Role: VCAM1 facilitates monocyte adhesion to inflamed endothelium in early atherosclerotic lesions.
Models:
Mechanism: Mediates α4β1-dependent leukocyte adhesion under shear stress .
Aged Mice: Elevated endothelial VCAM1 correlates with microglial activation and reduced neurogenesis.
Interventions:
Hypoxia-Driven Angiogenesis: VCAM1 regulates JunB/CXCL1 signaling in retinal endothelial cells, promoting pathological neovascularization in oxygen-induced retinopathy (OIR) models .
Therapeutic Targeting: Intravitreal VCAM1 siRNA reduces retinal neovascularization by 40% .
Kit | Sensitivity | Range (pg/mL) | Sample Types | Intra-Assay CV% |
---|---|---|---|---|
Proteintech KE10038 | 1.5 pg/mL | 15.6–1000 | Serum, plasma, supernatants | 4.3–7.5% |
R&D Systems MVC00 | 0.87 ng/mL | 0.93–8.58 | Serum, plasma, supernatants | 4.3–7.7% |
Complete VCAM1 knockout in mice results in embryonic lethality, demonstrating the critical developmental importance of this adhesion molecule . This lethal phenotype occurs because VCAM1 plays essential roles in placental development and embryonic cell migration. In contrast, conditional or partial knockdowns exhibit more nuanced phenotypes that allow for postnatal analysis .
Specifically, ER intrabody-mediated VCAM1 knockdown mice (iER-VCAM1) are viable but show:
Suppressed surface expression of VCAM1 in bone marrow
Altered distribution of immature B-cells between blood and bone marrow
Significantly higher white blood cell and lymphocyte counts in peripheral blood
Additionally, oligodendrocyte-specific VCAM1 knockout leads to reduced myelin thickness in the central nervous system, similar to phenotypes observed in α4 integrin mutant mice, which is a neuronal VCAM1 ligand .
Conditional knockout approaches provide significant advantages over complete knockouts when studying VCAM1 function due to the embryonic lethality of constitutive VCAM1 deletion . These approaches include:
ER intrabody-mediated knockdown:
Cell-type specific knockouts:
Domain-specific modifications:
These approaches make it possible to study VCAM1 functions that would otherwise be impossible to investigate due to early embryonic death in complete knockout models.
Several validated methods exist for quantifying VCAM1 expression in mouse samples:
Quantitative ELISA:
Immunofluorescence staining:
Flow cytometry:
Western blotting:
When investigating VCAM1's role in hematopoietic stem cell homing, researchers should design experiments that address the complex cellular interactions involved:
Live imaging approaches:
Use transgenic reporter lines to visualize HSPCs and VCAM1+ cells simultaneously
Implement intravital microscopy to track cell-cell interactions in real-time
Quantify interaction duration and retention outcomes as demonstrated in zebrafish models, where HSPCs interacting with VCAM1+ macrophages showed distinctive retention patterns
Interaction analysis parameters:
Measure interaction duration (average ~30 minutes in zebrafish models)
Categorize retention types based on interaction outcomes:
Genetic manipulation strategies:
Integrative analysis approach:
The integration of these approaches allows for comprehensive understanding of how VCAM1-expressing cells guide HSPC homing and retention in hematopoietic niches.
ER intrabody-mediated knockdown represents an innovative approach for VCAM1 functional studies with distinct methodological considerations compared to traditional genetic knockouts:
Advantages of ER intrabody approach:
Experimental validation requirements:
Key technical considerations:
Ensure proper design of the intrabody construct with ER retention signal
Use appropriate controls including wildtype and non-activated transgenic mice (e.g., iER-STOP-VCAM1)
Account for potential differences between heterozygous and homozygous models
Consider that even low residual VCAM1 expression (2-8%) may be sufficient for some functions
Potential limitations:
This approach represents a valuable compromise between maintaining viability and achieving functional knockdown, particularly suited for studying proteins like VCAM1 where complete loss is lethal.
Measuring and interpreting myelin thickness changes in VCAM1-deficient oligodendrocytes requires specialized techniques and careful analysis:
Recommended measurement techniques:
Electron microscopy (EM) remains the gold standard for myelin thickness quantification
Calculate g-ratio (axon diameter/total fiber diameter) as the standard measure of myelin thickness
Implement semi-automated analysis software to reduce bias in measurements
Include multiple CNS regions to account for regional differences in myelination
Complementary approaches:
Immunohistochemistry for myelin basic protein (MBP) and other myelin components
Western blot analysis of myelin proteins to assess expression levels
In vitro myelination assays to study oligodendrocyte-neuron interactions directly
Functional assessments (e.g., electrophysiology) to evaluate conduction velocity
Experimental controls and comparisons:
Include α4 integrin mutant mice as comparative models since they show similar phenotypes
Analyze CD69 expression levels, as this is downregulated when VCAM1 is knocked down
Use conditional knockouts in oligodendrocytes to isolate cell-autonomous effects
Include age-matched controls as myelination is developmentally regulated
Interpretation guidelines:
Consider that reduced myelin thickness may reflect initiation defects rather than maintenance issues
Evaluate both the percentage of myelinated axons and the thickness of individual myelin sheaths
Examine different developmental timepoints to distinguish between delayed versus permanently impaired myelination
Analyze the relationship between VCAM1 expression levels and myelination phenotype severity
These methodological considerations ensure accurate assessment of how VCAM1 deficiency affects the complex process of myelination.
VCAM1 operates within a complex network of adhesion molecules in hematopoietic niches, with coordinated functions that enable precise regulation of HSPC behavior:
VCAM1-ITGA4 (α4 integrin) axis:
Forms the primary adhesive interaction for HSPC retention
ITGA4 on HSPCs binds to VCAM1 on niche cells
Mutations in either partner produce similar homing and retention defects
This axis is evolutionarily conserved from zebrafish to mammals
Essential for both initial rolling on endothelium and stable retention
Cellular sources of VCAM1:
Functional coordination mechanisms:
Sequential adhesion process where initial endothelial contacts precede macrophage interactions
VCAM1+ macrophages exhibit patrolling behavior, especially at the dorsal caudal venous plexus
HSPCs interact with these macrophages for approximately 30 minutes on average
Successful interactions lead to either capillary entry (Type I retention) or endothelial pocket formation (Type II retention)
Temporal and spatial regulation:
VCAM1 expression begins in specific anatomical regions (cranial region, heart, CHT) at defined developmental timepoints
Macrophages from the rostral blood island migrate to hematopoietic territories and become VCAM1+
These VCAM1+ macrophages establish "retention hotspots" within the homing microenvironment
Understanding this coordinated system is essential for developing interventions that target specific aspects of HSPC homing and retention.
The relationship between VCAM1 and CD69 in regulating myelination represents a novel connection between immune-related molecules and CNS development:
Expression relationship:
Functional evidence:
Knockdown of CD69 in mice demonstrates its role in myelination
VCAM1 knockout in oligodendrocytes leads to decreased myelin thickness
Similar myelin defects are observed in α4 integrin mutant mice
This indicates a signaling pathway where neuronal α4 integrin interacts with oligodendrocyte VCAM1, which regulates CD69 expression to control myelination
Mechanistic model:
VCAM1 in oligodendrocytes likely responds to neuronal cues through α4 integrin binding
This interaction triggers intracellular signaling that maintains CD69 expression
CD69 then contributes to the initiation of myelination through mechanisms that remain to be fully elucidated
The entire pathway represents a novel form of neuron-oligodendrocyte communication
Broader implications:
Reveals unexpected functions for immune-related molecules in CNS development
Highlights the potential for identifying new therapeutic targets for demyelinating disorders
Suggests that other immune-related molecules may have unrecognized roles in the CNS
Establishes VCAM1 as a regulator of both the initiation of myelination and its ongoing regulation through CD69
This relationship demonstrates how molecules traditionally associated with immune function can play crucial roles in neural development.
Comparing heterozygous and homozygous VCAM1 knockdown models reveals important insights about dosage effects and phenotypic thresholds:
These comparisons highlight the value of intrabody approaches in generating models with varying degrees of functional impairment, allowing for more nuanced study of VCAM1 biology.
Live imaging of VCAM1+ cells and their interactions with HSPCs requires specialized techniques to capture dynamic cellular behaviors:
Antibody-based live labeling:
Anti-VCAM1 647 antibody can effectively label VCAM1+ macrophages in vivo
Intravascular antibody injection provides stable staining for at least 8 hours
This approach produces cell distribution patterns nearly identical to those revealed by standard immunofluorescence
Critical validation: Quantitative analysis should confirm that antibody labeling does not affect definitive hematopoiesis (e.g., using myb WISH analysis)
Transgenic reporter systems:
Imaging parameters and quantification:
Implement confocal or two-photon microscopy for optimal resolution
Conduct time-lapse imaging with intervals appropriate for capturing cell movement (typically 1-5 minute intervals)
Record for extended periods (6-8 hours) to capture the full range of interaction events
Quantify interaction metrics including:
Analytical framework:
Track >100 VCAM1+ macrophage-HSPC interactions for statistical validity
Classify retention events according to established categories (Types 0, I, and II)
Compare wildtype patterns with those in genetic mutants to establish causality
Correlate interaction patterns with functional outcomes in hematopoiesis
These methods enable researchers to visualize and quantify the dynamic cellular interactions that underlie HSPC homing and retention in vascular niches.
Optimizing ELISA assays for mouse VCAM1 across different sample types requires specific technical adjustments:
Sample-specific dilution protocols:
Recovery rates across sample types:
Sample Type | Average Recovery | Range |
---|---|---|
Cell culture supernatants | 100% | 89-112% |
Serum | 99% | 80-118% |
EDTA plasma | 100% | 81-117% |
Assay performance metrics:
Sample | Intra-Assay Precision | Inter-Assay Precision | ||
---|---|---|---|---|
Mean (ng/mL) | CV (%) | Mean (ng/mL) | CV (%) | |
1 | 0.93 | 7.5 | 0.86 | 5.8 |
2 | 2.61 | 6.5 | 2.46 | 4.5 |
3 | 8.58 | 4.7 | 8.46 | 7.7 |
Technical optimization strategies:
Create standard curves using four-parameter logistic (4-PL) curve-fitting for optimal accuracy
For linearization, plot log of mouse VCAM1 concentrations versus log of optical density
Ensure that natural mouse VCAM1 samples produce curves parallel to the standards
Multiple samples across different dilutions to confirm linearity within the assay range
Apply appropriate quality control measures including duplicate readings and controls
Adhering to these optimization parameters ensures reliable quantification of mouse VCAM1 across diverse experimental contexts.
Characterizing the developmental timing of VCAM1 expression across tissues requires integrated analytical approaches:
Developmental expression mapping:
Utilize resources like the ZFIN database to identify temporal expression patterns
In zebrafish, VCAM1 (vcam1b) expression is detected in the cranial region, heart, and caudal hematopoietic tissue (CHT) at approximately 30 hours post-fertilization
Track expression from embryonic stages through adulthood across multiple tissues
Combined temporal and spatial analysis:
Implement whole-mount in situ hybridization (WISH) to visualize expression domains
Use reporter transgenic lines to track expression dynamics in live animals
Combine with lineage tracing to identify the developmental origin of VCAM1+ cells
For example, some VCAM1+ macrophages originate from the rostral blood island at 18 hours post-fertilization and subsequently migrate to hematopoietic territories
Quantitative expression analysis:
Functional correlation approaches:
Conditional genetic systems (e.g., inducible Cre) to manipulate VCAM1 at specific developmental timepoints
Temporal inhibition (e.g., antibody blocking) to determine critical periods for VCAM1 function
Rescue experiments to determine when VCAM1 re-expression can reverse phenotypes
Cross-species comparison to identify conserved developmental timing of expression and function
These analytical approaches provide comprehensive understanding of when and where VCAM1 expression occurs, offering insights into its diverse developmental functions.
VCAM1 mouse models provide valuable insights into inflammatory and autoimmune conditions through several research applications:
Regulated knockdown models for chronic inflammation:
ER intrabody-mediated VCAM1 knockdown allows creation of viable mice with partial VCAM1 deficiency
These models can replicate aspects of inflammatory conditions without the confounding effects of embryonic lethality
Variable knockdown efficiency between heterozygous and homozygous models permits studying dose-dependent inflammatory responses
B-cell development and autoimmunity connections:
VCAM1 knockdown mice exhibit altered B-cell distribution between bone marrow and peripheral blood
This mimics aspects of autoimmune conditions where abnormal B-cell trafficking contributes to pathology
Analysis of these models can help delineate how VCAM1-dependent B-cell retention contributes to immune tolerance versus autoimmunity
Neuroinflammatory disorder insights:
VCAM1's role in myelination identified through oligodendrocyte-specific knockout models
Reduced myelin thickness in these models connects immune adhesion molecules to neuronal function
This provides a mechanistic link for understanding demyelinating disorders with inflammatory components
The unexpected relationship between VCAM1 and CD69 in regulating myelination reveals potential new therapeutic targets
Quantitative biomarker applications:
These mouse models bridge fundamental VCAM1 biology with disease mechanisms, offering platforms for testing therapeutic approaches targeting VCAM1-dependent processes.
The discovery of VCAM1's role in myelination has significant implications for demyelinating disease research:
Novel mechanistic insights:
VCAM1 in oligodendrocytes regulates myelin thickness through interaction with neuronal α4 integrin
This represents a previously unrecognized neuron-oligodendrocyte communication pathway
VCAM1 functions both in the initiation of myelination and in its regulation through CD69
This pathway operates in parallel with other known myelination regulators
Therapeutic target potential:
Modulation of VCAM1-α4 integrin interactions could influence myelin formation
The VCAM1-CD69 axis represents a new potential intervention point for promoting remyelination
As these molecules also function in the immune system, targeting approaches must consider both nervous system and immune effects
The ability to regulate CD69 through VCAM1 suggests potential for indirect therapeutic approaches
Disease modeling applications:
VCAM1 knockout in oligodendrocytes creates hypomyelination phenotypes that can model aspects of demyelinating disorders
Combined with inflammatory stimuli, these models could recapitulate both structural and immunological aspects of diseases like multiple sclerosis
The parallel α4 integrin mutant models offer complementary systems for studying this pathway
Translational research directions:
Investigation of VCAM1 and CD69 expression in human demyelinating disease samples
Development of small molecules or biologics that modulate specific aspects of the VCAM1-CD69 pathway
Creation of remyelination screening platforms based on VCAM1-dependent myelination mechanisms
Integration with other known remyelination pathways to develop combination approaches
This unexpected role for VCAM1 highlights how molecules traditionally associated with immune function can play crucial roles in neural development and potentially in demyelinating pathologies.
Integrating quantitative VCAM1 analysis into preclinical drug screening requires standardized methodologies and interpretive frameworks:
Assay platform selection and optimization:
ELISA provides quantitative measurement of soluble VCAM1 with high sensitivity (detection range 0.313-20 ng/mL)
Flow cytometry enables cell-specific analysis of surface VCAM1 expression
Immunohistochemistry allows tissue-specific localization of VCAM1+ cells
Each platform should be validated using the performance metrics established in assay development
Screening application framework:
Primary screening: Use ELISA to identify compounds that modulate VCAM1 levels in cell culture or ex vivo systems
Secondary validation: Apply flow cytometry to confirm cell-specific effects on surface expression
Tertiary analysis: Implement tissue-specific immunostaining to verify in vivo effects
Functional correlation: Connect VCAM1 modulation to relevant biological outcomes (e.g., B-cell distribution, myelination)
Screening data interpretation guidelines:
Parameter | Normal Range | Interpretation of Deviations |
---|---|---|
Serum VCAM1 | 0.86-8.46 ng/mL | Elevation indicates inflammatory activation |
Surface VCAM1 (bone marrow) | Cell-type dependent | Reduction may affect HSPC retention |
VCAM1+ macrophages | Tissue-specific distribution | Altered patterns may disrupt hematopoietic niches |
Myelin thickness | Context-dependent | Reduction correlates with VCAM1 deficiency |
Integration with established screening cascades:
This integrated approach enables drug developers to systematically evaluate how candidate compounds affect VCAM1 biology across multiple dimensions, from protein expression to functional outcomes.
Several emerging technologies hold promise for expanding our understanding of VCAM1 beyond current mouse models:
Advanced genetic engineering approaches:
CRISPR-Cas9 base editing for creating point mutations that selectively disrupt specific VCAM1 domains
Conditional degron systems for rapid, inducible VCAM1 protein degradation
Optogenetic control of VCAM1 expression for temporal and spatial precision
Single-cell CRISPR screens to identify cell-specific VCAM1 interaction partners
High-resolution imaging technologies:
Expansion microscopy combined with super-resolution techniques to visualize VCAM1-mediated cellular interactions at nanoscale resolution
Light-sheet microscopy for whole-organ imaging of VCAM1+ cell distributions and dynamics
Correlative light and electron microscopy to connect VCAM1 localization with ultrastructural features
Intravital imaging with faster acquisition rates to capture rapid VCAM1-dependent adhesion events
Single-cell and spatial transcriptomics:
Integrated single-cell RNA/ATAC-seq to identify transcriptional networks regulated by VCAM1
Spatial transcriptomics to map VCAM1-expressing cells and their microenvironments
Cellular indexing of transcriptomes and epitopes (CITE-seq) to correlate VCAM1 protein levels with gene expression profiles
Trajectory analysis to understand how VCAM1 influences cell fate decisions
Translational model systems:
Human induced pluripotent stem cell (iPSC)-derived organoids to study VCAM1 in human tissue contexts
Humanized mouse models to investigate human-specific aspects of VCAM1 biology
Patient-derived xenografts to study VCAM1 in disease contexts
Cross-species comparative studies to identify evolutionarily conserved versus divergent VCAM1 functions
These technologies promise to provide higher resolution, more dynamic, and more physiologically relevant insights into VCAM1 biology.
The unexpected connection between VCAM1 and CD69 in myelination suggests broader neuroimmune interactions that warrant further investigation:
Potential integrated neuroimmune signaling networks:
VCAM1-CD69 may represent one axis in a larger network of immune molecules functioning in the CNS
Investigation of other adhesion molecules (ICAM-1, selectins) that function alongside VCAM1 in immune contexts
Examination of whether other lymphocyte activation markers besides CD69 have CNS functions
Integration with known neuroimmune signaling pathways involving cytokines and their receptors
Developmental versus pathological contexts:
Determine whether VCAM1-CD69 functions primarily in developmental myelination or also in remyelination after injury
Investigate if inflammatory stimuli modulate this axis differently than developmental signals
Examine how this pathway may be dysregulated in neuroinflammatory conditions
Compare oligodendrocyte precursor cells from different developmental origins for their dependence on this pathway
Cell-specific roles beyond oligodendrocytes:
Explore whether VCAM1-CD69 signaling operates in other neural cell types (neurons, astrocytes, microglia)
Investigate potential roles in synapse formation or elimination, which also involve immune-like mechanisms
Examine whether this axis influences blood-brain barrier function or integrity
Determine if these molecules mediate interactions between immune cells and neural cells during inflammation
Molecular mechanism investigations:
Identify the signaling pathways downstream of VCAM1 that regulate CD69 expression
Determine how CD69 influences the myelination program at the transcriptional level
Investigate potential ligands for CD69 in the CNS context
Explore whether VCAM1-CD69 interacts with other known regulators of myelination
This research direction could fundamentally reshape our understanding of neuroimmune interactions and reveal new therapeutic targets for both neurological and immunological disorders.
Current mouse model findings suggest several unexplored VCAM1 functions that warrant investigation across different organ systems:
Neurodevelopmental processes beyond myelination:
Recent discovery of VCAM1's role in myelination suggests potential involvement in other aspects of neural development
Investigation of VCAM1 in axon guidance, neuronal migration, or synaptogenesis
Exploration of potential roles in adult neurogenesis or neural repair mechanisms
Examination of VCAM1 functions in different brain regions and neural circuits
Stem cell niches beyond hematopoietic system:
VCAM1's critical role in HSPC homing suggests potential functions in other stem cell niches
Investigation of VCAM1 in mesenchymal stem cell, neural stem cell, or intestinal stem cell biology
Exploration of whether "usher cell" functions exist for other tissue-specific stem cells
Examination of VCAM1's role in maintaining stemness versus promoting differentiation
Tissue-specific immune surveillance:
VCAM1+ macrophages show specialized patrolling behavior in hematopoietic tissues
Investigation of similar specialized VCAM1+ immune populations in other organs
Exploration of tissue-resident macrophage subsets expressing VCAM1 and their unique functions
Examination of how VCAM1 expression is regulated in response to tissue-specific stressors
Metabolic regulation and adaptation:
VCAM1's involvement in cell retention and trafficking may extend to metabolic processes
Investigation of VCAM1 in adipose tissue biology, where immune cells regulate metabolic homeostasis
Exploration of potential roles in pancreatic islet function or liver regeneration
Examination of how VCAM1-dependent cellular interactions influence tissue-specific metabolic adaptations
Vascular Cell Adhesion Molecule 1 (VCAM-1), also known as CD106, is a protein that plays a crucial role in the adhesion of leukocytes to the vascular endothelium. It is a member of the immunoglobulin superfamily and is encoded by the VCAM1 gene. In mice, the VCAM1 gene is located on chromosome 3.
VCAM-1 is a type I membrane protein that contains six or seven immunoglobulin domains. It is expressed on both large and small blood vessels, but only after the endothelial cells are stimulated by cytokines. The gene product is a cell surface sialoglycoprotein, and it is alternatively spliced into two known RNA transcripts that encode different isoforms in humans .
The primary function of VCAM-1 is to mediate the adhesion of lymphocytes, monocytes, eosinophils, and basophils to the vascular endothelium. It also plays a role in leukocyte-endothelial cell signal transduction. VCAM-1 is upregulated in response to inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-1 (IL-1), which increase gene transcription and stabilize messenger RNA (mRNA) .
VCAM-1 is implicated in various inflammatory diseases, including atherosclerosis and rheumatoid arthritis. It is also involved in the development of macular fibrosis in neovascular age-related macular degeneration (nAMD). In nAMD, VCAM-1 contributes to the development of macular fibrosis by modulating macrophage functions, including migration and profibrotic polarization .
Recombinant VCAM-1 (Mouse) is produced using recombinant DNA technology, which involves inserting the VCAM1 gene into an expression system to produce the protein in vitro. This recombinant protein is used in various research applications, including studies on cell adhesion, inflammation, and signal transduction.