Recombinant Pongo pygmaeus Rab GDP dissociation inhibitor alpha (GDI1)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: We will prioritize shipping the format currently in stock. However, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless otherwise requested. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, which can serve as a guideline for your reconstitution.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. If you require a specific tag type, please inform us, and we will prioritize its development.
Synonyms
GDI1; RABGDIARab GDP dissociation inhibitor alpha; Rab GDI alpha; Guanosine diphosphate dissociation inhibitor 1; GDI-1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-447
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Pongo pygmaeus (Bornean orangutan)
Target Names
GDI1
Target Protein Sequence
MDEEYDVIVL GTGLTECILS GIMSVNGKKV LHMDRNPYYG GESSSITPLE ELYKRFQLLE GPPESMGRGR DWNVDLIPKF LMANGQLVKM LLYTEVTRYL DFKVVEGSFV YKGGKIYKVP STETEALASN LMGMFEKRRF RKFLVFVANF DENDPKTFEG VDPQTTSMRD VYRKFDLGQD VIDFTGHALA LYRTDDYLDQ PCLETINRIK LYSESLARYG KSPYLYPLYG LGELPQGFAR LSAIYGGTYM LNKPVDDIIM ENGKVVGVKS EGEVARCKQL ICDPSYIPDR VRKAGQVIRI ICILSHPIKN TNDANSCQII IPQNQVNRKS DIYVCMISYA HNVAAQGKYI AIASTTVETT DPEKEVEPAL ELLEPIDQKF VAISDLYEPI DDGCESQVFC SCSYDATTHF ETTCNDIKDI YKRMAGTAFD FENMKRKQND VFGEAEQ
Uniprot No.

Target Background

Function
This protein regulates the GDP/GTP exchange reaction of most Rab proteins. It achieves this by inhibiting GDP dissociation and subsequent GTP binding. Furthermore, it promotes the dissociation of GDP-bound Rab proteins from membranes, thereby inhibiting their activation. Specifically, it promotes the membrane dissociation of RAB1A, RAB3A, RAB5A, and RAB10.
Database Links

UniGene: Pab.17785

Protein Families
Rab GDI family
Subcellular Location
Cytoplasm. Golgi apparatus, trans-Golgi network.

Q&A

What is GDI1 and what is its primary molecular function in cellular systems?

GDI1 (Rab GDP dissociation inhibitor alpha) regulates the GDP/GTP exchange reaction of most Rab proteins by inhibiting GDP dissociation. It functions as a critical regulator of intracellular vesicular trafficking mechanisms by controlling the activity of Rab proteins. Specifically, GDI1 functions to extract inactive GDP-bound Rab proteins from membranes by binding and solubilizing the geranylgeranyl anchor (a post-translational modification at C-terminal cysteine residues which anchors Rabs to membranes) . This extraction process is essential for the recycling of Rab proteins between membranes, enabling their participation in multiple rounds of vesicle transport. In mammals, GDI1 is expressed primarily in the brain and is necessary for the control of endocytic and exocytic pathways in neurons and astrocytes through the spatial and temporal control of numerous Rab proteins .

How is the structural organization of GDI1 related to its functional domains?

The GDI1 protein contains four sequence conserved regions (SCRs): SCR1, SCR2, SCR3A, and SCR3B, which are common to all members of the Rab-GDI/CHM superfamily . These domains have distinct functional roles:

  • SCR1 and SCR3B together form a Rab-binding platform at the apex of the GDI1 structure

  • SCR3A contains a mobile effector loop (MEL) which constitutes a membrane receptor binding site and a helix flanking the lipid binding pocket

  • SCR2 contains the C-terminus-binding region (CBR), which forms an essential interaction with the C-terminus of Rab proteins

This structural organization enables GDI1 to interact with multiple partners, including Rab proteins, membrane receptors, and lipid components, coordinating the complex process of membrane trafficking with high specificity.

What evolutionary conservation patterns exist for GDI1 across primates and other mammals?

While the search results don't provide specific information about Pongo pygmaeus GDI1 conservation, research indicates that GDI1 is highly conserved across species due to its fundamental role in cellular function. The functional complementation experiments involving human GDI1 expression in yeast models demonstrate biological conservation across vast evolutionary distances . A humanized yeast model system has successfully shown that Homo sapiens GDI1 (HsGDI1) can complement a temperature-sensitive allele of the orthologous Saccharomyces cerevisiae gene Gdi1 (ScGdi1) . This cross-species functionality highlights the remarkable evolutionary conservation of GDI1's core functions, suggesting that Pongo pygmaeus GDI1 would likely share high sequence identity and functional similarity with human GDI1.

How does GDI1 mechanistically extract Rab proteins from membranes?

The extraction of Rab proteins from membranes by GDI1 involves a sophisticated molecular mechanism centered around recognition of the geranylgeranyl modification on Rab proteins. GDI1 contains a specialized lipid binding pocket that accommodates the geranylgeranyl group, which is typically embedded in the membrane . The process begins with GDI1 recognizing and binding to GDP-bound (inactive) Rab proteins through its Rab-binding platform formed by SCR1 and SCR3B regions . Following initial binding, the C-terminus-binding region (CBR) in SCR2 interacts with the C-terminus of the Rab protein where the geranylgeranyl modification resides . This interaction facilitates the extraction of the lipid anchor from the membrane.

The mobile effector loop (MEL) in the SCR3A region is critical for this process, as it serves as both a membrane receptor binding site and contains structural elements that flank the lipid binding pocket . This coordinated action between multiple domains enables GDI1 to overcome the energetic barrier of removing the hydrophobic geranylgeranyl moiety from the membrane environment, thereby solubilizing the Rab protein in the cytosol.

What is the role of GDI1 in neuronal function and how does it relate to intellectual disability?

GDI1 plays a crucial role in neuronal function, primarily through its regulation of vesicular trafficking pathways essential for neurotransmitter release and synaptic plasticity. In mammals, GDI1 is predominantly expressed in the brain, where it regulates endocytic and exocytic pathways in neurons and astrocytes . GDI1-null mouse models exhibit significant deficits in short- and long-term synaptic plasticity, leading to behavioral phenotypes including alterations in hippocampus-dependent forms of short-term memory, spatial working memory, and associative fear-related memory .

In humans, loss-of-function variants in GDI1 can cause non-syndromic intellectual disability (ID), characterized by cognitive impairment without other symptoms or physical anomalies . This condition follows an X-linked semi-dominant inheritance pattern, with hemizygous males being most severely affected while female carriers may show milder or no symptoms . Specific pathogenic variants such as Leu92Pro, Gly237Val, and Arg423Pro have been clinically reported to cause intellectual disability . The mechanistic link between GDI1 dysfunction and intellectual disability likely involves impaired synaptic vesicle recycling and neurotransmitter release, disrupting normal neuronal communication and synaptic plasticity required for learning and memory.

How do post-translational modifications affect GDI1 function and Rab interactions?

Post-translational modifications play a critical role in regulating GDI1 function, particularly in its interaction with Rab proteins. The most significant modification concerning GDI1-Rab interactions is not on GDI1 itself but on its Rab protein partners. The C-terminal geranylgeranylation of Rab proteins is essential for their membrane association and subsequent interaction with GDI1 . This post-translational modification is catalyzed by Rab geranylgeranyl transferase (Rab-GGTase) .

For GDI1 to function properly, it must recognize and bind to this geranylgeranyl moiety on Rab proteins. The specific structure of GDI1, particularly its lipid-binding pocket, is designed to accommodate this hydrophobic modification, allowing GDI1 to extract Rab proteins from membranes and maintain them in a soluble state in the cytosol . Without proper geranylgeranylation of Rab proteins, GDI1 would be unable to perform its extraction function effectively, highlighting the interdependence of these proteins and their modifications in membrane trafficking pathways.

What expression systems are optimal for producing recombinant Pongo pygmaeus GDI1?

While the search results don't provide specific information on Pongo pygmaeus GDI1 expression systems, the successful expression of human GDI1 in various systems offers valuable guidance. For recombinant GDI1 expression, researchers should consider the following systems based on experimental requirements:

The choice of expression system should be guided by the intended application of the recombinant protein, balancing considerations of yield, purity, post-translational modifications, and functional requirements.

How can researchers assess the functional activity of recombinant GDI1 in vitro?

Several robust methods can be employed to assess the functional activity of recombinant GDI1 in vitro:

  • Yeast Complementation Assays: As demonstrated in the search results, a humanized yeast model system can be used where the recombinant GDI1 complements a temperature-sensitive allele of the orthologous yeast Gdi1 gene (ScGdi1) . Growth at restrictive temperatures indicates functional GDI1. This approach has been validated for testing the deleteriousness of GDI1 variants and can be adapted for Pongo pygmaeus GDI1.

  • GDP Dissociation Inhibition Assays: These assays directly measure GDI1's ability to inhibit the dissociation of GDP from Rab proteins. Typically, this involves pre-loading a Rab protein with labeled GDP (fluorescent or radioactive), adding GDI1, and then measuring the rate of GDP release in the presence of excess unlabeled GDP. Functional GDI1 will slow this exchange rate.

  • Membrane Extraction Assays: These assays evaluate GDI1's ability to extract Rab proteins from membranes. Using preparations of membrane-associated Rab proteins (either from cells or reconstituted systems), researchers can add recombinant GDI1 and measure the transfer of Rab proteins from the membrane fraction to the soluble fraction using techniques like Western blotting or fluorescence-based approaches.

  • Binding Affinity Measurements: Techniques such as isothermal titration calorimetry (ITC), surface plasmon resonance (SPR), or microscale thermophoresis (MST) can be used to quantitatively measure the binding affinity between recombinant GDI1 and various Rab proteins, providing insights into the specificity and strength of these interactions.

What methods are available for studying GDI1-Rab protein interactions?

Researchers can employ multiple complementary approaches to study GDI1-Rab protein interactions:

  • Variant Effect Mapping: As described in the search results, variant effect mapping combined with multiplexed functional assays can assess the effects of amino acid substitutions in GDI1 . This approach involves creating a library of single-codon GDI1 variants, expressing them in yeast cells carrying a temperature-sensitive Gdi1 allele, and measuring their ability to complement the temperature sensitivity through competitive growth and deep sequencing .

  • Co-immunoprecipitation: This technique can be used to pull down GDI1-Rab complexes from cell lysates, confirming their interaction in a cellular context. It can be particularly useful when studying the interaction of GDI1 with different Rab family members or when assessing how mutations affect these interactions.

  • Fluorescence-based Interaction Assays: Methods such as Förster resonance energy transfer (FRET) or fluorescence correlation spectroscopy (FCS) can provide real-time information about GDI1-Rab interactions in solution or even in living cells when using fluorescently tagged proteins.

  • Structural Studies: X-ray crystallography, cryo-electron microscopy, or nuclear magnetic resonance (NMR) spectroscopy can provide detailed structural information about the GDI1-Rab complex, revealing the precise molecular contacts involved in this interaction.

  • In vivo Tracking: Advanced microscopy techniques can be used to track the movement of fluorescently labeled GDI1 and Rab proteins in living cells, providing insights into their dynamic interactions and the spatiotemporal regulation of membrane trafficking processes.

How can variant effect mapping be used to predict pathogenicity of GDI1 mutations?

Variant effect mapping represents a powerful approach for predicting the pathogenicity of GDI1 mutations by systematically assessing the functional consequences of amino acid substitutions. The search results describe a comprehensive methodology that can be applied to GDI1 :

  • Library Generation: A pooled mutagenesis approach, such as Precision Oligo-Pool based Code Alteration (POPCode), can be used to create a library of single-codon GDI1 variants .

  • Functional Assay: The variant library is transformed into yeast cells carrying a temperature-sensitive Gdi1 allele, and the cells are grown competitively at restrictive temperatures. Variants that retain function will support growth, while deleterious variants will not .

  • Deep Sequencing Analysis: Pre- and post-selection yeast populations are sequenced to determine the relative abundance of each variant, with changes in frequency indicating functional impact .

  • Fitness Score Calculation: Log ratios are calculated and rescaled to define a "fitness score" that represents the ability of each variant to complement the yeast Gdi1 allele. Scores can be normalized such that 1 represents fully functional protein and 0 represents complete loss of function .

  • Imputation Pipeline: Machine learning approaches like Gradient Boosted Tree can be used to impute scores for variants with limited experimental data, based on features like amino acid substitution matrix scores and predictions from computational methods .

The resulting variant effect map can discriminate pathogenic from benign variants with higher precision than computational methods alone . For example, known pathogenic variants like Arg423Pro, Leu92Pro, and Gly237Val, as well as functionally validated deleterious variants like Tyr39Val, Glu233Ser, Met250Tyr, and Thr248Pro, can be correctly identified as damaging .

What is the correlation between GDI1 variant fitness scores and clinical severity in intellectual disability cases?

The correlation between GDI1 variant fitness scores and clinical severity in intellectual disability cases represents an area of ongoing research. The search results provide some insights into this relationship, though the limited number of clinically characterized GDI1 variants makes comprehensive correlation challenging.

The variant effect mapping studies indicate that known pathogenic variants like Arg423Pro show reduced fitness scores (0.64 ± 0.24) . Similarly, other clinically reported variants associated with intellectual disability, such as Leu92Pro and Gly237Val, also demonstrate reduced function in complementation assays . This suggests that the degree of functional impairment measured by fitness scores may correlate with clinical manifestations.

The search results also describe the X-linked semi-dominant inheritance pattern of GDI1-associated intellectual disability, where hemizygous males are most severely affected while female carriers may show milder or no symptoms . This pattern adds another layer of complexity to genotype-phenotype correlations, as the clinical manifestation depends not only on the specific variant but also on the sex of the individual.

What bioinformatic tools and resources are most effective for analyzing novel GDI1 variants?

For comprehensive analysis of novel GDI1 variants, researchers should employ a multi-layered approach combining experimental data with computational predictions:

  • Variant Effect Maps: As described in the search results, experimentally derived variant effect maps provide a powerful resource for assessing the functional impact of GDI1 variants . These maps can discriminate pathogenic from benign variants with higher precision than computational methods alone.

  • Computational Prediction Tools: Several algorithms can provide initial assessments of variant pathogenicity:

    • PolyPhen-2: Predicts the possible impact of amino acid substitutions on protein structure and function

    • PROVEAN: Predicts whether protein sequence variation affects protein function

    • BLOSUM substitution matrices: Evaluate the likelihood of amino acid substitutions based on evolutionary conservation

  • Structural Analysis: Mapping variants onto the three-dimensional structure of GDI1 can provide insights into how they might affect protein function. Particularly important are variants in the sequence conserved regions (SCRs), which show significantly lower average fitness scores in functional assays .

  • Population Databases: Resources like gnomAD can help identify variants that are observed in the general population (particularly in hemizygous males for X-linked genes like GDI1), which are less likely to be highly pathogenic .

  • Machine Learning Integration: As demonstrated in the search results, machine learning approaches like Gradient Boosted Tree can integrate multiple sources of evidence to predict the effects of variants with limited experimental data .

How can recombinant GDI1 be used to develop therapeutic strategies for GDI1-associated disorders?

The development of therapeutic strategies for GDI1-associated disorders represents an important research frontier. Recombinant GDI1 can play a pivotal role in these efforts through several approaches:

What are the emerging techniques for studying GDI1 in the context of neuronal function?

Emerging technologies are expanding our ability to study GDI1's role in neuronal function:

  • CRISPR-based Approaches: CRISPR/Cas9 genome editing allows precise modification of GDI1 in various model systems, from cell lines to animals. This facilitates the creation of isogenic cell lines differing only in specific GDI1 variants, enabling direct comparison of their effects on neuronal function.

  • Human iPSC-derived Neurons: Induced pluripotent stem cells from patients with GDI1 mutations can be differentiated into neurons, providing a human cellular context for studying GDI1 function. These models preserve the genetic background of affected individuals and allow for personalized studies of disease mechanisms.

  • Advanced Imaging Techniques: Super-resolution microscopy and live-cell imaging approaches enable visualization of GDI1-mediated Rab trafficking with unprecedented spatial and temporal resolution. These techniques can reveal subtle defects in vesicle transport and synaptic function associated with GDI1 mutations.

  • Optogenetic and Chemogenetic Tools: These approaches allow temporal control over GDI1 or Rab protein activity in specific neuronal populations, helping to dissect their roles in different aspects of neuronal function and synaptic plasticity.

  • Single-cell Transcriptomics and Proteomics: These technologies can identify cell type-specific effects of GDI1 dysfunction and characterize the downstream molecular consequences in neuronal subtypes, potentially revealing unexplored aspects of GDI1 biology.

How do interspecies differences in GDI1 inform our understanding of its evolution and function?

Comparative analysis of GDI1 across species provides valuable insights into its evolution and fundamental functions:

  • Functional Conservation: The successful complementation of yeast Gdi1 by human GDI1 demonstrates remarkable functional conservation across vast evolutionary distances . This conservation suggests that the core mechanisms of GDI1 function in regulating Rab proteins have been preserved throughout eukaryotic evolution, highlighting their fundamental importance to cellular processes.

  • Structural Insights: Comparing GDI1 sequences from different species, including Pongo pygmaeus, can identify ultra-conserved regions that likely represent critical functional domains. The sequence conserved regions (SCRs) identified in GDI1 are likely to be preserved across primates and other mammals, reflecting their essential roles in protein function .

  • Specialized Adaptations: Species-specific variations in GDI1 may reflect adaptations to particular cellular or tissue environments. For instance, variations in neuronal-specific functions might be more pronounced in species with more complex nervous systems. Comparative studies of primate GDI1, including Pongo pygmaeus, could reveal adaptations related to primate brain evolution.

  • Expression Patterns: While GDI1 is primarily expressed in the brain in mammals , the specific expression patterns within brain regions and cell types may vary across species. These differences could inform our understanding of how GDI1 function has been adapted to support species-specific neuronal functions.

Understanding these interspecies differences not only illuminates the evolutionary history of GDI1 but also helps identify which aspects of its function are most critical for therapeutic targeting and which might be more amenable to species-specific manipulation in model organisms.

What are the common challenges in producing functional recombinant GDI1 and how can they be addressed?

Researchers often encounter several challenges when producing recombinant GDI1. Here are the most common issues and their solutions:

  • Protein Solubility: GDI1 may form inclusion bodies when expressed at high levels, particularly in bacterial systems. This can be addressed by:

    • Optimizing expression temperature (typically lowering to 16-18°C)

    • Using solubility-enhancing fusion tags (e.g., MBP, SUMO)

    • Employing eukaryotic expression systems like yeast, which have shown success with GDI1 expression

    • Adding low concentrations of non-ionic detergents during purification

  • Protein Stability: Purified GDI1 may show limited stability during storage. Researchers can improve stability by:

    • Including glycerol (10-20%) in storage buffers

    • Testing different buffer conditions (pH, salt concentration)

    • Adding reducing agents to prevent disulfide bond formation

    • Storing aliquoted protein at -80°C to avoid freeze-thaw cycles

  • Functional Activity: Recombinant GDI1 may show reduced activity compared to native protein. To address this:

    • Ensure careful removal of any fusion tags that might interfere with function

    • Verify proper folding using techniques like circular dichroism

    • Include stabilizing co-factors during purification and storage

    • Use functional assays like yeast complementation to confirm activity

  • Species-Specific Considerations: When working with Pongo pygmaeus GDI1, species-specific codon optimization may be necessary for expression in heterologous systems. Additionally, any unique post-translational modifications should be considered when choosing an expression system.

How can researchers troubleshoot inconsistent results in GDI1 functional assays?

When encountering inconsistent results in GDI1 functional assays, researchers should systematically address potential sources of variability:

  • Protein Quality Assessment:

    • Verify protein purity by SDS-PAGE and mass spectrometry

    • Confirm proper folding using circular dichroism or limited proteolysis

    • Check for batch-to-batch consistency using activity assays

    • Ensure the absence of protein aggregation using dynamic light scattering

  • Assay Optimization:

    • For yeast complementation assays, control for cell density and growth phase carefully

    • In biochemical assays, optimize buffer conditions, temperature, and incubation times

    • Use appropriate positive and negative controls, including known functional and non-functional GDI1 variants

    • Consider the impact of different Rab proteins, as GDI1 may show varying affinities for different Rab family members

  • Experimental Design Improvements:

    • Perform biological and technical replicates to assess variability

    • Use internal standards where possible

    • Implement blinding procedures to eliminate bias

    • For high-throughput approaches like variant effect mapping, ensure sufficient sequencing depth (e.g., ~2 million reads per position as described in the search results)

  • Data Analysis Refinement:

    • Apply appropriate statistical methods to account for experimental variability

    • Consider using machine learning approaches for imputation of missing data, as described in the search results

    • Implement quality control metrics, such as only considering variants with standard deviation < 0.3 for high-confidence measurements

How does Pongo pygmaeus GDI1 compare structurally and functionally to human GDI1?

While the search results don't provide specific information comparing Pongo pygmaeus and human GDI1, evolutionary relationships between great apes and humans suggest high conservation. Based on general patterns of protein conservation between orangutans and humans:

  • Sequence Similarity: Pongo pygmaeus GDI1 likely shares >95% amino acid identity with human GDI1, given the close evolutionary relationship between orangutans and humans. This high conservation would be expected particularly in functional domains like the sequence conserved regions (SCRs) that are crucial for GDI1 function .

  • Structural Conservation: The three-dimensional structure of Pongo pygmaeus GDI1 would be expected to be virtually identical to human GDI1, especially in regions involved in Rab binding (SCR1 and SCR3B), membrane interaction (SCR3A), and the C-terminus-binding region (SCR2) .

  • Functional Equivalence: Given the essential cellular role of GDI1 and the successful complementation of yeast Gdi1 by human GDI1 , it is highly likely that Pongo pygmaeus GDI1 would be functionally equivalent to human GDI1 in most assays, including Rab protein extraction from membranes and regulation of vesicular trafficking.

  • Species-Specific Differences: Any differences between Pongo pygmaeus and human GDI1 would most likely be located in regions less critical for core functions, possibly affecting subtle aspects of protein regulation, binding kinetics, or interaction with species-specific partners.

The high expected conservation between Pongo pygmaeus and human GDI1 makes recombinant orangutan GDI1 a valuable research tool, potentially offering insights into conserved mechanisms while also highlighting any species-specific adaptations in primate GDI1 function.

What insights can comparative studies of GDI1 across primates provide about neurological disorders?

Comparative studies of GDI1 across primates, including Pongo pygmaeus, can yield valuable insights into neurological disorders through several mechanisms:

  • Identification of Critical Functional Residues: By comparing GDI1 sequences across primates, researchers can identify ultra-conserved residues that have been maintained throughout primate evolution. These residues likely represent functionally critical sites where mutations would be poorly tolerated and potentially pathogenic. Variant effect mapping approaches as described in the search results can then be applied to these residues to confirm their functional importance .

  • Natural Variants as Functional Probes: Naturally occurring sequence differences between primate GDI1 proteins can serve as "natural experiments" that reveal which amino acid changes are tolerated without disrupting function. This information can help interpret human variants of uncertain significance identified in clinical settings.

  • Species-Specific Neuronal Adaptations: Primates exhibit varying degrees of cognitive complexity. Comparing GDI1 function in different primate species might reveal adaptations that correlate with cognitive capabilities, potentially highlighting mechanisms relevant to intellectual disability. The search results note that GDI1-null mouse models show deficits in various forms of memory, suggesting a conserved role in cognitive function .

  • Differential Vulnerability: Some species may show different sensitivities to GDI1 dysfunction. Identifying factors that confer resilience or vulnerability could reveal potential therapeutic targets or compensatory mechanisms that could be leveraged for treatment strategies.

  • Evolutionary Medicine Perspective: Understanding how GDI1 function has been shaped by evolutionary pressures in different primate lineages can provide insights into why certain mutations cause disease in humans. This evolutionary medicine perspective might reveal why GDI1 dysfunction specifically affects cognitive function rather than other cellular processes.

What statistical approaches are most appropriate for analyzing GDI1 variant effect data?

The analysis of GDI1 variant effect data requires robust statistical approaches to ensure reliable interpretation. Based on the methodologies described in the search results, researchers should consider:

How can researchers integrate structural, functional, and clinical data for comprehensive GDI1 variant interpretation?

Developing a comprehensive framework for GDI1 variant interpretation requires the integration of multiple data types:

  • Multi-layer Data Integration:

    • Structural Mapping: Plot variant fitness scores onto the three-dimensional structure of GDI1 to visualize patterns and identify functional clusters. The search results describe how positional fitness scores revealed lower average fitness in sequence-conserved regions, consistent with their functional importance .

    • Functional Domain Correlation: Compare variant effects across different functional domains (e.g., SCR1, SCR2, SCR3A, SCR3B) to identify domain-specific sensitivity patterns .

    • Clinical Correlation: When available, link variant fitness scores with clinical data from patients with GDI1 mutations to establish genotype-phenotype relationships.

  • Decision Framework Development:

    • Bayesian Integration: Develop Bayesian models that combine prior probabilities based on computational predictions with experimental evidence from variant effect mapping.

    • Weighted Scoring Systems: Create weighted scoring systems that prioritize direct experimental evidence while incorporating computational predictions and population frequency data.

    • Machine Learning Classification: Train machine learning algorithms on known benign and pathogenic variants to classify variants of uncertain significance.

  • Visualization Tools:

    • Heat Maps: Generate heat maps of variant effects across the protein sequence, highlighting regions of particular sensitivity to mutation.

    • Interactive 3D Models: Develop interactive structural models that allow researchers to visualize variant effects in the context of protein structure and interactions.

    • Network Graphs: Create interaction networks showing relationships between GDI1 variants, Rab proteins, and clinical outcomes.

  • Standardized Reporting Framework:

    • Evidence Levels: Establish standardized evidence levels for different types of data (e.g., computational prediction, in vitro functional assay, animal model, clinical observation).

    • Confidence Scores: Develop confidence scores that reflect the quantity and quality of evidence supporting a particular interpretation.

    • Consensus Guidelines: Adapt existing variant interpretation guidelines (e.g., ACMG/AMP guidelines) specifically for GDI1 variants.

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.