PDCD1 Human, Active

Programmed Cell Death 1 Human Recombinant, Active
Shipped with Ice Packs
In Stock

Description

Protein Structure

PD-1 is a 268–288 amino acid monomer with:

  • IgV-like extracellular domain: Binds ligands PD-L1 and PD-L2 .

  • Transmembrane region: Anchors the protein to the cell membrane .

  • Intracellular tail: Contains immunoreceptor tyrosine-based inhibitory motif (ITIM) and switch motif (ITSM), which recruit SHP-1/2 phosphatases to suppress T-cell receptor (TCR) signaling .

FeatureDescriptionSource
Molecular Weight17–44 kDa (varies by glycosylation and tag)
TagsC-terminal 6×His, Fc, or polyhistidine tags for purification and detection
Expression SystemsHEK293 cells, Sf9 baculovirus, or CHO cells

Recombinant Production

PDCD1 Human, Active is produced via recombinant DNA technology, often in mammalian or insect cell lines:

ProducerTagMolecular WeightApplicationsSource
GenScriptC-terminal 6×His30–40 kDaPD-L1/PD-L2 binding assays
Prospec BiohIgG-His (C-terminal)44.0 kDaImmunological studies, ELISA
Active MotifNone (baculovirus)25–167 a.a.T-cell activation inhibition studies

Bioactivity Assays

Recombinant PD-1 demonstrates functional binding to ligands and therapeutic antibodies:

Ligand/TargetBinding Affinity (ED₅₀/ Kd)ApplicationSource
PD-L1 Fc Chimera24–390 ng/mL (linear range)ELISA-based binding assays
Nivolumab (Opdivo)0.1–3 ng/mLNeutralization assays
Keytruda (Pembrolizumab)0.017–1.37 ng/mLAntibody competition studies

Mechanism of Action

PD-1 engagement with PD-L1/PD-L2 suppresses T-cell responses by:

  • Inhibiting PI3K/Akt pathway: Reduces cytokine production (IFN-γ, IL-2, TNF-α) .

  • Promoting T-cell exhaustion: Observed in chronic infections and cancer .

Clinical Relevance

DiseasePD-1 Expression PatternTherapeutic TargetingSource
MelanomaHigh PD-L1 on tumor cellsPD-1 inhibitors (e.g., nivolumab)
HPV-Related LesionsIncreased PD-1 in low-grade CINLinked to immune evasion
Chronic InfectionsT-cell exhaustionPD-1 blockade to restore immunity

PDCD1 Polymorphisms

The PDCD1 -606G>A polymorphism (rs36084323) influences PD-1 expression:

  • A allele carriers: Higher PDCD1 mRNA and PD-1 protein in low-grade cervical lesions .

  • HPV interaction: PD-1 levels decrease in high-grade lesions with HPV, suggesting viral immune evasion .

miRNA Regulation

miR-15a-5p and miR-16-5p directly target PDCD1 mRNA, reducing PD-1 expression by ~70% in transfected models .

PD-1 Inhibitors in Cancer

Combination therapies (e.g., anti-PD-1 + anti-CTLA-4) enhance tumor-infiltrating CD8+ T cells:

TherapyMechanismClinical EfficacySource
Nivolumab + IpilimumabDual checkpoint blockade10-fold increase in tumor CD8+ T cells
PD-L1 expressionPredictive biomarkerHigher response in PD-L1+ tumors

Limitations in PD-1 Targeting

  • Resistance mechanisms: Tumor mutational burden (TMB) and neoantigen load influence response variability .

  • Toxicity: Anti-PD-1 therapies risk autoimmune side effects (e.g., colitis, thyroiditis) .

Emerging Strategies

  • Bispecific antibodies: Targeting PD-1 with tumor-specific antigens to enhance specificity .

  • miRNA therapeutics: Modulating miR-15a-5p/miR-16-5p to downregulate PD-1 in cancers .

Product Specs

Introduction

Programmed cell death protein 1 (PDCD1) is a protein found on the surface of immune cells. It plays a critical role in regulating immune responses by interacting with its ligand, PD-L1. PDCD1 is involved in suppressing immune responses, helping to prevent autoimmune reactions. Studies have shown that PDCD1 is expressed on various immune cells, including T cells and B cells, and its expression can be induced upon activation. Research using mice models has indicated a potential role for PDCD1 in T cell function and the suppression of autoimmunity. For instance, mice lacking the PDCD1 gene on a specific genetic background (BALB/c) developed heart muscle disease (dilated cardiomyopathy) and experienced heart failure. These findings suggest that PDCD1 may contribute to maintaining normal heart function by regulating immune responses in the heart.

Description

This product consists of the human PDCD1 protein, which has been produced in a lab setting using HEK293 cells. This particular format of PDCD1 is a single chain containing 383 amino acids, with a portion of the protein (amino acids 21-170) representing the active part. To facilitate purification and detection, a tag (hIgG-His-Tag) is attached to the end of the protein. The final purified product appears as a single band on a protein gel (SDS-PAGE), indicating a high level of purity. Glycosylation, a common modification in proteins, is present in this recombinant PDCD1.

Physical Appearance

The product is a clear, colorless liquid that has been sterilized by filtration.

Formulation

The PDCD1 protein is supplied in a solution containing phosphate buffered saline (pH 7.4) at a concentration of 0.5 mg/ml, along with 10% glycerol.

Stability

For short-term storage (up to 4 weeks), keep the product refrigerated at 4°C. For extended storage, freeze the PDCD1 solution at -20°C. To prevent potential damage from repeated freezing and thawing, it is recommended to divide the solution into smaller aliquots before freezing. Adding a carrier protein (0.1% HSA or BSA) can further enhance stability during long-term storage.

Purity

The purity of the PDCD1 protein is greater than 95%, as determined by SDS-PAGE analysis.

Biological Activity

The biological activity of this PDCD1 protein has been confirmed through its ability to bind to its ligand, PD-L1/B7-H1, in a functional ELISA assay.

Synonyms

Programmed cell death protein 1, CD279, hPD-1, SLEB2, hPD-l, hSLE1, PD-1, PD1, PDCD1.

Source

HEK293 Cells.

Amino Acid Sequence

PGWFLDSPDR PWNPPTFSPA LLVVTEGDNA TFTCSFSNTS ESFVLNWYRM SPSNQTDKLA AFPEDRSQPG QDCRFRVTQL PNGRDFHMSV VRARRNDSGT YLCGAISLAP KAQIKESLRA ELRVTERRAE VPTAHPSPSP RPAGQFQTLV LEPKSCDKTH TCPPCPAPEL LGGPSVFLFP PKPKDTLMIS RTPEVTCVVV DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS VLTVLHQDWL NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS RDELTKNQVS LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK

Q&A

What is PDCD1 and what is its role in immune regulation?

PDCD1 (Programmed Cell Death Protein 1), also known as PD-1 or CD279, is a cell surface receptor expressed on T cells and B cells that plays a crucial role in regulating immune responses. It functions by down-regulating the immune system and promoting self-tolerance by suppressing T cell inflammatory activity. This immune checkpoint protein guards against autoimmunity through two primary mechanisms: promoting apoptosis of antigen-specific T-cells in lymph nodes and reducing apoptosis in regulatory T cells that have suppressive functions. While this prevents autoimmune diseases, tumor cells can exploit this pathway to evade immune surveillance by overexpressing PD-L1, the ligand for PD-1 .

What is the molecular structure of human PD-1 protein?

Human PD-1 is a type I membrane protein consisting of 288 amino acids. Structurally, it belongs to the extended CD28/CTLA-4 family of T cell regulators and includes an extracellular IgV domain, a transmembrane region, and an intracellular tail. The intracellular domain contains two phosphorylation sites located within an immunoreceptor tyrosine-based inhibitory motif (ITIM) and an immunoreceptor tyrosine-based switch motif (ITSM). These structural features enable PD-1 to negatively regulate T-cell receptor (TCR) signals through the binding of SHP-1 and SHP-2 phosphatases to its cytoplasmic tail . When expressed as a recombinant protein with a polyhistidine tag, human PD-1 has a calculated molecular weight of approximately 16.8 kDa, but migrates as 31-44 kDa in SDS-PAGE due to glycosylation .

How do PD-1 binding interactions work with its ligands?

PD-1 binds to two ligands: PD-L1 (B7-H1) and PD-L2 (B7-DC). These binding interactions have different affinities that affect downstream signaling pathways. Based on bio-layer interferometry (BLI) assays, human PD-1 binds to PD-L1 with an affinity constant of approximately 38.9 nM, while it binds to PD-L2 with a higher affinity of about 16.3 nM . This differential binding may explain the varied biological effects observed in different tissues and immunological contexts. When PD-1 engages with its ligands, it triggers intracellular signaling that leads to the inhibition of T cell proliferation, cytokine production, and cytolytic activity, ultimately dampening the immune response .

What are the optimal storage and reconstitution conditions for recombinant human PD-1 protein?

For optimal stability and activity, lyophilized human PD-1 protein should be stored at -20°C or lower. When reconstituting the protein, it's critical to follow the specific instructions provided in the Certificate of Analysis. Most preparations are lyophilized from filtered solutions in PBS (pH 7.4) with trehalose as a protectant . Repeated freeze-thaw cycles should be strictly avoided as they can lead to protein degradation and loss of functional activity. Following reconstitution, the protein solution can typically be stored at 4°C for short-term use (1-2 weeks) or aliquoted and stored at -80°C for longer periods . Always verify protein integrity before use, especially for sensitive applications like binding assays or functional studies.

How can researchers verify the purity and activity of recombinant human PD-1?

Researchers can employ multiple complementary techniques to verify both the purity and activity of recombinant human PD-1:

  • Purity assessment: SDS-PAGE under reducing conditions is the standard method, with properly prepared human PD-1 showing >95% purity. The protein typically appears as a band between 31-44 kDa due to glycosylation, despite its calculated molecular weight of 16.8 kDa .

  • Structural integrity: Size-exclusion chromatography coupled with multi-angle light scattering (SEC-MALS) can verify that the molecular weight falls within the expected range of 25-40 kDa and confirm aggregation levels below 5% .

  • Functional activity verification: Conduct binding assays to confirm the protein's ability to interact with its known ligands. For example:

    • Immobilize human PD-1 at 2 μg/mL (100 μL/well) and test binding to PD-L2 with a linear detection range of 10-156 ng/mL

    • Perform bio-layer interferometry (BLI) to measure binding kinetics with PD-L1 (KD ≈ 38.9 nM) and PD-L2 (KD ≈ 16.3 nM)

    • Test interaction with therapeutic antibodies such as Nivolumab, which should bind with an affinity constant of approximately 5.09 nM

What methods can be used to study PD-1:PD-L1/PD-L2 interactions in vitro?

Several robust methods are available to study PD-1:PD-L1/PD-L2 interactions:

  • Enzyme-Linked Immunosorbent Assay (ELISA): Immobilize human PD-1 protein (2 μg/mL, 100 μL/well) and measure binding with labeled PD-L1 or PD-L2. This approach allows for quantitative measurement with linear detection ranges (e.g., 10-156 ng/mL for PD-L2 binding) .

  • Bio-Layer Interferometry (BLI): Load His-tagged PD-1 onto HIS1K biosensors to measure real-time binding kinetics with Fc-tagged PD-L1 or PD-L2. This yields affinity constants (KD) of approximately 38.9 nM for PD-L1 and 16.3 nM for PD-L2 .

  • Surface Plasmon Resonance (SPR): Capture antibodies like Nivolumab on CM5 chips via anti-human IgG Fc antibodies, then measure binding to human PD-1 with high sensitivity (KD ≈ 4.94 nM) .

  • Cell-Based Assays: Develop reporter cell lines expressing PD-1 and measure signaling outcomes when exposed to PD-L1/PD-L2 expressing cells, with or without blocking antibodies.

  • Microscopy Techniques: Use fluorescently labeled proteins to visualize binding interactions and co-localization in fixed or live cells through techniques like FRET (Förster Resonance Energy Transfer).

How do engineered high-affinity PD-1 variants compare to anti-PD-L1 antibodies in cancer immunotherapy?

Engineered high-affinity PD-1 variants represent a promising alternative to conventional anti-PD-L1 antibodies in cancer immunotherapy, with several important advantages:

  • Superior tumor penetration: High-affinity PD-1 variants (110 pM binding affinity) demonstrate enhanced tumor penetration compared to anti-PD-L1 antibodies, likely due to their smaller size .

  • Preserved immune cell populations: Unlike antibodies that can induce depletion of peripheral effector T cells through Fc-mediated mechanisms, engineered PD-1 variants do not trigger this unwanted effect .

  • Efficacy against large tumors: In syngeneic CT26 tumor models, high-affinity PD-1 variants maintained effectiveness against both small (50 mm³) and large tumors (150 mm³), whereas anti-PD-L1 antibodies completely lost activity against larger tumors .

  • Dual diagnostic and therapeutic potential: Beyond therapeutic applications, high-affinity PD-1 variants can be used as PET imaging tracers to non-invasively assess tumor PD-L1 expression status, enabling better patient stratification for immunotherapy .

These findings suggest that engineered high-affinity PD-1 variants may overcome key limitations of antibody-based approaches, particularly for treating advanced solid tumors where penetration is crucial for efficacy.

What are the key considerations when selecting anti-PD-1 antibodies for research applications?

Selecting the appropriate anti-PD-1 antibody for research requires careful consideration of several factors:

  • Species reactivity: Determine whether you need anti-mouse or anti-human PD-1 antibodies based on your experimental model. Use anti-mouse antibodies for mouse models expressing mouse PD-1 and anti-human antibodies for humanized mice or human cell cultures .

  • Antibody clone selection: For mouse models, consider the specific properties of available clones:

    • Clone 29F.1A12 has approximately 100-fold higher PD-1 blocking affinity than RMP1-14 and more closely models human therapeutic antibodies

    • RMP1-14 has the most extensive publication record for in vivo blocking but is limited to this application

    • Clones 29F.1A12 and J43 can be used for both in vivo blocking and additional applications like western blotting, immunohistochemistry, and flow cytometry

  • Mechanism of action: Confirm that the antibody functions through the desired mechanism—most anti-PD-1 antibodies work by sterically blocking PD-1:PD-L1/PD-L2 interactions rather than depleting PD-1-expressing cells .

  • Published validation: Review the publication record for each antibody clone to find examples using similar experimental approaches to your own. Search for specific combinations (e.g., "RMP1-14 MC38 BALB/c") to find the most relevant literature .

  • Antibody format: Consider specialized formats like recombinant chimeric antibodies with mouse constant regions to reduce immunogenicity in sensitive mouse models, or Fc-silenced versions that cannot bind to endogenous Fcγ receptors when Fc-mediated effects are undesirable .

How can tissue expression analysis of PDCD1 inform experimental design in immunology research?

Understanding the tissue expression pattern of PDCD1 is crucial for designing meaningful immunology experiments:

  • Target tissue selection: PDCD1 shows cytoplasmic expression in a subset of cells in lymphoid germinal centers, making lymphoid tissues like lymph nodes particularly relevant for studying PD-1 biology .

  • Control tissue considerations: When designing flow cytometry or immunohistochemistry experiments, select appropriate positive and negative control tissues based on known expression patterns. Tissues with minimal PDCD1 expression can serve as negative controls .

  • Single-cell analysis approach: Given that PDCD1 is expressed in only a subset of immune cells, single-cell approaches rather than bulk tissue analysis will provide more accurate insights into its biological functions .

  • Dynamic regulation assessment: Design experiments that capture the dynamic regulation of PDCD1 expression, which changes during immune cell activation, exhaustion, and in response to cytokines .

  • Correlation with functional outcomes: When studying PDCD1 expression, simultaneously assess functional parameters (cytokine production, proliferation, cytotoxicity) to establish correlations between expression levels and immune cell functionality .

This understanding helps researchers focus on relevant cell populations and tissues, design appropriate controls, and interpret results in the context of normal PDCD1 expression patterns.

What post-translational modifications occur in human PD-1 and how do they affect function?

Human PD-1 undergoes several post-translational modifications that significantly influence its function:

  • N-glycosylation: This is the most prominent modification, causing PD-1 to migrate at 31-44 kDa in SDS-PAGE despite its calculated molecular weight of 16.8 kDa . These glycosylation patterns can affect:

    • Protein stability and half-life in circulation

    • Binding affinity to PD-L1 and PD-L2

    • Interaction with other immune regulatory molecules

  • Phosphorylation: The intracellular domain contains phosphorylation sites within the immunoreceptor tyrosine-based inhibitory motif (ITIM) and immunoreceptor tyrosine-based switch motif (ITSM) . Phosphorylation at these sites is crucial for:

    • Recruitment of SHP-1 and SHP-2 phosphatases

    • Negative regulation of TCR signaling

    • Downstream inhibitory pathways that suppress T cell function

  • Ubiquitination: Although less studied, ubiquitination likely regulates PD-1 turnover and internalization, affecting its surface expression levels and duration of signaling.

Understanding these modifications is critical for producing functional recombinant PD-1 proteins for research, as expression systems that cannot properly perform these modifications may yield proteins with altered binding properties or biological activities.

How does PD-1 signaling mechanistically suppress T cell function?

PD-1 signaling suppresses T cell function through multiple mechanistic pathways:

  • Inhibition of TCR and CD28 signaling: When PD-1 binds to PD-L1/PD-L2, its intracellular domain becomes phosphorylated, recruiting SHP-1 and SHP-2 phosphatases. These phosphatases dephosphorylate and inactivate proximal signaling molecules downstream of the T cell receptor (TCR) and CD28 co-stimulatory receptor .

  • Metabolic reprogramming: PD-1 engagement leads to:

    • Inhibition of glycolysis

    • Promotion of fatty acid oxidation

    • Decreased mitochondrial function

    • Impaired amino acid metabolism

  • Cell cycle regulation: PD-1 signaling blocks cell cycle progression by:

    • Inhibiting cyclin-dependent kinases

    • Upregulating cell cycle inhibitors

    • Preventing expression of transcription factors needed for proliferation

  • Transcriptional reprogramming: PD-1 signaling alters the expression of multiple genes involved in:

    • Cytokine production and signaling

    • Effector function

    • Cell survival

    • Migration and tissue retention

  • Altered T cell differentiation: Chronic PD-1 signaling promotes the development of exhausted T cells with diminished effector functions and distinct transcriptional profiles compared to functional effector or memory T cells.

Understanding these mechanisms has been crucial for developing effective checkpoint inhibitor therapies that restore T cell function in cancer and chronic infections.

What are common issues in PD-1 protein preparation and how can they be addressed?

Researchers frequently encounter several challenges when working with recombinant human PD-1 protein:

  • Protein aggregation:

    • Issue: PD-1 proteins can form aggregates during expression, purification, or storage.

    • Solution: Maintain protein aggregation levels below 5% by optimizing buffer conditions, adding stabilizers like trehalose, avoiding freeze-thaw cycles, and using techniques like SEC-MALS to verify monomeric status .

  • Inconsistent glycosylation:

    • Issue: Variable glycosylation patterns between protein batches can affect functional studies.

    • Solution: Use consistent expression systems (preferably mammalian cells), verify glycosylation status by SDS-PAGE migration pattern (31-44 kDa for properly glycosylated PD-1), and include glycosylation analysis in quality control .

  • Endotoxin contamination:

    • Issue: Endotoxin contaminants can trigger immune responses that confound results.

    • Solution: Use low-endotoxin preparation methods, verify endotoxin levels (<1EU/mg), and consider specialized low-endotoxin formulations for sensitive applications .

  • Loss of binding activity:

    • Issue: Improper handling can lead to loss of ligand binding capacity.

    • Solution: Validate protein activity using binding assays with known ligands (PD-L1, PD-L2) or antibodies (e.g., Nivolumab), and establish acceptance criteria based on affinity constants (KD values) from techniques like BLI or SPR .

  • Poor reproducibility between experiments:

    • Issue: Different protein batches may perform inconsistently in experiments.

    • Solution: Implement comprehensive quality control testing, including purity assessment (>95% by SDS-PAGE), activity testing, and lot-to-lot comparison. Reserve reference samples from well-performing lots as benchmarks .

How can researchers effectively compare different anti-PD-1 therapeutic strategies in preclinical models?

To effectively compare anti-PD-1 therapeutic strategies in preclinical models, researchers should implement a systematic approach:

  • Standardized model selection:

    • Choose tumor models with validated PD-1/PD-L1 expression patterns

    • Consider both small (50 mm³) and large (150 mm³) tumor models to assess size-dependent efficacy differences

    • Include models with varying immune infiltration profiles to assess broader applicability

  • Therapeutic agent characterization:

    • Directly compare binding affinities using consistent methodology (e.g., BLI, SPR)

    • Verify target engagement in vivo using techniques like intravital microscopy or ex vivo analysis

    • Assess tissue/tumor penetration capability of different molecular formats

  • Comprehensive immune profiling:

    • Analyze changes in immune cell populations, particularly tracking whether peripheral effector T cells are depleted

    • Evaluate functional parameters (cytokine production, proliferation, cytotoxicity)

    • Perform single-cell RNA sequencing to capture heterogeneous responses

  • Multiple readouts beyond tumor size:

    • Measure survival endpoints

    • Assess immune memory formation through rechallenge studies

    • Evaluate abscopal effects in distant untreated tumors

    • Include PK/PD studies to correlate exposure with efficacy

  • Translational biomarkers:

    • Develop companion biomarkers that predict response

    • Include PD-L1 expression analysis using engineered PD-1 variants as imaging tracers

    • Correlate with emerging clinical biomarkers for relevance to human applications

This systematic approach allows for fair comparison between traditional antibody approaches and newer strategies like engineered high-affinity PD-1 variants, providing insights into their relative advantages in different contexts.

What are the most sensitive methods for detecting low-level PD-1 expression in research samples?

Detecting low-level PD-1 expression requires highly sensitive methods:

  • Enhanced flow cytometry approaches:

    • Use signal amplification systems like tyramide signal amplification (TSA)

    • Employ fluorophores with high quantum yield and minimal spectral overlap

    • Implement staining protocols with extended incubation times

    • Consider measuring both surface and intracellular PD-1 pools

  • Digital droplet PCR (ddPCR):

    • Provides absolute quantification with higher sensitivity than conventional qPCR

    • Can detect as few as 1-2 copies of PDCD1 mRNA per reaction

    • Particularly valuable for samples with limited material

  • Single-cell RNA sequencing:

    • Enables detection of PDCD1 transcript at the single-cell level

    • Provides context of expression within heterogeneous cell populations

    • Can be combined with protein detection through CITE-seq approaches

  • Proximity ligation assay (PLA):

    • Combines antibody specificity with rolling circle amplification

    • Offers 100-1000× higher sensitivity than conventional immunohistochemistry

    • Particularly useful for tissue sections with sparse PD-1 expression

  • High-sensitivity mass spectrometry:

    • Targeted approaches like selected reaction monitoring (SRM)

    • Parallel reaction monitoring (PRM) with internal standard peptides

    • Sample enrichment strategies (e.g., immunoprecipitation before MS analysis)

  • Engineered reporter systems:

    • Reporter cell lines with luciferase or fluorescent proteins under PD-1 promoter control

    • CRISPR activation systems targeting the endogenous PDCD1 locus

    • Particularly useful for measuring dynamic changes in expression

These methods can be selected based on sample type, available material, and the specific research question, with combinations often providing the most comprehensive assessment of PD-1 expression.

How might tissue-specific variations in PD-1 glycosylation impact therapeutic targeting strategies?

Variations in PD-1 glycosylation patterns across different tissues and disease states represent an emerging area of investigation with significant implications for therapeutic targeting:

  • Differential binding to natural ligands: Tissue-specific glycosylation may alter binding affinity to PD-L1 and PD-L2, potentially creating microenvironments where standard therapeutic doses are insufficient for complete pathway blockade .

  • Antibody epitope accessibility: Glycan structures can mask or expose different epitopes on the PD-1 protein, affecting the binding of therapeutic antibodies. This may explain why certain patients respond differently to different anti-PD-1 antibodies despite all targeting the same protein .

  • Pharmacokinetic considerations: Glycosylation affects protein half-life in circulation and tissue distribution patterns. Understanding these variations could inform dosing strategies and predict treatment durability across different cancer types.

  • Biomarker development: Characterizing tissue-specific glycoforms of PD-1 could yield novel biomarkers for patient stratification, moving beyond simple expression level assessment to more nuanced evaluation of functional PD-1 variants.

  • Next-generation therapeutics: Future therapeutic agents could be designed to target specific glycoforms of PD-1 that predominate in certain tumor microenvironments, potentially increasing specificity and reducing off-target effects.

Methodologically, this research direction will require advanced glycoproteomics approaches combined with functional assays to correlate glycosylation patterns with therapeutic response in different tissues and disease contexts.

What are the emerging approaches for studying PD-1 in the context of spatial immunology?

Spatial immunology approaches are revolutionizing our understanding of PD-1 biology by preserving the crucial information about cellular positioning and interactions:

  • Multiplexed imaging technologies:

    • Imaging mass cytometry can measure PD-1 alongside 40+ markers simultaneously on tissue sections

    • Multiplexed immunofluorescence with spectral unmixing allows visualization of PD-1 in relation to its ligands and other immune markers

    • CODEX (CO-Detection by indEXing) enables highly multiplexed imaging of PD-1+ cells in their native tissue context

  • Spatial transcriptomics:

    • Visium spatial gene expression platform can map PDCD1 transcription patterns across tissue sections

    • Slide-seq approaches provide near-single-cell resolution of transcript localization

    • In situ sequencing can detect PDCD1 mRNA directly within intact tissue architecture

  • 3D tissue imaging:

    • Tissue clearing techniques combined with light-sheet microscopy allow visualization of PD-1+ cells throughout intact organs

    • Serial section reconstruction creates detailed 3D maps of PD-1 expression in relation to vessels, stroma, and tumor cells

  • Live imaging approaches:

    • Intravital microscopy with fluorescently tagged PD-1 or PD-1 reporter systems

    • Two-photon microscopy to visualize PD-1+ T cell dynamics in living tissues

    • PD-1-specific nanobodies for real-time imaging of receptor engagement

  • Computational spatial analysis:

    • Neighborhood analysis to identify spatial interactions between PD-1+ cells and other cell types

    • Trajectory inference to map migration patterns of PD-1-expressing cells

    • Graph-based approaches to quantify cellular interaction networks

These approaches are particularly valuable for understanding the heterogeneity of PD-1 expression and function across different tissue microenvironments and could inform more precise therapeutic targeting strategies.

How can recombinant PD-1 proteins be effectively used as tools in high-throughput screening?

Recombinant human PD-1 proteins can serve as valuable tools in high-throughput screening applications:

  • Antibody discovery platforms:

    • Immobilize properly folded and glycosylated PD-1 protein (2 μg/mL) in microplate formats to screen antibody libraries

    • Implement competition assays with known ligands (PD-L1, PD-L2) to identify antibodies that block the interaction

    • Use BLI or SPR to rapidly characterize binding affinity and kinetics of hits (reference: Nivolumab binds with KD ≈ 5.09 nM)

  • Small molecule inhibitor screening:

    • Develop fluorescence polarization assays using labeled PD-1 and its ligands

    • Implement AlphaScreen or similar proximity-based detection methods

    • Establish robust Z-factors (>0.5) and signal-to-background ratios (>10) for reliable screening

  • Peptide mimetic development:

    • Use truncation libraries of PD-1 to identify minimal binding domains

    • Screen peptide libraries against PD-L1/PD-L2 binding pockets

    • Validate hits using the same binding assays established for the full-length protein

  • Engineered variant screening:

    • Display libraries of PD-1 variants (e.g., yeast or phage display) and select for desired properties

    • Screen for variants with enhanced affinity (reference: engineered variants with 110 pM affinity)

    • Validate promising candidates using multiple methodologies (ELISA, BLI, cell-based assays)

  • Quality control applications:

    • Implement automated PD-1:PD-L1/PD-L2 binding assays as quality control measures for therapeutic antibodies

    • Establish reference standards and acceptance criteria based on known binding parameters

    • Develop lot release assays with appropriate sensitivity and reproducibility

To ensure success in these applications, researchers should maintain protein quality (>95% purity, proper glycosylation, low aggregation) and implement appropriate controls to account for non-specific binding and matrix effects .

What are best practices for developing flow cytometry panels that include PD-1?

Developing effective flow cytometry panels that include PD-1 requires careful consideration of several factors:

  • Optimal clone selection:

    • For human samples, select validated anti-PD-1 clones with demonstrated specificity

    • For mouse studies, consider the different properties of available clones (e.g., 29F.1A12 has approximately 100-fold higher affinity than RMP1-14)

    • Verify that selected clones do not block the epitopes recognized by therapeutic antibodies if studying treatment effects

  • Panel design considerations:

    • Place PD-1 in a channel with appropriate sensitivity given its often intermediate expression level

    • Avoid spectral overlap with markers expected to be co-expressed with PD-1 (e.g., other exhaustion markers)

    • Include markers that define the parent population (e.g., CD3, CD4, CD8 for T cells)

    • Add functional markers (cytokines, activation markers) to correlate with PD-1 expression

  • Control strategies:

    • Include biological controls (PD-1 knockout or known negative populations)

    • Use fluorescence-minus-one (FMO) controls to set accurate gates

    • Include samples from healthy donors alongside disease samples

    • Consider including samples from patients treated with anti-PD-1 therapies as reference points

  • Sample preparation optimization:

    • Minimize time between sample collection and staining

    • Standardize fixation protocols if needed, as some epitopes may be fixation-sensitive

    • Determine whether surface staining alone is sufficient or if intracellular PD-1 pools should also be measured

    • Establish consistent gating strategies across experiments

  • Data analysis approaches:

    • Analyze PD-1 both as percent positive cells and mean fluorescence intensity

    • Consider density plots rather than simple dot plots for better visualization

    • Use dimensionality reduction techniques (tSNE, UMAP) to identify PD-1+ cell clusters

    • Correlate PD-1 expression with functional readouts and clinical parameters

Following these best practices ensures reliable and reproducible assessment of PD-1 expression across different experimental conditions and sample types.

What factors affect the reproducibility of PD-1:PD-L1 binding assays?

Several critical factors influence the reproducibility of PD-1:PD-L1 binding assays:

  • Protein quality and integrity:

    • Glycosylation status: Properly glycosylated PD-1 (appearing as 31-44 kDa in SDS-PAGE) is essential for native binding properties

    • Aggregation levels: Maintain protein aggregation below 5% as aggregates can confound binding measurements

    • Storage conditions: Avoid repeated freeze-thaw cycles and store according to manufacturer recommendations (-20°C or lower for lyophilized proteins)

  • Assay format considerations:

    • Immobilization strategy: For ELISA formats, use consistent protein coating concentration (2 μg/mL recommended) and buffer conditions

    • Biosensor selection: For BLI assays, HIS1K biosensors are suitable for His-tagged PD-1, while Protein A biosensors work well for Fc-fusion constructs

    • Detection methodology: Match detection sensitivity to the expected KD values (e.g., PD-1:PD-L1 KD ≈ 38.9 nM; PD-1:PD-L2 KD ≈ 16.3 nM)

  • Buffer and reaction conditions:

    • pH dependence: Binding interactions can vary significantly with pH; standardize to physiologically relevant conditions

    • Divalent cations: Some protein-protein interactions are influenced by calcium or magnesium levels

    • Blocking agents: Optimize blocking to minimize non-specific binding without interfering with the PD-1:PD-L1 interaction

  • Data analysis approaches:

    • Curve fitting models: Select appropriate models based on binding mechanism (1:1, heterogeneous ligand, etc.)

    • Reference subtraction: Implement consistent background subtraction methods

    • Replicate handling: Include sufficient technical and biological replicates (minimum triplicate measurements)

  • Standardization practices:

    • Reference standards: Include well-characterized controls in each experiment (e.g., Nivolumab binding to PD-1 with expected KD ≈ 5.09 nM)

    • Instrument calibration: Regularly calibrate analytical instruments according to manufacturer guidelines

    • Laboratory environmental conditions: Control temperature and humidity to minimize variation

By systematically addressing these factors, researchers can achieve the high reproducibility necessary for reliable comparative studies of PD-1:PD-L1 interactions.

How do different expression systems affect the properties of recombinant human PD-1?

Different expression systems produce recombinant human PD-1 with varying properties that can significantly impact research applications:

  • Mammalian expression systems (e.g., HEK293, CHO cells):

    • Produce PD-1 with native-like glycosylation patterns, resulting in 31-44 kDa apparent molecular weight on SDS-PAGE

    • Provide proper folding and post-translational modifications

    • Generate proteins with binding properties closely matching endogenous PD-1 (KD ≈ 38.9 nM for PD-L1, 16.3 nM for PD-L2)

    • Typically yield lower protein quantities but higher biological relevance

  • Insect cell systems (e.g., Sf9, High Five):

    • Produce proteins with simplified glycosylation patterns

    • May alter binding kinetics due to differences in glycan structures

    • Often provide higher yields than mammalian systems

    • Can be suitable for structural studies but may not fully recapitulate functional properties

  • Bacterial expression systems (e.g., E. coli):

    • Lack glycosylation machinery, producing non-glycosylated PD-1

    • Often require refolding from inclusion bodies

    • Significantly altered binding properties compared to native PD-1

    • Higher yield but lower functional relevance

    • May be suitable for applications where glycosylation is not critical

  • Yeast expression systems (e.g., Pichia pastoris):

    • Provide intermediate glycosylation complexity

    • Can be engineered for humanized glycosylation

    • Used successfully for directed evolution of high-affinity PD-1 variants

    • Balance between yield and functionality

  • Cell-free expression systems:

    • Rapid production without cell culture

    • Limited post-translational modifications

    • Useful for initial screening or structural studies

    • Not ideal for applications requiring native functionality

Product Science Overview

Structure and Function

PD-1 is a type I membrane protein consisting of 268 amino acids . It includes an extracellular IgV domain, a transmembrane region, and an intracellular tail . The intracellular tail contains two phosphorylation sites located in an immunoreceptor tyrosine-based inhibitory motif and an immunoreceptor tyrosine-based switch motif . These motifs suggest that PD-1 negatively regulates T cell receptor (TCR) signals by binding to SHP-1 and SHP-2 phosphatases upon ligand binding .

PD-1 has two known ligands: PD-L1 (B7-H1) and PD-L2 (B7-DC) . The interaction between PD-1 and its ligands leads to the inhibition of T cell activity in peripheral tissues during inflammation, thereby preventing autoimmunity .

Biological Activity

Recombinant human PD-1 (rhPD-1) is produced in HEK293 cells and is a polypeptide chain containing 149 amino acids with a C-terminal 6×His tag . It is a fully biologically active molecule with a molecular mass of 30-40 kDa as analyzed by reducing SDS-PAGE . The biological activity of rhPD-1 is demonstrated by its ability to bind PD-L1 Fc Chimera and Keytruda (an anti-PD-1 therapy) in a dose-dependent manner .

Role in Immune Regulation

PD-1 is expressed on T cells, B cells, monocytes, natural killer cells, and many tumor-infiltrating lymphocytes . It plays a critical role in maintaining immune homeostasis by promoting apoptosis of antigen-specific T cells in lymph nodes and reducing apoptosis in regulatory T cells . This dual mechanism helps in preventing autoimmune responses while also allowing the immune system to tolerate self-antigens .

Clinical Significance

The PD-1/PD-L1 pathway has become a significant target in cancer immunotherapy . PD-1 inhibitors, such as Keytruda, block the interaction between PD-1 and its ligands, thereby activating the immune system to attack tumor cells . This approach has shown promising results in treating various types of cancer, including melanoma, lung cancer, and renal cell carcinoma .

Production and Storage

Recombinant human PD-1 is lyophilized from a 0.2 μm filtered solution in PBS, 5% trehalose, and mannitol . Upon reconstitution, it remains stable for up to 1 week at 4°C or up to 3 months at -20°C . For long-term storage, it is recommended to add a carrier protein, such as 0.1% BSA, to avoid repeated freeze-thaw cycles .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.