RDH12 Human

Retinol Dehydrogenase 12 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biochemical Function and Catalytic Properties

RDH12 exhibits high catalytic efficiency for all-trans-retinal (kcat/Km ~900 min⁻¹·μM⁻¹) and lower activity for 11-cis and 9-cis-retinal . It also metabolizes lipid peroxidation aldehydes (e.g., C₉ aldehydes) but with reduced affinity compared to retinoids . Key interactions include:

  • CRBPI (cellular retinol-binding protein type I): Binds all-trans-retinol, restricting its oxidation by RDH12 .

  • CRALBP (cellular retinaldehyde-binding protein): Inhibits 11-cis-retinal reduction more effectively than 11-cis-retinol oxidation .

SubstrateKm (μM)kcat (min⁻¹)Catalytic Efficiency (kcat/Km)
All-trans-retinal0.5450900
11-cis-retinal2.2990450
9-cis-retinal101000100
4-Hydroxynonenal4.520044.4
Data adapted from

Genetic Mutations and Disease Mechanisms

Mutations in RDH12 disrupt retinoid metabolism, leading to toxic aldehyde accumulation and photoreceptor degeneration. Key findings include:

  • Autosomal Recessive Inheritance: Biallelic mutations (e.g., missense, deletions) cause early-onset LCA or RP. Over 20 mutations have been identified, with founder effects observed in p.L99I, p.T155I, and c.806_810delCCCTG variants .

  • Autosomal Dominant RP: Rare heterozygous frameshift mutations (e.g., c.759del p.Phe254Leufs*24) impair protein dimerization, causing dominant-negative effects .

Mutation TypeExamplesEffect on Enzyme ActivityDisease Association
Missensep.T49M, p.R161QLoss of stability/substrate bindingAR LCA/RP
Deletionc.806_810delCCCTGPremature termination codonAR LCA
Frameshiftc.759del p.Phe254Leufs*24Dominant-negative dimer disruptionAD RP

Clinical Features of RDH12-Associated Retinal Dystrophies

RDH12 mutations result in early-onset, progressive vision loss with distinct phenotypes:

FeatureObservationSource
Age of OnsetInfancy to early childhood (LCA); adolescence to adulthood (RP)
Visual Acuity20/40 to light perception; central vision loss dominates
Retinal ImagingMacular atrophy, parafoveal depigmentation, thin outer nuclear layer (ONL)
ElectrophysiologyNon-reproducible ERG; residual rod function in some cases
Pupillary Light ResponseReduced sensitivity to chromatic stimuli (e.g., 467 nm)

Key Pathological Findings:

  • Cone-Rod Dystrophy: Central photoreceptor loss with relative sparing of peripheral rods .

  • Lipid Peroxidation: Accumulation of 4-hydroxynonenal in photoreceptors, indicating oxidative stress .

Therapeutic Advances

Gene Therapy and Preclinical Models:

  • AAV-Mediated Replacement: Subretinal injection of AAV2/5-RDH12 in Rdh12−/− mice restored retinal reductase activity and protected against light-induced damage .

  • iPSC-Derived Retinal Organoids: Models of AD RP showed disrupted cone function, retinol biosynthesis, and photoreceptor shortening, validating disease mechanisms .

Challenges:

  • Mouse Models: Rdh12−/− mice lack overt retinal degeneration, limiting translation to humans .

  • Outcome Measures: Developing age-appropriate tests (e.g., multi-luminance mobility tests) for clinical trials .

Tissue Expression and Pathological Insights

RDH12 is predominantly expressed in photoreceptor inner segments . Its localization and interaction with retinoid-binding proteins are critical for detoxifying retinaldehydes. In RDH12-deficient cells, aldehyde accumulation triggers apoptosis, explaining the rapid progression of retinal degeneration .

Product Specs

Introduction
RDH12, a member of the short-chain Dehydrogenases / Reductases (SDR) family, is typically found in the brain, stomach, eye, skeletal muscle, and kidney. This NADPH-dependent retinal reductase primarily acts on 9-cis and all-trans-retinol. Additionally, RDH12 participates in metabolizing short-chain aldehydes but lacks steroid dehydrogenase activity.
Description
Recombinant human RDH12, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 303 amino acids (residues 39-316) with a molecular weight of 33.5 kDa. This protein is expressed with a 25 amino acid His-tag fused to the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The RDH12 solution (0.25 mg/ml) is supplied in a buffer containing 20 mM Tris-HCl (pH 8.0), 200 mM NaCl, 2 mM DTT, and 40% glycerol.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 90% by SDS-PAGE analysis.
Synonyms
Retinol dehydrogenase 12 (all-trans/9-cis/11-cis), LCA3, LCA13, SDR7C2, All-trans and 9-cis retinol dehydrogenase, short chain dehydrogenase/reductase family 7C, member 2, FLJ30273, EC 1.1.1.100.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMGKVVV ITGANTGIGK ETARELASRG ARVYIACRDV LKGESAASEI RVDTKNSQVL VRKLDLSDTK SIRAFAEGFL AEEKQLHILI NNAGVMMCPY SKTADGFETH LGVNHLGHFL LTYLLLERLK VSAPARVVNV SSVAHHIGKI PFHDLQSEKR YSRGFAYCHS KLANVLFTRE LAKRLQGTGV TTYAVHPGVV RSELVRHSSL LCLLWRLFSP FVKTAREGAQ TSLHCALAEG LEPLSGKYFS DCKRTWVSPR ARNNKTAERL WNVSCELLGI RWE

Q&A

What is the primary function of RDH12 in human photoreceptors?

RDH12 is an enzyme belonging to the short-chain dehydrogenases/reductases superfamily that is highly expressed in photoreceptor inner segments . Its primary function involves metabolizing retinoids, which are photosensitive molecules crucial for vision and cellular differentiation . The enzyme plays a significant role in clearing excessive retinal and other toxic aldehydes produced by light exposure, serving as a protective mechanism for photoreceptors .

RDH12 exhibits NAD(P)H-dependent reductive activity toward retinaldehydes and other aldehydes, essentially functioning as a retinaldehyde reductase that converts all-trans-retinal to all-trans-retinol within the photoreceptor inner segments. This activity is particularly important for detoxification of potentially harmful aldehydes that could otherwise lead to photoreceptor damage.

What is the genetic location and structure of the human RDH12 gene?

RDH12 is a relatively small gene located on chromosome 14 in humans . The gene contains multiple exons encoding a protein that functions in retinoid metabolism. Its genomic organization has been characterized through molecular genetic analysis which has facilitated the identification of disease-causing variants.

The coding sequence of RDH12 is conserved across species, highlighting its evolutionary importance in visual function. Expression studies have demonstrated that in addition to the retina, RDH12 transcripts can be detected at lower levels in other tissues, though its primary functional significance appears to be in the retina.

What clinical conditions are associated with RDH12 mutations in humans?

RDH12 mutations are associated with a wide spectrum of inherited retinal disorders showing significant phenotypic variability . The most common presentation is early-onset severe retinal dystrophy/Leber Congenital Amaurosis (EOSRD/LCA), characterized by severe visual impairment beginning in early childhood . Other associated conditions include:

  • Retinitis pigmentosa

  • Cone-rod dystrophy

  • Macular dystrophy

These conditions can be inherited in either autosomal recessive (most common) or autosomal dominant patterns . The specific clinical phenotype often correlates with the type and location of mutations within the RDH12 gene, with complete loss-of-function mutations typically resulting in more severe presentations.

What are the key phenotypic features of RDH12-associated retinal dystrophies?

RDH12-EOSRD/LCA presents with several distinctive clinical features that can help distinguish it from other forms of inherited retinal dystrophies . Key characteristics include:

  • Early-onset visual impairment, often in early childhood

  • Petal-shaped, coloboma-like macular atrophy

  • Variegated watercolour-like pattern in the macula

  • Peripapillary sparing (preservation of retina around the optic disc)

  • Often dense bone spicule pigmentation in the retinal periphery

These features create a recognizable clinical pattern that can guide genetic testing. The progression of disease typically involves early central vision loss followed by peripheral visual field constriction. Night blindness is common, reflecting the involvement of rod photoreceptors.

How is RDH12 expression distributed across human retinal tissue?

RDH12 is predominantly expressed in the inner segments of photoreceptors in the human retina . Expression studies utilizing techniques such as in situ hybridization and immunohistochemistry have demonstrated this localization pattern. In mice, Rdh12 mRNA has been detected exclusively in the retina, with no significant expression in the retinal pigment epithelium (RPE)/choroid/sclera fraction .

Unlike some other retinal dehydrogenases, RDH12 expression appears to be relatively specific to photoreceptors rather than distributing throughout multiple retinal cell types. Age-related changes in expression have been observed, with some studies showing increased expression in older retinal tissue, suggesting potential compensatory mechanisms during aging or in response to retinal stress .

What are the current experimental models for studying RDH12 function and pathology?

Several experimental models have been developed to study RDH12 function and associated pathologies:

  • Mouse Models: Rdh12-knockout mice have been created to study the physiological role of RDH12 and test potential therapies . These models have been valuable for AAV-mediated gene replacement studies, though they don't fully recapitulate the severe human phenotype.

  • Human iPSC-derived Retinal Organoids: Patient-specific hiPSC lines with RDH12 mutations have been differentiated into retinal organoids to model disease mechanisms . These organoids can be cultured for extended periods (up to 44 weeks) to observe developmental and degenerative phenotypes. The protocol typically involves:

    • Generating optic cups between day 21-32

    • Culturing with FGF2 for 7 days

    • Extended culture with retinoic acid (0.5μM) to obtain laminated retinal structures

  • Cell Culture Systems: HEK-293 cells transfected with expression constructs encoding FLAG-tagged versions of RDH12 have been used to examine protein function and antibody specificities .

  • RNA-seq Analysis: Transcriptomic profiling of RDH12-mutant organoids has revealed differential gene expression patterns that provide insights into pathophysiological mechanisms . The data from such studies are publicly available (e.g., through NCBI Gene Expression Omnibus under access codes like GSE271751).

These models collectively provide complementary approaches to understanding RDH12 biology and testing potential therapeutic interventions.

What gene therapy approaches are being investigated for RDH12-associated retinal dystrophies?

Gene therapy represents a promising approach for treating RDH12-associated retinal dystrophies. Current research focuses on the following strategies:

  • AAV-Mediated Gene Replacement: Studies have utilized AAV2/8 vectors containing a rhodopsin-kinase promoter (hRK) to drive expression of human RDH12 cDNA in photoreceptors . This approach has been tested in both wild-type and Rdh12-knockout mice to evaluate safety and efficacy.

  • Targeting Considerations: Given the progressive nature of RDH12-associated retinal degeneration, optimal therapeutic windows are being investigated. Gene therapy approaches must target areas of the retina most likely to respond, which requires precise structural-functional mapping to identify viable tissue .

  • Delivery Methods: Subretinal injections of viral vectors containing the functional RDH12 gene are being evaluated for their ability to transduce photoreceptors efficiently while minimizing procedural damage to the already compromised retina.

Ongoing preclinical investigations and natural history studies are establishing the foundation necessary for designing robust clinical trials . The development of these approaches has required careful consideration of vector design, dosing, and delivery techniques to maximize therapeutic potential.

What are the key considerations for designing clinical trials for RDH12 gene therapy?

Designing clinical trials for RDH12-associated retinal dystrophies requires addressing several challenging considerations:

  • Phenotypic Heterogeneity: The wide variability in RDH12-associated disease presentations necessitates careful patient selection and stratification . Trial designs must account for different rates of progression and baseline characteristics.

  • Outcome Measure Selection: Selecting appropriate endpoints remains challenging due to:

    • The need for structural-functional correlation metrics

    • Limited natural history data

    • Lack of validated patient-reported outcome measures specific to RDH12-associated disease

  • Innovative Trial Designs: Adaptive trial designs that allow modification based on interim results may be particularly valuable for rare conditions like RDH12-associated retinopathies .

  • Multi-stakeholder Involvement: Successful trial design requires collaboration among:

    • Patient organizations (e.g., Eyes on the Future, RDH12 Fund for Sight)

    • Academic and industry researchers

    • Regulatory agencies

    • Clinicians

  • Natural History Data: Comprehensive natural history studies are essential for establishing disease progression benchmarks and identifying suitable outcome measures . These studies help determine the timing of therapeutic intervention and define clinically meaningful changes.

A patient-centric approach incorporating outcomes most valued by the affected community is considered critical for designing trials that will generate meaningful data while addressing patients' most important concerns .

What are the molecular mechanisms underlying RDH12-associated retinal degeneration?

The pathophysiology of RDH12-associated retinal degeneration involves several interrelated mechanisms:

  • Impaired Retinoid Metabolism: Mutations in RDH12 disrupt the photoreceptors' ability to metabolize potentially toxic retinaldehydes, leading to accumulation of these compounds and subsequent cellular damage .

  • Oxidative Stress: RDH12 normally functions to detoxify light-generated aldehydes. Without this protective mechanism, photoreceptors experience increased oxidative damage, particularly following light exposure .

  • Photoreceptor Structural Abnormalities: Studies using electron microscopy of patient-derived retinal organoids have revealed ultrastructural abnormalities in photoreceptors with RDH12 mutations .

  • Differential Gene Expression: RNA-seq analysis of RDH12-mutant organoids has identified dysregulated pathways involved in:

    • Visual transduction

    • Photoreceptor maintenance

    • Cellular stress responses

  • Dominant vs. Recessive Mechanisms: While most cases follow an autosomal recessive inheritance pattern requiring two mutated alleles, dominant mutations may operate through different mechanisms, potentially involving dominant-negative effects or gain-of-function properties .

Understanding these molecular mechanisms is essential for developing targeted therapeutic approaches that address the specific pathological processes involved in RDH12-associated retinal degeneration.

What methodologies are recommended for measuring RDH12 protein expression and activity?

For researchers investigating RDH12, several methodological approaches have been validated for measuring expression and enzymatic activity:

  • Antibody Selection and Validation:

    • Affinity-purified polyclonal antibodies against specific sequences (e.g., 16 amino acids in the C-terminal portion) have been successfully used

    • Antibody specificity should be validated using overexpression systems with tagged proteins (e.g., FLAG-tagged RDH12)

    • Relative antibody affinities should be determined when comparing expression levels

  • Enzymatic Activity Assays:

    • NAD(P)H-dependent reduction of all-trans-retinal to all-trans-retinol can be measured spectrophotometrically

    • Conversion rates using radiolabeled or fluorescent substrates provide quantitative measures of enzymatic function

    • Mass spectrometry-based approaches allow for precise quantification of retinoid metabolites

  • RNA Expression Analysis:

    • qRT-PCR has been used to quantify Rdh11 and Rdh12 mRNA in different ocular tissues

    • Controls for tissue purity (e.g., rhodopsin for retina, Rpe65 for RPE) should be included

    • RNA-seq provides comprehensive transcriptomic profiles and can be analyzed using packages like DESeq2

  • Protein Visualization:

    • Immunohistochemistry with validated antibodies can determine cellular and subcellular localization

    • Immunoprecipitation followed by Western blotting allows for quantification of protein levels

These methodologies provide complementary approaches to characterizing RDH12 expression and function in various experimental contexts.

How do dominant RDH12 mutations differ mechanistically from recessive mutations?

Dominant and recessive RDH12 mutations appear to operate through distinct pathogenic mechanisms:

  • Recessive Mutations:

    • Typically result in loss-of-function through protein truncation, unstable protein, or catalytically inactive enzyme

    • Require mutations in both alleles to manifest disease

    • Often present with earlier and more severe phenotypes (EOSRD/LCA)

    • The normal allele in carriers is sufficient to maintain retinal function

  • Dominant Mutations:

    • May act through dominant-negative effects where mutant protein interferes with function of normal protein

    • Can potentially result in gain-of-function mechanisms

    • Often associated with later-onset presentations such as retinitis pigmentosa

    • Recent research with hiPSC-derived retinal organoids suggests that dominant RDH12 mutations impair photoreceptor development and function

  • Structural Impacts:

    • Recessive mutations often affect critical catalytic domains

    • Dominant mutations may alter protein-protein interactions or create toxic protein products

    • Specific structural changes can be studied using molecular modeling based on crystal structures of related dehydrogenases

RNA-seq analysis of retinal organoids has revealed that dominant RDH12 mutations lead to dysregulation of genes involved in photoreceptor development and maintenance pathways, suggesting broader impacts beyond simple enzymatic function .

What are the recommended outcome measures for clinical studies of RDH12-associated retinal dystrophies?

Based on recent multi-stakeholder workshops and research, several outcome measures have been identified as particularly relevant for clinical studies of RDH12-associated retinal diseases:

  • Functional Measures:

    • Visual acuity (though often limited utility in advanced disease)

    • Visual field testing (static and kinetic perimetry)

    • Full-field stimulus testing (FST)

    • Electroretinography (ERG) for remaining retinal function

    • Microperimetry for macular sensitivity

  • Structural Measures:

    • Optical coherence tomography (OCT) to assess retinal lamination and thickness

    • OCT angiography to evaluate vascular changes

    • Fundus autofluorescence to map RPE health and atrophic regions

    • Adaptive optics imaging for cellular-level assessment

  • Patient-Reported Outcomes:

    • Vision-specific quality of life measures

    • Functional vision assessments

    • Development of RDH12-specific PRO instruments is needed

  • Novel Endpoints:

    • Mobility courses under standardized lighting conditions

    • Reading performance metrics

    • Activities of daily living assessments

The selection of appropriate outcome measures must consider the specific phenotypic features of RDH12-associated disease, including the characteristic macular atrophy pattern and peripapillary sparing . Comprehensive natural history data is essential for informing the selection of these measures and interpreting changes observed in interventional studies.

Product Science Overview

Structure and Function

RDH12 is a membrane-associated enzyme predominantly expressed in the retina, specifically at the base of photoreceptor inner segments . It is composed of 316 amino acids and has a molecular mass of approximately 35 kDa . The enzyme contains two highly conserved motifs among SDRs: the cofactor-binding site and catalytic residues .

The primary function of RDH12 is to catalyze the reduction of all-trans-retinal and its isomers (9-cis-, 11-cis-, and 13-cis-retinal) to their corresponding retinols in the presence of NADPH . This reaction is essential for the regeneration of 11-cis-retinal, a critical component of the visual cycle. RDH12 also metabolizes both all-trans- and cis-retinols, making it a dual-specificity enzyme .

Expression and Localization

RDH12 is predominantly expressed in the eye but is also found in other tissues such as the kidney, brain, skeletal muscle, and stomach . In situ hybridization studies in monkey and mouse retinas have shown that RDH12 is localized at the base of photoreceptor inner segments .

Clinical Significance

Mutations in the RDH12 gene are associated with Leber congenital amaurosis (LCA) and early-onset severe retinal dystrophy (EOSRD) . These conditions are characterized by severe vision loss at an early age. RDH12 mutations account for approximately 3.4% to 10.5% of LCA cases . The enzyme’s role in the visual cycle makes it a potential target for gene therapy aimed at treating inherited retinal diseases .

Research and Therapeutic Potential

Recombinant RDH12 has been expressed in insect cells as a membrane protein with enzymatic properties similar to those of the native enzyme . Studies have shown that RDH12 activity is inhibited by retinoic acids, recombinant cellular retinol-binding protein 1 (CRBP1), and cellular retinaldehyde-binding protein (CRALBP) . Understanding the enzyme’s function and regulation is crucial for developing therapeutic strategies for retinal diseases associated with RDH12 mutations.

In conclusion, Retinol Dehydrogenase 12 is a vital enzyme in the visual cycle and retinoid metabolism. Its role in retinal health and disease makes it an important target for research and potential therapeutic interventions.

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.