Endotoxic Shock: Icam1 tm1Jcgr mice showed 100% survival after lethal LPS doses (vs. 20% in wild-type), with reduced neutrophil infiltration .
Experimental Autoimmune Encephalomyelitis (EAE):
Asthma: ICAM-1-deficient mice exhibited 60% reduced ILC2s in lungs and remission of papain-induced airway inflammation .
Cerebral Malaria (ECM):
Atherosclerosis: Double-knockout (ApoE−/−/Icam1−/−) mice showed 54–77% smaller lesions in aortic roots compared to ApoE−/− controls .
Leukocyte Trafficking: ICAM-1 deficiency reduces monocyte homing to atherosclerotic lesions by >50% .
Signal Transduction: ICAM-1 ligation activates p56lyn and MAPK pathways, amplifying pro-inflammatory chemokines like RANTES .
Paradoxical Phenotypes: Isoform-specific deletions reveal context-dependent roles (e.g., protective in ECM but pathogenic in EAE) .
Intercellular adhesion molecule 1 (ICAM1) is a protein that belongs to the ICAM family of proteins. This protein serves as a ligand for integrin alpha-L/beta-2, a leukocyte adhesion peptide. During leukocyte migration, ICAM1 facilitates the formation of endothelial apical assembly cups by activating ARHGEF26/SGEF & RHOG.
Recombinant Mouse ICAM1, expressed in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain. It comprises 466 amino acids (28-485 a.a), resulting in a molecular weight of 51.2kDa. The protein includes an 8 amino acid His-tag at the C-terminus and is purified using proprietary chromatographic techniques.
The ICAM1 solution is provided at a concentration of 0.5mg/ml and is formulated in Phosphate-Buffered Saline (pH 7.4) with 10% glycerol.
The purity of the protein is determined to be greater than 95.0% as assessed by SDS-PAGE analysis.
Intercellular adhesion molecule 1, ICAM-1, Major group rhinovirus receptor, CD54 antigen, ICAM1, BB2, CD54, P3.58
Sf9, Baculovirus cells.
QVSIHPREAF LPQGGSVQVN CSSSCKEDLS LGLETQWLKD ELESGPNWKL FELSEIGEDS SPLCFENCGT VQSSASATIT VYSFPESVEL RPLPAWQQVG KDLTLRCHVD GGAPRTQLSA VLLRGEEILS RQPVGGHPKD PKEITFTVLA SRGDHGANFS CRTELDLRPQ GLALFSNVSE ARSLRTFDLP ATIPKLDTPD LLEVGTQQKL FCSLEGLFPA SEARIYLELG GQMPTQESTN SSDSVSATAL VEVTEEFDRT LPLRCVLELA DQILETQRTL TVYNFSAPVL TLSQLEVSEG SQVTVKCEAH SGSKVVLLSG VEPRPPTPQV QFTLNASSED HKRSFFCSAA LEVAGKFLFK NQTLELHVLY GPRLDETDCL GNWTWQEGSQ QTLKCQAWGN PSPKMTCRRK ADGALLPIGV VKSVKQEMNG TYVCHAFSSH GNVTRNVYLT VLYHSQNNVE HHHHHH
ICAM1 (CD54) is an 85-110 kDa single-chain type 1 integral membrane glycoprotein with an extracellular domain comprising five immunoglobulin superfamily repeats, a transmembrane region, and a cytoplasmic domain. In mice, ICAM1 functions as a ligand for leukocyte adhesion proteins, particularly LFA-1 (integrin alpha-L/beta-2) and Mac-1 (CD11b/CD18). During leukocyte trans-endothelial migration, ICAM1 engagement promotes the assembly of endothelial apical cups through ARHGEF26/SGEF and RHOG activation. It also plays crucial roles in inflammatory responses, leukocyte recruitment, and can serve as a receptor for certain pathogens .
ICAM1 expression varies significantly across mouse tissues, with detectable levels in multiple organs. Western blot analyses show expression in spleen, lung, kidney, and heart tissues, with varying band intensities at approximately 56 kDa. ICAM1 is expressed by activated endothelial cells and detected on epithelial cells, fibroblasts, chondrocytes, and various immune cells including B lymphocytes, T lymphocytes (low expression), monocytes, macrophages, dendritic cells, and neutrophils. Expression levels typically increase during inflammation. In the brain, ICAM1 expression increases after transient middle cerebral artery occlusion (tMCAO), particularly in endothelial cells .
Membrane-bound ICAM1 is the full-length protein anchored to the cell surface, mediating cell-cell adhesion and signaling. Soluble ICAM1 is a cleaved form that lacks the transmembrane and cytoplasmic domains and can be detected in circulation. In mouse models, soluble ICAM1 can be measured in serum, plasma, or cell culture medium using ELISA kits. While membrane-bound ICAM1 directly participates in cellular interactions, soluble ICAM1 serves as a biomarker for inflammatory conditions and may competitively inhibit cell adhesion mediated by the membrane-bound form .
Several complementary techniques should be employed for comprehensive ICAM1 detection:
Western blotting: Use monoclonal antibodies like 1A29 (ab171123) at 1:250 dilution with chemiluminescent detection. Predicted band size is 57 kDa, with observed band typically at 56 kDa.
Immunohistochemistry: For paraffin-embedded tissues, use 1:100 dilution of anti-ICAM1 antibody with biotin-conjugated secondary antibody and SA-HRP, followed by DAB detection and hematoxylin counterstaining.
Flow cytometry: Harvest cells, adjust to 1-5×10^6 cells/ml, fix with 2% paraformaldehyde, block with 2% BSA-PBS, and incubate with ICAM1 antibody at 1 μg/test, followed by fluorescent secondary antibody.
ELISA: Use commercial Mouse ICAM-1 ELISA kits for quantitative measurement in serum, plasma, or cell culture medium .
For effective ICAM1 knockout or chimeric mouse models:
Complete ICAM1 knockout: Generate or obtain established ICAM1 knockout mice with global deletion of the gene. Validate knockout by genotyping and protein expression analysis across multiple tissues.
Chimeric models: To distinguish between the roles of ICAM1 in different cell populations, create chimeric models through bone marrow transplantation:
M+/H+ (ICAM1 expressed on both marrow-derived and heart cells)
M+/H− (ICAM1 expressed only on marrow-derived cells)
M−/H+ (ICAM1 expressed only on heart and other tissues, not on marrow-derived cells)
For chimeric models, irradiate recipient mice (typically 9-11 Gy) and inject bone marrow cells from donor mice. Allow 6-8 weeks for reconstitution before experimental use. Verify chimerism by flow cytometry of peripheral blood cells .
Critical controls for ICAM1 functional studies include:
Genotype controls: Include wild-type, heterozygous, and homozygous knockout mice.
Antibody controls: For blocking experiments, include isotype control antibodies and verify antibody specificity using ICAM1 knockout tissues.
Leukocyte depletion controls: When studying ICAM1-dependent leukocyte functions, include cyclophosphamide pretreatment to decrease leukocyte count.
Stimulation controls: Include both baseline and stimulated conditions (e.g., LPS, cytokines) to observe ICAM1 upregulation.
Chimeric controls: For bone marrow transplantation studies, include all relevant combinations (M+/H+, M+/H−, M−/H+) to distinguish tissue-specific effects from bone marrow-derived cell effects .
Cardiac ICAM1 plays a critical role in mediating decreased left ventricular contractility during systemic inflammation. In endotoxemic mice, LPS injection significantly increases cardiac ICAM1 expression and decreases in vivo measures of left ventricular contractility (end-systolic elastance decreased by 58±4%, [dP/dt max]/EDV decreased by 60±6%). This mechanism is leukocyte-dependent, as demonstrated by:
Cyclophosphamide pretreatment to decrease leukocyte count prevents LPS-induced decreases in contractility
ICAM1 knockout mice show protection against LPS-induced cardiac dysfunction
Chimeric mouse experiments reveal that cardiac ICAM1 (not leukocyte ICAM1) is necessary for this effect
The mechanism involves ICAM1-mediated signaling that alters intracellular Ca²⁺ transients in cardiomyocytes, ultimately leading to decreased contractility. This provides a direct mechanistic link between cardiac inflammation and cardiac dysfunction in systemic inflammatory conditions .
For comprehensive assessment of ICAM1-dependent cardiac function:
In vivo hemodynamic measurements: Use a volume-conductance micromanometer catheter to measure:
End-systolic elastance (Ees)
Preload-adjusted maximal power
Preload recruitable stroke work
[dP/dt max]/EDV ratio
ICAM1 expression analysis:
Quantify cardiac ICAM1 mRNA by RT-PCR
Measure protein levels by Western blot
Visualize tissue expression by immunohistochemistry
Calcium handling assessment:
Isolate cardiomyocytes and measure Ca²⁺ transients using fluorescent indicators
Assess sarcomere shortening in conjunction with Ca²⁺ measurements
Inflammatory markers:
Quantify cardiac inflammatory cell infiltration by flow cytometry
Measure cardiac cytokine production
These measurements should be performed in both wild-type and ICAM1 knockout mice, with and without inflammatory stimuli, to establish ICAM1-dependent effects .
ICAM1 plays a crucial role in circulating tumor cell (CTC) cluster formation and subsequent metastasis through multiple mechanisms:
Homotypic clustering: ICAM1 mediates homophilic ICAM1-ICAM1 interactions between tumor cells, promoting CTC aggregation rather than single cell dissemination. This clustering increases metastatic efficiency by 20-100 times compared to single cells.
Trans-endothelial migration: ICAM1 facilitates heterotypic tumor-endothelial adhesion, enabling CTCs to adhere to and migrate through the endothelial barrier to form distant metastases.
Pathway activation: ICAM1 promotes metastasis by activating cellular pathways related to cell cycle progression and stemness properties.
Expression upregulation: In triple-negative breast cancer (TNBC) models, ICAM1 expression increases by 200-fold in lung metastases compared to primary tumors.
Metastatic dependency: Depletion of ICAM1 significantly abrogates lung colonization of TNBC cells by inhibiting homotypic tumor cell-tumor cell cluster formation.
This multifaceted role makes ICAM1 a potential therapeutic target for preventing metastasis initiation, particularly in TNBC .
To identify and characterize ICAM1-mediated interactions in tumor progression:
Single-cell RNA sequencing:
Compare primary tumors and metastatic lesions to identify differential ICAM1 expression
Analyze cellular heterogeneity and ICAM1-associated pathways
Protein-protein interaction analysis:
Use machine learning-based algorithms to identify ICAM1 regions responsible for homophilic and heterophilic interactions
Perform mutagenesis analyses to validate these interaction domains
In vivo metastasis models:
Develop ICAM1 knockdown or knockout tumor cell lines
Use patient-derived xenografts (PDXs) to assess ICAM1 expression changes during metastasis
Track CTC cluster formation in circulation using microfluidic devices
Therapeutic intervention studies:
Test ICAM1 blocking antibodies or peptides for inhibition of:
CTC cluster formation
Tumor cell transendothelial migration
Lung metastasis formation
These approaches provide complementary insights into the multifaceted roles of ICAM1 in tumor progression and metastasis .
Following transient middle cerebral artery occlusion (tMCAO) in mice, ICAM1 expression in the brain undergoes significant changes:
Temporal progression: ICAM1 expression increases in the ipsilateral (affected) hemisphere after tMCAO, with expression patterns changing over time during the acute and subacute phases of stroke.
Cellular localization: Endothelial cells show particularly prominent upregulation of ICAM1 expression in the ischemic region, serving as an indicator of the inflammatory response in the cerebral vasculature.
Regional differences: The expression is most pronounced in the core and penumbra of the ischemic lesion, with less expression in more distal regions.
Regulatory factors: The endothelial expression of ICAM1 appears to be regulated by inflammatory cytokines, particularly interleukin-1 (IL-1). Studies with IL-1 knockout mice show decreased ICAM1-immunopositive vessels after tMCAO compared to wild-type mice.
These changes in ICAM1 expression contribute to the pathophysiology of ischemic stroke by facilitating leukocyte adhesion and transmigration across the blood-brain barrier, potentially exacerbating neuronal cell death and neurodegeneration .
For optimal detection of cerebrovascular ICAM1 changes in mouse models:
Immunohistochemistry:
Use thin (5-7 μm) brain sections with specific anti-ICAM1 antibodies
Quantify vessel-associated ICAM1 staining in multiple fields
Compare ipsilateral (affected) versus contralateral hemispheres
Co-stain with endothelial markers (CD31/PECAM-1) for localization
Western blotting:
Separate analysis of ipsilateral and contralateral hemispheres
Include time course studies (6h, 12h, 24h, 3d, 7d post-stroke)
Use microdissection to isolate specific brain regions when possible
Flow cytometry:
Isolate brain microvascular endothelial cells using enzymatic digestion
Analyze ICAM1 expression levels on endothelial (CD31+) populations
Compare with other cell types to confirm endothelial specificity
In vivo imaging:
Use labeled anti-ICAM1 antibodies with intravital microscopy
Track dynamic changes in ICAM1 expression and leukocyte-endothelial interactions
These complementary approaches provide comprehensive spatial, temporal, and quantitative data on cerebrovascular ICAM1 expression changes following ischemic stroke .
To resolve inconsistent ICAM1 antibody staining in mouse tissues:
Fixation optimization:
For immunohistochemistry: Test different fixatives (4% paraformaldehyde vs. methanol vs. acetone)
For flow cytometry: Compare 2% paraformaldehyde fixation with alternative methods
For Western blotting: Ensure consistent sample preparation with appropriate protease inhibitors
Antigen retrieval methods:
Compare heat-induced epitope retrieval (citrate buffer pH 6.0 vs. EDTA buffer pH 9.0)
Evaluate enzymatic retrieval methods (proteinase K, trypsin)
Optimize retrieval times and temperatures based on tissue type
Antibody validation:
Test multiple anti-ICAM1 antibodies (e.g., clone 1A29 vs. clone 15.2)
Include ICAM1 knockout tissues as negative controls
Use positive control tissues with known ICAM1 expression (activated endothelium, inflamed tissues)
Detection system optimization:
For IHC: Compare biotin-streptavidin-HRP systems with polymer detection systems
For IF: Test different fluorophores and mounting media to minimize autofluorescence
For Western blot: Evaluate different secondary antibodies and chemiluminescent substrates
Blocking protocol refinement:
When analyzing ICAM1 expression by flow cytometry in mouse models:
Sample preparation:
Consistent cell concentration (1-5×10^6 cells/ml)
Optimal fixation (2% paraformaldehyde)
Thorough washing to remove fixative
Effective blocking (2% BSA-PBS for 30 minutes)
Antibody selection and titration:
Use validated anti-mouse ICAM1 antibodies (e.g., clone 1A29)
Determine optimal antibody concentration (typically 1 μg/test)
Select appropriate conjugated secondary antibodies (e.g., Dylight 488)
Include isotype controls matched to primary antibody
Multiparameter analysis:
Include cell-specific markers to identify ICAM1 on different populations
Compensate properly when using multiple fluorophores
Use viability dye to exclude dead cells
Controls and validation:
Unstained, single-color, and fluorescence minus one (FMO) controls
Include samples from ICAM1 knockout mice when available
Use positive controls (LPS-stimulated cells) and negative controls
Analysis considerations:
ICAM1 can be targeted therapeutically in mouse disease models through multiple approaches:
Antibody-based strategies:
Blocking antibodies against ICAM1 to inhibit leukocyte adhesion and transmigration
Antibody-drug conjugates to deliver therapeutic payloads to ICAM1-expressing cells
Bi-specific antibodies targeting ICAM1 and disease-specific antigens
Peptide inhibitors:
Design peptides targeting homophilic ICAM1-ICAM1 interaction domains
Develop LFA-1 mimetic peptides that block ICAM1-LFA-1 interactions
Create peptide-based nanoparticles for targeted drug delivery
Small molecule inhibitors:
Screen for compounds that disrupt ICAM1 protein-protein interactions
Develop inhibitors of signaling pathways that upregulate ICAM1 expression
Genetic approaches:
Use siRNA or antisense oligonucleotides to downregulate ICAM1 expression
Apply CRISPR/Cas9 for tissue-specific ICAM1 knockout
Employ adenoviral vectors encoding ICAM1 antagonists (e.g., soluble ICAM1)
These approaches have shown efficacy in various disease models including cancer metastasis, where blocking ICAM1 interactions significantly inhibits CTC cluster formation, tumor cell transendothelial migration, and lung metastasis .
Emerging technologies revolutionizing ICAM1 research in mouse models include:
Single-cell multi-omics:
Integrated single-cell RNA-seq and proteomics to correlate ICAM1 transcription with protein expression
Spatial transcriptomics to map ICAM1 expression patterns in tissue microenvironments
Single-cell ATAC-seq to identify regulatory elements controlling ICAM1 expression
Advanced imaging techniques:
Intravital microscopy with long-term imaging capabilities to track ICAM1-dependent cell interactions
Super-resolution microscopy to visualize ICAM1 clustering and distribution at the nanoscale
Correlative light and electron microscopy to link ICAM1 expression with ultrastructural features
Biomimetic models:
Organ-on-chip technologies incorporating ICAM1-expressing endothelial cells
3D bioprinting of tissues with controlled ICAM1 expression patterns
Microfluidic devices that recapitulate ICAM1-dependent processes like leukocyte extravasation
Computational approaches:
Machine learning algorithms to identify ICAM1 interaction domains and predict binding partners
Systems biology modeling of ICAM1-dependent inflammatory networks
AI-assisted image analysis for quantifying ICAM1 expression and function
These technologies provide unprecedented resolution and throughput for studying ICAM1 biology, enabling researchers to address previously intractable questions about ICAM1's role in health and disease .
ICAM-1 is composed of 532 amino acids and has a molecular mass ranging from 90 to 110 kDa . The protein is heavily glycosylated and its extracellular domain is composed of multiple loops created by disulfide bridges . This structure allows ICAM-1 to bind to integrins, specifically CD11a/CD18 (LFA-1) and CD11b/CD18 (Mac-1) . These interactions are essential for the adhesion and transmigration of leukocytes across the endothelium during immune responses .
ICAM-1 is typically expressed at low levels on the surface of endothelial cells and leukocytes. However, its expression can be significantly upregulated in response to pro-inflammatory cytokines such as interleukin-1 (IL-1) and tumor necrosis factor (TNF) . This upregulation enhances the ability of leukocytes to adhere to endothelial cells and migrate to sites of inflammation .
ICAM-1 is involved in various immune processes, including the activation of T-cells. It functions as a costimulatory molecule on antigen-presenting cells to activate MHC class II restricted T-cells, and on other cell types in association with MHC class I to activate cytotoxic T-cells . Additionally, ICAM-1 serves as a receptor for human rhinovirus, facilitating the entry of the virus into respiratory epithelial cells .
Recombinant ICAM-1, such as the mouse recombinant version, is produced using recombinant DNA technology. This involves inserting the gene encoding ICAM-1 into a suitable expression system, such as bacteria or mammalian cells, to produce the protein in large quantities. Recombinant ICAM-1 is used in various research applications, including studies on cell adhesion, immune response, and inflammation .
The recombinant form of ICAM-1 is widely used in research to study its role in immune responses and inflammatory processes. It is also utilized in the development of therapeutic agents targeting ICAM-1 interactions to treat inflammatory diseases and conditions involving excessive leukocyte adhesion .