EMR3 is a member of the class B seven-span transmembrane (TM7) receptor family, also known as Adhesion G protein-coupled receptor E3 (ADGRE3). It is predominantly expressed by cells of the immune system, with highest expression levels observed in neutrophils, monocytes, and macrophages . The protein structure is characterized by:
An extended extracellular region with variable N-terminal epidermal growth factor (EGF)-like domains
A TM7 domain connected via a mucin-like spacer domain
EMR3 is notably absent on lymphocytes, hematopoietic stem cells, and myeloid progenitors, but studies suggest it becomes upregulated during late stages of neutrophil differentiation, potentially serving as a marker for terminally differentiated cells .
EMR3 antibodies are available in multiple conjugated and unconjugated formats to suit different experimental needs:
| Antibody Format | Catalog Examples | Host Species | Clone | Applications |
|---|---|---|---|---|
| Unconjugated | MAB8496 | Mouse | 908235 | Flow Cytometry, IHC |
| PE-conjugated | FAB8496P | Mouse | 908235 | Flow Cytometry |
| APC-conjugated | 366105/366106 | Mouse | QA20A59 | Flow Cytometry |
| DyLight 680 | NB100-65935FR | Armenian Hamster | 3D7 | Flow Cytometry |
| PE/Atto594 | NB10065935PEATT594 | Armenian Hamster | 3D7 | Flow Cytometry |
| Polyclonal | PA5-102056, BS-14588R | Rabbit | - | WB, IHC, IF |
EMR3 antibodies have demonstrated utility across multiple experimental platforms:
Flow cytometry represents the primary application for most EMR3 antibodies. The search results show validated protocols for:
Detection of EMR3 in human peripheral blood monocytes
Identification of EMR3-expressing cells in transfection studies
Differentiation of granulocyte populations
Typical staining protocols involve 5 μL per million cells in 100 μL staining volume or 5 μL per 100 μL of whole blood , though optimal dilutions should be experimentally determined for each specific application and antibody.
Several EMR3 antibodies have been validated for immunohistochemistry on:
Paraffin-embedded tissue sections
Human lung cancer tissue
Glioblastoma samples
For instance, the MAB8496 antibody has been used at 5 μg/mL for 1 hour at room temperature in combination with appropriate secondary antibodies and detection systems .
EMR3's physiological role remains incompletely understood, but current evidence suggests:
Potential involvement in myeloid-myeloid interactions during immune and inflammatory responses
Possible involvement in cellular adhesion processes given its classification within adhesion GPCRs
Particularly noteworthy is EMR3's emerging role in cancer research:
Upregulation in glioblastoma multiforme (GBM) correlates with poor patient survival
Knocking down EMR3 in GBM cells decreases cellular invasion by greater than 3-fold, though it has no impact on cellular proliferation
EMR3 protein is variably expressed in human glioblastoma tissues and cell lines
The study by Kahn et al. demonstrated that:
EMR3 was robustly expressed in 3/8 primary glioblastoma samples
Weakly expressed in 1/8 samples
These findings suggest EMR3 may be a potential mediator of invasive phenotypes in glioblastoma and a possible therapeutic target.
For successful EMR3 detection, researchers should consider:
Working concentration: Typically 2.5-5 μg/mL for most applications, though this varies by antibody and application
Buffer systems: Most antibodies perform optimally in phosphate-buffered solutions (PBS) at pH 7.2
Antigen retrieval for IHC: Heat-induced epitope retrieval using basic retrieval reagents has been successfully employed with some antibodies
Storage conditions: Generally stored at 4°C, with specific formulations containing stabilizers such as BSA and preservatives like sodium azide
To ensure experimental validity when using EMR3 antibodies:
Isotype controls are essential, especially for flow cytometry applications
Positive controls: Human peripheral blood monocytes and neutrophils serve as reliable positive controls
Negative controls: Lymphocytes, which do not express EMR3, serve as appropriate negative controls
Based on current knowledge and limitations, several research directions merit further investigation:
Elucidation of EMR3 ligands, which remain unknown despite the receptor's discovery and characterization
Further exploration of EMR3 as a potential therapeutic target in glioblastoma
Investigation of EMR3's role in other cancer types beyond GBM
Development of therapeutic antibodies targeting EMR3 for potential cancer treatment
Expanded understanding of EMR3's physiological role in neutrophil differentiation and function
What is EMR3 and what is its biological significance in immune function?
EMR3 (EGF module containing, mucin-like hormone receptor-3) is a member of the class B seven-span transmembrane (TM7) receptor family, also classified as an adhesion G protein-coupled receptor (Adhesion G protein-coupled receptor E3). This protein is characterized by an extended extracellular region with N-terminal epidermal growth factor (EGF)-like domains coupled to a TM7 domain via a mucin-like spacer domain .
EMR3 is predominantly expressed by cells of the immune system and plays a significant role in myeloid-myeloid interactions during immune and inflammatory responses. Unlike other EGF-TM7 family members, EMR3 has a distinctive expression pattern, making it valuable for studying myeloid cell function and differentiation .
Which immune cell populations express EMR3 and how does its expression pattern differ from other EGF-TM7 family members?
EMR3 demonstrates a specific expression pattern across myeloid lineage cells:
| Cell Type | EMR3 Expression Level |
|---|---|
| Granulocytes | Highest |
| Neutrophils | High |
| CD16+ (mature) Monocytes | High |
| CD16- Monocytes | Dim/Low |
| Myeloid Dendritic Cells | Dim/Low |
| Macrophages | Positive |
| Lymphocytes | Negative |
| Plasmacytoid DC | Negative |
| CD34+ Hematopoietic Stem Cells | Negative |
A distinguishing characteristic of EMR3 compared to other EGF-TM7 receptors (like CD97 and EMR2) is that EMR3 is not expressed on CD34+CD33−/CD38− committed hematopoietic stem cells or CD34+CD33+/CD38+ progenitors in bone marrow. Instead, EMR3 is specifically upregulated during late stages of granulopoiesis, making it the first EGF-TM7 family member found predominantly on granulocytes .
What are the principal applications of EMR3 antibodies in immunological research?
EMR3 antibodies have several key research applications:
Flow Cytometry: For identifying and characterizing myeloid cell populations, particularly granulocytes and monocyte subsets. Most protocols recommend using 0.25 µg per 10^6 cells .
Immunohistochemistry: For detecting EMR3 in tissue sections, particularly in studies of inflammatory conditions and cancer. Optimal concentrations range from 5-25 µg/mL for paraffin-embedded sections .
CyTOF (Mass Cytometry): EMR3 antibodies can be labeled for use in high-dimensional immunophenotyping .
Functional Studies: For investigating myeloid cell differentiation and immune responses, including blocking studies to understand EMR3's role in cellular interactions .
Cancer Research: Particularly in studying glioblastoma, where EMR3 expression correlates with invasiveness and patient outcomes .
How does EMR3 contribute to invasive phenotypes in glioblastoma multiforme and what evidence supports its potential as a therapeutic target?
EMR3 has been identified as a potential mediator of invasive behavior in glioblastoma multiforme (GBM). Research evidence showing this relationship includes:
Microarray analysis through The Cancer Genome Atlas (TCGA) demonstrated a significant survival benefit for GBM patients with tumors showing EMR3 downregulation (P<0.001) .
Immunohistochemistry on primary GBM tissue samples revealed variable EMR3 expression patterns: robust expression (>25% of tumor cells) in 37.5% (3/8) of samples, weak expression (1-5% of tumor cells) in 12.5% (1/8), and no detectable expression in 50% (4/8) .
siRNA knockdown experiments demonstrated that suppressing EMR3 expression in SF767 glioblastoma cells had a greater than 3-fold reduction in cellular invasion compared to controls (3.42 vs 1, p<0.05), while having no impact on cellular proliferation .
Western blot and RT-PCR analysis showed variable EMR3 expression across different glioma cell lines, with SF-767 showing high levels, U87 showing low levels, and U251 and G55 showing no expression .
These findings suggest that EMR3 could serve as both a prognostic marker and a potential therapeutic target in GBM, particularly for addressing the invasive characteristics that make GBM treatment challenging.
What methodological approaches have successfully demonstrated EMR3 expression during myeloid cell differentiation?
Several experimental approaches have been employed to characterize EMR3 expression during myeloid differentiation:
In vitro differentiation models: Studies using HL-60 cells (a promyelocytic cell line) and CD34+ progenitor cells have demonstrated that EMR3 is specifically upregulated during late granulopoiesis, unlike other EGF-TM7 receptors that appear earlier in differentiation .
Flow cytometric analysis: Using antibodies like clone 908235 or 3D7, researchers have mapped EMR3 expression across the myeloid differentiation spectrum. This approach revealed that EMR3 is absent on early progenitors but increases as cells progress toward terminal differentiation, particularly in the granulocyte lineage .
Bone marrow examination: Comparative analysis of bone marrow populations has shown that while CD97 and EMR2 are expressed on CD34+ progenitors, EMR3 is specifically absent on these early cells and appears only in more differentiated stages .
Correlation with maturation markers: EMR3 expression has been shown to correlate with the expression of CD16 on monocytes, with more mature CD16+ monocytes expressing higher levels of EMR3 compared to CD16- monocytes .
These methodological approaches collectively demonstrate that EMR3 serves as a marker for more terminally differentiated myeloid cells, particularly granulocytes.
What structural features differentiate EMR3 from other EGF-TM7 family members at the protein level?
EMR3, like other EGF-TM7 receptors, is expressed at the cell surface as a heterodimeric molecule consisting of:
A long extracellular α-chain containing EGF-like domains at its N-terminus
A membrane-spanning β-chain
The protein undergoes autocatalytic processing at the G-protein-coupled receptor-proteolytic site (GPS) proximal to the first transmembrane segment, which cleaves the polypeptide into these two subunits that noncovalently associate at the cell surface .
EMR3's distinguishing features include:
Its specific amino acid sequence (human EMR3 spans Met1-Tyr652, Accession # Q9BY15)
Its unique expression pattern compared to other family members (CD97, EMR1, EMR2, and EMR4)
Its close genetic linkage to the gene encoding EGF-like molecule containing mucin-like hormone receptor 2 on chromosome 19
While all EGF-TM7 family members share a similar domain organization, their ligand specificity and expression patterns differ significantly, with EMR3 being unique in its predominant expression on mature granulocytes and its absence from hematopoietic stem cells .
What are the optimal protocols for detecting EMR3 via flow cytometry in various cell populations?
Based on established research protocols, the following methodological approach is recommended for EMR3 detection by flow cytometry:
Protocol for detection in peripheral blood monocytes:
Sample preparation:
Isolate peripheral blood monocytes using standard density gradient separation
Wash cells in phosphate-buffered saline (PBS) containing 0.5% bovine serum albumin
Staining procedure:
Use 0.25 µg of anti-EMR3 antibody (e.g., clone 908235) per 10^6 cells
For two-color analysis, co-stain with Mouse Anti-Human CD14 APC-conjugated Monoclonal Antibody (e.g., FAB3832A)
Include appropriate isotype controls (e.g., Mouse IgG Flow Cytometry Isotype Control, MAB0041)
For indirect staining, use a secondary antibody such as Phycoerythrin-conjugated Anti-Mouse IgG (e.g., F0102B)
Alternatively, directly conjugated antibodies can be used (e.g., PE-conjugated Anti-Human EMR3, FAB8496P)
Analysis parameters:
For detection in transfected cell lines (validation studies):
What tissue preparation methods yield optimal results for EMR3 immunohistochemistry in clinical samples?
Based on published protocols, the following tissue preparation methods have been successfully employed for EMR3 immunohistochemistry:
Tissue fixation and embedding:
Use immersion fixation in formalin
Process tissues through standard paraffin embedding protocols
Section preparation:
Cut paraffin sections at 4-6 μm thickness
Mount sections on positively charged slides
Antigen retrieval (critical step):
Perform heat-induced epitope retrieval using Antigen Retrieval Reagent-Basic (e.g., CTS013)
This step is essential for optimal EMR3 detection in fixed tissues
Staining protocol:
Apply Mouse Anti-Human EMR3 Monoclonal Antibody (e.g., MAB8496) at 5 µg/mL
Incubate for 1 hour at room temperature
Follow with incubation with an appropriate detection system (e.g., Anti-Mouse IgG VisUCyte™ HRP Polymer Antibody, VC001)
Develop with DAB (3,3'-diaminobenzidine) chromogen (brown)
Counterstain with hematoxylin (blue)
Expected results:
This protocol has been successfully used to demonstrate EMR3 expression in both normal tissues (for studying myeloid cells) and pathological specimens (particularly in cancer research).
How should researchers validate the specificity of EMR3 antibodies in experimental systems?
Comprehensive validation of EMR3 antibodies should include these methodological approaches:
Positive and negative cell line controls:
Correlation with transcript levels:
Flow cytometry validation:
Blocking and competition studies:
Pre-incubate antibodies with recombinant EMR3 protein
Observe elimination or reduction of staining
Cross-reactivity assessment:
Test antibodies on cells expressing other EGF-TM7 family members
Confirm specificity for EMR3 versus CD97, EMR1, EMR2, or EMR4
Multiple antibody comparison:
These validation approaches are essential for ensuring reliable and reproducible results in EMR3 research.
What critical controls should be incorporated in EMR3 antibody-based experimental designs?
Rigorous experimental design for EMR3 studies should include these controls:
Isotype controls:
Cellular controls:
Expression knockdown controls:
Tissue controls:
Known positive tissues (e.g., normal neutrophil-rich tissues)
Serial sections with primary antibody omission
Secondary antibody controls:
Sections/cells treated with secondary antibody only
Transfection controls for overexpression studies:
Multiple detection method confirmation:
Correlation between different techniques (e.g., flow cytometry, IHC, Western blot)
Cross-validation with different antibody clones targeting EMR3
These controls help ensure the reliability and reproducibility of experimental results involving EMR3 antibodies and minimize the risk of false positive or negative findings.