PDE6H Human

Phosphodiesterase 6H cGMP-Specific Cone Gamma Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to PDE6H

The PDE6H gene encodes the inhibitory gamma subunit (PDE6γ’) of cone-specific phosphodiesterase (PDE6), a key enzyme in phototransduction within retinal cone cells . Located on chromosome 12p13, this gene is critical for converting cyclic guanosine monophosphate (cGMP) to 5’-GMP, enabling membrane channel closure and visual signal transmission . Its expression is retina-specific, with prominent activity in photoreceptor cones responsible for color vision and daylight vision .

Role in Phototransduction

PDE6 in cones is a heterotetramer composed of two catalytic subunits (α/β) and two inhibitory subunits (γ). The PDE6γ’ subunit regulates enzyme activity by modulating cGMP hydrolysis, a process essential for adapting to varying light intensities . Disruption of this subunit impairs phototransduction, leading to vision disorders .

ComponentFunctionLocalization
PDE6γ’ (PDE6H)Inhibitory regulation of cGMP-PDE activityCone photoreceptor outer segments and synaptic terminals
Catalytic α/β subunitsHydrolysis of cGMPCone photoreceptor outer segments

Associated Diseases

Mutations in PDE6H are linked to:

  • Incomplete achromatopsia (ACHM6): A rare recessive disorder characterized by reduced color vision, photophobia, and nystagmus .

  • Retinal cone dystrophy type 3A (RCD3A): Progressive cone dysfunction leading to vision loss .

DiseaseGenetic CauseFunctional Impact
ACHM6Nonsense mutation (e.g., Ser12Ter/S12X)Nonfunctional PDE6γ’ subunit, impaired cGMP-PDE activity
RCD3AMissense/nonsense mutationsCone-specific phototransduction defects

Cancer Biology

Recent studies highlight PDE6H’s role in cancer metabolism:

  • PDE6H knockdown in colorectal cancer cells (e.g., HCT116) induces G1 cell cycle arrest, reduces mTORC1 signaling, and suppresses mitochondrial function .

  • PDE6H knockout increases intracellular cGMP, alters purine metabolism, and inhibits tumor growth in xenograft models .

StudyKey FindingsSpecies/Model
PDE6H in cancer cells G1 arrest, reduced mTORC1, mitochondrial dysfunctionHuman colorectal cancer
Sildenafil treatment Slowed tumor growth, no additive effect in PDE6H-deficient tumorsHCT116 xenografts

Human vs. Mouse Models

  • Human PDE6H mutations cause achromatopsia/RCD3A due to loss of cone-specific PDE6γ’ .

  • Mouse PDE6H knockout shows no retinal dysfunction; rod-derived PDE6G compensates for cone PDE6γ’ deficiency .

SpeciesPDE6H Deficiency PhenotypeMechanism
HumanCone dysfunction, vision lossNo compensatory PDE6G in cones
MouseNormal retinal functionPDE6G substitutes for PDE6H

Tissue Specificity

PDE6H is primarily expressed in the retina, with minimal detection in other tissues .

TissueExpression LevelSource
RetinaHighHuman Protein Atlas
Brain/CerebellumLow/UndetectableHuman Protein Atlas

Thyroid Hormone (TH) Regulation

In non-mammalian models (e.g., flounder), pde6h expression peaks during metamorphosis and is regulated by TH signaling via thyroid hormone receptors (TRs) .

StagePDE6H ExpressionRegulation
Metamorphosis peakHighTH-dependent activation
Adult tissuesEye-specificTRαA-TRE interaction

Product Specs

Introduction
PDE6H is a member of the rod/cone cGMP-PDE gamma subunit family. This family is responsible for regulating the levels, location, and duration of action of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) by controlling their degradation rate. PDE6H specifically inhibits the cone-specific cGMP phosphodiesterase, a tetramer composed of two catalytic chains (alpha and beta) and two inhibitory chains (gamma). This protein is primarily found in the retina and plays a crucial role in transmitting and amplifying visual signals. Mutations in PDE6H have been linked to retinal cone dystrophy type 3A.
Description
Recombinant PDE6H Human, produced in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 106 amino acids (with amino acids 1-83 being part of the PDE6H sequence) and has a molecular mass of 11.5 kDa. For purification purposes, a 23 amino acid His-tag is fused to the N-terminus. The protein is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The PDE6H protein solution has a concentration of 0.5 mg/ml and is supplied in a buffer containing 20mM Tris-HCl (pH 8.0), 0.2M NaCl, 40% glycerol, 2mM DTT, and 0.1mM PMSF.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein such as HSA or BSA (0.1%) is advisable for long-term storage. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the PDE6H protein is determined by SDS-PAGE analysis and is greater than 80%.
Synonyms
Phosphodiesterase 6H, CGMP-Specific, Cone, Gamma, Retinal Cone Rhodopsin-Sensitive CGMP 3',5'-Cyclic Phosphodiesterase, Subunit Gamma, EC 3.1.4.35, EC 3.1.4.17, RCD3, GMP-PDE Gamma, ACHM6, PDE6H.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMSDNTTL PAPASNQGPT TPRKGPPKFK QRQTRQFKSK PPKKGVKGFG DDIPGMEGLG TDITVICPWE AFSHLELHEL AQFGII.

Q&A

What is the precise molecular function of PDE6H in human tissues?

PDE6H encodes the inhibitory (gamma) subunit of cone-specific cGMP phosphodiesterase, which functions as a tetramer composed of two catalytic chains (alpha and beta) and two inhibitory chains (gamma). This protein actively participates in the transmission and amplification of visual signals and is specifically expressed in the retina .

Methodologically, researchers can investigate PDE6H function through:

  • Immunohistochemistry of retinal tissue using PDE6H-specific antibodies

  • Measurement of cGMP levels in cells with modulated PDE6H expression

  • Analysis of protein-protein interactions between PDE6H and other phototransduction components

What genetic and protein identifiers are associated with human PDE6H?

Human PDE6H is identified by multiple database entries across research platforms:

DatabaseIdentifierAdditional Information
HGNC8790Official gene naming authority
NCBI Gene5149Gene sequence and variation data
EnsemblENSG00000139053Genome annotation resource
OMIM601190Disease associations and inheritance
UniProtKB/Swiss-ProtQ13956Protein sequence and structure

Researchers should use these standardized identifiers when reporting PDE6H experiments to maintain consistency across studies .

What is the tissue expression profile of PDE6H in humans?

Methodological approach for expression analysis:

  • RNA-seq and single-cell transcriptomics for tissue-specific expression

  • Immunohistochemistry with validated antibodies

  • Western blot analysis of protein expression in various tissues

  • qRT-PCR for quantitative expression measurement

The Human Protein Atlas demonstrates predominant PDE6H expression in retinal tissue, with minimal expression in other tissues , making it a relatively specific biomarker for cone photoreceptors in normal physiology.

How can researchers effectively knock down or knock out PDE6H in experimental models?

For successful PDE6H modulation, researchers have employed several techniques with varying efficiency:

siRNA knockdown approach:

  • Design siRNAs targeting conserved regions of PDE6H mRNA

  • Transfect target cells (e.g., HCT116) with siRNA using lipofection

  • Confirm knockdown efficiency via qRT-PCR and Western blot

  • Assess phenotypic changes 48-72 hours post-transfection

CRISPR-Cas9 knockout strategy:

  • Design guide RNAs targeting early exons of PDE6H

  • Clone into appropriate CRISPR vectors

  • Generate stable knockout cell lines through transfection and selection

  • Validate knockout through sequencing and protein expression analysis

Research has demonstrated successful PDE6H knockdown and knockout in cancer cell lines, revealing its role in cell cycle progression and metabolism .

What experimental approaches effectively measure changes in cGMP levels following PDE6H modulation?

Since PDE6H regulates cGMP-specific phosphodiesterase activity, measuring cGMP is critical:

Methodological options include:

  • Enzyme-linked immunosorbent assay (ELISA) for cGMP quantification

    • Commercial kits available with detection limits ~0.1 pmol/ml

    • Sample cells must be lysed in acidic conditions to prevent cGMP degradation

  • Radioimmunoassay (RIA)

    • Higher sensitivity but requires radioisotope handling facilities

  • HPLC-MS/MS analysis

    • Most precise quantification method

    • Can simultaneously measure multiple nucleotides

  • Fluorescent biosensors

    • Allow real-time, live-cell monitoring of cGMP fluctuations

    • Require genetic manipulation to express the sensor

Studies have confirmed that PDE6H knockout results in increased intracellular cGMP levels in experimental models .

How should researchers design experiments to investigate PDE6H's emerging role in cancer?

Based on recent discoveries, a comprehensive experimental design should include:

  • Expression analysis:

    • Analyze PDE6H expression across cancer cell lines and patient samples

    • Compare with matched normal tissues

    • Correlate expression with clinical outcomes

  • Functional studies:

    • Perform PDE6H knockdown/knockout in multiple cancer cell lines

    • Assess impacts on:

      • Cell proliferation and viability

      • Cell cycle progression (particularly G1 arrest)

      • Apoptosis markers

      • Migration and invasion capacity

  • Mechanistic investigations:

    • Measure cGMP levels and downstream signaling

    • Assess mTORC1 pathway activity

    • Analyze mitochondrial function

    • Perform metabolomic profiling

  • In vivo validation:

    • Generate xenograft models with PDE6H-knockout cancer cells

    • Test PDE6 inhibitors like sildenafil on tumor growth

    • Monitor survival outcomes

Recent research has demonstrated that PDE6H deletion, as well as treatment with the PDE5/6 inhibitor sildenafil, significantly slowed tumor growth and improved survival in xenograft models .

What human diseases are definitively linked to PDE6H mutations?

PDE6H mutations have been associated with two main retinal disorders:

DiseaseInheritance PatternClinical FeaturesKey Mutation Types
Retinal Cone Dystrophy 3A (RCD3A)Autosomal recessiveProgressive cone dysfunction, reduced central vision, color vision defectsMissense, nonsense, frameshift
AchromatopsiaAutosomal recessiveCongenital color blindness, photophobia, reduced visual acuity, nystagmusPrimarily nonsense mutations

Researchers investigating PDE6H in retinal diseases should employ:

  • Targeted gene sequencing or whole-exome sequencing

  • Functional validation of identified variants

  • Phenotype-genotype correlation analysis

  • Model systems to recapitulate disease mechanisms .

How can researchers experimentally differentiate between the impact of PDE6H mutations versus other phototransduction genes?

To disambiguate PDE6H-specific effects from other phototransduction genes:

Methodological approach:

  • Generate isogenic cell lines with various phototransduction gene mutations

  • Create animal models with conditional, tissue-specific knockouts

  • Perform rescue experiments where wild-type PDE6H is reintroduced

  • Conduct comparative transcriptomics and proteomics

  • Use pharmacological inhibitors with varying specificities

Cross-species studies have revealed significant differences in photoreceptor protein inventory between species, highlighting the importance of human-specific studies when possible .

How does PDE6H knockout affect cancer cell metabolism beyond cGMP regulation?

Recent research has uncovered broader metabolic impacts of PDE6H modulation:

Key findings from metabolic studies:

  • PDE6H knockout modifies levels of nucleotides and key energy metabolism intermediates

  • Both knockdown and knockout of PDE6H result in suppression of mitochondrial function

  • These metabolic changes appear independent of the PKG pathway

  • Changes resemble aspects of the "dark retina response" seen in photoreceptors

Methodological approaches for metabolic analysis:

  • Targeted and untargeted metabolomics

  • Seahorse XF analysis for mitochondrial function

  • 13C metabolic flux analysis

  • Isotope tracing experiments

  • Analysis of metabolic gene expression

What molecular mechanisms link PDE6H to mTORC1 signaling in cancer cells?

PDE6H knockdown reduces mTORC1 signaling in cancer cell lines through pathways that researchers can investigate using:

Experimental approaches:

  • Western blot analysis of phospho-S6K, phospho-4EBP1, and other mTORC1 effectors

  • Pharmacological manipulation with rapamycin and other mTOR inhibitors

  • Genetic manipulation of upstream regulators (TSC1/2, AMPK, etc.)

  • Comparative analysis with other cGMP-modulating interventions

  • Co-immunoprecipitation to identify novel protein interactions

This PDE6H-mTORC1 connection may partly explain the effects on cancer cell proliferation and offers potential for combined therapeutic targeting .

How might PDE6H inhibition be optimized as a cancer therapeutic approach?

Building on the discovery that PDE6H deletion and sildenafil treatment slow tumor growth:

Strategic research considerations:

  • Develop high-throughput screening for PDE6H-specific inhibitors

  • Test combinations with established cancer therapies

  • Evaluate cancer type-specific responses based on PDE6H expression

  • Investigate potential resistance mechanisms

  • Assess toxicity profiles, particularly related to visual function

Methodological approach for drug development:

  • Structure-based design using crystal structure information

  • Fragment-based drug discovery

  • High-throughput screening of compound libraries

  • Animal testing of lead compounds

  • Biomarker development for patient stratification

Sildenafil treatment did not show additive effects on slowing PDE6H-deficient tumor growth, suggesting both interventions act through the same pathway .

How can researchers reconcile contradictory data regarding PDE6H function across different experimental systems?

Contradictions in PDE6H research may arise from:

Methodological considerations for resolving contradictions:

  • Standardize experimental conditions and cell lines

  • Compare acute versus chronic PDE6H inhibition

  • Distinguish between genetic knockout and pharmacological inhibition

  • Account for compensatory mechanisms in long-term studies

  • Consider species- and tissue-specific differences in PDE6H function

  • Validate findings across multiple model systems

  • Use inducible systems to control timing of PDE6H modulation

Targeted ablation studies in mice have revealed significant cross-species differences in photoreceptor protein isoform inventory, highlighting potential limitations in extrapolating from animal models to humans .

What are the optimal experimental controls when studying PDE6H in cancer cells that don't normally express visual transduction proteins?

When studying PDE6H in non-retinal contexts:

Recommended control strategy:

  • Include multiple non-targeting control siRNAs/sgRNAs

  • Generate rescue cell lines re-expressing wild-type PDE6H

  • Use pharmacological inhibitors alongside genetic approaches

  • Include siRNAs targeting related PDE family members

  • Validate findings in multiple cell lines with varying baseline PDE6H expression

  • Consider inducible systems to control the timing of PDE6H modulation

Studies have identified PDE6H as a controller of cell cycle progression in HCT116 cells, despite this being a colorectal cancer cell line rather than retinal tissue .

How can researchers effectively study the interplay between PDE6H and the PKG-independent pathways it influences?

To elucidate PKG-independent mechanisms:

Experimental design considerations:

  • Use PKG inhibitors (e.g., KT5823) alongside PDE6H modulation

  • Generate PKG knockout cells with PDE6H modulation

  • Directly measure cGMP-PKG pathway activation

  • Perform phosphoproteomic analysis to identify novel targets

  • Conduct interactome studies to identify PDE6H binding partners

  • Use cGMP analogs that selectively activate or inhibit specific pathways

Research has demonstrated that PDE6H depletion results in metabolic changes that are independent of the PKG pathway, suggesting novel mechanisms of action that warrant further investigation .

Product Science Overview

Structure and Function

PDE6H is part of a tetrameric complex composed of two catalytic chains (alpha and beta) and two inhibitory chains (gamma). The gamma subunit, encoded by the PDE6H gene, plays a vital role in regulating the activity of the catalytic subunits by inhibiting their function in the absence of light. This regulation is essential for the proper transmission and amplification of visual signals in the retina .

Expression and Localization

The PDE6H gene is specifically expressed in the retina, where it is involved in the phototransduction pathway. Phototransduction is the process by which light is converted into electrical signals in the photoreceptor cells of the retina. The gamma subunit of PDE6H is particularly important in cone cells, which are responsible for color vision and function best in bright light conditions .

Clinical Significance

Mutations in the PDE6H gene have been associated with retinal cone dystrophy type 3A (RCD3A) and achromatopsia. RCD3A is a condition characterized by the progressive loss of cone photoreceptor function, leading to decreased visual acuity and color vision defects. Achromatopsia is a condition where individuals have little to no color vision and experience photophobia and nystagmus .

Research and Applications

Human recombinant PDE6H is used in various research applications to study its role in visual signal transduction and its involvement in retinal diseases. Recombinant proteins are produced through genetic engineering techniques, allowing researchers to investigate the protein’s structure, function, and interactions in a controlled environment .

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 2024 Thebiotek. All Rights Reserved.