ICAM-1 Human

Intercellular Adhesion Molecule-1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of ICAM-1 Human

Intercellular Adhesion Molecule 1 (ICAM-1), also designated CD54, is a transmembrane glycoprotein encoded by the ICAM1 gene in humans. It belongs to the immunoglobulin superfamily and is constitutively expressed at low levels on endothelial cells, epithelial cells, and immune cells. ICAM-1 is dynamically upregulated by proinflammatory cytokines (e.g., TNF-α, IFN-γ, IL-1β) and plays a pivotal role in leukocyte adhesion, transendothelial migration (TEM), and immune synapse formation .

Biological Functions

ICAM-1 regulates diverse physiological and pathological processes:

Immune Regulation

  • Leukocyte trafficking: Mediates firm adhesion of leukocytes to endothelial cells via LFA-1/Mac-1 integrins .

  • T-cell activation: Enhances immune synapse formation between dendritic cells and T cells .

  • Phagocytosis: Expressed on macrophages to facilitate apoptotic cell clearance .

Inflammatory Signaling

  • Outside-in signaling: Cytoplasmic domain interactions with actin cytoskeleton modulate cell migration and ROS production .

  • Blood-brain barrier (BBB): Endothelial ICAM-1 facilitates leukocyte transmigration during neuroinflammation .

Viral Pathogenesis

  • Serves as a receptor for rhinoviruses (major common cold pathogens) .

Disease Associations

ConditionRole of ICAM-1Reference
CancerOverexpressed in thyroid, breast, and pancreatic cancers; correlates with metastasis
AtherosclerosisPromotes monocyte adhesion to vascular endothelium
NeuroinflammationBBB disruption via leukocyte infiltration in depression and schizophrenia

Therapeutic Targeting

ApproachMechanismStatus
CAR T-cell therapyICAM-1-specific CAR T cells eliminate advanced thyroid cancer in preclinical models
Monoclonal antibodiesR6.5 antibody blocks LFA-1/ICAM-1 interaction in autoimmune diseases
Small-molecule inhibitorsTarget ICAM-1/LFA-1 binding in inflammatory disorders

Plasmid for ICAM-1 Expression

The pCDM8-ICAM-1 plasmid (Addgene #8632) enables functional ICAM-1 expression in mammalian cells :

ParameterDetails
Vector backbonepCDM8
Insert size1,800 bp
Host strainMC1061/P3 (Ampicillin/Tetracycline resistance)
ApplicationsLeukocyte adhesion assays, viral entry studies, and endothelial cell modeling

Recent Research Advances

  1. Epithelial Repair: ICAM-1 promotes wound healing by coordinating epithelial cell migration and matrix remodeling .

  2. Tumor Microenvironment: High ICAM-1 expression in cancer-associated fibroblasts enhances immunosuppression and chemoresistance .

  3. Psychiatric Disorders: Elevated soluble ICAM-1 (sICAM-1) levels correlate with BBB dysfunction in major depression .

Challenges and Future Directions

  • Therapeutic limitations: Early ICAM-1 inhibitors failed in clinical trials due to paradoxical worsening of inflammation .

  • Precision targeting: Engineering ICAM-1 CAR T cells with tunable affinity to avoid off-tumor toxicity .

  • Biomarker potential: sICAM-1 as a prognostic marker for cancer metastasis and neuroinflammatory diseases .

Product Specs

Introduction
ICAM-1, also known as CD54, is a transmembrane glycoprotein found on various cell types, including immune and non-immune cells. It plays a crucial role in cell signaling, particularly in the immune response, by facilitating adhesion between cells like leukocytes and endothelial cells. This interaction is vital for leukocyte migration to inflammatory sites. ICAM-1 binds to specific integrins, which are proteins on leukocytes, enabling this adhesion. Notably, rhinoviruses utilize ICAM-1 as a receptor for cell entry. ICAM-1 expression is upregulated by inflammatory signals like IL-1 and TNFα, leading to increased leukocyte recruitment to areas of inflammation. Elevated levels of ICAM-1 are linked to conditions like subarachnoid hemorrhage, highlighting its role in inflammatory processes. A soluble form of ICAM-1 is detectable in plasma and serves as a marker for inflammation.
Description
This product consists of the recombinant human ICAM-1 protein, expressed in E. coli, with a hexahistidine tag at its amino terminus. The protein spans amino acids 28-483, totaling 455 amino acids. Purification is achieved through proprietary chromatographic methods.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
The ICAM-1 protein is provided at a concentration of 0.2 mg/ml in a buffer solution containing PBS, 50% glycerol, and 2mM imidazole.
Applications
ICAM-1 serves as a positive control in various immunological techniques including Western blotting, ELISA, and immunoprecipitation. However, it's important to note that the biological activity of this specific product remains untested.
Stability
For short-term storage (1-2 weeks), maintain the product at 4°C. For long-term storage, keep it frozen at -20°C. Repeated freeze-thaw cycles should be avoided to preserve protein integrity.
Purity
The purity of this product is greater than 95% as assessed by SDS-PAGE analysis.
Synonyms
Intercellular adhesion molecule 1, ICAM-1, Major group rhinovirus receptor, CD54 antigen, ICAM1, BB2, CD54, P3.58.
Source
Escherichia Coli.

Q&A

What is ICAM-1 and what are its primary functions in human physiology?

ICAM-1 (Intercellular Adhesion Molecule-1) is a cell surface glycoprotein expressed in vascular endothelial cells, immune cells, and glial cells of the nervous system . Its primary physiological functions include:

  • Supporting cell adhesion interactions critical for leukocyte extravasation (the movement of leukocytes from circulation to sites of inflammation)

  • Mediating immune cell migration and activation

  • Contributing to normal cell-to-cell communication

  • Serving as a receptor for 90% of human rhinovirus serotypes that bind to domain 1 of ICAM-1

  • Participating in inflammatory signaling pathways

In the human nervous system, ICAM-1 is expressed in endothelial cells residing in white and gray matter of the forebrain during early development . The molecule plays a complex role in both promoting necessary inflammatory responses and potentially contributing to pathological processes when dysregulated.

How does ICAM-1 expression change during normal human aging?

Research demonstrates significant age-related changes in ICAM-1 expression, particularly in the human brain. A study examining ICAM-1 immunoreactivity in the orbitofrontal cortex found:

  • The area fraction of ICAM-1 immunoreactivity was 120% higher (p < 0.0001) in older subjects (60-86 years) compared to younger subjects (27-54 years)

  • This increase was predominantly localized to extravascular ICAM-1 immunoreactivity associated with GFAP-IR astrocytes

  • A smaller but still age-dependent increase occurred in vascular ICAM-1 immunoreactivity

These findings suggest that normal aging is accompanied by a dramatic increase in extravascular ICAM-1 associated with astrocytes in the orbitofrontal cortex, which may contribute to enhanced risk for brain inflammatory processes during aging . This age-related increase parallels the increased expression of the astrocytic protein GFAP, which shows significant elevation in the human brain around age 60 and beyond .

What is the relationship between ICAM-1 and inflammatory processes in human tissues?

ICAM-1 is intimately involved in inflammatory processes across multiple human tissues. Key aspects of this relationship include:

  • ICAM-1 is upregulated in various inflammatory processes

  • In vascular endothelium, increased ICAM-1 expression facilitates leukocyte adhesion and extravasation to sites of inflammation

  • Astrocytes and microglia express ICAM-1 after brain trauma

  • Inflammatory processes in the brain increase with aging, corresponding with elevated ICAM-1 levels

  • In experimental models, mRNA levels for ICAM-1 and inflammatory cytokines increase more in aged subjects than in younger ones, paralleling increased GFAP immunostaining in astrocytes

The bidirectional relationship between ICAM-1 and inflammation means that while ICAM-1 can promote inflammatory processes, it may also serve as a barrier to further inflammation in certain contexts . This dual role makes ICAM-1 a complex target for therapeutic intervention in inflammatory conditions.

How can researchers differentiate between vascular and extravascular ICAM-1 immunoreactivity in human tissue samples?

Differentiating between vascular and extravascular ICAM-1 immunoreactivity requires precise methodological approaches:

  • Dual immunostaining technique: Use simultaneous immunostaining with antibodies to ICAM-1 and markers of vascular structures (such as CD31) or astrocytes (such as GFAP)

  • Quantification approach:

    • Collect frozen slabs of target brain regions and cut into 20 μm-thick sections

    • Mount sections onto gelatin-coated slides, vacuum dry, and store at -80°C

    • Immunostain evenly spaced sections (400 μm apart) with antibodies to ICAM-1

    • For co-localization studies, use fluorescent probes simultaneously to study GFAP and ICAM-1 immunoreactivities in single sections

  • Analysis protocol:

    • Quantify the total area fraction of ICAM-1 immunoreactivity

    • Separately measure the fraction of vascular and extravascular ICAM-1 immunoreactivity

    • Examine the association of extravascular ICAM-1 immunoreactivity to GFAP-IR astrocytes

  • Validation controls:

    • Include omission of primary or secondary antibodies to confirm absence of non-specific immunostaining

    • Suppress specific immunostaining by pre-incubating the primary antibody with recombinant human ICAM-1

    • Include the same number of sections from comparative groups in each immunostaining experiment to minimize variability

This methodological approach allows researchers to accurately distinguish and quantify ICAM-1 expression in different cellular compartments, essential for understanding its diverse roles in normal physiology and pathological conditions.

What are the domain-specific functions of human ICAM-1 in viral infections, particularly rhinovirus?

Human ICAM-1 contains multiple structural domains with specific functions, particularly in the context of viral infections:

  • Domain 1: Approximately 90% of human rhinovirus serotypes bind specifically to domain 1 of ICAM-1 as their cellular receptor . This domain-specific interaction makes it an attractive target for therapeutic development.

  • Domain-specific blocking: Research has demonstrated that antibodies specifically targeting domain 1 of human ICAM-1, such as the mouse anti-human ICAM-1 antibody 14C11, can:

    • Prevent entry of major groups of rhinoviruses (HRV16 and HRV14)

    • Reduce cellular inflammation and pro-inflammatory cytokine induction

    • Decrease viral load in vivo

    • Diminish Th2 cytokine/chemokine production in models of HRV-induced asthma exacerbation

  • Specificity for viral binding vs. physiological function: Interestingly, domain 1-specific antibodies like 14C11 can block viral entry without preventing physiologically important cell adhesion mediated by ICAM-1/LFA-1 interactions . This suggests that the epitope targeted by such antibodies is specific for viral entry mechanisms.

Understanding these domain-specific functions is crucial for developing targeted antiviral therapies that block pathogen entry while preserving normal cellular functions of ICAM-1, thus potentially avoiding unwanted side effects.

How does ICAM-1 expression in glial cells change during aging, and what are the implications for neuroinflammation?

Research on ICAM-1 expression in glial cells during aging reveals significant changes with important implications for neuroinflammation:

  • Age-related increases: Studies demonstrate a dramatic increase in extravascular ICAM-1 immunoreactivity associated with GFAP-immunoreactive astrocytes in the orbitofrontal cortex during normal aging

  • Cellular localization:

    • In all subjects, brain blood vessels show ICAM-1 immunoreactivity

    • Older subjects exhibit significantly more extravascular patches of ICAM-1 immunoreactivity

    • These extravascular ICAM-1 patches are in register with GFAP-IR astrocytes

  • Quantitative changes:

    • 120% higher area fraction of ICAM-1 immunoreactivity in older subjects (60-86 years) compared to younger subjects (27-54 years)

    • This increase is primarily localized to extravascular ICAM-1 associated with astrocytes

  • Implications for neuroinflammation:

    • May contribute to enhanced risk for brain inflammatory processes during aging

    • Could potentially represent a compensatory mechanism, as extravascular ICAM-1 might also function as a barrier to further inflammation

    • Parallels other age-related inflammatory changes, as inflammatory processes in the brain generally increase with aging

  • Association with neurodegenerative disorders: ICAM-1 increases are observed in age-related neurodegenerative diseases, suggesting a potential mechanistic link between age-related ICAM-1 changes and pathological processes

These findings highlight the complex role of ICAM-1 in age-related neuroinflammatory processes and suggest that targeting ICAM-1 might be a potential approach for modulating age-related inflammatory changes in the brain.

What are the most reliable techniques for quantifying ICAM-1 expression in human tissue samples?

Reliable quantification of ICAM-1 expression in human tissue samples requires systematic approaches:

  • Immunohistochemistry with digital image analysis:

    • Collect tissue sections of consistent thickness (e.g., 20 μm)

    • Immunostain with validated anti-ICAM-1 antibodies

    • Capture digital images using standardized acquisition parameters

    • Quantify the area fraction of ICAM-1 immunoreactivity using image analysis software

    • Distinguish between vascular and extravascular ICAM-1 staining patterns

  • Dual immunofluorescence for cellular co-localization:

    • Use fluorescent probes for ICAM-1 and cell-specific markers (e.g., GFAP for astrocytes)

    • Perform confocal microscopy to establish precise cellular localization

    • Quantify co-localization using specialized software

  • PCR-based quantification methods:

    • Extract RNA from fresh or properly preserved tissue samples

    • Perform quantitative real-time PCR using ICAM-1-specific primers

    • Normalize to appropriate housekeeping genes

    • Use this approach to complement protein-level analyses

  • Key methodological considerations:

    • Include appropriate negative controls (omission of primary or secondary antibodies)

    • Use competitive blocking with recombinant human ICAM-1 to confirm antibody specificity

    • Balance experimental groups in each immunostaining experiment to minimize inter-batch variability

    • Perform blinded analysis to prevent bias

  • Microarray or RNA-sequencing approaches:

    • For larger-scale expression studies, utilize microarray data as demonstrated in studies of ICAM-1 in lung cancer research

    • Apply bioinformatics tools like those used by researchers examining DEGs in lung cancer (DAVID, Cytoscape, Metascape, etc.)

These methodologies offer complementary approaches to quantifying ICAM-1 expression at both the protein and mRNA levels, providing robust data for understanding ICAM-1's role in various physiological and pathological contexts.

How can researchers effectively design experiments to study ICAM-1's role in human rhinovirus infection models?

Designing effective experiments to study ICAM-1's role in human rhinovirus infection requires careful consideration of multiple factors:

  • Selection of appropriate virus serotypes:

    • Include representatives from major rhinovirus groups that bind ICAM-1 (approximately 90% of identified serotypes bind domain 1 of human ICAM-1)

    • Consider using both HRV16 and HRV14 for broader coverage of major rhinovirus groups

  • In vitro experimental approaches:

    • Develop cell culture systems expressing human ICAM-1

    • Use domain-specific antibodies to block viral binding (e.g., antibody 14C11 that specifically binds domain 1 of human ICAM-1)

    • Assess viral entry, replication, and cellular responses

    • Evaluate antibody specificity by testing ability to prevent cell adhesion via human ICAM-1/LFA-1 interactions

  • In vivo model development:

    • Utilize appropriate animal models that express human ICAM-1 or humanized ICAM-1

    • Consider both topical and systemic administration of ICAM-1-targeting interventions

    • Measure multiple outcome parameters including:

      • Cellular inflammation

      • Pro-inflammatory cytokine induction

      • Virus load

      • For asthma exacerbation models, assess Th2 cytokine/chemokine production

  • Binding specificity evaluation:

    • Test antibodies against multiple targets including human ICAM-1, human ICAM-2, human ICAM-3, human VCAM-1, and mouse control proteins to ensure specificity

    • Characterize binding epitopes to distinguish viral-entry blocking from interference with physiological functions

  • Methodological controls:

    • Include isotype control antibodies

    • Perform dose-response studies

    • Evaluate potential toxicity of interventions

    • Compare effects against established antiviral compounds when available

These experimental design considerations enable researchers to comprehensively evaluate ICAM-1's role in rhinovirus infection and develop targeted interventions that specifically block viral entry while preserving ICAM-1's physiological functions.

What bioinformatics approaches are most useful for analyzing ICAM-1 expression data across different human tissues and disease states?

Effective bioinformatics approaches for analyzing ICAM-1 expression across tissues and disease states include:

  • Microarray and RNA-seq data analysis:

    • Download relevant datasets from repositories like NCBI Gene Expression Synthesis Database (NCBI-GEO)

    • Apply selection criteria for datasets such as:

      • Adequate sample size (greater than 50)

      • Appropriate sample types (disease tissue and matched controls)

      • Absence of confounding interventions

      • Specific focus on RNA expression differences

    • Process data using standardized tools like Morpheus for identifying differentially expressed genes (DEGs)

  • Functional annotation and pathway analysis:

    • Utilize multiple complementary tools including:

      • DAVID for functional annotation

      • Cytoscape for network visualization

      • Metascape for comprehensive biometric information extraction and visualization

      • UCSC Genome Browser for genomic context

      • cBioportal for cancer genomics data

      • BioCyc and Panther for pathway enrichment analysis

    • Focus on Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis with significance cutoff (p-value <0.01)

  • Protein-protein interaction network analysis:

    • Develop comprehensive DEG protein-protein interaction (PPI) networks using STRING database

    • Identify central genes and their interactions with ICAM-1

    • Perform module analysis to discover functional gene clusters

  • Integration of multi-omics data:

    • Combine transcriptomic data with:

      • Proteomic profiles

      • Epigenetic information

      • Clinical parameters

    • Use integrated approaches to identify biomarkers and therapeutic targets

  • Visualization and interpretation:

    • Generate enrichment score rankings [-log10(p-value)]

    • Create visual representations of gene relationships

    • Develop dynamic views linking genes to biological functions and pathways

These bioinformatics approaches provide powerful tools for analyzing ICAM-1 expression patterns across diverse tissues and disease states, enabling researchers to identify novel associations, potential biomarkers, and therapeutic targets involving ICAM-1.

How might targeting ICAM-1 with domain-specific antibodies present advantages over traditional antiviral approaches?

Domain-specific anti-ICAM-1 antibodies offer several distinct advantages over traditional antiviral approaches:

  • Receptor rather than virus targeting:

    • Traditional antivirals often target viral components that can rapidly mutate

    • ICAM-1 domain-specific antibodies target the host receptor, which is genetically stable

    • This approach potentially reduces the risk of developing viral resistance

  • Broad-spectrum activity:

    • Approximately 90% of rhinovirus serotypes (major group) use domain 1 of ICAM-1 as their cellular receptor

    • Domain-specific antibodies like 14C11 can block multiple rhinovirus serotypes (demonstrated with both HRV16 and HRV14)

    • This broad coverage addresses the challenge of developing antivirals against the 100+ identified rhinovirus serotypes

  • Reduction of downstream inflammation:

    • Studies show domain-specific antibodies not only prevent viral entry but also:

      • Reduce cellular inflammation

      • Decrease pro-inflammatory cytokine induction

      • Lower virus load in vivo

      • Diminish Th2 cytokine/chemokine production in models of rhinovirus-induced asthma exacerbation

  • Preservation of physiological functions:

    • Crucially, domain 1-specific antibodies like 14C11 can block viral entry without interfering with important physiological functions

    • For example, 14C11 does not prevent cell adhesion via human ICAM-1/LFA-1 interactions in vitro

    • This selective inhibition suggests the epitope targeted is specific for viral entry mechanisms

  • Potential for topical or systemic administration:

    • Research demonstrates efficacy with both topical and systemic administration of anti-ICAM-1 antibodies

    • This flexibility in delivery methods enhances clinical applicability

These advantages highlight the potential of domain-specific anti-ICAM-1 antibodies as a novel therapeutic approach for rhinovirus infections and associated conditions like asthma and COPD exacerbations, addressing significant unmet medical needs.

What are the implications of age-related changes in ICAM-1 expression for neurodegenerative disease research?

Age-related changes in ICAM-1 expression have significant implications for neurodegenerative disease research:

  • Potential mechanistic link to neurodegeneration:

    • ICAM-1 is increased in age-related neurodegenerative diseases

    • The 120% higher area fraction of ICAM-1 immunoreactivity in older subjects may create a neuroinflammatory environment that contributes to neurodegenerative processes

    • Age-related increases in extravascular ICAM-1 associated with astrocytes might represent a predisposing factor for neurodegenerative diseases

  • Regional vulnerability:

    • Studies showing age-related ICAM-1 changes in the orbitofrontal cortex align with this region's vulnerability in neurodegeneration

    • The orbitofrontal cortex shows propensity to age-related alterations in neuronal activity and volume reductions

    • These changes are particularly prominent in subjects with psychiatric and neurodegenerative disorders

  • Relationship to astrocytic changes:

    • Age-related increases in astrocytic GFAP immunoreactivity in the cerebral cortex parallel ICAM-1 changes

    • This suggests coordinated glial responses during aging that may influence neurodegeneration

    • Understanding this relationship could provide insights into the role of astrocyte-mediated inflammation in disease progression

  • Biomarker potential:

    • Changes in ICAM-1 expression might serve as biomarkers for early neurodegenerative changes

    • The spatial association between ICAM-1 immunoreactivity and GFAP-IR astrocytes could be used to develop imaging or fluid biomarkers

    • Age-specific reference ranges for ICAM-1 expression would be needed for accurate interpretation

  • Therapeutic implications:

    • Targeting age-related ICAM-1 changes might represent a novel approach to preventing or modifying neurodegenerative processes

    • Interventions could aim to normalize ICAM-1 expression or block its inflammatory effects

    • The complex role of ICAM-1 (potentially both promoting inflammation and serving as a barrier) requires careful consideration in therapeutic development

These implications highlight the importance of understanding age-related ICAM-1 changes for developing new approaches to diagnose, monitor, and treat neurodegenerative diseases, particularly those with significant inflammatory components.

How does ICAM-1 function as a marker for cancer stem cells, and what are the implications for cancer research?

ICAM-1's role as a cancer stem cell marker has important implications for cancer research:

  • Cancer stem cell identification:

    • Studies have shown that ICAM-1 functions as a marker of both human and mouse liver cancer stem cells

    • This finding provides a potential cellular target for identifying and isolating cancer stem cell populations

    • The expression pattern of ICAM-1 in cancer stem cells may differ from its expression in bulk tumor cells

  • Involvement in metastatic processes:

    • ICAM-1 is involved in the metastasis of liver cancer

    • This suggests that ICAM-1-expressing cancer stem cells may have enhanced metastatic potential

    • Understanding this mechanism could lead to strategies for preventing metastatic spread

  • Integration with other cancer pathways:

    • Research approaches integrating ICAM-1 with other cancer-related genes has identified important associations

    • For example, studies examining lung cancer have developed comprehensive DEG protein-protein interaction networks and module analyses to identify central genes including ICAM-1

    • These networks help place ICAM-1 in the broader context of cancer-related signaling pathways

  • Biomarker development:

    • The identification of ICAM-1 as a cancer stem cell marker supports its potential use as a biomarker

    • Bioinformatics approaches involving tools like DAVID, Cytoscape, Metascape, UCSC, cBioportal, BioCyc, and Panther can help identify the most relevant contexts for ICAM-1 as a biomarker

    • Combining ICAM-1 with other markers may improve specificity and sensitivity for cancer detection and monitoring

  • Therapeutic targeting strategies:

    • ICAM-1-positive cancer stem cells represent a potential therapeutic target

    • Approaches similar to those developed for blocking viral entry via ICAM-1 could potentially be adapted for targeting cancer stem cells

    • Domain-specific antibodies or other targeted therapies might selectively affect cancer stem cells while sparing normal stem cells

Understanding ICAM-1's function in cancer stem cells provides new opportunities for cancer diagnosis, prognosis, and treatment, particularly for addressing the challenges of metastasis and recurrence that are often attributed to cancer stem cell populations.

What are the most promising future research directions for understanding ICAM-1's role in human health and disease?

Several promising research directions emerge from current understanding of ICAM-1:

  • Domain-specific targeting for therapeutic development:

    • Further exploration of domain-specific antibodies that can block pathological ICAM-1 interactions while preserving physiological functions

    • Expansion of the approach demonstrated with rhinovirus to other pathogens that utilize ICAM-1

    • Development of small molecule inhibitors targeting specific ICAM-1 domains

  • Age-related changes and neuroinflammation:

    • More comprehensive mapping of age-related ICAM-1 changes across different brain regions

    • Longitudinal studies correlating ICAM-1 changes with cognitive and neurological outcomes

    • Investigation of interventions that might modulate age-related ICAM-1 increases to prevent neurodegenerative processes

  • Cancer stem cell targeting:

    • Further characterization of ICAM-1's role in cancer stem cell biology beyond liver cancer

    • Development of ICAM-1-targeted approaches for eliminating cancer stem cells

    • Integration of ICAM-1 targeting with other cancer therapies

  • Advanced bioinformatic integration:

    • Application of machine learning and artificial intelligence to better understand ICAM-1's position in complex biological networks

    • Multi-omics approaches integrating genomic, transcriptomic, proteomic, and metabolomic data

    • Systems biology frameworks to model ICAM-1's diverse roles

  • Precision medicine applications:

    • Development of ICAM-1-based biomarkers for personalized disease risk assessment

    • Tailoring of ICAM-1-targeted therapies based on individual expression patterns

    • Integration of ICAM-1 status into comprehensive patient profiles for personalized treatment approaches

Product Science Overview

Discovery and Structure

ICAM-1 was discovered in the 1980s and identified as a ligand for the β2 integrin lymphocyte function-associated antigen (LFA)-1 (CD11a/CD18) . Structurally, ICAM-1 is characterized by heavy glycosylation and consists of an amino-terminus extracellular domain, a single transmembrane domain, and a carboxy-terminus cytoplasmic domain . The extracellular domain is composed of multiple loops created by disulfide bridges, and the dominant secondary structure is the beta sheet .

Expression and Regulation

ICAM-1 is typically expressed on endothelial cells and cells of the immune system, such as leukocytes . Its expression can be induced by cytokines like interleukin-1 (IL-1) and tumor necrosis factor (TNF) . Upon cytokine stimulation, the concentration of ICAM-1 on cell membranes increases significantly .

Function and Role in Inflammation

ICAM-1 is essential for stabilizing cell-cell interactions and facilitating leukocyte endothelial transmigration . It acts as a ligand for LFA-1, a receptor found on leukocytes, enabling them to bind to endothelial cells and transmigrate into tissues . This process is crucial for the immune response, particularly in inflammation and immune surveillance .

Clinical Significance

ICAM-1 plays a central role in various inflammatory diseases, including ulcerative colitis and rheumatoid arthritis . It is also implicated in neuroinflammatory conditions such as Parkinson’s disease, where it influences ferroptosis, an iron-dependent form of cell death . The interaction between ICAM-1, glial cells, and T cells is a subject of ongoing research, with potential implications for novel therapeutic interventions .

Recombinant ICAM-1

Human recombinant ICAM-1 is produced using recombinant DNA technology, which involves inserting the ICAM1 gene into a suitable expression system, such as bacteria or mammalian cells. This allows for the large-scale production of ICAM-1 for research and therapeutic purposes. Recombinant ICAM-1 is used in various studies to understand its role in immune responses and to develop potential treatments for inflammatory and neurodegenerative diseases.

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 2024 Thebiotek. All Rights Reserved.