The generation of the SEMA4D recombinant monoclonal antibody involves a series of steps. Initially, the SEMA4D monoclonal antibody is harvested and its gene sequence is determined. A vector containing the SEMA4D monoclonal antibody gene is subsequently constructed and transfected into a host cell line for culturing. The synthesis of the SEMA4D monoclonal antibody employs a recombinant human SEMA4D protein as an immunogen. Finally, the SEMA4D recombinant monoclonal antibody is purified through affinity chromatography and analyzed for specificity using ELISA and FC assays. It only reacts with human SEMA4D protein.
SEMA4D is a protein that belongs to the semaphorin family, which plays a crucial role in cell signaling and regulation of various physiological processes, including cell migration, axon guidance, and immune responses. SEMA4D acts as both a ligand and a receptor, interacting with a variety of other proteins, including plexin B1, CD72, CD100, and integrins. These interactions activate intracellular signaling pathways, leading to changes in cell behavior, such as cell adhesion, cell migration, proliferation, and survival. In the immune system, SEMA4D plays a role in regulating immune cell function. It can also modulate immune cell activity by regulating cytokine production, phagocytosis, and antigen presentation. SEMA4D has been implicated in various physiological and pathological processes, including cancer, inflammation, cardiovascular diseases, and neurological disorders.
The production of the SEMA4D recombinant monoclonal antibody involves a multi-step process. Initially, the SEMA4D monoclonal antibody is harvested, and its genetic sequence is determined. Subsequently, a vector containing the SEMA4D monoclonal antibody gene is constructed and transfected into a host cell line for cultivation. The synthesis of the SEMA4D monoclonal antibody utilizes a recombinant human SEMA4D protein as an immunogen. Finally, the SEMA4D recombinant monoclonal antibody undergoes purification through affinity chromatography and is analyzed for specificity using ELISA and FC assays. It exhibits reactivity solely with human SEMA4D protein.
SEMA4D is a protein that belongs to the semaphorin family, which plays a critical role in cellular signaling and regulation of diverse physiological processes, including cell migration, axon guidance, and immune responses. SEMA4D acts as both a ligand and a receptor, engaging in interactions with a range of other proteins, including plexin B1, CD72, CD100, and integrins. These interactions activate intracellular signaling pathways, leading to modifications in cell behavior, such as cell adhesion, cell migration, proliferation, and survival. Within the immune system, SEMA4D plays a role in modulating immune cell function. It can also influence immune cell activity by regulating cytokine production, phagocytosis, and antigen presentation. SEMA4D has been implicated in various physiological and pathological processes, including cancer, inflammation, cardiovascular diseases, and neurological disorders.
SEMA4D is expressed on the cell surface as a disulfide-linked homodimer with a molecular weight of approximately 300-kDa. The protein contains a large N-terminal β-propeller "sema" domain, followed by an Ig-like domain, a lysine-rich domain, a transmembrane domain, and a cytoplasmic tail with consensus tyrosine and serine phosphorylation sites. Upon cell activation, SEMA4D can be proteolytically cleaved to generate a physiologically active 240-kDa soluble form (sSEMA4D) .
Successful antibody design requires targeting accessible epitopes within the extracellular portion of SEMA4D, particularly within the sema domain that mediates receptor interactions. For experimental applications, researchers should consider whether their antibody recognizes both membrane-bound and soluble forms, as this will significantly impact experimental outcomes when studying tissues where proteolytic shedding occurs .
SEMA4D expression varies significantly across cell types and activation states. In platelets, resting cells express detectable surface SEMA4D that transiently doubles upon activation with phorbol 12-myristate 13-acetate (PMA), followed by a decline to nearly undetectable levels due to proteolytic shedding . In immune cells, SEMA4D is upregulated upon activation, particularly in T cells.
For experimental design, researchers should: (1) determine baseline expression in their target cell population through flow cytometry; (2) characterize expression kinetics following relevant stimuli; (3) quantify proportions of membrane-bound versus soluble SEMA4D using complementary approaches such as flow cytometry and ELISA; and (4) assess potential regulation by metalloprotease inhibitors if shedding is a critical experimental parameter .
SEMA4D interacts with three main receptors: Plexin-B1 (high-affinity), Plexin-B2, and CD72 (lower-affinity). Effective anti-SEMA4D antibodies should block binding to all three receptors. Receptor blockade can be measured using flow cytometry-based binding assays where His-tagged SEMA4D is pre-incubated with anti-SEMA4D antibodies at various concentrations, then added to receptor-expressing cells. Bound SEMA4D is detected with anti-His-APC antibodies, with decreasing fluorescence indicating successful blocking .
When designing experiments to measure receptor blocking, researchers should:
Establish receptor expression systems for each receptor (e.g., PLXNB1-transfected 293 cells)
Determine antibody potency as EC50 values for blocking each receptor interaction
Include appropriate isotype controls to distinguish specific from non-specific effects
Consider downstream signaling assays to confirm functional receptor blockade
For VX15/2503, the mean EC50 for blocking SEMA4D-Plexin-B1 interaction is approximately 1.2 nM, providing a benchmark for comparative studies .
SEMA4D receptor signaling differs based on the specific receptor engaged. For CD72, SEMA4D binding disrupts CD72's association with the tyrosine phosphatase SHP-1. In resting platelets and B cells, CD72 complexes with SHP-1, maintaining its inhibitory effects. SEMA4D binding causes dissociation of this complex, releasing SHP-1-mediated inhibition and enhancing signaling through phosphotyrosine pathways .
For Plexin-B1, SEMA4D binding activates small GTPases and downstream pathways affecting cytoskeletal organization, cellular migration, and vascular permeability. In endothelial cells, this signaling can disrupt tight junctions forming the blood-brain barrier .
To experimentally assess antibody effects on these pathways, researchers can:
Perform co-immunoprecipitation studies to evaluate CD72/SHP-1 complex integrity
Measure phosphorylation status of downstream signaling molecules
Employ cell migration and permeability assays to assess functional outcomes
Use receptor-specific cell systems to distinguish effects on different signaling pathways
Generating antibodies with cross-species reactivity to SEMA4D presents challenges due to high protein conservation and immunological tolerance. The most successful approach involves immunizing SEMA4D-deficient (knockout) mice to bypass tolerance mechanisms. As demonstrated with the development of mAb 67-2 (the parent antibody for humanized VX15/2503), this strategy yielded antibodies that react with mouse, rat, primate, and human SEMA4D .
Methodological steps include:
Immunization of SEMA4D knockout mice with recombinant SEMA4D
Screening approximately 96 parental hybridomas for cross-reactivity with both mouse and human SEMA4D
Characterization through various assays including ELISA, competition assays, flow cytometry, and immunohistochemistry
Selection of lead candidates based on affinity, species cross-reactivity, and functional blocking activity
Humanization of murine antibodies for potential therapeutic development
Attribute | Analytical Method | Acceptance Criteria |
---|---|---|
Antigen Specificity | ELISA, Flow cytometry, IHC | High specificity for SEMA4D across species; no binding to irrelevant proteins |
Affinity | Biacore, Cell-based flow cytometry | KD for recombinant SEMA4D: 1-5 nM; KD for cell-associated SEMA4D: ~0.45 nM |
Receptor Blocking Potency | Flow cytometry-based binding assay | EC50 for PLXNB1 blocking: ~1.2 nM |
Species Cross-reactivity | ELISA, Flow cytometry using species-specific cells | Consistent binding across mouse, rat, rabbit, cynomolgus macaque, marmoset, rhesus macaque, and human |
Effector Functions | CDC, ADCC assays | Minimal effector functions for research applications to avoid confounding cell depletion |
Epitope Specificity | Competition assays | Consistent epitope recognition across batches |
Researchers should validate each antibody lot against these parameters before experimental use to ensure reproducible results. For antibodies intended for in vivo studies, additional testing for endotoxin levels, aggregation, and formulation stability should be performed .
Anti-SEMA4D antibodies have demonstrated efficacy in preserving blood-brain barrier (BBB) integrity in models of neuroinflammation. SEMA4D signaling through Plexin-B1 on endothelial cells disrupts tight junctions, and antibody blockade prevents this disruption .
Methodological approaches to assess BBB integrity include:
In vivo permeability assays:
Intravenous injection of tracers (Evans blue, fluorescent dextrans)
Quantification of tracer extravasation into brain parenchyma
Comparison between anti-SEMA4D treated and control groups
Ex vivo histological assessment:
Immunostaining for tight junction proteins (ZO-1, claudin-5, occludin)
Quantitative image analysis of junction continuity and expression levels
Co-localization studies with endothelial markers
In vitro barrier models:
Transendothelial electrical resistance (TEER) measurements using brain endothelial monolayers
Permeability coefficients for labeled molecules of various sizes
Assessment of tight junction protein localization and phosphorylation status
These methodologies allow researchers to determine both the functional and molecular effects of anti-SEMA4D treatment on BBB integrity, providing insight into potential therapeutic applications in multiple sclerosis and other neuroinflammatory conditions .
Anti-SEMA4D antibodies can reverse the inhibitory effects of SEMA4D on OPC survival, migration, and differentiation, making them valuable tools for studying remyelination processes. Comprehensive experimental approaches include:
In vitro OPC cultures:
Primary OPC isolation from neonatal rodent brains
Assessment of survival (TUNEL assay, MTT assay)
Differentiation analysis (immunostaining for myelin basic protein, proteolipid protein)
Migration assays (transwell, scratch assays) with/without recombinant SEMA4D and anti-SEMA4D antibodies
Ex vivo slice cultures:
Organotypic brain slice cultures
Focal demyelination with lysolecithin
Treatment with anti-SEMA4D antibodies
Analysis of OPC recruitment and remyelination
In vivo demyelination models:
Cuprizone-induced demyelination
Lysolecithin focal demyelination
Measurement of OPC migration to lesion sites
Quantitative assessment of remyelination (electron microscopy, myelin staining)
Analysis of functional recovery (electrophysiology, behavioral testing)
These approaches collectively provide robust evidence for the role of anti-SEMA4D antibodies in promoting remyelination by modulating OPC biology in developmental and pathological contexts .
SEMA4D can guide migration and trigger cytoskeletal changes in endothelial, tumor, and immune cells within the tumor microenvironment (TME). Anti-SEMA4D antibodies can modulate these effects, potentially enhancing anti-tumor immunity. Key experimental approaches include:
Immune cell migration and infiltration:
Transwell migration assays for monocytes, T cells, and dendritic cells
In vivo tracking of labeled immune cells in tumor models
Immunohistochemical quantification of tumor-infiltrating lymphocytes
Flow cytometric analysis of immune cell populations within tumors
Immune cell activation and function:
T cell proliferation assays
Cytokine production profile (ELISA, intracellular cytokine staining)
Cytotoxic T cell killing assays against tumor targets
Assessment of dendritic cell maturation markers
Tumor vasculature assessment:
CD31 immunostaining for vessel density
Pericyte coverage (α-SMA co-staining)
Vessel perfusion and permeability
Analysis of hypoxic regions within tumors
These assays allow researchers to comprehensively evaluate the multifaceted effects of anti-SEMA4D antibodies on the immunological landscape of the TME, providing insights into their potential as immunomodulatory agents in cancer therapy .
When designing combination studies with anti-SEMA4D antibodies and other immunotherapeutics, researchers should consider:
Mechanism of action complementarity:
Map the molecular pathways affected by each agent
Identify potential synergistic or antagonistic interactions
Determine optimal timing for each intervention based on mechanistic understanding
Dosing and scheduling optimization:
Perform dose-response studies for each agent alone
Test sequential versus concurrent administration
Establish appropriate dosing intervals based on pharmacokinetics
Comprehensive outcome assessment:
Analyze changes in immune cell populations by flow cytometry
Evaluate both direct anti-tumor effects and immune-mediated responses
Assess development of immunological memory
Monitor potential immune-related adverse events
Control groups and experimental design:
Include single-agent arms for each therapy
Use appropriate isotype controls
Consider genetic approaches (e.g., SEMA4D knockout models) as complementary evidence
Design experiments with sufficient statistical power for subgroup analyses
These considerations ensure robust evaluation of combination approaches and facilitate translational research toward clinical applications .
SEMA4D is expressed on platelets and influences thrombus formation, making it a relevant target for thrombosis research. Comprehensive experimental approaches include:
In vitro platelet function assays:
Aggregometry using various agonists (collagen, ADP, thrombin)
Flow chamber adhesion and aggregation under shear stress
Clot retraction and stability assessments
Platelet spreading on extracellular matrix proteins
SEMA4D shedding analysis:
Flow cytometry to monitor surface SEMA4D expression kinetics
ELISA to quantify soluble SEMA4D release
Effects of metalloprotease inhibitors on shedding
Comparison between wild-type and ADAM17-deficient platelets
In vivo thrombosis models:
FeCl₃-induced carotid artery thrombosis
Laser-induced cremaster arteriole injury
Intravital microscopy for real-time thrombus formation
Assessment of time to occlusion and stability of formed thrombi
Signaling studies:
Co-immunoprecipitation of CD72/SHP-1 complexes
Phosphotyrosine profiling following platelet activation
Analysis of signaling changes with anti-SEMA4D treatment
When designing these experiments, researchers should consider that SEMA4D appears to particularly affect collagen-induced platelet responses, suggesting a specific role in GPVI-mediated signaling pathways .
SEMA4D appears to play dual roles in vascular responses to injury through distinct mechanisms depending on its membrane-bound or soluble state:
Membrane-bound SEMA4D on platelets:
Promotes initial thrombus formation through homotypic interactions with other platelets
Interacts with CD72 on neighboring platelets, causing dissociation of CD72-SHP-1 complexes
Releases inhibitory effects of SHP-1 on phosphotyrosine-based signaling downstream of collagen receptor GPVI
Enhances platelet activation in a contact-dependent manner
Shed soluble SEMA4D (sSEMA4D):
Becomes available to interact with receptors on endothelial cells (primarily Plexin-B1)
Affects monocyte migration and inflammatory responses
Mediates angiogenic responses in endothelial cells
Continues to interact with platelets in a paracrine fashion
This dual functionality creates a spatiotemporal regulation mechanism: initially membrane-bound SEMA4D promotes local thrombus formation, while subsequently shed sSEMA4D influences the broader vascular environment and inflammatory response. Researchers studying these mechanisms should design experiments that can distinguish between these two phases and the transition between them .
Experimental Control/Validation | Implementation Method | Purpose |
---|---|---|
Isotype Control Antibodies | Include matched isotype control at equivalent concentrations | Distinguish specific from non-specific effects |
Genetic Controls | Use SEMA4D knockout cells/animals as negative controls | Confirm antibody specificity and validate phenotypes |
Epitope Competition | Pre-incubate with recombinant SEMA4D | Demonstrate binding specificity |
Positive Controls | Include known SEMA4D-dependent phenotypes | Ensure system responsiveness |
Receptor Blockade Validation | Flow cytometry-based receptor binding assays | Confirm functional blocking activity |
Batch Consistency | Test multiple antibody lots for consistent affinity and function | Eliminate batch variation as a confounder |
Concentration Titration | Use multiple antibody concentrations | Establish dose-response relationships |
Pharmacokinetic Assessment | In vivo studies to determine half-life and tissue penetration | Ensure appropriate dosing schedules |
Multiple Detection Methods | Combine flow cytometry, IHC, Western blotting | Cross-validate findings across platforms |
These controls and validation steps ensure experimental rigor and facilitate reproducibility across different research groups and experimental systems .
Translating findings between species presents challenges due to potential differences in SEMA4D biology. Researchers can address these challenges through:
Cross-species antibody selection:
Use antibodies like VX15/2503 that recognize SEMA4D across species
Validate comparable binding affinities for mouse, rat, primate, and human SEMA4D
Confirm similar receptor-blocking potencies across species
Comparative biology approaches:
Directly compare SEMA4D expression patterns across species
Conduct side-by-side functional assays with cells from different species
Identify conserved versus divergent signaling pathways
Humanized models:
Utilize humanized mouse models where appropriate
Consider xenograft approaches for specific applications
Complement with ex vivo human tissue studies
Validation in multiple species:
Confirm key findings in at least two different species
Include non-human primate studies as a translational bridge
Correlate with available human data from clinical samples
The cross-species reactivity of well-characterized antibodies like VX15/2503 (KD values ranging from 1.5-5.1 nM across species) provides a valuable tool for such translational research, allowing direct comparison of SEMA4D biology across experimental systems .
Anti-SEMA4D antibodies provide a powerful tool for investigating the neurovascular-immune axis in neurological disorders. Advanced research approaches include:
Multi-parameter intravital imaging:
Two-photon microscopy of cranial windows
Simultaneous tracking of labeled immune cells, vascular integrity, and neural activity
Real-time assessment of anti-SEMA4D antibody effects on cellular interactions
Cell-specific conditional approaches:
Compare global anti-SEMA4D treatment with cell-specific SEMA4D deletion
Use Cre-lox systems to delete SEMA4D receptors from specific cell populations
Determine the relative contribution of different cell types to observed phenotypes
Multi-omics integration:
Perform transcriptomic, proteomic, and metabolomic analyses on isolated cell populations
Construct network models of SEMA4D-dependent signaling changes
Identify convergent pathways affected by anti-SEMA4D treatment
Temporal intervention studies:
Administer anti-SEMA4D antibodies at different disease stages
Determine windows of opportunity for therapeutic intervention
Assess acute versus chronic effects on disease progression
These sophisticated approaches allow researchers to dissect the complex roles of SEMA4D in maintaining and disrupting the neurovascular unit, with particular relevance to multiple sclerosis, stroke, and other neuroinflammatory conditions .
Emerging technologies for single-cell resolution studies of SEMA4D biology include:
Single-cell RNA sequencing:
Transcriptional profiling of individual cells after anti-SEMA4D treatment
Identification of cell-specific response signatures
Trajectory analysis of developmental or differentiation processes
Mass cytometry (CyTOF):
High-dimensional phenotyping of immune and neural cell populations
Simultaneous measurement of multiple signaling pathways
Integration of cellular phenotypes with functional states
Spatial transcriptomics and proteomics:
Mapping SEMA4D expression and signaling within tissue architecture
Visualization of cellular interactions at receptor interfaces
Correlation of molecular profiles with histopathological features
Live-cell biosensors:
FRET-based reporters for real-time signaling dynamics
Optogenetic control of SEMA4D signaling components
Single-molecule tracking of SEMA4D-receptor interactions
Engineered organoid systems:
Brain organoids incorporating vascular and immune components
Controlled perturbation of SEMA4D signaling in a human tissue context
Assessment of developmental and disease-relevant processes
These cutting-edge approaches will provide unprecedented insights into the cellular and molecular mechanisms of SEMA4D function and the effects of antibody-mediated intervention, advancing both basic research and therapeutic development .