PTPRC recombinant monoclonal antibodies target CD45, also known as protein tyrosine phosphatase receptor type C (PTPRC), a transmembrane protein encoded by the PTPRC gene located on chromosome 1q31-32. CD45 consists of approximately 1,258 amino acids with a molecular weight ranging from 180 to 220 kDa due to extensive glycosylation and post-translational modifications, including phosphorylation and disulfide bond formation . As a transmembrane protein, CD45 is primarily localized to the plasma membrane of hematopoietic cells, where it plays essential roles in cell signaling, activation, and differentiation .
The PTPRC protein undergoes alternative splicing to generate multiple isoforms, including CD45RA (ABC and BC isoforms) and CD45RB, which are expressed differentially across immune cell populations. The CD45RA isoform is a protein of 205-220 kDa that represents a specific variant of the human leukocyte common antigen . These isoforms arise from differential splicing of three exons (A, B, and C), generating at least five distinct variants: ABC, AB, BC, B, and O . The distribution of these isoforms varies significantly across immune cell types and developmental stages, providing valuable markers for distinguishing cell subpopulations.
CD45 functions as a critical regulator of immune cell activation and signaling, particularly in T and B lymphocytes. It acts as a key tyrosine phosphatase that modulates signaling pathways downstream of antigen receptors, cytokine receptors, and co-stimulatory molecules . By dephosphorylating specific tyrosine residues on signaling molecules, CD45 helps maintain the balance of intracellular signaling cascades critical for immune cell responses .
In T-cell biology, CD45 is essential for T-cell activation through the antigen receptor and serves as a positive regulator of T-cell coactivation upon binding to DPP4 . Upon T-cell activation, CD45 recruits and dephosphorylates proteins such as SKAP1 and FYN, influencing downstream signaling events . Additionally, it dephosphorylates LYN, thereby modulating LYN activity and further influencing cellular signaling pathways .
CD45 expression is found on all nucleated hematopoietic cells, including T cells, B cells, natural killer cells, monocytes, macrophages, and granulocytes . The differential expression of CD45 isoforms is physiologically significant, with CD45RA expressed on 40-50% of peripheral CD4 T-cells, 50% of peripheral CD8 T-cells, B-cells, and leukemic B-cell lines . T-cells expressing CD45RA are considered naïve or virgin T-cells, while those expressing CD45RO represent memory T-cells, allowing for functional distinction between immune cell subsets .
The production of PTPRC recombinant monoclonal antibodies represents a sophisticated biotechnological process that ensures high specificity and consistency. The initial step involves introducing PTPRC antibody genes into plasmid vectors, followed by the transfection of these engineered plasmids into suitable host cells for expression using exogenous protein expression technology . Following successful expression, the antibodies undergo purification through affinity chromatography to isolate the target protein with high purity .
Advanced methodologies such as phage display technology have further enhanced the development of PTPRC antibodies. This approach allows for efficient screening of human single-chain variable fragments (scFvs) against specific antigens, enabling the production of fully human IgG1 monoclonal antibodies with superior specificity and reduced immunogenicity . The process typically involves:
Identification of potential antigenic peptides through bioinformatic analysis
Synthesis of selected peptides for antibody screening
Screening of human scFv libraries through phage display
Selection of high-affinity fragments for full antibody production
Expression and purification of complete monoclonal antibodies
Validation through various analytical techniques
These antibodies undergo rigorous validation for specific applications, including ELISA, flow cytometry, and immunohistochemistry, ensuring their reliability for research purposes .
PTPRC recombinant monoclonal antibodies are available in various formats and classifications, each designed for specific applications and targeting different epitopes of the CD45 protein. The following table presents a comprehensive overview of different PTPRC antibody types currently available:
These antibodies vary in their specificity toward different CD45 isoforms, making them valuable tools for distinguishing between cell populations. For instance, antibodies targeting CD45RA can effectively identify naïve T-cells and B-cells, while those against CD45RB target a different subset of immune cells . The selection of the appropriate antibody depends on the specific research question, target cell population, and intended application.
PTPRC recombinant monoclonal antibodies offer numerous advantages over traditional monoclonal antibodies produced through hybridoma technology. The recombinant approach provides:
Increased sensitivity: Recombinant antibodies typically demonstrate enhanced binding affinities, with some PTPRC antibodies exhibiting nanomolar binding constants as confirmed by surface plasmon resonance (SPR) studies . This increased sensitivity enables the detection of lower antigen concentrations, enhancing research outcomes.
Confirmed specificity: Through genetic engineering, recombinant antibodies can be designed to target specific epitopes with exceptional precision. This results in minimized cross-reactivity and more reliable experimental data. For example, recombinant anti-PTPRC antibodies have demonstrated the ability to selectively bind to tumor T lymphocytes expressing specific TCR segments without binding to other lymphocytes or blood cell components .
High repeatability: The genetically defined nature of recombinant antibodies ensures consistent performance across experiments, enhancing reproducibility in research settings.
Animal-free production: Many recombinant antibody production systems eliminate or significantly reduce the need for animal immunization, addressing ethical concerns associated with traditional antibody production methods .
Customizable features: Recombinant technology allows for the incorporation of specific tags, conjugates, or modifications to enhance functionality. For instance, PTPRC antibodies can be conjugated with fluorochromes like PE-Cy5 for flow cytometry applications or biotin for detection systems .
A major advantage of recombinant monoclonal antibodies is their exceptional batch-to-batch consistency, which addresses one of the significant limitations of traditional hybridoma-derived antibodies. This consistency stems from several factors:
Defined genetic sequence: Recombinant antibodies are produced from a known genetic sequence, eliminating the variability introduced by hybridoma drift or inconsistent animal immune responses.
Controlled expression systems: Production in well-characterized expression systems allows for standardized culture conditions and protein expression.
Standardized purification protocols: Consistent purification methods ensure uniform antibody quality across different manufacturing batches.
Long-term sustainable supply: The genetic information encoding the antibody can be stored indefinitely, ensuring continuous availability of identical antibodies for longitudinal studies.
Reduced lot-to-lot variation: Studies have demonstrated that recombinant PTPRC antibodies maintain consistent binding properties and specificity across different production lots, enhancing experimental reproducibility .
For researchers, these quality factors translate into more reliable experimental results, reduced need for antibody validation across batches, and improved data comparability across different research studies or time points.
PTPRC recombinant monoclonal antibodies serve as versatile tools across various laboratory techniques. Their applications include:
Flow Cytometry (FC): PTPRC antibodies are extensively used in flow cytometry for immunophenotyping of hematopoietic cells. Different isoform-specific antibodies can distinguish between naïve and memory T-cells, making them valuable for studying immune cell development and activation states . Recommended dilutions for flow cytometry typically range from 1:20 to 1:500, depending on the specific antibody and experimental conditions .
Western Blotting (WB): These antibodies can detect CD45 protein in cell or tissue lysates, enabling quantitative analysis of protein expression. The high specificity of recombinant antibodies reduces background signal and improves data quality .
Immunohistochemistry (IHC) and Immunocytochemistry (ICC): PTPRC antibodies are used to visualize CD45 expression in tissue sections or fixed cells, aiding in the identification of immune cell infiltration in tissues and the characterization of hematopoietic malignancies .
Enzyme-Linked Immunosorbent Assay (ELISA): Certain PTPRC recombinant antibodies are validated for ELISA applications, providing quantitative measurement of CD45 levels in biological samples .
Immunoprecipitation (IP): Some clones, such as UCH-L1, are suitable for immunoprecipitation studies, allowing researchers to isolate CD45 and its binding partners for further analysis .
Protocol considerations for optimal results with PTPRC recombinant monoclonal antibodies include:
Proper sample preparation to preserve epitope integrity
Appropriate blocking steps to minimize non-specific binding
Careful titration to determine optimal antibody concentration
Selection of suitable detection systems based on the application
Inclusion of appropriate positive and negative controls
Beyond basic research applications, PTPRC recombinant monoclonal antibodies hold significant value in clinical research contexts:
Diagnostic Applications: CD45RA antibodies are useful in differentiating T-cell lymphomas (typically CD45RO positive) from B-cell lymphomas (typically CD45RA positive), aiding in the accurate classification of hematological malignancies . This distinction is critical for proper diagnosis and treatment selection.
Immune Monitoring: These antibodies facilitate the monitoring of immune cell populations in various disease states, including immunodeficiencies, autoimmune disorders, and infectious diseases. For instance, tracking naïve versus memory T-cell ratios provides insights into immune status and response to therapies.
Therapeutic Target Identification: Research using PTPRC antibodies has contributed to the identification of potential therapeutic targets in immune-related disorders. Studies have demonstrated that targeting specific V segments of the TCR beta chain with human monoclonal antibodies represents a potential therapeutic option for patients with mature T-cell neoplasms .
Biomarker Development: CD45 isoform expression patterns serve as potential biomarkers for disease progression or response to therapy in various conditions, with PTPRC antibodies enabling their reliable detection and quantification.
Cell Therapy Research: In the evolving field of cell-based therapies, these antibodies aid in the characterization and quality control of therapeutic cell products, ensuring their safety and efficacy.
The clinical research applications of PTPRC recombinant monoclonal antibodies continue to expand as our understanding of immune cell biology and its role in disease pathogenesis deepens.
Recent research utilizing PTPRC recombinant monoclonal antibodies has significantly enhanced our understanding of immune cell biology and pathology:
The development of PTPRC recombinant monoclonal antibodies has opened new avenues for potential therapeutic applications:
Targeted Therapy for T-cell Neoplasms: The development of fully human IgG1 monoclonal antibodies targeting specific V segments of the TCR beta chain represents a promising therapeutic direction for patients with mature T-cell neoplasms . This approach offers the potential for highly specific treatments that target malignant T-cell clones while sparing healthy immune cells.
Immunomodulatory Applications: Given CD45's role in regulating immune cell activation, antibodies that modulate its function could potentially be used to treat autoimmune disorders or enhance immune responses in certain contexts.
Diagnostic Applications in Lymphoma Classification: CD45RA antibodies have demonstrated utility in differentiating T-cell lymphomas from B-cell lymphomas, potentially improving diagnostic accuracy and treatment selection .
Future research directions in this field include:
Development of antibody-drug conjugates targeting specific CD45 isoforms for targeted therapy of hematological malignancies
Engineering bispecific antibodies that simultaneously target CD45 and other immune checkpoints to enhance anti-tumor immune responses
Further exploration of CD45's role in various immune disorders to identify new therapeutic targets
Refinement of recombinant antibody production methods to improve yield, reduce costs, and enhance accessibility for research and clinical applications
Investigation of CD45 as a potential target for immunotherapy approaches in solid tumors with significant immune cell infiltration
The production of the PTPRC recombinant monoclonal antibody involves the initial step of introducing PTPRC antibody genes into plasmid vectors. These engineered plasmids are subsequently introduced into appropriate host cells for expression using exogenous protein expression technology. Following this, the PTPRC recombinant monoclonal antibody undergoes a purification process using affinity chromatography. It has undergone stringent validation for specific applications, including ELISA and FC. Notably, this antibody exhibits exclusive recognition of the human PTPRC protein.
PTPRC (CD45) plays a crucial role in regulating immune cell activation and signaling. It fine-tunes immune responses by modulating the phosphorylation status of key signaling molecules, ensuring proper immune cell development, antigen recognition, activation, and immune response regulation. Dysregulation of PTPRC can lead to immune disorders and compromised immune function.
PTPRC monoclonal antibodies have been validated for multiple research applications including Western Blot (WB), Immunohistochemistry (IHC), Immunocytochemistry (ICC), Immunofluorescence (IF), and Flow Cytometry . When selecting a specific antibody clone for your experiment, verify that it has been specifically validated for your intended application.
For optimal experimental design:
Flow cytometry applications typically use 1:100-1:500 dilutions
IHC and IF applications generally start with 1:200 dilutions, adjusting based on signal-to-noise ratio
For western blotting, reducing conditions are typically suitable unless specifically contraindicated
Validation with appropriate positive controls (lymphoid tissues) is essential regardless of application
The antibody validation should include specificity testing, especially when investigating novel tissue types or experimental conditions not previously reported in literature.
Based on published literature and experimental evidence, PTPRC expression has been documented in multiple tissue types:
When designing experiments, include appropriate positive and negative control tissues. Lymphoid tissues consistently show strong PTPRC expression and serve as reliable positive controls, while epithelial tissues typically show minimal expression and can function as negative controls. Researchers should note that positive staining in placenta has been confirmed by multiple sources, though this may be confined to specific cell populations within the tissue .
To maximize antibody stability and performance, implement these evidence-based storage protocols:
Long-term storage: Store antibodies at -20°C for up to one year . For preservation beyond one year, consider dividing into smaller aliquots to minimize freeze-thaw cycles.
Short-term storage: For frequent use within a one-month period, keep antibodies at 4°C to avoid repeated freezing and thawing .
Freeze-thaw management: Each freeze-thaw cycle typically reduces antibody activity by approximately 10-15%. After 5 cycles, significant degradation may occur, compromising experimental results.
Aliquoting strategy: Upon receipt, immediately divide antibodies into small working aliquots (10-20 μl) before freezing to prevent multiple freeze-thaw cycles of the stock solution.
Cryoprotectant addition: Some researchers report improved stability by adding 50% glycerol to antibody preparations, though this requires adjustment of working dilutions in downstream applications.
When working with the antibody, always allow it to thaw completely at room temperature or 4°C rather than using heat, which can permanently denature the protein structure .
Implementing rigorous validation protocols is essential for confirming antibody specificity:
Positive and negative tissue controls: Use lymphoid tissues as positive controls and tissues with minimal PTPRC expression as negative controls.
Antibody titration: Perform a dilution series (e.g., 1:50, 1:100, 1:200, 1:500) to identify the optimal concentration that maximizes specific signal while minimizing background.
Blocking peptide validation: Pre-incubate the antibody with a blocking peptide corresponding to the immunogen. Specific staining should be significantly reduced or eliminated.
Genetic validation: Utilize CRISPR-Cas9 knockout or siRNA knockdown models as definitive negative controls. This approach has been documented in the literature for PTPRD and is applicable to PTPRC validation as well .
Multi-antibody approach: Use two or more antibodies targeting different epitopes of PTPRC to confirm consistent staining patterns.
For unexpected staining patterns, particularly in tissues like placenta where researchers have reported positive staining , consult current literature to determine whether this represents true biological expression rather than non-specific binding. When encountering positive staining in blood samples, this is consistent with expected PTPRC expression patterns as documented in multiple sources .
Tissue-specific optimization is critical for successful PTPRC detection:
For lymphoid tissues (high expression):
Shorter primary antibody incubation times (1-2 hours at room temperature)
Lower antibody concentrations (1:200-1:500 dilutions)
Standard antigen retrieval methods (citrate buffer, pH 6.0)
For placental tissue (variable expression):
Extended antigen retrieval may be necessary
Higher antibody concentrations (1:50-1:100)
Extended primary antibody incubation (overnight at 4°C)
For liver tissue:
Enhanced blocking of endogenous peroxidase activity
Careful selection of visualization system to minimize background
Comparison with CD45 mRNA expression data for validation
When examining tissues with potential low-level expression, signal amplification systems such as polymer-based detection methods can significantly improve sensitivity without compromising specificity. For frozen tissue sections, researchers have specifically asked about placenta applications, and suppliers have confirmed that the antibody works in this context .
PTPRC undergoes alternative splicing to generate multiple isoforms (CD45RA, CD45RB, CD45RC, CD45RO) with distinct functional properties:
Epitope selection considerations:
Antibodies targeting constant regions detect all isoforms
Antibodies recognizing variable exon products (A, B, C) are isoform-specific
Review epitope mapping data from manufacturers to determine specificity
Experimental validation methodology:
Use cell lines with known isoform expression profiles as controls
Employ recombinant isoform proteins for Western blot comparison
Design multi-color flow cytometry panels including established isoform markers
Perform RT-PCR with isoform-specific primers as complementary validation
Flow cytometry analysis recommendations:
When differentiating naive vs. memory T cells (CD45RA vs. CD45RO), include lineage markers
Apply appropriate compensation controls for multi-color experiments
Consider fluorescence-minus-one (FMO) controls for accurate gating
From the search results, researchers have specifically asked about isoform reactivity , indicating this is an important consideration in experimental design. For studies requiring precise isoform discrimination, combining multiple antibody clones targeting different epitopes significantly enhances specificity and confidence in results.
Based on multiple queries in the search results regarding placental applications , this represents an area of significant research interest:
Tissue preparation considerations:
Fresh frozen sections: Better preserve antigenicity but have poorer morphology
FFPE sections: Provide superior morphology but may require more aggressive antigen retrieval
Section thickness: 5-7 μm sections typically provide optimal results
Fixation protocols: For frozen sections, post-fixing in cold acetone for 10 minutes often yields good results
Antigen retrieval optimization:
For FFPE placental tissue, heat-induced epitope retrieval using citrate buffer (pH 6.0) for 20 minutes
Enzymatic retrieval using proteinase K can be effective but may compromise tissue morphology
Pressure cooker-based retrieval may improve results for challenging samples
Result interpretation guidelines:
PTPRC expression in placenta is primarily associated with tissue-resident immune cells
Positive staining should be correlated with cell morphology and distribution patterns
Serial sections stained with markers for different immune cell populations help identify specific PTPRC-positive cell types
According to multiple sources cited in the search results, PTPRC expression in placenta has been confirmed , so positive staining in this tissue type represents a valid finding when properly controlled. Researchers should document the specific placental compartments (decidual, villous, etc.) where staining is observed, as expression may be compartmentalized.
When facing discrepancies between different detection methods:
Technical considerations:
Western blot detects denatured protein while IHC/IF detect proteins in their native conformation
Epitope accessibility differs significantly between methods
Fixation protocols for IHC may mask certain epitopes
Protein extraction methods for Western blot may favor certain protein populations
Systematic troubleshooting approach:
Test multiple antibody clones targeting different epitopes
Employ different protein extraction methods for Western blot
Try multiple fixation and antigen retrieval protocols for IHC
Consider that proteolytic processing may occur differently across sample types
Reconciliation strategies:
Use phosphatase activity assays as functional validation
Correlate protein detection with mRNA expression
Employ genetic models (knockouts, knockdowns) as definitive controls
Consider subcellular fractionation before Western blotting
Researchers have reported successful PTPRC detection in both Western blot and IHC applications , though protocol optimization may be necessary depending on the specific tissue being examined. For placental tissue specifically, researchers have validated positive PTPRC expression through both methodologies .
Based on researcher inquiries in the search results , there is significant interest in antibody conjugation strategies:
Pre-conjugation requirements:
BSA-free formulation: The antibody must be free of carrier proteins like BSA that would interfere with conjugation chemistry
Buffer exchange: Replace sodium azide-containing buffers with PBS or another compatible buffer
Concentration: Optimal antibody concentration for conjugation is typically 1-5 mg/ml
Conjugation chemistry options:
NHS-ester biotinylation: Most common approach targeting primary amines (lysine residues)
Maleimide-based conjugation: For site-specific labeling through reduced disulfide bonds
Click chemistry: For site-specific conjugation with minimal impact on binding properties
Optimization parameters:
Biotin:antibody ratio: Test multiple ratios (10:1, 20:1, 30:1) to determine optimal conjugation without compromising binding
Reaction conditions: pH 7.2-8.0, temperature, and duration significantly impact conjugation efficiency
Purification method: Dialysis, gel filtration, or spin columns to remove unreacted biotin
Post-conjugation storage:
Validation requirements:
Compare reactivity pre- and post-conjugation
Determine optimal working concentration (typically higher than unconjugated antibody)
Verify detection specificity in relevant experimental systems
The search results specifically mention researchers' interest in BSA-free formulations for biotin conjugation purposes , confirming this as an important consideration for advanced applications of PTPRC antibodies.
For challenging samples requiring enhanced sensitivity and specificity:
Signal amplification systems:
Tyramide Signal Amplification (TSA): Increases sensitivity by 10-100 fold
Polymer-based detection: Provides enhanced sensitivity without increased background
QDot-conjugated secondaries: Offer higher photostability and brighter signals
Background reduction strategies:
Extended blocking (2-3 hours) using combined blocking agents (serum, BSA, casein)
Autofluorescence quenching with Sudan Black B for fluorescence applications
Fc receptor blocking for immune cell-rich tissues
Optimization of antibody concentrations through careful titration
Advanced microscopy techniques:
Confocal microscopy with spectral unmixing to distinguish signal from autofluorescence
Super-resolution microscopy for subcellular localization studies
Digital image analysis for objective quantification of signal intensity
Sample pre-enrichment:
Cell sorting of specific populations before analysis
Laser capture microdissection for tissue region isolation
Subcellular fractionation for Western blot applications
Researchers have successfully detected PTPRC in challenging tissues like placenta , demonstrating that with appropriate optimization, reliable detection is achievable even in tissues with potentially low expression or complex compositions.