SEMA4D Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

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.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

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.

Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your orders. Delivery time may vary depending on the purchase method or location. Please contact your local distributor for specific delivery details.
Synonyms
Semaphorin-4D (A8) (BB18) (GR3) (CD antigen CD100), SEMA4D, C9orf164 CD100 SEMAJ
Target Names
Uniprot No.

Target Background

Function
SEMA4D serves as a cell surface receptor for PLXNB1 and PLXNB2, playing a pivotal role in cell-cell signaling. It regulates GABAergic synapse development, promoting the formation of inhibitory synapses in a PLXNB1-dependent manner. SEMA4D modulates the complexity and arborization of developing neurites in hippocampal neurons by activating PLXNB1. Its interaction with PLXNB1 mediates the activation of RHOA. SEMA4D promotes the migration of cerebellar granule cells. It also plays a role in the immune system, inducing B-cells to aggregate and enhancing their viability (in vitro). Additionally, SEMA4D induces endothelial cell migration through the activation of PTK2B/PYK2, SRC, and the phosphatidylinositol 3-kinase-AKT pathway.
Gene References Into Functions
  1. Combined detection of tumor-associated macrophages markers, CD68 and Sema4D, in gastric carcinoma tissue shows potential to predict the trend of gastric carcinoma progression. PMID: 29434448
  2. SEMA4D coordinates with VEGF during angiogenesis via plexin-B1 in epithelial ovarian cancer. PMID: 29308068
  3. Knockdown of Sema4D in Head and neck squamous cell carcinomas (HNSCCs) cells inhibited tumor growth and decreased the number of osteoclasts in a mouse xenograft model. Taken together, IGF-I-driven production of Sema4D in HNSCCs promotes osteoclastogenesis and bone invasion. PMID: 28656278
  4. The results obtained provide significant insights into understanding of Seam4D effects in lymphoid cells. PMID: 28508207
  5. This study describes the transfection, purification, and visualization of Fc-tagged SEMA4D (semaphorin 4D) recombinant protein. PMID: 27787840
  6. This research details the application of in vitro migration and tubulogenesis assays and the directed in vivo angiogenesis assay (DIVAA) in the measurement of the angiogenic potential of cell-derived and soluble SEMA4D. PMID: 27787869
  7. The positive expression of both Sema4D and PlexinB1 was found to be an independent risk factor for a worse survival in colorectal cancer. PMID: 27456345
  8. Serum levels of soluble SEMA4D were elevated in patients with ANCA-associated vasculitis. Cell-surface expression of SEMA4D was downregulated, a consequence of proteolytic cleavage of membrane SEMA4D. Soluble SEMA4D exerted pro-inflammatory effects on endothelial cells. Membranous SEMA4D on neutrophils bound to plexin B2 on endothelial cells, and this interaction decreased NET formation. PMID: 28416516
  9. A novel genome-wide significant African-specific locus for BMI (SEMA4D, rs80068415) was identified. A novel variant in SEMA4D was significantly associated with body mass index. Carriers of the C allele were 4.6 BMI units heavier than carriers of the T allele. PMID: 28296344
  10. This study highlights a critical pathogenic engagement of Semaphorin 4D produced by gamma delta T cells in the development of medication-related osteonecrosis of the jaw. PMID: 27720716
  11. This research identified FGL2, GAL, SEMA4D, SEMA7A, and IDO1 as new candidate genes that could be involved in MSCs-mediated immunomodulation. FGL2, GAL, SEMA4D, SEMA7A, and IDO1 genes appeared to be differentially transcribed in the different MSC populations. Moreover, these genes were not similarly modulated following MSCs-exposure to inflammatory signals. PMID: 28336906
  12. Interferon-alpha-induced CD100 expression on naive CD8(+) T cells enhances antiviral responses to hepatitis C infection through CD72 signal transduction. PMID: 28222623
  13. This review summarizes current findings on neuroimmune Sema4A and Sema4D molecules in chronic inflammation underlying many diseases and discusses their positive or negative impacts on the implicated molecular and cellular processes. PMID: 27554682
  14. A novel reverse signaling pathway acting through Tiam1 and Rac that promotes aggressive behavior in OSCC expressing S4D and PB1 was identified. PMID: 28038319
  15. Sema 4D was found to be the direct target of miR-214 and was negatively regulated by miR-214 in ovarian cancer cells. PMID: 26718213
  16. Tax and semaphorin 4D released from lymphocytes infected with human lymphotropic virus type 1 inhibit neurite growth in a neuron cell line. PMID: 26389656
  17. This assay specifically and reproducibly measured cSEMA4D saturation and expression levels. Evaluation of the SEMA4D-specific PD markers were critical in determining the clinical saturation threshold of cSEMA4D by VX15/2503. PMID: 26566052
  18. Serum sSema4D levels are increased in patients with atrial fibrillation and are independently associated with atrial remodeling. PMID: 26417899
  19. Semaphorin 4D Promotes Skeletal Metastasis in Breast Cancer. PMID: 26910109
  20. This study describes a novel immunosuppressive role for Sema4D in head and neck squamous cell carcinoma through induction of myeloid derived suppressor cells. PMID: 26740106
  21. Plexin-B1 induces cutaneous squamous cell carcinoma cell proliferation, migration, and invasion by interacting with Sema4D. Plexin-B1 might serve as a useful biomarker and/or as a novel therapeutic target for cSCC. PMID: 26051877
  22. Results show that decreased expression of Sema4D, plexin-B1 and -B2 was associated with local recurrence and poor prognosis of breast neoplasm. PMID: 26035216
  23. Blocking of CD100, plexin B1 and/or B2 in adhesion experiments have shown that both CD100 and plexins act as adhesion molecules involved in monocyte-endothelial cell binding. PMID: 26275342
  24. This study suggests that HIF-1alpha and Sema4D expression correlates with histological tumor type, TNM stage, and lymphatic metastasis in colorectal carcinoma. PMID: 25717256
  25. A positive feedback loop involving sSema4D/IL-6 and TNFalpha/ADAMTS-4 may contribute to the pathogenesis of rheumatoid arthritis. PMID: 25707877
  26. Sema4D contributes to enhanced invasion and tumor progression through increased motility of cervical cancer and VEGF-C/-D-mediated lymphangiogenesis. PMID: 24603190
  27. SEMA4D might possibly serve as a reliable tool for early and accurate prediction of EOC poor prognosis. PMID: 24289594
  28. The results suggest that the contribution of Sema4D in platelets applies to ITAM-containing receptors as a class. PMID: 24131822
  29. Sema4D could play an important role in promoting tumor proliferation, migration and metastasis in the NSCLC, by influencing the Akt protein phosphorylation. Inhibition of Sema4D may be a useful approach for the treatment of NSCLC. PMID: 25135716
  30. Data suggest that CD100 seems to be involved in hepatitis C virus (HCV) clearance by natural killer (NK) cells. PMID: 25108441
  31. The Semaphorin 4D-Plexin-B1-RhoA signaling axis recruits pericytes and regulates vascular permeability through endothelial production of PDGF-B and ANGPTL4. PMID: 24114199
  32. Elevated plasma soluble Sema4D/CD100 levels are associated with disease severity in patients of hemorrhagic fever with renal syndrome. PMID: 24040126
  33. CD100 may have a role in atherosclerotic plaque development, and may possibly be employed in targeted treatments of these atheromas. PMID: 24098722
  34. There was an increased level of plasma soluble Sema4 in the Sema4D(high) population of T-cells suggesting a potential role of these T-cells in heart failure. PMID: 23741311
  35. The membrane-proximal cytoplasmic domain of Sema4D contains a binding site for calmodulin within the polybasic region Arg762-Lys779, that regulates Sema4D exodomain shedding in platelets. PMID: 23564909
  36. Copy number loss of the Sema4D gene region may play a role in the etiology of acetabular dysplasia. PMID: 23335257
  37. Lycorine hydrochloride suppressed the expression of several key angiogenic genes, including VE-cadherin and Sema4D, and reduced Akt phosphorylation in Hey1B cells. PMID: 23376478
  38. CD72 mRNA expression level correlates with Sema4D expression in peripheral blood mononuclear cells in immune thrombocytopenia. PMID: 22111667
  39. CD100 protein levels were highly dysregulated around 10 weeks of gestation in first and second miscarriage placentas. The CD100 soluble form was produced and immediately shed from placental tissue in all samples. PMID: 22606231
  40. The expression of semaphorin 4D (SEMA4D), which is under the control of the HIF-family of transcription factors, cooperates with VEGF to promote tumor growth and vascularity in oral squamous cell carcinoma (OSCC). PMID: 22652457
  41. Sema4D, the ligand for Plexin B1, suppresses c-Met activation and migration and promotes melanocyte survival and growth. PMID: 22189792
  42. Sema4D potentiates the invasiveness of pancreatic cancer cells. The binding of Sema4D to plexinB1 induced small GTPase Ras homolog gene family, member A activation and resulted in the phosphorylation of MAPK and Akt. PMID: 21812859
  43. From the data obtained in this study, SEMA4D may have a role in more aggressive and potentially metastatic breast tumors. PMID: 21925246
  44. Rho-mediated activation of PI(4)P5K and lipid second messengers is necessary for promotion of angiogenesis by Semaphorin 4D. PMID: 21538148
  45. Dysregulations in CD100 expression and release could play a role in SSc development and/or maintenance. PMID: 21244334
  46. Crystal structures of cognate complexes of the semaphorin-binding regions of plexins B1 and A2 with semaphorin ectodomains (human PLXNB1(1-2)-SEMA4D(ecto) and murine PlxnA2(1-4)-Sema6A(ecto)), plus unliganded structures of PlxnA2(1-4) and Sema6A(ecto). PMID: 20877282
  47. CD100-CD72 interaction can be the mechanism by which NK cell communicate with B cells. PMID: 17786190
  48. Leukemic & normal CD5+ B cells express CD100; upon interaction between CD100 & Plexin-B1, both increase their proliferative activity & life span. CD100/Plexin-B1 crosstalk is not malignancy related but reproduces a mechanism used by normal CD5+ B cells. PMID: 12406905
  49. Soluble CD100 induces a progressive decrease in process extension of oligodendrocytes, followed by their death and the death of multipotent neural precursors. PMID: 14707103
  50. Up-modulation of the survival receptor CD100 is restricted to proliferating B-cell leukemia cells. PMID: 15613544

Show More

Hide All

Database Links

HGNC: 10732

OMIM: 601866

KEGG: hsa:10507

STRING: 9606.ENSP00000348822

UniGene: Hs.494406

Protein Families
Semaphorin family
Subcellular Location
Cell membrane; Single-pass type I membrane protein.
Tissue Specificity
Strongly expressed in skeletal muscle, peripheral blood lymphocytes, spleen, and thymus and also expressed at lower levels in testes, brain, kidney, small intestine, prostate, heart, placenta, lung and pancreas, but not in colon and liver.

Q&A

What is the molecular structure of SEMA4D and how does it relate to antibody design?

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 .

How is SEMA4D expression regulated in different cell types relevant to research applications?

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 .

How do anti-SEMA4D antibodies affect the interactions with different receptors, and what methodologies can measure this interference?

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 .

What signaling cascades are initiated by SEMA4D-receptor interactions that can be modulated by antibody treatment?

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

What strategies overcome immunological tolerance when generating high-affinity anti-SEMA4D antibodies with cross-species reactivity?

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

What are the critical quality attributes and analytical methods for characterizing anti-SEMA4D antibodies for research applications?

AttributeAnalytical MethodAcceptance Criteria
Antigen SpecificityELISA, Flow cytometry, IHCHigh specificity for SEMA4D across species; no binding to irrelevant proteins
AffinityBiacore, Cell-based flow cytometryKD for recombinant SEMA4D: 1-5 nM; KD for cell-associated SEMA4D: ~0.45 nM
Receptor Blocking PotencyFlow cytometry-based binding assayEC50 for PLXNB1 blocking: ~1.2 nM
Species Cross-reactivityELISA, Flow cytometry using species-specific cellsConsistent binding across mouse, rat, rabbit, cynomolgus macaque, marmoset, rhesus macaque, and human
Effector FunctionsCDC, ADCC assaysMinimal effector functions for research applications to avoid confounding cell depletion
Epitope SpecificityCompetition assaysConsistent 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 .

How can anti-SEMA4D antibodies be used to study blood-brain barrier integrity and what specific methodologies provide quantitative assessment?

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 .

What experimental approaches best demonstrate the effects of anti-SEMA4D antibodies on oligodendrocyte precursor cell (OPC) migration and differentiation?

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 .

How do anti-SEMA4D antibodies affect immune cell functions in the tumor microenvironment, and what assays best evaluate these effects?

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 .

What considerations are important when designing experiments to evaluate anti-SEMA4D antibodies in combination with other immunotherapeutic approaches?

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 .

How can researchers effectively study the role of SEMA4D in platelet function and thrombus formation using anti-SEMA4D antibodies?

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 .

What molecular mechanisms explain the dual functions of membrane-bound versus shed SEMA4D in vascular biology?

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 .

What controls and validation steps are critical when using anti-SEMA4D antibodies to ensure experimental rigor and reproducibility?

Experimental Control/ValidationImplementation MethodPurpose
Isotype Control AntibodiesInclude matched isotype control at equivalent concentrationsDistinguish specific from non-specific effects
Genetic ControlsUse SEMA4D knockout cells/animals as negative controlsConfirm antibody specificity and validate phenotypes
Epitope CompetitionPre-incubate with recombinant SEMA4DDemonstrate binding specificity
Positive ControlsInclude known SEMA4D-dependent phenotypesEnsure system responsiveness
Receptor Blockade ValidationFlow cytometry-based receptor binding assaysConfirm functional blocking activity
Batch ConsistencyTest multiple antibody lots for consistent affinity and functionEliminate batch variation as a confounder
Concentration TitrationUse multiple antibody concentrationsEstablish dose-response relationships
Pharmacokinetic AssessmentIn vivo studies to determine half-life and tissue penetrationEnsure appropriate dosing schedules
Multiple Detection MethodsCombine flow cytometry, IHC, Western blottingCross-validate findings across platforms

These controls and validation steps ensure experimental rigor and facilitate reproducibility across different research groups and experimental systems .

How can researchers overcome challenges in studying species-specific SEMA4D effects when translating findings between rodent models and human 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 .

How can researchers utilize anti-SEMA4D antibodies to investigate the complex interplay between neural, vascular, and immune systems in neurological disorders?

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 .

What emerging technologies and methods can enhance the study of SEMA4D functions and anti-SEMA4D antibody effects at single-cell resolution?

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 .

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 2024 Thebiotek. All Rights Reserved.