STRING: 3702.AT3G52310.1
ABCG2 (ATP-binding cassette transporter G2), also known as Breast Cancer Resistance Protein 1 (Bcrp1), is a membrane transporter molecule that spans the cell membrane six times. It functions as either homo or hetero dimers linked by a short intracellular flexible linker region. While initially identified in a breast cancer cell line and associated with drug resistance, ABCG2 has emerged as a critical marker for primitive stem cells, particularly hematopoietic stem cells. ABCG2 plays a significant role in the efflux of various substrates, including chemotherapeutic drugs and fluorescent dyes like Hoechst 33342 and Rhodamine 123 .
ABCG2 is expressed in various cell types, with particularly high expression in:
Hematopoietic stem cells (particularly in the CD34+ lineage-negative bone marrow fraction)
Cancer cell lines including JAR human choriocarcinoma cells
A549 human lung carcinoma cells
RPMI 8226 cells
Detection methods include flow cytometry, immunocytochemistry, and Western blot. For immunocytochemistry, ABCG2 can be detected using monoclonal antibodies (like clone 5D3) at concentrations of approximately 8-10 μg/mL, followed by fluorescently conjugated secondary antibodies such as NorthernLights™ 557-conjugated Anti-Mouse IgG .
ABCG2 antibodies are primarily used for:
Identification and isolation of stem cell populations, particularly the "side-population" that fails to retain intracellular staining dyes
Analysis of cancer cell resistance mechanisms
Flow cytometric analysis of surface expression
Immunocytochemistry for localization studies
Western blotting for protein expression quantification
Cell lysate analysis
These applications have proven valuable in cancer research, particularly for studying malignant behaviors of hepatocellular carcinoma and investigating the role of ABCG2 in mitochondrial function .
Recent research has uncovered that ABCG2 localizes not only to the plasma membrane but also to mitochondria, affecting 5-aminolevulinic acid-mediated protoporphyrin IX accumulation. This dual localization necessitates specialized experimental approaches:
Researchers should employ subcellular fractionation techniques to separate mitochondrial and plasma membrane fractions before Western blot analysis
Confocal microscopy with mitochondrial co-staining is recommended for accurate localization studies
When investigating ABCG2 function, researchers should account for both plasma membrane efflux activity and mitochondrial-specific functions
Experimental protocols analyzing ABCG2 inhibition should monitor effects on both membrane transport and mitochondrial metabolism
Several methodological considerations affect ABCG2 antibody specificity:
Antibody clone selection is critical—the 5D3 clone shows high specificity for human ABCG2
Fixation methods significantly impact epitope accessibility; immersion fixation is recommended for immunocytochemistry applications
Incubation time and temperature affect binding efficiency (3 hours at room temperature yields optimal results for immunocytochemistry)
Background fluorescence must be carefully controlled using appropriate isotype controls (e.g., MAB0041 for mouse IgG2B antibodies)
Cell type-specific expression levels necessitate optimization of antibody concentration (typically 8-10 μg/mL for cancer cell lines)
ABCG2 has emerged as a potential cancer stem cell marker, particularly in hepatocellular carcinoma. When designing experiments to investigate this relationship:
Use multiparameter flow cytometry to correlate ABCG2 expression with other established cancer stem cell markers
Employ functional assays (sphere formation, tumor initiation in xenograft models) to validate stemness properties of ABCG2+ populations
Analyze relationship between ABCG2 expression levels and clinical parameters including tumor progression, metastasis, and treatment resistance
Consider combinatorial targeting approaches that address both ABCG2-mediated drug efflux and stemness-associated signaling pathways
Implement lineage tracing techniques to track the fate of ABCG2+ cells during tumor development and treatment
CD27 is a co-stimulatory receptor belonging to the Tumor Necrosis Factor receptor family. It is expressed on T cells, particularly tumor-infiltrating lymphocytes (TILs), and plays a crucial role in:
T cell activation and proliferation
Cytotoxic T cell function enhancement
Memory T cell formation and maintenance
NK cell activation
CD27 requires higher-order receptor cross-linking for effective signaling and activation. Agonistic antibodies against CD27 can stimulate these pathways, potentially enhancing anti-tumor immune responses by reactivating tumor-infiltrated and tumor-reactive T cells .
CD27 agonist antibodies enhance anti-tumor immunity through multiple mechanisms:
Direct stimulation of CD27 signaling on CD8+ T cells, increasing their activation, proliferation, and cytotoxic function
Enhancement of NK cell activity, as evidenced by increased KLRG1 expression following anti-CD27 treatment
Promotion of IFNγ and chemokine release from activated T and NK cells
Indirect activation of macrophages through IFNγ signaling, improving their phagocytic capability
Synergistic activity when combined with direct tumor-targeting antibodies like anti-CD20 (for B-cell lymphomas) or anti-gp75 (for melanoma)
Several methodological approaches can assess CD27 antibody functionality:
Flow cytometric analysis of activation markers (CD62L, CD44) on CD8+ T cells
Quantification of effector T cell populations following antibody treatment
Assessment of NK cell activation via KLRG1 expression
Measurement of cytokine production (particularly IFNγ) in response to treatment
Analysis of macrophage infiltration and activation in the tumor microenvironment
Evaluation of T cell proliferation using CFSE dilution or other proliferation assays
Bispecific antibodies targeting both CD27 and tumor antigens represent an advanced approach to cancer immunotherapy. The CD27xEGFR bispecific antibody exemplifies this strategy:
The antibody simultaneously binds CD27 on T cells and EGFR on tumor cells
This dual binding induces cancer cell-localized crosslinking and activation of CD27
The Fc-silent domain minimizes potential toxicity by reducing Fc gamma receptor-mediated binding
This design achieves EGFR-restricted co-stimulation of T cells
The approach results in enhanced T cell proliferation, activation marker expression, cytotoxicity, and IFNγ release
Experimental validation shows augmented T cell cytotoxicity in artificial antigen-presenting carcinoma cell line models
The combination of anti-CD27 and anti-CD20 antibodies demonstrates superior efficacy compared to either treatment alone. This synergy operates through a multi-step process:
Anti-CD27 stimulates CD8+ T cells and NK cells to produce IFNγ and chemokines
Released chemokines recruit myeloid cells, particularly macrophages, to the tumor site
IFNγ activates these macrophages, enhancing their phagocytic capabilities
Activated macrophages more efficiently perform anti-CD20-dependent phagocytosis of tumor cells
This coordinated immune response leads to improved tumor clearance and survival rates
This mechanism has been validated across multiple lymphoma models, including studies in huCD27 transgenic mice using the anti-huCD27 antibody varlilumab .
Rigorous experimental design for CD27 antibody research requires several key controls:
Fc receptor knockout models (FcγRIII−/−) to distinguish direct CD27 stimulation from Fc-mediated effects
SCID (severe combined immune deficiency) mice to determine whether NK activation occurs directly or via T cell-dependent mechanisms
Isotype-matched control antibodies to establish baseline immune activation
Single agent controls (anti-CD27 alone, tumor-targeting antibody alone) to accurately assess combinatorial effects
Multiple tumor models with varying CD27 and target antigen expression profiles
Time-course studies to capture dynamic changes in immune cell populations and activation states
Cytokine neutralization experiments to confirm the role of specific mediators (e.g., IFNγ) in observed effects
Optimization strategies for CD27-targeted immunotherapy include:
Engineering antibodies with enhanced receptor clustering capabilities to improve CD27 signaling
Developing tumor-targeted delivery approaches to concentrate CD27 stimulation in the tumor microenvironment
Combining CD27 agonists with other immunomodulatory agents (checkpoint inhibitors, cytokines)
Exploring trispecific antibody formats to engage multiple immune activating receptors simultaneously
Using single-cell RNA sequencing to identify optimal combination partners based on immune cell transcriptional profiles
Implementing predictive biomarkers to identify patients most likely to respond to CD27-targeted therapy
Developing smaller antibody formats with improved tumor penetration capabilities
The recent discovery of SC27, a broadly neutralizing antibody against all COVID-19 variants, provides valuable insights for antibody engineering across research domains:
Researchers identified and isolated this plasma antibody from a single patient as part of a study on hybrid immunity
The antibody works by recognizing and blocking the virus's spike protein across all variants
Advanced technology allowed researchers to determine the exact molecular sequence of the antibody
This breakthrough demonstrates how targeting conserved epitopes can overcome viral mutation challenges
The approach suggests similar strategies could be applied to other rapidly mutating pathogens or cancer targets
The research emphasizes the value of studying exceptional responders to identify antibodies with unique properties
Recent technical innovations have enhanced antibody research capabilities:
Single-cell RNA sequencing now allows detailed mechanistic analysis of antibody effects on diverse cell populations
Advanced protein engineering techniques enable creation of bispecific and multispecific antibody formats with novel functionalities
Structural biology approaches inform rational antibody design targeting specific epitopes
Humanized mouse models (like huCD27 transgenic mice) facilitate translation of findings toward human applications
High-throughput screening methodologies accelerate identification of antibodies with desired properties
Computational approaches predict antibody-antigen interactions and guide optimization efforts