BEST1 Human

Bestrophin 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Electrophysiological Properties

BEST1 mediates calcium-dependent chloride currents in RPE cells, essential for:

  • Light Response: Maintains electrochemical gradients critical for photoreceptor function .

  • Volume Regulation: Swelling-activated chloride efflux in RPE cells, distinct from LRRC8A-dependent VRACs .

  • Bicarbonate Transport: High permeability to bicarbonate ions, influencing pH homeostasis .

Tissue-Specific Expression

TissueExpression LevelPrimary Localization
Retinal Pigment Epithelium (RPE)HighBasolateral plasma membrane
TestisVariableSperm cells (species-dependent)
TracheaModerateEpithelial cells
Source: *

Disease Associations

Over 250 BEST1 mutations cause bestrophinopathies, including:

  • Best Vitelliform Macular Dystrophy (BVMD): Autosomal dominant, characterized by vitelliform macular lesions.

  • Autosomal Recessive Bestrophinopathy (ARB): Severe visual impairment from biallelic mutations.

  • Retinitis Pigmentosa (RP): Rare association with BEST1 mutations .

Mutation Impact

MutationFunctional EffectClinical Outcome
P274R (ARB)Complete loss of channel activitySevere vision loss, retinal atrophy
I201T (BVMD)Partial reduction in Cl⁻ currentsMilder macular degeneration
Source: *

Species-Specific Variability

  • Humans vs. Mice: BEST1 expression is highest in RPE in humans but predominant in testis in mice .

  • Functional Rescue: Human and mouse BEST1 are functionally interchangeable in Xenopus oocytes .

Gene Therapy Strategies

ApproachMechanismOutcome
Viral Gene SupplementationDelivering wild-type BEST1 to RPE cellsRestores Cl⁻ currents in patient-derived iPSC-RPE
CRISPR/Cas9 KnockdownReducing endogenous mutant BEST1 expressionCombines with gene augmentation for gain-of-function mutations

Preclinical Models

  • iPSC-RPE Models: Patient-specific induced pluripotent stem cell-derived RPE cells recapitulate disease phenotypes and enable functional rescue studies .

  • Canine Models: Adeno-associated virus (AAV)-mediated gene therapy corrects retinal microdetachment in BEST1-mutant dogs .

Biochemical Properties

The recombinant BEST1 protein (PRO-1900) is produced in E. coli as a non-glycosylated, His-tagged polypeptide (residues 292–585, 36 kDa) . Key features:

  • Purity: >90% via SDS-PAGE.

  • Storage: Stable at -20°C; avoid freeze-thaw cycles.

  • Applications: Used in structural and functional studies, including lipid bilayer assays .

Emerging Clinical Spectrum

Recent studies expand BEST1-associated phenotypes:

  • Stargardt Disease Phenocopy: A novel in-frame deletion-insertion variant (c.233dupT) mimics Stargardt disease, highlighting diagnostic challenges .

  • Intrafamilial Diversity: Autosomal dominant mutations exhibit variable expressivity, necessitating comprehensive genetic analysis .

Research Gaps and Future Directions

  • Mechanistic Links: Elucidating how BEST1 mutations disrupt RPE-photoreceptor interactions.

  • Therapeutic Optimization: Developing AAV vectors with enhanced tropism for RPE cells.

  • Biomarkers: Identifying non-invasive markers for early disease detection.

Product Specs

Introduction
Bestrophin 1 (BEST1) belongs to the bestrophin gene family and functions as a calcium-activated chloride-ion channel in epithelial cells. This small gene family is known for its proteins with a highly conserved N-terminus containing 4-6 transmembrane domains. BEST1 exhibits high permeability to bicarbonate.
Description
Recombinant human BEST1 protein, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It comprises 317 amino acids (residues 292-585) and has a molecular weight of 36 kDa. The protein includes a 23 amino acid His-Tag fused at the N-terminus and undergoes purification through proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The BEST1 solution is provided at a concentration of 1 mg/ml and contains 20 mM Tris-HCl buffer (pH 8.0), 0.4 M Urea, and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), keep at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Minimize repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 90.0% using SDS-PAGE analysis.
Synonyms
Bestrophin-1, TU15B, Vitelliform macular dystrophy protein 2, BEST1, VMD2, ARB, BEST, BMD, RP50, TU15B, Bestrophin-1 isoform 1, Bestrophin 1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSEQLINPF GEDDDDFETN WIVDRNLQVS LLAVDEMHQD LPRMEPDMYW NKPEPQPPYT AASAQFRRAS FMGSTFNISL NKEEMEFQPN QEDEEDAHAG IIGRFLGLQS HDHHPPRANS RTKLLWPKRE SLLHEGLPKN HKAAKQNVRG QEDNKAWKLK AVDAFKSAPL YQRPGYYSAP QTPLSPTPMF FPLEPSAPSK LHSVTGIDTK DKSLKTVSSG AKKSFELLSE SDGALMEHPE VSQVRRKTVE FNLTDMPEIP ENHLKEPLEQ SPTNIHTTLK DHMDPYWALE NRDEAHS.

Q&A

What is the molecular function of BEST1 in human cells?

BEST1 (bestrophin-1) functions primarily as a calcium-activated and volume-regulated chloride channel. It is an integral membrane protein with critical roles in maintaining ionic homeostasis in epithelial cells. Current research indicates that BEST1 not only mediates chloride conductance but also interacts with calcium signaling pathways, particularly in the retinal pigment epithelium (RPE) . Methodologically, researchers have characterized these functions through patch-clamp electrophysiology and calcium imaging techniques in both native tissues and heterologous expression systems.

How does BEST1 protein expression differ across human tissues?

While BEST1 is predominantly expressed in the retinal pigment epithelium (RPE) in humans, significant expression has been documented in other tissues. In mice, the highest endogenous Best1 expression is observed in testis, which has proven valuable for experimental studies. Researchers investigating tissue-specific expression patterns should employ quantitative RT-PCR and immunohistochemistry with validated antibodies, as protein expression levels can vary substantially despite similar mRNA transcription levels . This differential expression pattern has important implications for understanding tissue-specific manifestations of BEST1 mutations.

What are the molecular mechanisms underlying Best vitelliform macular dystrophy?

Best vitelliform macular dystrophy (BVMD) results primarily from missense mutations in the BEST1 gene that lead to reduced protein stability and impaired chloride channel function. Research has demonstrated that these mutations cause:

  • Electrophysiological dysfunction of the retinal pigment epithelium

  • Accumulation of vitelliform (egg yolk-like) material between the RPE and photoreceptor layer

  • Progressive death of overlying photoreceptor cells

  • Irreversible central vision loss

The molecular pathophysiology involves protein misfolding, altered channel activity, and disrupted calcium homeostasis. Experimental approaches to study these mechanisms include patient-derived iPSC-RPE cells, which provide a human-relevant model system that maintains the genetic background of affected individuals .

How do different BEST1 mutations correlate with clinical phenotypes?

BEST1 mutations exhibit variable expressivity and incomplete penetrance, presenting significant challenges for genotype-phenotype correlation studies. The Y227N mutation, for example, causes autosomal dominant Best disease in humans but shows different tissue manifestations in mouse models . When designing correlation studies, researchers should:

  • Document detailed clinical characterization including electro-oculogram (EOG) findings

  • Perform comprehensive genetic analysis including potential modifier genes

  • Utilize multiple model systems to validate phenotypic effects

  • Consider protein stability measurements alongside functional studies

This multi-factorial approach helps address the complexity of BEST1-related disorders, which can present as autosomal dominant or recessive conditions with varying severity.

What are the advantages and limitations of iPSC-derived RPE models for studying BEST1 mutations?

iPSC-derived RPE cells offer significant advantages for BEST1 research:

Advantages:

  • Recapitulate patient-specific genetic background including potential modifier genes

  • Develop morphological and biochemical characteristics similar to native RPE

  • Allow direct testing of mutation-specific therapeutics

  • Enable high-throughput drug screening approaches

Limitations:

  • Variability between iPSC lines and differentiation protocols

  • Absence of interactions with choroid and photoreceptors

  • Time-consuming and resource-intensive generation process

  • Potential epigenetic differences from native RPE

Methodologically, researchers can enhance iPSC-RPE models by implementing BEST1 reporter constructs containing fluorescent markers (GFP or tdTomato) driven by the BEST1 promoter, which facilitates monitoring of BEST1 expression and response to potential therapeutics .

Why do BEST1 knock-in mouse models often fail to replicate human ocular phenotypes?

The divergence between human BEST1-associated macular dystrophy and mouse models presents a complex research question. Current evidence suggests several factors contribute to this discrepancy:

  • Anatomical differences: Mice lack a defined macula and have a rod-dominant retina compared to humans

  • Species-specific protein interactions: Different binding partners or regulatory mechanisms may exist

  • Compensatory mechanisms: Alternative chloride channels may mitigate BEST1 dysfunction in mice

  • Temporal factors: The relatively short lifespan of mice may preclude development of late-onset pathology

Despite these limitations, mouse models remain valuable for studying protein stability and exploring potential therapeutics. The Best1 Y227N knock-in mouse, while lacking ocular abnormalities, demonstrates significant protein degradation in testis with consequent effects on sperm motility and function, providing an in vivo system to evaluate therapies aimed at restoring protein stability .

How can BEST1 reporter constructs be designed for high-throughput drug screening?

Designing effective BEST1 reporter constructs requires careful consideration of regulatory elements and signal optimization. A methodological approach includes:

  • Promoter selection: Utilize the native BEST1 promoter to maintain physiological expression patterns and levels

  • UTR integration: Include both 5' and 3' untranslated regions to preserve post-transcriptional regulation

  • Reporter selection: Incorporate bright fluorescent proteins (GFP or tdTomato) with appropriate cellular localization signals

  • Vector optimization: Design constructs that are compatible with AAV delivery systems for efficient transduction

For high-throughput screening applications, these constructs should be stably integrated into iPSC-derived RPE cells from patients with Best disease. This enables quantitative assessment of drug effects on BEST1 expression through automated fluorescence microscopy or flow cytometry, significantly accelerating therapeutic discovery .

What approaches are most effective for measuring BEST1 protein stability in experimental systems?

Assessing BEST1 protein stability is crucial for understanding mutation effects and evaluating potential therapeutics. Recommended methodological approaches include:

  • Pulse-chase analysis: Monitor protein half-life through metabolic labeling with radioactive or non-radioactive amino acids

  • Cycloheximide chase assays: Block protein synthesis and measure degradation rates via western blotting

  • Ubiquitination assessment: Quantify mono- and poly-ubiquitinated BEST1 species as indicators of protein targeted for degradation

  • Proteasomal inhibition studies: Determine the contribution of proteasomal versus lysosomal degradation pathways

Research on the Y227N mutation demonstrates that these methods can effectively detect reduced protein stability even when mRNA transcription remains normal. In the Best1 Y227N mouse model, significantly reduced testicular BEST1 protein levels were observed despite similar wild-type and mutant transcript levels, highlighting the importance of post-translational mechanisms in disease pathology .

How can animal models of BEST1 mutations inform therapeutic development despite phenotypic differences?

Despite phenotypic differences between human and animal BEST1-associated diseases, animal models provide valuable platforms for therapeutic development through:

  • Mechanism validation: Confirming shared molecular mechanisms such as protein instability across species

  • Biomarker identification: Identifying measurable indicators of therapeutic efficacy (e.g., protein expression levels)

  • Pharmacological screening: Testing compounds that modulate protein stability and function in vivo

  • Dosing and safety assessments: Determining therapeutic windows and potential toxicities before human trials

The Best1 Y227N mouse model demonstrates reproducible pathology in testis tissue, providing a valuable system for evaluating therapies aimed at restoring protein stability. This approach has precedent in other BEST1 models, such as the canine model of autosomal recessive bestrophinopathy, which has successfully been used to demonstrate efficacy of both pharmacological intervention and gene augmentation therapy .

What are the considerations for developing BEST1 gene therapy approaches?

Development of gene-based therapeutics for BEST1-associated disorders requires addressing several critical factors:

  • Mutation mechanism: Different approaches are needed for haploinsufficiency (gene augmentation) versus dominant-negative mutations (gene editing or silencing)

  • Delivery system: AAV vectors show promise for RPE targeting, with AAV2 demonstrating efficacy in canine models

  • Expression regulation: Native promoter elements and UTRs should be incorporated to maintain physiological expression patterns

  • Safety monitoring: BEST1 reporter constructs can provide real-time assessment of expression levels and localization

For dominant mutations like Y227N, combination approaches may be necessary, potentially including both gene silencing of the mutant allele and supplementation with wild-type BEST1. These strategies can be evaluated in iPSC-derived RPE cells before advancing to animal models, creating a translational pipeline for therapy development .

How should researchers interpret contradictory findings between in vitro and in vivo BEST1 studies?

Contradictory findings between experimental systems are common in BEST1 research and require systematic approaches to resolution:

  • Context evaluation: Consider differences in cellular environment, protein interactions, and compensation mechanisms

  • Technical validation: Verify antibody specificity, assay sensitivity, and experimental conditions across systems

  • Species-specific factors: Acknowledge evolutionary differences in channel function and tissue expression

  • Temporal considerations: Account for developmental and aging effects on phenotype manifestation

The discrepancy between the severe ocular phenotype in humans with Y227N mutations and the lack of ocular pathology in corresponding mouse models exemplifies this challenge. Rather than undermining the validity of either system, these contradictions highlight the complex, context-dependent nature of BEST1 biology and emphasize the importance of multi-system approaches to research .

What statistical approaches are appropriate for analyzing BEST1 expression and functional data?

Given the complex and often variable nature of BEST1 data, robust statistical approaches are essential:

  • Paired designs: When comparing wild-type and mutant BEST1, use matched samples when possible

  • Normalization strategies: Account for total protein content and housekeeping gene expression

  • Multiple timepoints: Analyze kinetic data rather than single timepoints for stability studies

  • Power analysis: Ensure sufficient sample sizes to detect potentially subtle effects of mutations or treatments

For protein stability studies, quantitative approaches should include calculation of half-life values and degradation rates rather than simple presence/absence comparisons. Additionally, researchers should explicitly report both transcriptional and translational analyses to differentiate mRNA-level effects from protein-level effects .

How might BEST1 function in non-ocular tissues inform understanding of macular dystrophy mechanisms?

The significant expression and functional relevance of BEST1 in non-ocular tissues, particularly testis, offers novel perspectives on disease mechanisms:

  • The reproductive phenotype in Best1 Y227N mice suggests shared vulnerability to protein destabilization across tissues

  • Comparative analysis of tissue-specific interactomes may reveal protective factors present in mouse eyes but absent in human macula

  • Investigation of membrane trafficking and quality control mechanisms across tissues could identify targetable pathways for therapeutic intervention

  • Cross-tissue biomarker development might enable non-invasive monitoring of therapeutic efficacy

This integrated approach leverages the strength of each model system, using the reproductive phenotype in mice to develop and test stability-enhancing compounds that may benefit ocular disease in humans, despite the absence of an ocular phenotype in the mouse model.

What emerging technologies hold promise for advancing BEST1 research?

Several cutting-edge technologies are poised to accelerate BEST1 research:

  • CRISPR-based approaches: Prime editing and base editing technologies enable precise correction of point mutations without double-strand breaks

  • Organoid models: Retinal organoids containing functional RPE-photoreceptor interfaces provide three-dimensional disease models

  • Single-cell transcriptomics: Reveals cell-type specific responses to BEST1 mutations and potential compensatory mechanisms

  • Computational protein structure prediction: Tools like AlphaFold2 enable more accurate modeling of mutation effects on channel structure and function

Integration of these approaches with established experimental systems will likely provide deeper insights into BEST1 biology and accelerate therapeutic development for associated disorders.

Product Science Overview

Discovery and Function

The bestrophin family was first identified in humans by linking a mutation in the BEST1 gene with Best vitelliform macular dystrophy (BVMD), a degenerative retinal disease . Mutations in the BEST1 gene have been identified as the primary cause for at least five different degenerative retinal diseases .

Bestrophin-1 is predominantly expressed in the retinal pigment epithelium (RPE), a layer of cells in the eye that is vital for vision . When stimulated by calcium ions, Bestrophin-1 opens up to allow chloride ions to flow into and out of the cell . This chloride ion flow is essential for maintaining the health of the human eye and is believed to be involved in the eye’s response to light .

Structural Insights

Bestrophin-1 has been studied extensively to understand its structure and function. The protein’s structure has been resolved in complex with an Fab antibody fragment, chloride, and calcium . This structural information has provided insights into how Bestrophin-1 functions as a calcium-activated chloride channel.

Clinical Significance

Mutations in the BEST1 gene lead to various retinal degenerative diseases, which can result in progressive vision loss and even blindness . Over two hundred disease-causing mutations have been identified in the BEST1 gene . These mutations impair Bestrophin-1’s ability to mediate calcium-dependent chloride currents in the RPE, which is crucial for normal visual function .

Research and Therapeutic Approaches

Recent research has focused on understanding the molecular mechanisms of BEST1 mutations and their impact on Bestrophin-1 function . Using patient-specific induced pluripotent stem cell (iPSC)-based disease models, researchers have been able to analyze the effects of specific BEST1 mutations and explore potential therapeutic approaches . One promising approach involves viral gene supplementation to rescue the function of mutant Bestrophin-1 in patient-derived RPE cells .

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.