CD11b (integrin αM, ITGAM) is a 165–170 kDa transmembrane glycoprotein that non-covalently associates with CD18 (β2 integrin) to form the Mac-1 complex (αMβ2). Key functional and structural attributes include:
Property | Details |
---|---|
Ligands | ICAM-1 (CD54), ICAM-2 (CD102), iC3b, fibrinogen |
Cellular Expression | Monocytes, macrophages, granulocytes, NK cells, dendritic cells, microglia |
Gene ID (Human/Mouse) | 3684 (Human) / 16409 (Mouse) |
Key Domains | I/A domain (ligand-binding), β-propeller domain |
CD11b mediates adhesion, phagocytosis, and immune cell recruitment . Its dysregulation is linked to autoimmune diseases and cancer progression .
Two widely used clones, M1/70 (mouse-reactive) and ICRF44 (human-reactive), are highlighted below:
Clone | Species Reactivity | Applications | Target Cells |
---|---|---|---|
M1/70 | Mouse, cross-reactive to human | Flow cytometry, IHC (frozen sections), inhibition assays | Splenocytes, bone marrow cells, peritoneal macrophages |
ICRF44 | Human | Flow cytometry, adhesion/transmigration assays | Peripheral blood monocytes, granulocytes, activated lymphocytes |
M1/70: Blocks iC3b binding to Mac-1 and reduces myeloid cell infiltration in irradiated tumors .
ICRF44: Inhibits leukocyte adhesion to endothelial cells by targeting ICAM-1 interactions .
Radiation Therapy Enhancement: Systemic administration of CD11b antibodies (M1/70) in murine squamous cell carcinoma models reduced infiltration of S100A8+ and MMP-9+ myeloid cells, leading to tumor shrinkage (12/16 cures vs. 7/19 in controls) .
Mechanism: Antibodies inhibited bone marrow-derived cell adhesion to endothelial monolayers by 60–80% and abolished chemotaxis toward CXCL12 and C5a .
Glioblastoma Imaging: 89Zr-labeled anti-CD11b antibodies enabled specific detection of tumor-associated myeloid cells (SUVmean = 2.60 ± 0.24 in tumors vs. 0.6 ± 0.11 in contralateral brain) .
Biodistribution: High uptake in spleen (14.2% ID/g) and lymph nodes (9.8% ID/g) confirmed myeloid specificity .
BG34-200 Ligand: Binding to CD11b’s I domain triggered phagocytosis and differentiation of tumor-associated monocytes into dendritic cells, enhancing T-cell activation in osteosarcoma and melanoma models .
CD11b is a 165-kDa adhesion glycoprotein that associates with the 95-kDa integrin β2 (CD18) to form the CD11b/CD18 complex, also known as Mac-1 or CR3. It functions as a type I transmembrane glycoprotein encoded by ITGAM (Integrin alpha M). CD11b plays crucial roles in cell-cell and cell-substrate interactions and serves as a receptor for iC3b, CD54 (ICAM-1), CD102 (ICAM-2), and CD50 (ICAM-3) . It is expressed on activated lymphocytes, monocytes, granulocytes, and a subset of NK cells, making it a vital marker for studying immune cell populations and functions . Additionally, CD11b is implicated in various adhesive interactions of myeloid cells and mediates the uptake of complement-coated particles and pathogens, positioning it as a significant target for immunological research .
Different clones of anti-CD11b antibodies recognize distinct epitopes on the CD11b molecule, resulting in varying functional effects:
ICRF44 (human CD11b): Specifically binds to human CD11b and significantly inhibits polymorphonuclear leukocyte aggregation in response to fMLP .
M1/70 (mouse/rat CD11b): Binds to mouse/rat CD11b and reportedly blocks cell adherence and C3bi binding but does not block cell-mediated lysis .
ED7, ED8, and 1B6c (rat CD11b): These induce strong homotypic aggregation of granulocytes. Cross-blocking experiments show ED7 and ED8 recognize identical or closely related epitopes .
OX-42 (rat CD11b): Unlike ED7, ED8, and 1B6c, OX-42 has no proaggregatory effect and belongs to a group of inhibitory anti-CR3 mAbs .
These differences in epitope recognition explain why some anti-CD11b antibodies enhance cell aggregation while others inhibit it, highlighting the importance of clone selection based on experimental objectives .
Biotin conjugation to CD11b antibodies provides significant advantages for research applications without substantially altering antibody specificity when properly optimized. The conjugation process typically involves binding biotin to the antibody under optimum conditions, followed by removal of unreacted biotin . This modification enables secondary detection through avidin-biotin systems, allowing for signal amplification in techniques such as flow cytometry, immunohistochemistry, and immunoprecipitation.
When working with biotinylated anti-CD11b antibodies, researchers should consider:
The conjugation ratio of biotin to antibody must be optimized to maintain antibody functionality while providing sufficient biotin for detection.
Pre-diluted biotinylated antibodies (such as those from BD Biosciences) are designed for use at recommended volumes per test, typically with 1 × 10^6 cells in a 100-μl experimental sample .
An appropriate isotype control (also biotin-conjugated) should be used at the same concentration as the test antibody to control for non-specific binding .
For optimal flow cytometry results with biotinylated CD11b antibodies, researchers should follow this methodological approach:
Sample preparation: Prepare single-cell suspensions at a concentration of 1 × 10^6 cells in 100 μl of appropriate buffer (typically PBS with 1-2% serum proteins and 0.1% sodium azide) .
Titration: Even for pre-diluted antibodies, titration is recommended to determine optimal concentration for your specific experimental conditions .
Staining procedure:
Controls:
Instrument settings: Adjust fluorescence parameters based on the specific fluorochrome conjugated to your avidin/streptavidin. Refer to multicolor flow cytometry guidelines for optimal settings .
Analysis considerations: When analyzing myeloid populations, use additional markers to differentiate between monocytes, granulocytes, and other CD11b-expressing cell types for comprehensive phenotyping.
Cross-blocking experiments are essential for determining the epitope specificity of anti-CD11b antibodies. Based on published methodologies, the following protocol is recommended:
Cell preparation: Isolate granulocytes or other CD11b-expressing cells and prepare at 1×10^6 cells/ml in appropriate buffer .
Primary blocking step:
Secondary binding step:
Detection step:
Data interpretation:
This approach allowed researchers to identify at least three different epitopes on the rat CD11b molecule, with some antibodies (ED7 and ED8) recognizing closely related epitopes while others (1B6c) recognized non-related epitopes .
For effective use of biotinylated CD11b antibodies in immunohistochemistry:
Tissue preparation:
Antigen retrieval:
For paraffin sections, perform heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)
Optimization of retrieval conditions may be necessary for specific tissue types
Blocking steps:
Block endogenous peroxidase activity with 0.3-3% hydrogen peroxide
Block endogenous biotin using avidin/biotin blocking kit for tissues with high endogenous biotin
Block non-specific binding with serum or protein block
Primary antibody incubation:
Detection systems:
For direct detection: Apply streptavidin-HRP or streptavidin-AP
For signal amplification: Use ABC (Avidin-Biotin Complex) method
Develop with appropriate substrate (DAB for HRP, Fast Red for AP)
Counterstaining and mounting:
Counterstain with hematoxylin
Dehydrate, clear, and mount with permanent mounting medium
This protocol is appropriate for detecting CD11b expression in human samples using rabbit recombinant monoclonal antibodies conjugated to biotin, such as the EPR1344 clone .
CD11b-neutralizing antibodies have proven valuable in studying and potentially modulating myeloid cell recruitment to irradiated tumors. The following methodology has been established:
Experimental design:
Assessment of tumor response:
Analysis of myeloid cell infiltration:
Functional assays:
This approach has demonstrated that CD11b antibodies can significantly enhance antitumor response to radiation by inhibiting myeloid cell recruitment to irradiated tumors, thereby preventing the restoration of vasculature and tumor regrowth .
When investigating how different CD11b antibody clones affect neutrophil aggregation and adhesion, researchers should follow these methodological considerations:
Clone selection and characterization:
Aggregation assays:
Static conditions: Incubate neutrophils with antibodies (10-20 μg/ml) at 37°C and assess aggregation at multiple timepoints (30 min, 2 hr, 4 hr, 24 hr)
Hanging drop assay: Use this method to exclude contact with plastic surfaces and isolate direct cellular effects
Quantification: Use both visual scoring systems and automated image analysis
Mechanistic investigations:
Controls and comparisons:
This approach revealed that:
Some antibodies (ED7, ED8, 1B6c) induce strong homotypic aggregation
Others (OX-42) have no proaggregatory effect
Fab fragments can induce aggregation, which is enhanced by cross-linking
At least three different functional epitopes exist on rat CD11b
CD11b antibodies can significantly influence CD11b/CD18 complex functions through various mechanisms, which can be investigated using these methodological approaches:
Neutrophil migration assays:
Transwell migration: Pretreat neutrophils with CD11b antibodies and measure migration toward chemoattractants
Under-agarose migration: Assess directional migration in the presence of antibodies
Intravital microscopy: Examine neutrophil recruitment in vivo following antibody administration
Compare effects: Different epitope-targeting antibodies may enhance or inhibit migration
Phagocytosis assessment:
Complement-mediated phagocytosis: Measure uptake of iC3b-opsonized particles
Flow cytometry-based assays: Use fluorescent particles to quantify phagocytic capacity
Live cell imaging: Monitor real-time phagocytic events in the presence of antibodies
Western blotting: Assess activation of downstream signaling pathways
Molecular mechanisms:
Research has demonstrated that CD11b/CD18 complex:
Regulates neutrophil migration through interaction with endothelial adhesion molecules
Mediates uptake of complement-coated particles and pathogens
Recognizes fibrinogen, factor X, and ICAM1
Controls production of neutrophil superoxide ions
May regulate phagocytosis-induced apoptosis in extravasated neutrophils
When working with biotinylated CD11b antibodies, researchers commonly encounter the following issues and solutions:
High background in detection systems:
Reduced antibody binding efficiency:
Inconsistent signal intensity:
Cross-reactivity issues:
Sodium azide interference:
Cause: Sodium azide in antibody preparations can inhibit enzymatic reactions
Solution: For HRP or AP-based detection systems, dialyze antibody or use commercial preparations without azide
Safety note: Dilute azide compounds in running water before disposal to prevent accumulation of toxic hydrazoic acid
Inconsistent results across species:
To validate biotinylated CD11b antibodies before experimental use:
Flow cytometry validation:
Western blot validation:
Confirm antibody recognizes a protein of appropriate molecular weight (165-kDa)
Include positive controls (neutrophil/monocyte lysates) and negative controls
Perform peptide competition assays to verify specificity
Functional validation:
Cross-reactivity assessment:
Epitope mapping:
Knockout/knockdown controls:
When designing experiments to study CD11b+ cell populations in complex tissues, researchers should consider these critical parameters:
Sample preparation optimization:
Tissue digestion: Optimize enzymatic digestion protocols to maintain CD11b epitope integrity
Single-cell preparation: Use gentle mechanical dissociation methods to preserve surface marker expression
Fixation conditions: If fixation is required, validate that the fixative doesn't alter antibody binding
Multi-parameter analysis strategy:
Panel design: Combine CD11b with other markers to identify specific myeloid subpopulations:
Neutrophils: CD11b+CD66b+
Monocytes: CD11b+CD14+
Macrophages: CD11b+CD68+
Myeloid-derived suppressor cells: CD11b+CD33+HLA-DR-
Functional markers: Include markers of activation (CD80/86) or immunosuppression (PD-L1)
Technical controls:
Cross-validation approaches:
Complementary techniques: Validate flow cytometry findings with immunohistochemistry to preserve spatial context
Functional validation: Correlate phenotypic findings with functional assays (e.g., phagocytosis, cytokine production)
Single-cell technologies: Consider scRNA-seq to correlate CD11b expression with transcriptional profiles
Microenvironmental considerations:
Data analysis frameworks:
Advances in antibody engineering present several promising directions for enhancing CD11b antibodies in therapeutic contexts:
Epitope-specific targeting improvements:
Development of antibodies targeting specific functional domains of CD11b could provide more precise control over biological functions
Engineering antibodies that selectively inhibit pathological functions while preserving beneficial immune surveillance
Utilizing cross-blocking studies to identify epitopes associated with specific functions for targeted modification
Format diversification strategies:
Bispecific antibodies: Combining CD11b targeting with tumor-specific antigens to enhance tumor-directed immune responses
Antibody-drug conjugates: Delivering cytotoxic payloads specifically to CD11b+ myeloid cells in pathological microenvironments
Single-domain antibodies: Developing smaller formats with improved tissue penetration for targeting tissue-resident myeloid cells
Functional modification approaches:
Combination therapy optimization:
Refining CD11b antibody use in combination with radiation therapy to maximize tumor control while minimizing toxicity
Identifying synergistic combinations with checkpoint inhibitors or other immunomodulatory agents
Developing biomarkers to predict which patients would benefit most from CD11b-targeted interventions
Translation of preclinical findings:
Building on the significant enhancement of antitumor response observed when CD11b antibodies are administered following radiation
Addressing challenges in translating antibody efficacy across species (given the known cross-reactivity patterns)
Developing humanized antibodies suitable for clinical development based on functionally characterized murine antibodies
Emerging methodologies for investigating CD11b+ cell heterogeneity and plasticity include:
Single-cell multi-omics approaches:
Single-cell RNA-seq + protein: CITE-seq combining CD11b antibody detection with transcriptome analysis
Spatial transcriptomics: Mapping CD11b+ cell transcriptional states within their tissue contexts
Epigenetic profiling: Assessing chromatin accessibility in CD11b+ cells to understand developmental plasticity
Proteomics integration: Combining CD11b-based cell sorting with deep proteomic analysis
Advanced imaging technologies:
Multiplexed imaging: Using technologies like CODEX or Imaging Mass Cytometry to analyze multiple markers on CD11b+ cells
Intravital microscopy: Real-time tracking of CD11b+ cells in living organisms
Super-resolution microscopy: Examining nanoscale organization of CD11b/CD18 complexes
Dynamic imaging: Capturing temporal changes in CD11b activation states during cellular responses
Functional heterogeneity assessment:
Single-cell functional assays: Measuring phagocytosis, ROS production, and cytokine secretion at single-cell level
CyTOF-based functional profiling: Simultaneously detecting multiple functional markers in CD11b+ populations
Live-cell biosensors: Developing reporters for real-time monitoring of CD11b activation
Metabolic phenotyping: Characterizing metabolic states of different CD11b+ subpopulations
Genetic manipulation techniques:
CRISPR-based screening: Identifying genes regulating CD11b function in different contexts
Lineage tracing: Tracking developmental trajectories of CD11b+ cells in disease progression
Conditional knockout models: Generating temporal and cell-type specific CD11b deletion
Reporter systems: Creating mice with fluorescent proteins under CD11b promoter control
Computational integration frameworks:
Machine learning algorithms: Identifying novel CD11b+ cell subsets from high-dimensional data
Trajectory inference: Mapping developmental relationships between CD11b+ populations
Systems biology approaches: Modeling CD11b signaling networks in different disease contexts
Multi-scale integration: Connecting molecular, cellular, and tissue-level data on CD11b function
These emerging methodologies promise to reveal unprecedented insights into the roles of diverse CD11b+ cell populations in health and disease, potentially leading to more targeted therapeutic approaches.
Researchers can integrate CD11b antibody-based approaches with other immunomodulatory strategies through:
Rational combination therapy design:
With radiation therapy: Building on established enhancement of radiation response by CD11b antibodies
With checkpoint inhibitors: Combining myeloid-targeting via CD11b with T-cell activation through PD-1/CTLA-4 blockade
With targeted therapies: Addressing multiple components of the tumor microenvironment simultaneously
Sequential approaches: Determining optimal timing of CD11b antibody administration relative to other treatments
Cellular therapy enhancements:
CAR-T cell combinations: Using CD11b antibodies to modify myeloid suppression in CAR-T environments
Adoptive cell therapy optimization: Preconditioning with CD11b antibodies to improve cellular therapy efficacy
Ex vivo manipulation: Treating isolated CD11b+ cells to reprogram their function before reinfusion
Engineered cellular products: Developing CD11b-targeted CAR-macrophages for improved phagocytosis of cancer cells
Biomarker-guided strategy selection:
Predictive biomarkers: Identifying markers of CD11b+ cell activity that predict response to immunotherapy
Pharmacodynamic monitoring: Using CD11b expression patterns to track treatment effects
Resistance mechanisms: Understanding how CD11b+ cells contribute to therapy resistance
Patient stratification: Selecting patients most likely to benefit from CD11b-targeted approaches
Delivery system innovations:
Nanoparticle-based delivery: Targeting CD11b antibodies specifically to tumor-associated myeloid cells
Bispecific approaches: Developing molecules targeting both CD11b and tumor-specific antigens
Local delivery strategies: Administering CD11b antibodies directly to tumor sites
Controlled release formulations: Providing sustained CD11b modulation through innovative delivery platforms
Translational research frameworks:
Integrated biomarker programs: Collecting comprehensive immune monitoring data during clinical trials
Patient-derived models: Testing CD11b antibody combinations in models preserving patient immune complexity
Early-phase trial design: Incorporating immune pharmacodynamic endpoints in dose-finding studies
Multi-center collaborations: Establishing standardized protocols for CD11b assessment across clinical sites
The biotin-conjugated Mouse Anti-Human CD11b Antibody has several applications in research and diagnostics:
The antibody is purified by affinity chromatography and conjugated with biotin under optimal conditions . It is formulated in a phosphate-buffered solution with a pH of 7.2, containing 0.09% sodium azide as a preservative . The recommended storage conditions are between 2°C and 8°C, and the solution should not be frozen .