A phase 1/2 trial (NCT03374657) evaluated AAV8-RLBP1, an adeno-associated virus vector delivering functional RLBP1 into the retina. Key outcomes:
In untreated eyes, only 2/12 showed dark adaptation recovery vs. 8–11/12 in treated eyes .
Mouse Models: Rlbp1-knockout mice exhibited delayed dark adaptation. Subretinal injection of scAAV8-pRLBP1-hRLBP1 restored visual function, with effects sustained for 1 year .
iPSC-Derived RPE Models: Patient-specific models of Bothnia dystrophy, RPA, and NFRCD replicated disease phenotypes (e.g., retinoid accumulation). Gene therapy restored CRALBP expression and retinoid metabolism .
Visual Cycle Dysfunction: RLBP1 deficiency disrupts 11-cis-retinal regeneration, causing photoreceptor stress and apoptosis .
Therapeutic Challenges: Variable disease progression (e.g., RPA vs. NFRCD) necessitates tailored endpoints. Dark adaptation kinetics are validated as primary efficacy metrics .
RLBP1 (Retinaldehyde Binding Protein 1), also known as CRALBP (Cellular Retinaldehyde-Binding Protein), is a 36-kD water-soluble protein that functions as a critical transporter of 11-cis-retinal or 11-cis-retinaldehyde, which serve as its physiologic ligands. The protein plays an essential role as a functional element of the visual cycle, facilitating the regeneration of visual pigments following light exposure . This regeneration process is crucial for maintaining continuous visual function, particularly under conditions requiring adaptation to changing light levels. In the retinal pigment epithelium (RPE) and Müller cells of the retina, RLBP1 facilitates the visual cycle by binding to 11-cis-retinoids during their processing, thus enabling proper photoreceptor function.
Human recombinant RLBP1 produced in E. coli is a single, non-glycosylated polypeptide chain containing 340 amino acids (positions 1-317 of the native sequence) with a molecular mass of 38.9 kDa. When engineered for research purposes, it is typically fused to a 23 amino acid His-tag at the N-terminus to facilitate purification . The complete amino acid sequence has been characterized and includes regions responsible for retinoid binding. The protein's tertiary structure enables it to form a specific binding pocket that accommodates the 11-cis-retinal molecule, positioning it appropriately for transport within the visual cycle pathway.
Mutations in the RLBP1 gene are associated with several inherited retinal dystrophies that demonstrate clinical heterogeneity despite originating from the same gene. These conditions include:
Retinitis punctata albescens (RPA)
Bothnia dystrophy (BD)
Severe rod-cone dystrophy
All these conditions fall under the broader category of autosomal recessive retinitis pigmentosa (RP). These disorders are characterized by progressive vision loss, with a distinctive feature being extremely prolonged dark adaptation (approximately six hours), reflecting the critical role of RLBP1 in the visual cycle . The severity and progression rate vary among patients, with some forms presenting more severe phenotypes than others.
For optimal experimental outcomes when working with recombinant RLBP1, researchers should follow these methodological guidelines:
Formulation requirements: The protein should be maintained in a sterile filtered clear solution containing 20mM Tris-HCl buffer (pH 8.0), 0.4M Urea, and 10% glycerol at a concentration of 1mg/ml .
Short-term storage: For experiments that will utilize the entire vial within 2-4 weeks, store at 4°C.
Long-term storage: For extended storage periods, keep the protein frozen at -20°C. To maintain stability during long-term storage, addition of a carrier protein (0.1% HSA or BSA) is strongly recommended .
Critical handling precautions: Avoid multiple freeze-thaw cycles as they significantly compromise protein integrity and activity. When planning experiments, aliquot the protein solution upon first thaw to minimize repeated freezing and thawing cycles .
Quality control: Prior to experimental use, verify protein integrity by SDS-PAGE analysis, with acceptable purity being greater than 90% .
These handling procedures ensure that experimental results reflect the true biological properties of RLBP1 rather than artifacts introduced by improper sample management.
Several cellular models have proven valuable for investigating RLBP1 function and associated disease mechanisms:
iPSC-derived RPE cells: Patient-specific induced pluripotent stem cell (iPSC)-derived retinal pigment epithelium provides an excellent model system for studying RLBP1-associated retinal dystrophies. This approach allows researchers to generate RPE cells from patients with different clinical subtypes of RLBP1-associated diseases, enabling correlation between genotype and cellular phenotype .
Fibroblast reprogramming methodology: Researchers have successfully cultured skin fibroblasts from patients with different RLBP1-associated clinical subtypes (Bothnia dystrophy, classical RPA, and severe NFRCD) and reprogrammed them into iPSCs. These cells can then be differentiated into RPE cells that recapitulate disease phenotypes .
Quality control for iPSC models: When establishing iPSC models, researchers should confirm Sendai vector clearance by RT-PCR, assess pluripotency by qPCR, verify trilineage differentiation through teratoma assays, and ensure genetic integrity via karyotype analyses. Crucially, the causative RLBP1 variants must be confirmed through appropriate genetic testing methods .
Transfected cell lines: For specific biochemical studies, COS-7 cells transfected with RLBP1 expression constructs provide a simplified system for investigating protein expression and basic functional characteristics .
These cellular models enable detailed investigation of molecular mechanisms underlying RLBP1-associated diseases and provide platforms for testing potential therapeutic approaches.
Recent research has unveiled the unexpected presence of two CRALBP isoforms encoded by the RLBP1 gene, a finding that significantly impacts our understanding of visual cycle biology and therapeutic development:
Discovery context: The presence of dual isoforms was unexpectedly identified during development of an AAV2/5 vector carrying RLBP1 under control of the CAG promoter for gene therapy applications. Western blot analysis consistently revealed two distinct bands of CRALBP protein despite using an intronless RLBP1 cassette .
Validation of dual isoforms: The presence of these two isoforms was confirmed under various experimental conditions, including different lysis buffers and both denaturing and native protein analysis methods. Both monoclonal and polyclonal anti-CRALBP antibodies detected the dual bands, confirming this was not an artifact of the detection method .
Evolutionary conservation: Importantly, these isoforms were found to be differentially expressed in both human and murine retinal tissue, suggesting evolutionary conservation and functional significance. This indicates the dual isoform expression is a native characteristic of RLBP1 expression rather than an artificial outcome of recombinant systems .
Mechanistic origin: Evidence suggests the smaller isoform arises from initiation at a second methionine codon within the RLBP1 mRNA. Both isoforms appear to play roles in the visual cycle, though their specific functional differences are still being characterized .
Therapeutic implications: This discovery has critical implications for gene therapy approaches, as it suggests that effective RLBP1 replacement strategies must replicate the native dual isoform expression pattern. Some previous gene therapy vectors that produced only a single CRALBP band may not fully recapitulate natural RLBP1 function .
This finding represents a paradigm shift in RLBP1 research, suggesting that previous studies may have overlooked the complexity of CRALBP expression and function in the visual cycle.
Research has revealed important correlations between specific RLBP1 genetic variants and clinical manifestations:
Bothnia dystrophy (BD): Associated with compound heterozygous mutations including c.333T>G (p.Tyr111*) in exon 5 and c.700C>T (p.Arg234Trp) in exon 8. This phenotype typically presents with moderate severity .
Classical retinitis punctata albescens (RPA): Linked to compound heterozygous mutations including c.25C>T (p.Arg9Cys) in exon 4 and c.333T>G in exon 5. This form generally exhibits intermediate severity .
Newfoundland rod-cone dystrophy (NFRCD): Associated with homozygous exon 7-9 deletion, resulting in a more severe clinical presentation with earlier onset and rapid progression .
Genotype-phenotype correlation: The functionality of RLBP1 iPSC-derived RPE correlates with clinical severity. RPE derived from patients with different mutations demonstrates varying degrees of functional impairment, with NFRCD-associated mutations showing the most severe cellular dysfunction .
Age-related progression: Cross-sectional studies have shown that visual acuity impairment (measured in logMAR) correlates with patient age, with poorer visual acuity associated with older age (correlation coefficient: 0.606 for right eye, -0.578 for left eye; P < 0.001). Similarly, Humphrey visual field mean deviation (HVF MD) values decrease with age (correlation coefficient: -0.672 for right eye, -0.654 for left eye; P < 0.001) .
Understanding these genotype-phenotype correlations is essential for patient stratification in clinical trials and for developing mutation-specific therapeutic approaches.
Based on natural history studies of RLBP1-associated retinal dystrophies, the following outcome measures have demonstrated reliability and clinical relevance:
Best-corrected visual acuity (BCVA): Measured in logMAR (ETDRS letter scores), this provides a standardized assessment of central visual function. In RLBP1 patients, BCVA ranges from -0.2 to 1.3 logMAR at baseline, with variability reflecting disease progression stages .
Dark adaptation (DA) kinetics: This is an exceptionally important and distinctive measure for RLBP1-associated diseases. Patients consistently show extremely prolonged dark adaptation rod recovery of approximately six hours at both tested wavelengths (450 and 632 nm), making this a particularly sensitive marker for RLBP1 dysfunction .
Contrast sensitivity (CS): RLBP1 patients typically demonstrate poor contrast sensitivity, making this a valuable functional assessment .
Humphrey visual fields (HVF): These tests reveal characteristic field defects that progress with disease advancement and correlate with patient age .
Central foveal thickness: Structural assessment via optical coherence tomography reveals prominent thinning in central foveal thickness in RLBP1 patients, providing an objective anatomical measure .
Full-field flicker electroretinograms: These provide objective measurements of retinal function and can track disease progression independently of patient-reported outcomes .
Importantly, five-year prospective natural history studies have demonstrated high test-retest repeatability across all anatomic and functional endpoints, confirming the reliability of these measures for clinical research . While cross-sectional analyses show correlations between functional measures and age, longitudinal studies suggest relatively slow progression over a five-year observation period, suggesting longer duration studies or more sensitive metrics may be needed for interventional clinical trials .
Designing effective gene therapy strategies for RLBP1-associated retinal diseases requires careful consideration of several factors:
These considerations highlight the complexity of developing gene therapies for RLBP1-associated diseases and the importance of thorough preclinical validation before clinical application.
Researchers investigating RLBP1 face several methodological challenges that require careful experimental design:
Detecting dual isoforms: Standard western blot analyses may not consistently resolve the two CRALBP isoforms, leading to misinterpretation of results. Researchers must optimize protein separation conditions (gel percentage, running time) and use appropriate antibodies. Under denaturing conditions, monoclonal antibodies may detect both bands, while under native conditions, polyclonal antibodies provide better resolution of the two isoforms .
Isoform misinterpretation: The presence of a second CRALBP band has previously been misattributed to protein degradation or proteolysis. Researchers should implement controls to distinguish native isoforms from degradation products, including fresh sample preparation and use of protease inhibitors .
Expression system considerations: When expressing recombinant RLBP1, researchers must consider that E. coli-based systems produce non-glycosylated protein, which may differ functionally from native RLBP1. For studies requiring post-translational modifications, mammalian expression systems may be more appropriate .
Stability challenges: RLBP1 protein exhibits limited stability, requiring careful storage conditions. For long-term storage, addition of carrier proteins (0.1% HSA or BSA) is recommended, and multiple freeze-thaw cycles must be avoided .
Functional assays: Assessing RLBP1 function requires specialized assays of retinoid binding and transport. The 11-cis-retinal binding capacity of RLBP1 is particularly challenging to measure due to the light-sensitive nature of retinoids, requiring all procedures to be conducted under dim red light conditions.
These methodological challenges highlight the need for standardized protocols and careful experimental design when studying RLBP1, particularly as research expands to investigate the newly discovered dual isoforms.
Several promising research directions are emerging for RLBP1-associated retinal diseases:
Isoform-specific functions: Investigating the distinct functional roles of the two CRALBP isoforms represents a critical research direction. Understanding how these isoforms differ in their binding properties, cellular localization, and contributions to the visual cycle could provide new insights into disease mechanisms and therapeutic targets .
Advanced gene therapy approaches: Developing next-generation gene therapy vectors that ensure balanced expression of both CRALBP isoforms and achieve broader retinal coverage could improve therapeutic outcomes. Exploration of alternative delivery methods or vector designs that enhance transduction efficiency while maintaining physiologically relevant expression patterns will be important .
Pharmacological approaches: Identifying small molecules that could stabilize mutant RLBP1 protein or enhance the function of remaining RLBP1 in patients with partial loss-of-function mutations could provide alternatives to gene therapy for some patients.
Combined therapy approaches: Investigating whether combining gene therapy with neuroprotective or anti-inflammatory approaches could enhance outcomes, particularly for patients with advanced disease where photoreceptor degeneration has already occurred.
Biomarker development: The identification of sensitive biomarkers that can detect disease progression over shorter time periods would facilitate clinical trials. Current natural history studies suggest relatively slow progression over five years, complicating the design of interventional studies with manageable durations .
Patient stratification strategies: Developing improved methods for patient stratification based on genetic variants, age of onset, and functional characteristics could enhance clinical trial design and potentially lead to more personalized therapeutic approaches .
These research directions reflect the evolving understanding of RLBP1 biology and the need for therapeutic approaches that address the complexity of RLBP1-associated retinal diseases.
The cellular retinol-binding protein (CRBP) was first discovered in 1973 from lung tissues by Bashor et al. There are three categories of cellular retinol-binding proteins: cellular retinol-binding protein, cellular retinoic acid-binding protein, and cellular retinaldehyde-binding protein (CRALBP) . CRALBP was first discovered in 1977 after it was purified from retina and retinal pigment epithelial cells .
The RLBP1 gene is located on human chromosome 15, specifically on 15q26.1 . The gene is composed of 8 exons and 7 introns . The protein itself contains 360 amino acid residues .
RLBP1 is primarily involved in the visual cycle, where it acts as an 11-cis-retinal acceptor. This protein facilitates the enzymatic isomerization of all 11-trans-retinal to 11-cis-retinal, which is crucial for the function of rod and cone cells in the retina . The protein carries 11-cis-retinaldehyde or 11-cis-retinal as its physiological ligands .
While RLBP1 is predominantly found in the retina and retinal pigment epithelial cells, it is also expressed in other cell types. It is majorly found in the iris, cornea, ciliary epithelium, Muller cells, the pineal gland, and oligodendrocytes of the optic nerve and brain . The function of RLBP1 in cells not related to the eyes is not yet fully understood .
Mutations in the RLBP1 gene have been associated with several retinal dystrophies, including Bothnia retinal dystrophy, fundus albipunctatus, and Newfoundland rod-cone dystrophy . These conditions are characterized by progressive vision loss and other visual impairments. Studies have shown that recombinant CRALBP containing certain mutations, such as the R150Q substitution, lacks the ability to bind 11-cis-retinaldehyde, leading to disruptions in retinal vitamin A metabolism .
Human recombinant RLBP1 is produced using recombinant DNA technology, which involves inserting the human RLBP1 gene into a suitable expression system, such as bacteria or yeast. This allows for the production of large quantities of the protein for research and therapeutic purposes. Recombinant RLBP1 is used in various studies to understand its function, structure, and role in retinal diseases.