies can be used to:

  • Validate vector production quality by confirming protein expression in vitro

  • Determine optimal viral titers by correlating dose with protein expression levels

  • Assess the longevity of transgene expression in long-term animal studies

Experimental Design Considerations:

  • Include appropriate controls:

    • Uninjected eyes

    • Eyes injected with control vectors (e.g., AAV5-GFP)

    • Time-matched diseased controls

  • Design time-course studies to monitor expression kinetics

  • Consider cross-species reactivity of antibodies when translating from animal models to human applications

What controls should be included when validating GUCY2D antibody specificity?

Rigorous validation of GUCY2D antibody specificity requires comprehensive controls:

Positive Controls:

  • Recombinant GUCY2D protein or cells overexpressing GUCY2D

  • Tissue known to express high levels of GUCY2D (retina)

  • Cell lines with confirmed GUCY2D expression

Negative Controls:

  • Tissues from GUCY2D knockout animals (GCDKO or GC1KO mice)

  • Cell lines that do not express GUCY2D

  • Tissues where GUCY2D is not expected (non-retinal tissues)

Technical Controls:

  • Primary antibody omission control

  • Isotype control (non-specific IgG from the same species)

  • Blocking peptide competition assay using the immunogenic peptide (e.g., synthetic peptide between AA 540-570)

  • Comparison of results with multiple antibodies targeting different GUCY2D epitopes

Validation Experiments:

  • Western blot to confirm single band at expected molecular weight (~120 kDa)

  • Immunoprecipitation followed by mass spectrometry to confirm target identity

  • RNA interference (siRNA) or CRISPR knockout of GUCY2D followed by antibody staining

  • Correlation of protein detection with mRNA expression (RT-PCR or RNA-seq)

Documentation:

  • Record detailed experimental conditions (antibody lot, dilution, incubation time/temperature)

  • Document all validation experiments with appropriate positive and negative controls

  • Note any non-specific binding or background observed under different conditions

What techniques are recommended for troubleshooting inconsistent GUCY2D antibody results?

When facing inconsistent results with GUCY2D antibodies, systematic troubleshooting is essential:

Sample Preparation Issues:

  • Verify protein integrity with general protein stains or housekeeping antibodies

  • Check for proteolytic degradation by using fresh samples and complete protease inhibitor cocktails

  • For membrane proteins like GUCY2D, ensure adequate solubilization with appropriate detergents

  • Consider the impact of fixation methods on epitope preservation in immunohistochemistry

Antibody-Related Factors:

  • Test multiple antibody dilutions (1:200 to 1:2000 range for Western blot)

  • Evaluate different antibody lots or sources

  • Consider antibodies targeting different GUCY2D epitopes

  • Verify antibody stability and storage conditions (-20°C long-term; 4°C short-term)

  • Minimize freeze-thaw cycles as they can degrade antibody quality

Protocol Optimization:

  • Adjust blocking conditions (3% nonfat dry milk in TBST has been validated)

  • Modify incubation times and temperatures

  • Try different detection methods (chemiluminescence vs. fluorescence)

  • For Western blot, experiment with transfer conditions for this high molecular weight protein

  • For IHC, test various antigen retrieval methods

Systematic Approach to Troubleshooting:

  • Change only one variable at a time

  • Document all modifications and results

  • Include positive controls in every experiment

  • Consider tissue-specific factors that might affect GUCY2D detection

How can researchers differentiate between endogenous and therapeutic GUCY2D in gene therapy studies?

Distinguishing between endogenous and therapeutic GUCY2D in gene therapy studies presents unique challenges that require specialized approaches:

Epitope-Tagging Strategies:

  • Use epitope-tagged therapeutic GUCY2D constructs (e.g., FLAG, HA, or myc tags)

  • Employ antibodies against the epitope tag for specific detection of the therapeutic protein

  • Perform dual labeling with tag-specific and pan-GUCY2D antibodies to compare distribution patterns

Species-Specific Detection:

  • Design antibodies that specifically recognize human GUCY2D but not mouse Gucy2e (or vice versa)

  • Use this species specificity to distinguish AAV-delivered human GUCY2D from endogenous mouse protein in preclinical studies

  • Validate species specificity using Western blot or immunostaining on human and mouse samples separately

Expression Pattern Analysis:

  • Compare treated and untreated regions within the same retina

  • Quantify GUCY2D levels to identify overexpression beyond endogenous levels

  • Analyze spatial distribution patterns that might differ between endogenous and therapeutic expression

Functional Discrimination:

  • Use knockout models (GCDKO or GC1KO mice) where any detected GUCY2D must be therapeutically derived

  • Correlate protein detection with functional recovery measured by ERG

  • Employ activity-based assays to measure guanylate cyclase function in treated vs. untreated areas

Timing-Based Approaches:

  • In models with rapid degeneration, establish the timeline of endogenous GUCY2D loss

  • Conduct studies at timepoints when endogenous protein should be absent

  • Design time-course experiments to track therapeutic protein expression over time

These strategies are particularly important in the context of ongoing clinical trials using AAV5-hGRK1-GUCY2D (NCT03920007), where validating the expression and function of the therapeutic gene product is essential for understanding efficacy mechanisms .

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.