Genome-wide Cell Surface Protein Interaction Screening Service

Discover receptor–ligand pairs, identify cell-surface drug targets, and clarify mechanism of action (MoA) with a genome-scale screening workflow designed for extracellular protein interactions.

Our service supports end-to-end delivery—from project design and primary screening to hit prioritization, confirmation, and optional orthogonal validation—so you can move quickly from a biological question to actionable candidates.

Best for

  • Receptor–ligand pairing (ligand-to-receptor or receptor-to-ligand)
  • Deorphanization of orphan receptors
  • Target identification for antibodies, ADCs, bispecifics, and Fc-fusion therapeutics
  • Cell–cell communication studies and surface biomarker discovery
  • Pathogen/toxin/particle receptor discovery (binding-based projects)

Submit Your Inquiry

What Is Genome-wide Receptor–Ligand Screening on the Cell Surface?

Genome-wide cell surface interaction screening is a high-throughput approach to identify specific extracellular binding partners at scale—typically focusing on membrane receptors, GPI-anchored proteins, and other surface-accessible proteins, and/or secreted ligands depending on the library design.

Unlike many intracellular PPI methods, this screening strategy is built around surface-accessible, extracellular interactions, making it especially suitable for:

  • Full-length membrane targets (platform-dependent)
  • Low-affinity extracellular interactions (assay-dependent)
  • Rapid candidate triage for downstream functional work

Note: "Genome-wide" coverage is library-dependent. We define the target space with you during project setup and align screening strategy, controls, and validation to your scientific goal.

Scientific Questions for Cell Surface Interactome Mapping (Target ID & Deorphanization)

This service is designed to answer questions such as:

  • What receptor binds my ligand or secreted protein?
  • What is the natural ligand for an orphan receptor (deorphanization)?
  • What is the cell-surface target of my antibody/ADC/bispecific/Fc-fusion?
  • Which surface proteins mediate binding/entry for a pathogen, toxin, or delivery particle?
  • Which receptor–ligand interactions may explain a phenotype or pathway response?

Advantages of Genome-wide Cell Surface Interaction Screening for Low-Affinity PPIs

Genome-scale discovery, not guesswork

Screen large target spaces (library-dependent) to uncover novel receptor–ligand pairs.

Built for extracellular biology

Optimized for surface-accessible interactions and receptor–ligand binding rather than intracellular complexes.

Designed to capture low-affinity interactions

Assay formats can be tuned for weaker extracellular binding (project-dependent).

Clear evidence for decision-making

Ranked hit lists with supporting evidence help you quickly select candidates for validation and functional studies.

Optional orthogonal validation

Confirm binding using independent methods (e.g., flow cytometry, SPR/BLI, ELISA) to strengthen confidence.

Technical Services
Service Scope Workflow and Platform Tech Comparison Sample Requirements Deliverables FAQ Get a Custom Proposal

Target Space & Library Coverage Options (Secretome / Receptome / Cell-surface)  

Choose the target space based on your scientific question. Library size and coverage are platform-dependent and will be confirmed during feasibility review.

Secretome-to-surface screening

Best for: finding the receptor(s) of a secreted ligand, cytokine, growth factor, or Fc-fusion.

Input: purified ligand / Fc-fusion / tagged bait.

Output: ranked list of candidate surface binders with control-filtered evidence.

Receptome / cell-surface library screening

Best for: deorphanizing receptors or identifying which surface targets bind your bait.

Input: ligand, antibody, or engineered binder.

Output: prioritized receptor candidates with confirmation-ready readouts.

Custom sub-libraries (family-focused)

Best for: faster iteration or hypothesis-driven screening (e.g., cytokine receptors, immune checkpoints, GPCR subsets).

Input/Output: defined case-by-case for speed and cost efficiency.

Screening Strategies and Assay Formats for Cell Surface Binding Discovery

Assay format is selected based on bait type, expected binding strength, and whether membrane-protein context is required. Available options include:

  • Primary binding screen (discovery stage)
    Designed to capture candidate interactions at scale with control-based filtering. Formats may include cell-based binding readouts, plate-based binding, or other platform-dependent high-throughput readouts.
  • Confirmation screen (specificity stage)
    Re-testing top hits under increased stringency and with additional negative controls to remove artifacts and common non-specific binders.
  • Specificity-enhancing designs (optional)
    Competitive binding (blocking/competition), concentration series, and replicate-based calling to improve confidence—especially useful for low-affinity extracellular interactions.

For each project, we define clear hit-calling criteria (signal-to-background, reproducibility, and control separation) before screening begins.

Workflow: Primary Screen → Hit Ranking → Confirmation → Validation

Workflow for Isothermal Titration Calorimetry
1

Project consultation & feasibility review

Define goals, target space, bait format, assay strategy, controls, and success criteria.

2

Pilot setup (when needed)

Confirm bait integrity/behavior and establish assay conditions and control performance.

3

Primary genome-wide screen

Run the screen against the defined target space and capture primary signals.

4

Hit ranking and triage

Generate a prioritized candidate list based on signal strength, reproducibility, and specificity.

5

Confirmation (secondary screening)

Re-test top candidates with additional stringency and controls to remove artifacts.

6

Optional orthogonal validation

Validate binding using independent methods (flow cytometry, SPR/BLI, ELISA) to strengthen confidence.

7

Final reporting

Deliver ranked hits, evidence summaries, and recommended next steps.

Controls, QC & False-Positive Filtering (Bait vs Control, Replicates, Thresholds)

To ensure confident hit calling, we apply a control-first QC strategy:

  • Bait vs control design: appropriate negative controls (e.g., irrelevant bait/isotype/mock) to remove non-specific binders.
  • Replicates: prioritize candidates that reproduce across runs and reduce stochastic artifacts.
  • Pre-defined thresholds: hits are called using objective criteria such as signal-to-background and control separation, set before screening.
  • Optional specificity checks: competition/blocking and increased stringency during confirmation when suitable.

This approach yields a ranked candidate set with clear separation from background.

Orthogonal Validation for Cell Surface Binding (Flow, SPR/BLI, ELISA)

Orthogonal validation confirms prioritized hits using an independent assay to strengthen confidence:

  • Flow cytometry: verifies target-dependent cell binding and supports competition-based specificity checks.
  • SPR/BLI: confirms direct binding and provides kinetic/affinity characterization when reagents are available.
  • ELISA / plate-based assays: practical confirmation for well-behaved bait–target pairs and comparative binding tests.

Output: validation plots/data with control context and an interpreted conclusion for each tested hit.

Technology Comparison: Genome-wide Screening vs. Traditional Methods

Criteria Genome-wide Cell Surface Screening Co-IP SPR Protein Microarrays
Application Receptor–ligand pairing, target discovery, low-affinity interactions Protein–protein interaction validation Kinetics and affinity analysis High-throughput screening, antibody profiling
Sensitivity High (avidity-based binding for weak interactions) Moderate (dependent on antibody quality) High (real-time detection) Moderate (best for high-affinity targets)
Specificity High (bait vs control, competition assays) Moderate (prone to background contamination) High (precise affinity measurement) Moderate (depends on antibody selection)
Confirmation Methods Flow cytometry, SPR, BLI, ELISA (optional validation) Western blot (WB) Real-time kinetic/affinity measurement (SPR) Microarray analysis
Advantages Scalable, high-throughput, identifies low-affinity interactions, built-in validation Effective for validating known interactions, stable complexes Ideal for precise affinity and kinetics studies High throughput, but lacks in vivo context

How to Submit Samples for Cell Surface Screening

Accepted bait formats

Bait type Typical use Recommended info to provide
Recombinant ligand / ECD protein Receptor discovery, ligand–receptor pairing Sequence/construct, tag/Fc format, expected MW, species
Fc-fusion / engineered binder Cell-surface binding discovery, cross-reactivity checks Fc type, tag, glycosylation notes (if known), target hypothesis
Antibody (IgG / mAb) Target identification (MoA), surface target mapping Isotype, species, concentration, known binding behavior

Sample quality & handling

  • Purity/behavior: submit well-behaved material (low aggregation preferred). If available, include SEC or SDS-PAGE evidence.
  • Concentration: provide sufficient concentration for screening and confirmation (final requirements are assay-dependent).
  • Storage/shipping: ship frozen on dry ice (or as instructed) and avoid repeated freeze–thaw cycles.
  • Buffer compatibility: share the full buffer recipe. Avoid harsh detergents, high glycerol, or unknown additives unless confirmed.

Required project information

  • Project goal (receptor discovery / deorphanization / target ID)
  • Species and any required cross-species testing
  • Target space preference (secretome / receptome / custom)
  • Controls (recommended: irrelevant bait/isotype control; optional: competition blocker)
  • Experimental groups (if comparing conditions)

Applications: Receptor–Ligand Pairing, Cell–Cell Communication & Target Discovery

Receptor–Ligand Pairing

Map ligand-to-receptor interactions to accelerate pathway elucidation and target selection.

Orphan Receptor Deorphanization

Identify candidate ligands for orphan receptors to unlock biology and therapeutic opportunities.

Target Identification (MoA) for Biologics

Determine binding targets for antibodies, ADCs, and engineered proteins to support MoA and de-risk off-target concerns.

Cell–Cell Communication Mapping

Prioritize extracellular interaction pairs relevant to immune signaling, inflammation, and tumor microenvironment biology.

Cell-surface Target Discovery & Biomarker Identification

Nominate surface-accessible targets for therapeutic development or diagnostic applications.

Pathogen/Toxin/Particle Receptor Discovery

Identify surface factors involved in binding and entry (project scope defined by assay format and biosafety requirements).

Deliverables: Data Packages and Reports

  • Ranked hit list (primary and confirmed candidates where applicable)
  • Evidence summary (signals, control comparisons, reproducibility indicators)
  • Plots and tables supporting decision-making (hit ranking visuals as appropriate)
  • Optional validation data (flow/SPR/BLI/ELISA outputs if included)
  • Final report summarizing methods, QC, results, and recommended next steps
Two-panel hit ranking figure with bait vs control signals for top hits, error bars for replicates, and a threshold line plus overall signal distribution.

Primary Screen Hit Ranking

Primary screen hit ranking showing clear bait–control separation and a defined calling threshold (mean ± SD, n=3).

Two-panel binding validation figure with a semi-log dose–response curve and a competitor inhibition curve including negative controls and replicate error bars.

Confirmation / Competition Binding

Dose-dependent binding and competition inhibition confirm specific target engagement (mean ± SD, n=3).

Flow cytometry figure with FSC/SSC and singlet gating plots plus an overlay histogram comparing target-positive, mock, and competition conditions with a positive gate.

Flow Cytometry Cell-binding Validation

Flow cytometry validation shows a binding shift in target-positive cells and signal reduction with competition.

Volcano plot of log2 fold change versus -log10 p-value comparing bait to control, showing threshold lines and labeled bait plus top enriched hits.

Interactome Specificity Volcano Plot(Bait vs Control)

Volcano plot highlights significantly enriched hits in bait vs control with defined log2FC and p-value thresholds.

Q&A for Genome-wide Cell Surface Interaction Screening

How does genome-wide screening capture low-affinity extracellular interactions?

Many cell-surface interactions are weak and transient (often µM-range K_D with fast off-rates). We enhance detection using avidity-based binding (e.g., multimerized baits such as Fc-fusions or streptavidin-clustered tags), improving signal-to-noise and enabling interactions that may be missed by Co-IP or standard SPR.

Can this service identify targets for bispecific antibodies or ADCs?

Yes. Using the therapeutic molecule as the bait against a cell-surface library, we can identify the primary target and assess potential off-target binding to other surface-accessible proteins.

Why use cell-based screening instead of purified protein microarrays?

Cell-based formats present receptors in a more native context, supporting proper folding and key PTMs (e.g., glycosylation). This is especially important for GPCRs, multi-pass membrane proteins, and ion channels, which can lose epitopes when purified or immobilized.

What distinguishes a "hit" from non-specific background binding?

We use a control-first hit-calling strategy:

  • Statistical enrichment: Z-score or log2 fold change vs isotype/mock controls
  • Specificity support (when applicable): dose dependence and/or competition blocking

This helps filter common "sticky" binders and technical artifacts.

Is the screening limited to human protein libraries?

Human-focused libraries are standard, but custom species libraries (e.g., mouse, rat, NHP) can be supported for cross-species reactivity studies. Pathogen-related target spaces may also be possible depending on scope and feasibility.

Can whole cells or viral particles be used as bait?

In some projects, yes. The workflow can be adapted for particle-to-cell screening (e.g., AAV/lentiviral vectors, exosomes), provided a suitable detection strategy is available and feasibility is confirmed.

Online Inquiry