The His-tagged VSNL1 is expressed in E. coli and purified using immobilized metal affinity chromatography (IMAC). Stability is preserved in lyophilized form, with recommended storage at -20°C in PBS (pH 7.4) containing 10% glycerol .
VSNL1 modulates intracellular calcium signaling pathways, influencing:
Neuronal Plasticity: Enhances cAMP levels, promoting dendrite branching in hippocampal neurons .
Cancer Progression: Binds collagen type X alpha 1 (COL10A1) to drive colorectal cancer cell proliferation and invasion .
Endocrine Regulation: Regulates insulin secretion in pancreatic β-cells via cAMP-dependent mechanisms .
In Alzheimer’s disease, VSNL1 is linked to amyloid plaque formation and tau phosphorylation, with elevated CSF levels correlating with cognitive decline .
VSNL1 Human, His is widely used in:
Calcium Sensing: Binds Ca²⁺ via EF-hand domains, altering conformation to regulate adenylyl cyclase .
cAMP/cGMP Pathways: Overexpression increases cAMP, while knockdown reduces neurite outgrowth .
Disease-Associated Proteins: Associates with amyloid-β plaques in Alzheimer’s disease and KCNJ5 mutations in aldosterone-producing adenomas .
VSNL1 (visinin-like 1), also known as VILIP-1, is a member of the neuronal EF-hand Ca²⁺-sensor protein family. It plays crucial roles in neuronal function by influencing cytosolic cyclic adenosine monophosphate (cAMP) levels, cell migration, exocytotic processes, and neuronal differentiation . The protein was initially identified during developmental screening as upregulated in the chick visual cortex, showing enhanced expression during neuronal differentiation and axonal pathfinding .
Experimentally, VSNL1 has been demonstrated to regulate cAMP signaling in neurons - knockdown experiments show decreased cAMP levels and reduced dendrite branching, while overexpression produces opposite effects . This strongly suggests VSNL1's involvement in neuronal morphology development through cAMP-dependent pathways.
Methodologically, researchers study VSNL1 function using:
RNA interference (siRNA knockdown)
Overexpression studies
Pharmacological manipulation of associated pathways
Morphological analysis of neurite development
cAMP level quantification
When designing experiments with His-tagged VSNL1, researchers should consider:
Tag position effects: N-terminal versus C-terminal His-tags may differentially impact protein folding and function
Purification strategy: Optimizing imidazole concentration gradients during elution to maintain protein activity
Tag interference assessment: Comparing tagged and untagged protein activity in functional assays
Calcium-binding validation: Ensuring the His-tag doesn't interfere with VSNL1's calcium-sensing properties
Structure confirmation: Using circular dichroism or other structural analysis to verify proper folding
Multiple studies have demonstrated significant associations between VSNL1 single-nucleotide polymorphisms (SNPs) and schizophrenia. Both intergenic SNPs (rs394874 and rs424827) located on chromosome 2p24.3 and intragenic SNPs (rs650275, rs834504, rs1519477, rs12470654, rs1033297, and rs11677051) show significant association with schizophrenia (p=0.0080, p=0.0097, p=0.0178, p=0.0190, p=0.0210, p=0.0054, p=0.0384 and p=0.0053, respectively) .
Haplotype analysis has further confirmed these case-control results, revealing one haplotype (CGTCTAAG) with higher frequency in schizophrenia patients (27.7% vs 24.0% in controls, χ²=4.034, p=0.0446) and a protective haplotype (TACTCGGA) occurring less frequently in patients (23.7% vs 28.2% in controls, χ²=6.155, p=0.0131) .
Additionally, VSNL1 SNPs associate with performance on the Wisconsin Card Sorting Test, a sensitive measure of frontal cortical function that is typically impaired in schizophrenia . This suggests VSNL1's potential role in cognitive aspects of the disorder.
For researchers studying this association, methodological approaches should include:
Genotyping with high-quality platforms like Illumina BeadChip
Adequate sample sizes for detecting subtle genetic effects
Control for population stratification
Functional validation of SNP effects on VSNL1 expression/activity
Investigation of specific endophenotypes rather than broad diagnoses
VSNL1/VILIP-1 has been implicated in neurodegenerative processes, particularly Alzheimer's disease (AD), with evidence suggesting it may contribute to AD pathophysiology through altered expression or by affecting processes related to synapse or neuronal loss .
To effectively study VSNL1's role in neurodegeneration, researchers should implement a multifaceted approach:
Expression analysis:
Protein quantification:
Co-expression network analysis:
Functional studies:
Assess VSNL1's impact on calcium homeostasis in neuronal models
Investigate effects on synaptic function and neuronal survival
Examine interaction with known pathological processes (amyloid, tau)
When quantifying VSNL1 expression in human brain samples, researchers should consider several methodological factors to ensure accurate and reliable results:
Sample collection and preservation:
mRNA quantification:
Protein quantification:
Statistical analysis:
Apply linear regression models to control for technical covariates
Consider age stratification for co-expression analysis
Test for interactions between demographic and technical factors
A comprehensive experimental design might include:
Multiple brain regions (not limited to frontal cortex)
Balanced representation across demographic variables (79% male, 85% Caucasian in reference studies)
Detailed recording of technical parameters (mean pH: 6.7±0.3; mean RIN: 8.0±0.73)
To effectively analyze VSNL1's effects on neuronal morphology, researchers should employ sophisticated quantitative approaches that capture both gross and subtle changes in neuronal structure:
Cell culture models:
Molecular manipulation techniques:
Morphological analysis methods:
Sholl analysis: Quantify dendritic branching by counting intersections with concentric circles at increasing radii (20-100μm) from the soma
NeuronJ plugin with ImageJ software: Measure neurite number and length (specifically neurites longer than one cell diameter)
Time-course analysis: Examine neurons at different time points post-transfection (e.g., 48 hours)
Imaging and quantification:
Use consistent image acquisition parameters
Analyze sufficient numbers of neurons per condition
Blind the analysis process to prevent bias
Include appropriate statistical comparisons
This experimental design allows researchers to determine whether VSNL1 affects:
Total dendritic length
Branching complexity
Branch point distribution
Growth dynamics
Response to pharmacological manipulation of cAMP pathway
Contradictions in VSNL1 co-expression networks between age groups represent a complex research challenge requiring sophisticated approaches:
Age-stratified analysis:
Technical considerations:
Statistical approaches:
Use weighted gene co-expression network analysis (WGCNA)
Apply multivariate models to account for confounding variables
Implement network comparison statistics to quantify differences
Biological validation:
Confirm key relationships through independent techniques
Test whether identified differences are functionally relevant
Investigate if changes reflect altered cellular composition vs. molecular interactions
Experimental testing:
Manipulate VSNL1 expression in young vs. aged neuronal cultures
Assess impact on identified co-expression partners
Determine whether age-dependent effects are mechanistically different
Demographic Variable | Study Population Values |
---|---|
Age range (years) | 16-91 |
Sex distribution | Male (79%), Female (21%) |
Race distribution | Caucasian (85%), African-American (15%) |
Mean PMI (hours) | 17.2 ± 5.9 |
Mean pH | 6.7 ± 0.3 |
Mean RIN | 8.0 ± 0.73 |
This comprehensive approach can help determine whether apparent contradictions stem from biological reality or methodological differences, with important implications for understanding VSNL1's role in normal aging and pathological conditions.
To effectively capture VSNL1's calcium-dependent regulation of cAMP signaling, researchers should implement a comprehensive experimental design that integrates multiple approaches:
Real-time monitoring system:
FRET-based cAMP sensors for continuous measurement
Simultaneous calcium imaging using spectrally distinct indicators
Time-resolved correlation between calcium fluctuations and cAMP dynamics
Genetic manipulation approach:
Pharmacological dissection:
Downstream readouts:
Analytical considerations:
Dose-response relationships for calcium dependency
Temporal dynamics of calcium-cAMP coupling
Regional differences in neuronal responsiveness
Developmental stage influence on signaling efficiency
This multifaceted approach will reveal not just whether VSNL1 regulates cAMP levels in response to calcium, but also the quantitative parameters, spatiotemporal dynamics, and downstream consequences of this regulation in neuronal contexts.
Optimizing VSNL1 as a biomarker for neurological disorders requires addressing several methodological challenges:
Assay development considerations:
Clinical validation approach:
Define precise neurological conditions where VSNL1 has diagnostic value
Design studies with adequate statistical power for biomarker validation
Include longitudinal sampling to assess prognostic capabilities
Correlate with established biomarkers and clinical outcomes
Genetic influence assessment:
Technical challenges:
Ensure antibody specificity across VSNL1 isoforms
Address potential confounding factors (medications, comorbidities)
Establish age-appropriate reference ranges
Standardize sample collection and processing procedures
Analytical considerations:
Determine appropriate cutoff values for clinical decision-making
Evaluate biomarker performance in combination panels
Develop algorithms integrating VSNL1 with other markers
Assess cost-effectiveness of implementation
This systematic approach will help establish whether VSNL1 meets criteria for clinical utility as a biomarker while addressing the technical and biological complexities inherent in neurological disorder diagnostics.
To elucidate VSNL1's role in synaptic pathology related to Alzheimer's disease (AD), researchers should implement a comprehensive methodological framework:
Co-expression network analysis approach:
Cellular and subcellular localization studies:
Assess VSNL1 distribution at synapses using super-resolution microscopy
Examine colocalization with synaptic markers in control vs. AD tissue
Track calcium-dependent translocation in neuronal models
Compare subcellular distribution in different stages of AD pathology
Functional impact assessment:
Measure effects of VSNL1 manipulation on synaptic transmission
Evaluate influence on synaptic plasticity mechanisms
Assess interaction with amyloid-β and tau pathology
Quantify impact on dendritic spine morphology and density
Model systems approach:
Human post-mortem tissue with varying Braak stages
iPSC-derived neurons from AD patients vs. controls
Transgenic AD mouse models with VSNL1 overexpression/knockdown
Organoid models to capture complex cellular interactions
Intervention studies:
Test whether modulating VSNL1 function affects synaptic resilience
Assess potential for calcium signaling-based therapeutic approaches
Determine critical windows for intervention in disease progression
Evaluate combination approaches targeting multiple pathways
This multifaceted approach will help determine whether VSNL1 represents a contributor to AD-related synaptic dysfunction, a compensatory response, or a biomarker of the underlying pathological process, with important implications for therapeutic development.
VILIP-1 is expressed in neuronal pericaria, dendrites, and some axons . It is a full-length human protein expressed in Escherichia coli with a His tag at the C-terminus, which facilitates its purification and detection . The recombinant form of VILIP-1 is typically used in various biochemical assays, including SDS-PAGE and sELISA .
VILIP-1 has emerged as a potential biomarker for various neurological conditions. Its levels in blood and cerebrospinal fluid (CSF) are being studied for their correlation with neurodegenerative diseases . This makes VILIP-1 a valuable target for research in understanding and diagnosing these conditions.