Tight Junction Regulation: CLMP enhances transepithelial resistance in MDCK cells, critical for maintaining epithelial barrier integrity .
Smooth Muscle Coordination: Facilitates gap junction communication via Connexin43/45 in intestinal and ureteral smooth muscle cells, enabling synchronized peristalsis .
Intestinal Development: Essential for normal small intestine elongation and rotation. CLMP deficiency causes congenital short-bowel syndrome (CSBS) in humans, characterized by malrotation and functional obstructions .
Adipocyte Differentiation: Linked to adipocyte maturation and obesity development .
Synaptic Regulation: In hippocampal neurons, CLMP interacts with AMPA/kainate receptors, modulating excitatory synapse development and seizure susceptibility .
Neuroinflammation: Upregulated in brain endothelial cells during multiple sclerosis (MS), promoting leukocyte migration across the blood-brain barrier .
Mechanism: Loss of CLMP disrupts Connexin43/45 expression, impairing calcium signaling and smooth muscle coordination .
Multiple Sclerosis: CLMP is overexpressed in active MS lesions. CLMP⁺ B cells and monocytes are elevated in patients, and antibody blockade reduces leukocyte migration across brain barriers .
Obesity: CLMP expression in white adipose tissue correlates with adipocyte maturation .
Cognitive Deficits: One CSBS patient exhibited mild mental retardation, suggesting a role in brain development .
| Parameter | Details |
|---|---|
| Source | Escherichia coli |
| Purity | >90% (SDS-PAGE) |
| Storage | -20°C with 0.1% HSA/BSA; avoid freeze-thaw |
CLMP (coxsackie and adenovirus receptor–like membrane protein) is a cell adhesion molecule that has emerged as a potentially significant cell trafficking molecule involved in immune cell migration across brain barriers. Research indicates that CLMP plays a crucial role in the pathophysiology of neuroinflammatory conditions, particularly multiple sclerosis (MS). The significance of CLMP lies in its demonstrated upregulation on brain endothelial cells under inflammatory conditions and its potential as a therapeutic target for preventing immune cell infiltration into the central nervous system .
CLMP was identified through RNA sequencing of human brain endothelial cells (HBECs) and human meningeal endothelial cells (HMECs), where it showed significant differential expression under inflammatory conditions. This molecule performs cell-cell adhesion functions, making it particularly relevant in understanding immune cell trafficking mechanisms across the blood-brain barrier .
In patients with neuroinflammatory conditions such as multiple sclerosis, CLMP expression patterns show significant differences compared to healthy controls. Flow cytometry analysis of peripheral blood mononuclear cells (PBMCs) has demonstrated an increased frequency of CLMP+ B lymphocytes and monocytes in MS patients compared to healthy controls. Additionally, immunohistochemical analysis of brain tissue from MS patients shows strong upregulation of CLMP on the surface of CNS vessels in active MS lesions compared to control tissue .
At the cellular level, quantitative PCR and protein analyses have confirmed that CLMP is significantly upregulated in inflamed endothelial cells both in vitro and in situ in MS lesions. This differential expression pattern makes CLMP an important biomarker for understanding neuroinflammatory disease progression and a potential target for therapeutic interventions .
When designing experiments to study CLMP function in neuroinflammatory conditions, researchers should consider a multi-modal approach that combines in vitro, ex vivo, and in situ methods. Primary cultures of human brain endothelial cells (HBECs) and human meningeal endothelial cells (HMECs) exposed to inflammatory cytokines (TNFα and IFNγ) provide excellent in vitro models for studying CLMP upregulation under controlled inflammatory conditions .
For more complex functional studies, in vitro migration assays using HBECs and HMECs can effectively evaluate the role of CLMP in immune cell trafficking. These systems allow for the application of blocking antibodies against CLMP to assess migration inhibition potential. Additionally, flow cytometry analysis of PBMCs from patients with neuroinflammatory conditions compared to healthy controls offers valuable insights into CLMP expression on different immune cell subsets. For in situ validation, immunohistochemical analyses of post-mortem brain specimens from patients and controls remain the gold standard .
True experimental designs with appropriate controls are essential for establishing causality in CLMP function studies. As experimental research is best suited for explanatory research examining cause-effect relationships, careful manipulation of independent variables (such as inflammatory cytokine exposure) while controlling for extraneous variables is critical .
Accurate quantification of CLMP expression in human tissues requires careful consideration of multiple methodological approaches. A comprehensive protocol should include:
RNA-level quantification: RNA sequencing or quantitative real-time PCR with appropriate housekeeping gene controls should be employed. For RNA sequencing, ensure adequate sequencing depth (>20 million reads per sample) and appropriate bioinformatic analysis pipelines to identify differential expression .
Protein-level quantification: Western blot analysis with densitometry and immunofluorescence microscopy with standardized imaging parameters are recommended. Flow cytometry offers additional advantages for cellular resolution of CLMP expression on specific immune cell subsets .
Tissue-specific protocols: For CNS tissue, proper fixation methods that preserve both antigenicity and tissue architecture are essential. Paraffin-embedded sections typically require antigen retrieval steps optimized specifically for CLMP detection .
Control selection: Include both positive controls (tissues known to express CLMP) and negative controls (primary antibody omission) in each experimental batch to ensure specificity of staining .
Quantification methods: For immunohistochemistry, employ digital image analysis with standardized thresholding to minimize observer bias. For flow cytometry, consistent gating strategies and fluorescence minus one (FMO) controls are essential for accurate quantification .
Addressing data contradictions in CLMP research requires a systematic approach to identify the sources of discrepancies and resolve them methodologically. When faced with contradictory findings across different experimental platforms, researchers should:
Perform cross-platform validation: If gene expression analysis indicates CLMP upregulation but protein analysis does not confirm this finding, validate with a third method. For example, supplement RNA sequencing and Western blot with quantitative immunohistochemistry or flow cytometry .
Examine temporal dynamics: Contradictions may arise from different temporal sampling points. CLMP expression may vary during disease progression or inflammatory response time course. Implementing time-series experiments can help resolve these contradictions .
Consider post-transcriptional regulation: Discrepancies between mRNA and protein levels may indicate post-transcriptional regulation. Investigate miRNA regulation or protein degradation pathways that might affect CLMP expression .
Analyze cellular heterogeneity: Bulk tissue analysis may mask cell-specific changes. Single-cell approaches can resolve contradictions by identifying which specific cell populations are altering CLMP expression .
Apply statistical methods for contradiction resolution: When analyzing contradictory datasets, employ statistical methods specifically designed to handle contradictions in documents, such as those developed for detecting self-contradictions in long documents across multiple domains .
For comparative analyses between patient groups and controls:
Use non-parametric tests (Mann-Whitney U or Kruskal-Wallis) when data do not meet normality assumptions, which is common with flow cytometry data .
Apply FDR (False Discovery Rate) correction for multiple comparisons to control for Type I errors when examining CLMP across numerous immune cell subsets .
For correlation analyses:
Use Spearman's rank correlation for associations between CLMP expression and clinical parameters, as these relationships are often non-linear .
Consider partial correlation analyses to control for confounding variables such as treatment status or disease duration .
For predictive modeling:
Employ regression models with appropriate variable selection methods when using CLMP expression to predict disease outcomes .
Consider machine learning approaches for complex pattern recognition in multi-parameter datasets that include CLMP along with other biomarkers .
For longitudinal data:
Apply mixed-effects models to account for within-subject correlations in repeated measures designs studying CLMP expression over time .
Use time-series analysis methods when evaluating CLMP expression dynamics during disease progression or treatment response .
Validating CLMP's role in immune cell migration requires a comprehensive methodological approach that combines functional assays with specific inhibition or enhancement of CLMP activity. The most effective methods include:
In vitro transendothelial migration assays: Using primary human brain endothelial cells (HBECs) and meningeal endothelial cells (HMECs) grown on permeable supports, researchers can measure immune cell migration across these barriers in the presence or absence of CLMP-blocking antibodies. This approach has successfully demonstrated that blocking CLMP reduces immune cell migration across human brain and meningeal endothelial cells in vitro .
CRISPR/Cas9 gene editing: Generating CLMP-knockout endothelial cell lines allows for direct assessment of CLMP's necessity in facilitating immune cell migration. Comparison of migration rates across wild-type versus CLMP-knockout barriers provides strong functional evidence for CLMP's role .
Ex vivo tissue explant systems: Using fresh human brain tissue explants in culture systems permits evaluation of immune cell migration in a more complex microenvironment while maintaining the ability to manipulate CLMP function through antibody blocking or recombinant protein addition .
Live-cell imaging techniques: Time-lapse microscopy of fluorescently labeled immune cells interacting with endothelial monolayers allows visualization and quantification of the specific steps of transmigration that are affected by CLMP manipulation .
Flow chamber assays: These systems allow for controlled shear stress conditions that mimic blood flow, providing a more physiologically relevant context for studying CLMP's role in immune cell adhesion and migration under flow conditions .
Controlling for human error in CLMP experimental procedures requires implementation of the Human Error Assessment & Reduction Technique (HEART) methodology, which systematically identifies potential sources of error and establishes mitigating protocols. For CLMP research, consider the following approach:
Classify tasks by generic human unreliability: Categorize each experimental procedure (e.g., cell culture, Western blotting, flow cytometry) according to HEART's generic task types to estimate baseline error probabilities .
Identify error-producing conditions (EPCs): For CLMP experiments, common EPCs include complex procedures like multicolor flow cytometry, time pressure during time-sensitive protein extraction, and potential for misinterpretation of ambiguous immunohistochemistry staining patterns .
Implement error reduction strategies:
| Experimental Procedure | Potential Error Sources | Error Reduction Strategy | Estimated Error Reduction |
|---|---|---|---|
| CLMP antibody staining | Antibody concentration variability | Standardized preparation protocols with verification steps | 80-90% |
| Flow cytometry analysis | Inconsistent gating | Pre-established gating templates and dual-analyst verification | 75-85% |
| Western blot quantification | Subjectivity in band intensity assessment | Automated densitometry with standard curves | 70-80% |
| Migration assays | Variable cell counting | Automated cell counters with manual verification | 85-95% |
| RNA extraction timing | Degradation due to processing delays | Standardized timeline protocols with sample tracking | 80-90% |
Calculate the assessed impact for each error-producing condition using the formula: Assessed Impact = 1 + [(Multiplier - 1) × Assessed Proportion of Effect] .
Documentation and training: Maintain detailed records of procedure adjustments and ensure all researchers receive standardized training on error-prone steps in CLMP protocols .
This systematic approach not only reduces errors but also increases reproducibility of CLMP research findings across different laboratories and studies.
Interpreting contradictory findings regarding CLMP expression across different neuroinflammatory conditions requires a nuanced approach that considers multiple factors:
Disease heterogeneity: Neuroinflammatory conditions like MS have distinct pathological subtypes. Researchers should stratify analyses by disease subtype (e.g., relapsing-remitting MS vs. primary progressive MS) and lesion type (active vs. chronic) when comparing CLMP expression patterns .
Temporal dynamics: CLMP expression likely varies across disease stages. Contradictory findings may reflect different disease timepoints rather than true discrepancies. Longitudinal sampling and clear documentation of disease duration are essential for proper interpretation .
Regional specificity: Different CNS regions may exhibit distinct CLMP expression patterns. Carefully document and compare anatomical regions when evaluating contradictory findings between studies .
Methodological differences: Contradictions often arise from different detection methods or analytical approaches. Create standardized protocols that specify:
Meta-analytical approach: When faced with multiple contradictory studies, perform meta-analyses that account for methodological differences and weight findings based on study quality and sample size .
Functional validation: Resolve contradictions by moving beyond descriptive findings to functional studies that test CLMP's causal role in specific disease contexts through intervention studies (e.g., blocking antibodies, genetic manipulation) .
Translating CLMP research findings from laboratory discoveries to clinical applications requires careful consideration of several critical factors:
Target validation: Before developing CLMP-targeted therapies, researchers must thoroughly validate CLMP's role in human disease through multiple approaches:
Therapeutic approach selection: Based on CLMP's function in immune cell migration, potential therapeutic strategies include:
Biomarker development: CLMP expression may serve as a biomarker for:
Safety considerations: As CLMP likely plays physiological roles beyond neuroinflammation, researchers must assess:
Clinical trial design: When advancing to human studies, researchers should consider:
Advanced imaging technologies offer unprecedented opportunities to elucidate CLMP distribution and function in human tissues with enhanced spatial and temporal resolution. Researchers should consider implementing:
Super-resolution microscopy: Techniques such as STORM (Stochastic Optical Reconstruction Microscopy) and STED (Stimulated Emission Depletion) microscopy overcome the diffraction limit of conventional microscopy, enabling visualization of CLMP nanoscale organization at cell-cell contacts. This reveals clustering patterns that may be critical for function but invisible to conventional microscopy .
Intravital multiphoton microscopy: For animal models, this technique allows real-time visualization of CLMP-expressing cells in their native environment, providing insights into dynamic processes such as immune cell extravasation across the blood-brain barrier. Though not directly applicable to human subjects, these findings inform human tissue analysis .
Multiplexed immunofluorescence: Technologies like CODEX or Imaging Mass Cytometry enable simultaneous detection of CLMP alongside dozens of other markers in the same tissue section, revealing complex cellular interactions and microenvironment factors influencing CLMP function .
3D tissue clearing and light-sheet microscopy: These approaches allow imaging of CLMP distribution throughout intact tissue volumes rather than thin sections, providing comprehensive spatial information about CLMP expression relative to anatomical structures like blood vessels and inflammatory lesions .
Correlative light and electron microscopy (CLEM): This technique bridges the resolution gap between fluorescence microscopy and electron microscopy, enabling researchers to visualize CLMP subcellular localization at the ultrastructural level while maintaining the context of its distribution pattern at the cellular level .
Real-time biosensors: Developing CLMP-specific biosensors would allow monitoring of dynamic changes in CLMP conformation, activation, or interaction with binding partners in response to inflammatory stimuli .
Single-cell technologies are revolutionizing our understanding of cellular heterogeneity in health and disease. For CLMP research, several emerging methodological approaches offer powerful new insights:
Single-cell RNA sequencing (scRNA-seq): This technology reveals CLMP expression patterns across thousands of individual cells, identifying specific cell populations that up- or down-regulate CLMP under different conditions. When applied to peripheral blood and CNS samples from patients with neuroinflammatory diseases, scRNA-seq can reveal previously unrecognized cellular sources of CLMP .
Single-cell ATAC-seq: By profiling open chromatin regions in individual cells, this method reveals the epigenetic regulation of CLMP expression, identifying cell-specific transcription factor binding sites and regulatory elements that control CLMP transcription .
Spatial transcriptomics: Technologies like Slide-seq and MERFISH combine the throughput of scRNA-seq with spatial information, mapping CLMP expression in the context of tissue architecture. This is particularly valuable for understanding CLMP expression at the blood-brain barrier and in perivascular inflammatory infiltrates .
CyTOF (mass cytometry): This high-dimensional flow cytometry approach uses metal-conjugated antibodies to simultaneously measure CLMP alongside dozens of other proteins at the single-cell level, enabling comprehensive phenotyping of CLMP-expressing cells in human blood and CSF samples .
CRISPR screening at single-cell resolution: New approaches combining CRISPR perturbations with single-cell readouts enable systematic identification of genes regulating CLMP expression or function, providing insights into molecular pathways controlling CLMP in different cell types .
Live-cell imaging with single-molecule tracking: This approach allows visualization of individual CLMP molecules in living cells, revealing dynamics of CLMP movement, clustering, and interactions that are invisible to bulk measurements .
The CXADR gene encodes a protein that is a receptor for group B coxsackieviruses and subgroup C adenoviruses . The protein is composed of several isoforms due to alternative splicing, which allows it to perform diverse functions in different tissues . The primary structure of CXADR includes an extracellular domain, a single transmembrane region, and a cytoplasmic tail .
CXADR is a component of the epithelial apical junction complex and functions as a homophilic cell adhesion molecule . It is essential for the integrity of tight junctions and is involved in the transepithelial migration of leukocytes . CXADR interacts with JAML, a transmembrane protein on leukocytes, to mediate the activation of gamma-delta T-cells, which are crucial for tissue homeostasis and repair .
CXADR acts as a receptor for adenovirus type C and Coxsackievirus B1 to B6 . This interaction facilitates the entry of these viruses into host cells, making CXADR a critical factor in the pathogenesis of viral infections . The receptor’s role in viral entry has made it a target for therapeutic interventions aimed at preventing or treating infections caused by these viruses.
Recombinant human CXADR is produced using various expression systems to study its structure, function, and interactions . This recombinant protein is used in research to understand its role in cell adhesion, viral entry, and immune response. It is also utilized in the development of antiviral therapies and gene transfer techniques .