MS4A1 (Membrane Spanning 4-Domains A1) encodes the CD20 protein, a 33.1 kDa transmembrane receptor critical for B-cell maturation, calcium signaling, and receptor-mediated apoptosis . The recombinant monoclonal antibody binds specifically to CD20, enabling precise detection and modulation of B-cell populations.
Reactivity: Primarily targets human CD20 but cross-reacts with non-human primate (e.g., chimpanzee, baboon), canine, and porcine species .
Epitope: Recognizes the cytoplasmic domain of CD20 or extracellular regions, depending on the clone .
Applications: Flow cytometry, immunohistochemistry (IHC), enzyme-linked immunosorbent assay (ELISA), and immunophenotyping of lymphomas .
The antibody is synthesized via a multi-step process:
Immunization: Recombinant human MS4A1 protein is used to immunize animals (e.g., rabbits, mice) .
B-Cell Isolation: Antigen-specific B cells are harvested from immunized animals .
Gene Amplification: Reverse transcription and PCR amplify antibody variable region genes .
Expression: Genes are cloned into expression vectors and transfected into host cells (e.g., CHO cells) .
Purification: Affinity chromatography yields high-purity antibodies .
Calcium Signaling: CD20 facilitates store-operated calcium entry, critical for B-cell activation and differentiation .
Apoptosis Regulation: Antibody binding may induce caspase-3 activation, promoting apoptosis in malignant B-cells .
Leukemia/Lymphoma Detection: Identifies B-cell lineage in peripheral blood and lymphoid tissues .
Hodgkin’s Disease: Detects Reed-Sternberg cells in lymphocyte-predominant Hodgkin’s lymphoma .
Non-B Cell Reactivity: Rarely binds T-cell lymphomas or Reed-Sternberg cells .
Species Specificity: Limited to human and select non-human primates in most products .
Parameter | Cusabio (CSB-RA015007A1HU) | Bio-Techne (NBP3-24154H) | ARP1 (L26 + IGEL/773) |
---|---|---|---|
Host | N/A | Rabbit | Mouse |
Detection Method | ELISA | IHC, Flow Cytometry | IHC, Flow Cytometry |
Sensitivity | High (ELISA-validated) | Moderate | Moderate |
Cross-Reactivity | Human, Dog | Human, Primate | Human, Porcine, Rat |
Emerging methods, such as single-cell PCR and minigene technology, enable rapid generation of recombinant antibodies from antigen-specific B cells, reducing reliance on hybridoma screening . These innovations may expand access to high-affinity MS4A1 antibodies for therapeutic applications, such as targeting B-cell malignancies.
The MS4A1 recombinant monoclonal antibody is produced through a sophisticated process involving the acquisition of MS4A1 antibody genes, their integration into suitable host cells, and the utilization of a cellular expression and translation system for the synthesis of the MS4A1 antibodies. This method offers significant advantages, including enhanced purity and stability of the resulting MS4A1 recombinant monoclonal antibodies, as well as improved antibody affinity and specificity. The MS4A1 recombinant monoclonal antibody undergoes purification using affinity chromatography and is subjected to rigorous testing through ELISA, IHC, and IF assays to ensure its quality. It is important to note that this antibody exclusively targets the human MS4A1 protein.
The MS4A1 protein (CD20) plays a critical role as a cell surface marker on B cells, contributing to B cell activation. This makes it a key target for therapeutic interventions in B cell-related diseases and autoimmune conditions.
MS4A1 (membrane spanning 4-domains A1), commonly known as CD20, is a 33.1 kDa non-glycosylated membrane protein consisting of 297 amino acid residues in humans. The protein contains four transmembrane domains with both the N- and C-termini located intracellularly. Its significance stems from its specific expression pattern and functional importance in B cell biology. MS4A1 is primarily expressed on the surface of B lymphocytes throughout most stages of B cell development, from pre-B cells to memory B cells, but is notably absent on plasma cells and early pro-B cells . This expression profile makes it an ideal target for studying B cell development, activation, and for therapeutic interventions in B cell-related disorders.
MS4A1 regulates transmembrane calcium conductance, potentially functioning as a component of store-operated calcium channels, and plays critical roles in cell cycle progression and B-cell proliferation . The protein is found in lipid rafts, though the intensity of this association can vary depending on extracellular triggering that employs CD20 conformational changes . These structural and functional characteristics make MS4A1 an excellent target for recombinant monoclonal antibodies used in both research and clinical applications.
MS4A1 recombinant monoclonal antibodies offer significant advantages over conventional monoclonal antibodies for research applications. The recombinant production process involves precise genetic engineering that results in enhanced purity, stability, and batch-to-batch consistency compared to traditional hybridoma-derived antibodies . This manufacturing precision translates to improved affinity and specificity during experimental applications.
Recombinant MS4A1 antibodies are generated through a controlled process where antibody genes are amplified using specific primers designed for antibody constant regions, inserted into expression vectors, and then transfected into host cells . This genetic manipulation allows for customization of antibody properties, such as isotype switching or modification of the Fc region to alter effector functions, providing researchers with precisely tailored tools for specific experimental needs. Additionally, the recombinant production eliminates potential contamination with animal proteins or pathogens that might be present in hybridoma-derived antibodies, reducing experimental variability and improving reproducibility across different research groups .
Different recombinant monoclonal antibody clones target distinct epitopes on the MS4A1 protein, primarily focusing on the large extracellular loop between the third and fourth transmembrane domains. The extracellular loops contain multiple immunogenic epitopes that can be recognized by distinct antibody clones. Epitope specificity significantly impacts experimental and therapeutic outcomes due to differential effects on MS4A1 function, B cell signaling, and cell death mechanisms.
For research applications, understanding epitope specificity is crucial as it determines antibody functionality in various assays. Antibodies binding to certain epitopes may be excellent for flow cytometry but perform poorly in Western blotting due to epitope conformation changes during sample processing . Additionally, some epitopes may be conserved across species while others are species-specific, affecting cross-reactivity potential. Researchers should carefully select clones based on the intended application and consider whether conformational or linear epitopes are required for their specific experimental design. Documentation from manufacturers typically indicates validated applications for each clone, but preliminary testing is recommended when applying antibodies to novel experimental systems or unconventional applications .
The production of MS4A1 recombinant monoclonal antibodies follows a sophisticated molecular biological workflow that begins with B cell isolation from immunized animals, where recombinant human MS4A1 protein serves as the immunogen. Total RNA is extracted from these B cells and reverse-transcribed into cDNA . The antibody genes encoding both heavy and light chains are then amplified using PCR with primers designed specifically for antibody constant regions .
These amplified genes are subsequently inserted into expression vectors containing appropriate promoters and selection markers. The recombinant vectors are then transfected into host cells (commonly CHO, HEK293, or other mammalian cell lines) for antibody expression . Following transfection, the cells are cultured to facilitate antibody production, and the antibodies are harvested from the culture supernatant. The crude antibody preparation undergoes purification using affinity chromatography (typically Protein A or G), followed by additional chromatographic steps to ensure high purity .
The production method significantly impacts antibody characteristics, including glycosylation patterns, binding affinity, and stability. Recombinant production allows precise control over these parameters, resulting in antibodies with enhanced consistency compared to hybridoma-derived alternatives . Additionally, the recombinant approach enables antibody engineering, such as humanization or affinity maturation, to improve specificity and reduce immunogenicity in certain applications.
When selecting MS4A1 recombinant monoclonal antibodies for research, investigators should assess several critical quality parameters to ensure experimental reliability. The following evaluation framework is recommended:
Quality Parameter | Assessment Method | Acceptance Criteria |
---|---|---|
Specificity | ELISA, Western blot, Flow cytometry | Single band/peak at expected molecular weight; minimal cross-reactivity |
Sensitivity | Titration in relevant assay | Consistent detection at defined concentration threshold |
Affinity | Surface Plasmon Resonance | KD value appropriate for intended application |
Epitope mapping | Competitive binding assays | Defined epitope location that suits experimental needs |
Isotype verification | ELISA | Confirmation of expected isotype (e.g., IgG1, IgG2a) |
Endotoxin levels | LAL assay | <1 EU/mg for cell-based assays |
Aggregation | SEC-HPLC | <5% aggregates |
Functional activity | Cell-based assays | Application-specific functional readout |
Researchers should prioritize antibodies that have been validated for their specific application (e.g., flow cytometry, immunohistochemistry) . Additionally, lot-to-lot consistency should be verified through comparison of critical quality attributes between different manufacturing batches. For MS4A1 antibodies specifically, validation in B cell systems with known expression patterns provides an important functional confirmation of antibody performance . Documentation of validation data, including positive and negative controls, enhances confidence in antibody selection decisions.
Validating the specificity of MS4A1 recombinant monoclonal antibodies requires a comprehensive approach utilizing multiple complementary techniques. First, researchers should perform Western blot analysis using both MS4A1-positive (e.g., B lymphocyte cell lines like Raji or Daudi) and MS4A1-negative (e.g., T cell lines or MS4A1-knockout cells) samples to confirm specific detection of the expected 33-37 kDa band exclusively in positive samples .
Flow cytometry validation should be conducted using mixed lymphocyte populations, ensuring the antibody selectively labels B lymphocytes (CD19+) but not T lymphocytes (CD3+) in peripheral blood mononuclear cells . The staining pattern should match the expected surface membrane distribution of MS4A1. Immunohistochemistry on lymphoid tissues should demonstrate specific staining of B cell follicles with appropriate negative controls .
For advanced validation, competitive binding assays can be performed using unlabeled and labeled versions of the same antibody, or with antibodies known to target different MS4A1 epitopes. Additionally, RNA interference or CRISPR-Cas9 mediated knockdown/knockout of MS4A1 in B cell lines provides definitive confirmation of antibody specificity, as the signal should be reduced or eliminated in these genetic models . Cross-reactivity testing against related family members (other MS4A proteins) is also recommended for comprehensive specificity assessment, particularly when working with novel antibody clones or in comparative studies analyzing multiple B cell surface markers.
For optimal flow cytometric analysis using MS4A1 recombinant monoclonal antibodies, researchers should follow a systematic protocol that maximizes signal-to-noise ratio while preserving cellular viability. Begin with freshly isolated cells (peripheral blood, lymphoid tissue, or cultured B cells) and maintain them at 4°C throughout processing to prevent antibody internalization or capping. For peripheral blood samples, use 20 μl of properly titrated MS4A1 antibody per 100 μl of whole blood or 10^6 cells in suspension .
Surface staining should be performed in PBS supplemented with 1-2% BSA or FBS and 0.1% sodium azide (FACS buffer) for 20-30 minutes at 4°C in the dark. Include appropriate compensation controls when using multiple fluorochrome-conjugated antibodies. To enhance discrimination between B cell subpopulations, combine MS4A1 antibodies with other B cell markers such as CD19, CD27 (memory B cells), IgD (naive B cells), and CD38 (plasma cells) .
The MS4A1 marker facilitates identification of immature B cells, memory B cells, naive B cells, large intestine lamina propria lymphocytes, and some plasma cells . When analyzing MS4A1 expression levels (rather than simply positive/negative discrimination), calibration beads should be used to standardize fluorescence intensity across experiments. For intracellular staining protocols (examining internalized MS4A1 following receptor engagement), fixation with 2% paraformaldehyde followed by permeabilization with 0.1% saponin is recommended, with appropriate adjustment of gating strategies to account for autofluorescence changes after fixation.
Optimizing immunohistochemistry protocols for MS4A1 recombinant monoclonal antibodies requires careful attention to fixation, antigen retrieval, and detection systems. Formalin-fixed paraffin-embedded (FFPE) tissues should undergo heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0), with the optimal buffer determined empirically for each antibody clone . Retrieval conditions typically involve 20 minutes at 95-98°C, followed by gradual cooling to room temperature.
For MS4A1 antibodies, dilution optimization is critical and generally falls within the range of 1:20-1:200 for IHC applications . Researchers should perform titration experiments on positive control tissues (tonsil or lymph node sections) to determine the optimal dilution that provides specific staining with minimal background. Blocking endogenous peroxidase (3% H₂O₂ for 10 minutes) and nonspecific binding (5-10% normal serum from the same species as the secondary antibody) are essential steps before primary antibody incubation.
Detection systems should be selected based on the required sensitivity, with polymer-based detection generally offering superior results for MS4A1 visualization compared to traditional avidin-biotin complexes. For dual or multiplex staining to examine MS4A1 in relation to other markers, sequential immunostaining with appropriate blocking steps between antibody applications is recommended. Counterstaining with hematoxylin should be brief (30-60 seconds) to avoid obscuring membrane staining. Digital image analysis can enhance quantitative assessment of MS4A1 expression patterns, particularly when examining heterogeneous B cell populations within complex tissue microenvironments .
Designing experiments to study MS4A1-mediated signaling pathways using recombinant monoclonal antibodies requires careful consideration of antibody format, experimental timing, and downstream readouts. The MS4A1 protein is involved in regulating transmembrane calcium conductance and influences B cell activation, proliferation, and cell cycle progression . When investigating these pathways, researchers should consider the following experimental design elements:
First, antibody format significantly impacts signaling outcomes. F(ab')₂ fragments (lacking Fc regions) should be used to isolate direct MS4A1 signaling effects from Fc receptor-mediated responses. Antibody immobilization (plate-bound or bead-bound) generally enhances cross-linking and signaling compared to soluble antibodies . For temporal analysis of signaling cascades, researchers should establish detailed time-course experiments ranging from seconds (for calcium flux) to hours (for transcriptional changes).
Downstream readouts should include:
Immediate responses: Calcium flux measured by flow cytometry with indo-1 or fluo-4 dyes
Early signaling: Phosphorylation of proximal kinases (Lyn, Syk) assessed by phospho-specific antibodies in Western blotting or phospho-flow cytometry
Intermediate responses: Akt/ERK activation and NF-κB translocation
Late responses: Cell cycle analysis, proliferation (CFSE dilution), and survival/apoptosis markers
Control experiments should include isotype-matched antibodies, MS4A1-negative cell populations, and pharmacological inhibitors of specific signaling nodes to validate pathway specificity . For comprehensive pathway analysis, consider combining antibody-mediated MS4A1 engagement with other B cell receptor stimulation (anti-IgM) to examine co-stimulatory or regulatory interactions between these receptor systems.
Researchers working with MS4A1 recombinant monoclonal antibodies frequently encounter several technical challenges that can compromise experimental results. The following table outlines common issues and evidence-based solutions:
Additionally, MS4A1 detection can be complicated by its variable phosphorylation state, which affects its molecular weight (33-37 kDa range) . To address this, researchers should optimize sample preparation protocols to maintain consistent phosphorylation states or use phosphatase treatments when appropriate. For applications requiring detection of MS4A1 in tissue sections, heat-induced epitope retrieval optimization is critical, with different antibody clones potentially requiring distinct retrieval conditions .
Working with challenging samples or limited material requires specialized approaches to maximize MS4A1 detection sensitivity while preserving sample integrity. For rare cell populations or minimal clinical specimens, consider implementing the following optimization strategies:
For flow cytometry applications with limited cells, employ a panel-based approach that includes MS4A1 alongside other B cell markers (CD19, CD27, CD38) to enable precise gating strategies with fewer cells . Reducing assay volumes (using microplate formats or limited sample volume protocols) and optimizing antibody concentrations through careful titration can improve detection while conserving sample material. High-sensitivity flow cytometers with enhanced signal resolution capabilities may also be employed for challenging samples.
When working with formalin-fixed paraffin-embedded (FFPE) tissues that may have compromised epitopes, implement a systematic optimization of antigen retrieval conditions, testing both heat-induced epitope retrieval (HIER) and enzymatic retrieval methods at various durations . For particularly difficult samples, tyramide signal amplification or polymer-based detection systems can significantly enhance sensitivity without increasing background.
For protein analysis from limited material, consider using capillary-based immunoassay platforms (e.g., Simple Western) that require substantially less sample than traditional Western blotting while providing greater quantitative precision . Additionally, multiplexed antibody labeling strategies enable simultaneous detection of MS4A1 alongside other proteins of interest, maximizing the information obtained from each sample. Cryopreservation protocols should be carefully optimized to maintain MS4A1 epitope integrity during long-term storage, with validation studies comparing fresh and frozen samples recommended before processing valuable specimens.
Minimizing background and non-specific binding in multiplex immunofluorescence assays with MS4A1 recombinant monoclonal antibodies requires meticulous attention to blocking, antibody selection, and protocol sequencing. Implement a multi-step blocking protocol beginning with 5-10% normal serum (matched to secondary antibody species) supplemented with 0.1-0.3% Triton X-100 for 1-2 hours at room temperature . For tissues rich in endogenous biotin (e.g., lymphoid tissues), include an avidin-biotin blocking step if biotin-based detection systems are employed.
Antibody selection and preparation significantly impact non-specific binding. Use directly conjugated primary antibodies when possible to eliminate secondary antibody cross-reactivity. If unconjugated primaries are necessary, select secondary antibodies pre-adsorbed against potentially cross-reactive species . Dilute antibodies in blocking buffer and centrifuge at 10,000g for 5 minutes immediately before use to remove potential aggregates that contribute to background.
For multiplex panels including MS4A1, carefully plan the staining sequence based on antibody characteristics. Begin with the weakest signal/lowest expression target, followed by progressively stronger markers. Between antibody applications, incorporate stringent washing steps (4-5 washes with PBS containing 0.05-0.1% Tween-20) . When multiple antibodies from the same species must be used, employ tyramide signal amplification with microwave treatment between rounds to inactivate previous antibodies while preserving antigenicity.
Autofluorescence, particularly prominent in lymphoid tissues, can be mitigated through brief treatment with 0.1-1% Sudan Black B or commercial autofluorescence quenchers following immunostaining but before mounting. For quantitative analysis, include appropriate single-stained controls for spectral unmixing and fluorescence compensation, especially critical when examining MS4A1 co-expression with other B cell markers that may have overlapping cellular distributions .
MS4A1 recombinant monoclonal antibodies offer sophisticated tools for investigating B cell biology throughout developmental progression, activation states, and differentiation pathways. For developmental studies, researchers can implement flow cytometric analyses of bone marrow, fetal liver, or in vitro differentiation cultures using MS4A1 antibodies in combination with stage-specific markers (CD19, CD10, CD34, IgM) . This approach enables precise delineation of MS4A1 acquisition during the transition from pro-B to pre-B stages and subsequent expression modulation during maturation.
For activation studies, MS4A1 antibodies can serve dual roles as both detection reagents and functional modulators. When used as detection tools, they help characterize expression changes following stimulation with various activators (anti-IgM, CD40L, TLR ligands) . As functional modulators, MS4A1 antibodies can be employed to study calcium signaling alterations, with different epitope-binding clones producing distinct functional outcomes. Time-course experiments tracking MS4A1 redistribution into lipid rafts following B cell receptor engagement provide insights into the molecular organization of activation-induced signaling complexes .
In differentiation studies, the progressive downregulation of MS4A1 during plasma cell differentiation can be monitored using flow cytometry or imaging approaches. Single-cell analysis combining MS4A1 detection with transcription factor measurement (Blimp-1, XBP-1) enables correlation of protein expression changes with transcriptional reprogramming events . For in vivo tracking of B cell responses, adoptive transfer experiments using MS4A1-expressing B cells followed by recombinant antibody staining in recovered populations allows assessment of antigen-specific responses and memory formation within complex lymphoid microenvironments.
Imaging studies of B cell localization and trafficking benefit from specialized methodological approaches when employing MS4A1 recombinant monoclonal antibodies. For tissue-based imaging, multiplex immunofluorescence represents a powerful technique, combining MS4A1 detection with markers of lymphoid architecture (FDC networks, T cell zones) and activation states (proliferation markers, activation-induced cytidine deaminase) . Optimal results require careful antibody titration (typically 1:20-1:200 dilutions) and heat-induced epitope retrieval with either citrate (pH 6.0) or EDTA (pH 9.0) buffers .
For dynamic imaging of B cell trafficking, intravital microscopy using fluorescently conjugated MS4A1 Fab fragments minimizes potential functional perturbation while maintaining detection sensitivity. Two-photon microscopy of explanted lymph nodes or intravital imaging through surgically exposed lymphoid tissues allows real-time visualization of MS4A1-positive B cells interacting with antigen-presenting cells and follicular structures . To enhance signal durability during extended imaging sessions, consider photoconvertible fluorescent protein systems in combination with MS4A1 antibody labeling.
Super-resolution microscopy techniques provide unprecedented insights into MS4A1 molecular organization on the B cell surface. Stimulated emission depletion (STED) or stochastic optical reconstruction microscopy (STORM) approaches reveal nanoscale clustering patterns of MS4A1 following various stimulation conditions . For these applications, directly conjugated recombinant antibody fragments optimize spatial resolution by minimizing the distance between fluorophore and target. When designing imaging panels, carefully consider fluorophore selection to enable spectral separation while matching fluorophore brightness with target abundance – MS4A1's relatively high expression level permits the use of less bright fluorophores, reserving brighter options for less abundant markers .
Integrating MS4A1 recombinant monoclonal antibody data with multi-omics approaches creates a comprehensive framework for understanding B cell states across physiological and pathological conditions. Begin by implementing index sorting during flow cytometry, where MS4A1 expression and other protein markers are recorded for each individual cell before downstream single-cell sequencing . This strategy enables direct correlation between protein expression profiles and transcriptomic signatures at single-cell resolution.
For spatial analysis of B cell organization within tissues, combine MS4A1 immunohistochemistry with spatial transcriptomics platforms. Following digital image analysis of MS4A1 staining patterns, overlay the protein expression data with spatially resolved transcriptomes to contextualize B cell phenotypes within their microenvironmental niches . This approach is particularly valuable for studying lymphoid malignancies, autoimmune disorders, and normal germinal center reactions.
To connect MS4A1 surface expression with downstream signaling activities, implement phospho-flow cytometry panels that simultaneously detect MS4A1 alongside phosphorylated signaling molecules (p-SYK, p-BTK, p-ERK) . These multi-parameter data sets can be analyzed using dimensionality reduction techniques such as t-SNE or UMAP to identify distinct B cell populations and activation states. For integrative analysis across multiple data types, implement computational frameworks that align protein expression data (MS4A1 levels by flow cytometry), transcriptomic profiles, and functional readouts (calcium flux, proliferation) .
The resulting integrated datasets enable sophisticated characterization of B cell heterogeneity in both normal development and disease states. For example, correlating MS4A1 expression levels with transcriptomic changes during B cell lymphoma progression can identify cellular subpopulations with distinct therapeutic vulnerabilities. Similarly, in autoimmune conditions, integrated analysis may reveal associations between MS4A1 expression patterns, B cell receptor repertoire characteristics, and autoantibody production capabilities .