sCD40L Human, His mediates immune and inflammatory responses through CD40 receptor interactions:
Endothelial effects: sCD40L (5 µg/mL) decreases endothelial nitric oxide synthase (eNOS) mRNA by 61% and increases superoxide anion (O₂⁻) production by 76% in human coronary artery endothelial cells (HCAECs) .
Metabolic syndrome correlation: Serum sCD40L levels rise with BMI (r = 0.283, P < 0.001) and waist circumference (r = 0.239, P = 0.003) .
sCD40L Human, His is used to:
Sickle cell anemia (SCA): sCD40L levels are 30-fold higher in HbSS plasma vs. controls, correlating with tissue factor (TF) elevation (R² = 0.600) and ICAM-1 overexpression .
Obesity/metabolic syndrome: sCD40L levels correlate with hsCRP (r = 0.090, P = 0.278) and white blood cell count (r = 0.168, P = 0.042) .
sCD40L Human, His is a biomarker and therapeutic target in:
CD40 ligand (CD40L, also known as CD154, TRAP, or gp39) is a 261 amino acid type II transmembrane glycoprotein belonging to the TNF family. It exists in both membrane-bound (mCD40L) and soluble (sCD40L) forms. The soluble form is primarily derived from activated platelets through proteolytic cleavage of the membrane-bound form. While membrane-bound CD40L is predominantly expressed on activated CD4+ T lymphocytes, the soluble form circulates in plasma and can act on distant cells expressing the CD40 receptor. Structurally, recombinant His-tagged human sCD40L typically contains amino acids 116-261 of the native protein with an N-terminal His-tag to facilitate purification and detection .
The biological activity of sCD40L depends on its trimerization, as the trimeric form has the most potent biological activity through oligomerization of cell surface CD40, which is a common feature of TNF receptor family members .
In pathological conditions, elevated sCD40L levels are observed in several diseases:
In cancer patients, tumor microenvironments can stimulate increased production of sCD40L
In acute coronary syndrome, platelet activation leads to elevated sCD40L levels
In inflammatory and autoimmune disorders, activated immune cells contribute to elevated sCD40L levels
During blood component storage, especially platelet concentrates, sCD40L accumulates over time
sCD40L signaling is primarily mediated through its interaction with CD40, a type I transmembrane glycoprotein belonging to the TNF receptor family. This interaction triggers several downstream signaling pathways:
Recruitment of TNF receptor-associated factors (TRAFs): Upon binding of sCD40L to CD40, different TRAF proteins (particularly TRAF2) are recruited to the receptor complex in a cell-type and stimulus-dependent manner .
Activation of small GTPases: sCD40L stimulation induces the activation of the small GTPase Rac1, which leads to downstream signaling events .
MAPK pathway activation: The sCD40L-CD40 interaction activates p38 MAPK and ERK1/2, which contribute to various cellular responses including platelet shape change and actin polymerization .
NF-κB pathway activation: sCD40L-CD40 interaction leads to phosphorylation of IκBα and subsequent nuclear translocation of NF-κB, promoting inflammatory gene expression .
Notably, in platelets, sCD40L can also bind to the integrin αIIbβ3 through its KGD sequence, activating a distinct signaling pathway that contributes to platelet aggregation and thrombus stability .
sCD40L has emerged as a valuable biomarker in several disease states, with methodological considerations for its measurement and interpretation:
Cancer:
Studies have shown significantly elevated serum sCD40L levels in cancer patients compared to healthy donors. These elevated levels have been associated with immunosuppressive effects, suggesting sCD40L as a potential biomarker for cancer-related immune dysfunction .
Acute Coronary Syndrome (ACS):
sCD40L has significant predictive ability for ACS, particularly for STEMI and NSTEMI. ROC curve analysis has demonstrated that sCD40L can distinguish ACS patients from healthy controls with reasonable sensitivity and specificity, although its predictive value for unstable angina was not statistically significant in some studies .
Rheumatic Diseases (RDs):
Meta-analysis data indicates that sCD40L and sCD40 concentrations are significantly higher in RD patients compared to healthy controls. sCD40 concentrations, in particular, are significantly higher in RD patients with active disease compared to those with inactive disease, suggesting its potential utility in monitoring disease activity .
Hypercholesterolemia in Children:
Studies have found significantly higher levels of sCD40L in children and adolescents with hypercholesterolemia compared to healthy controls, suggesting it could serve as an early marker of cardiovascular risk in pediatric populations .
When using sCD40L as a biomarker, researchers should consider:
Standardization of collection methods (EDTA vs. serum tubes)
Time to processing (as platelets can release additional sCD40L ex vivo)
Storage conditions of samples (-80°C is recommended)
Use of validated ELISA kits with established reference ranges
Controlling for potential confounding factors (medication use, comorbidities)
sCD40L plays a complex and sometimes contradictory role in cancer immunology, with significant effects on the tumor microenvironment:
Immunosuppressive effects:
Research has demonstrated that cancer patients have significantly elevated serum sCD40L levels compared to healthy donors. This elevation contributes to immunosuppression through several mechanisms:
Expansion of myeloid-derived suppressor cells (MDSCs): Cancer patients show a larger population of MDSCs (defined as CD33+HLA-DR− cells) with higher CD40 expression. sCD40L enriches these MDSCs and enhances their immunosuppressive functions .
Inhibition of T-cell responses: When T cells are cocultured with MDSCs, their proliferation and IFN-γ production decrease. Addition of sCD40L further inhibits T-cell proliferation .
Expansion of regulatory T cells (Tregs): sCD40L promotes the expansion of CD4+CD25highFoxp3+ regulatory T cells, which suppress anti-tumor immune responses .
Induction of immunosuppressive cytokines: sCD40L stimulates the production of IL-10 and IL-6, cytokines that can promote immunosuppression .
Enhancement of PD-1 expression: sCD40L induces greater programmed death-1 (PD-1) expression on T cells from cancer patients compared to healthy donors, potentially contributing to T-cell exhaustion .
These findings suggest that elevated sCD40L in the tumor microenvironment may contribute to tumor immune evasion and resistance to immunotherapy. Targeting the sCD40L-CD40 pathway could potentially restore anti-tumor immunity in certain contexts .
sCD40L significantly contributes to cardiovascular pathology through multiple mechanisms:
Endothelial dysfunction: sCD40L decreases endothelial nitric oxide synthase (eNOS) mRNA and protein levels, reduces eNOS enzyme activity, and decreases cellular NO production. Simultaneously, it increases superoxide anion (O2−) production through enhanced NADPH oxidase activity and mitochondrial dysfunction. This leads to impaired endothelium-dependent vasorelaxation, a hallmark of early vascular disease .
Enhanced platelet activation and aggregation: sCD40L acts as a platelet primer, significantly enhancing platelet activation and aggregation in response to conventional agonists. This effect is mediated through:
Thrombus formation: In experimental models, sCD40L exacerbates thrombus formation and leukocyte infiltration in wild-type mice but not in CD40−/− mice, demonstrating the CD40-dependency of this effect. The sCD40L-enhanced platelet activation is mediated by its KGD sequence, a known αIIbβ3 binding motif .
Inflammatory signaling: sCD40L activates MAPKs p38 and ERK1/2 as well as IκBα, enhancing NF-κB nuclear translocation and promoting inflammatory responses in vascular cells .
Biomarker for acute coronary syndrome (ACS): Elevated sCD40L levels serve as a prognostic marker for ACS patients, with significantly higher concentrations in ACS patients compared to control groups. ROC curve analysis has demonstrated that sCD40L has significant predictive ability for ACS subtypes, particularly STEMI and NSTEMI .
These mechanisms highlight why elevated circulating levels of sCD40L are associated with increased cardiovascular risk and adverse outcomes in patients with existing cardiovascular disease .
For accurate measurement of sCD40L in biological samples, researchers should follow these methodological recommendations:
Sample Collection and Processing:
Blood collection tubes: Serum collection tubes (without additives) are commonly used, although EDTA tubes may be preferred for specific applications. Note that the choice of collection tube can affect measured levels .
Sample processing time: Process samples within 30 minutes of collection to minimize ex vivo release of sCD40L from platelets. If immediate processing is not possible, document the time to processing and control for this variable .
Centrifugation protocol: For plasma, centrifuge at 1500-2000g for 15 minutes at room temperature. For platelet-poor plasma (recommended for some applications), perform a second centrifugation at 10,000g for 10 minutes .
Storage conditions: Store samples at −80°C until analysis. Avoid repeated freeze-thaw cycles as they can affect sCD40L stability. For long-term storage, aliquoting samples is recommended .
Measurement Techniques:
ELISA (enzyme-linked immunosorbent assay): This is the most common method for sCD40L quantification.
Multiplex assays: Can be used to simultaneously measure sCD40L alongside other cytokines or biomarkers .
Flow cytometry: For cellular expression of CD40L rather than soluble forms.
Data Analysis Considerations:
Statistical approaches:
Reporting results:
Quality control:
Proper preparation and storage of recombinant His-tagged sCD40L is critical for maintaining its biological activity:
Reconstitution:
Lyophilized recombinant His-tagged sCD40L should be reconstituted in sterile ultrapure water (18MΩ-cm) to a concentration of at least 100 μg/mL, which can then be further diluted to working concentrations in appropriate buffers .
For cell culture applications, dilute the stock solution in serum-free medium immediately before use. Avoid repeated freeze-thaw cycles of diluted solutions .
For in vivo applications, ensure the preparation is endotoxin-free and dilute in sterile PBS or other appropriate physiological buffers .
Storage Conditions:
Short-term storage (lyophilized): Lyophilized sCD40L is generally stable at room temperature for up to 3 weeks, but should ideally be stored desiccated below -18°C for longer periods .
Post-reconstitution storage (short-term): Store at 4°C for use within 2-7 days .
Post-reconstitution storage (long-term): Store below -18°C and add a carrier protein (0.1% HSA or BSA) to enhance stability. Aliquot to avoid repeated freeze-thaw cycles .
Avoid freeze-thaw cycles: Each cycle can result in loss of biological activity. It is recommended to create single-use aliquots upon reconstitution .
Quality Control:
Purity assessment:
Activity verification:
Protein concentration determination:
Working Concentration Ranges:
For most in vitro experiments, sCD40L is typically used at concentrations ranging from 10 ng/mL to 5 μg/mL, with 100-1000 ng/mL being the most common range for observing biological effects in cell culture systems .
Several experimental models have proven valuable for investigating different aspects of sCD40L function:
In Vitro Models:
Primary Human Cell Cultures:
Human coronary artery endothelial cells (HCAECs): Ideal for studying endothelial dysfunction, as these cells express CD40 and respond to sCD40L with altered eNOS expression and activity .
Peripheral blood mononuclear cells (PBMCs): Useful for investigating immune cell responses to sCD40L, including effects on T-cell activation, MDSC function, and cytokine production .
Isolated platelets: Excellent for studying sCD40L's effects on platelet activation, aggregation, and thrombus formation .
Cell Lines:
Co-culture Systems:
T cell-MDSC co-cultures: Valuable for studying sCD40L's immunosuppressive effects in cancer models .
Platelet-endothelial cell co-cultures: For investigating vascular inflammation and thrombosis.
Three-dimensional culture systems: More physiologically relevant than monolayer cultures for studying complex cellular interactions .
In Vivo Models:
Mouse Models:
CD40L knockout (CD40L-/-) mice: Essential for establishing the specificity of observed effects to CD40L signaling .
CD40 knockout (CD40-/-) mice: Complementary to CD40L-/- models for confirming receptor specificity .
Atherosclerosis models (ApoE-/- or LDLR-/- mice): Useful for studying sCD40L's role in vascular inflammation and atherosclerotic plaque development.
Cancer xenograft models: For investigating sCD40L's effects on tumor growth and the tumor microenvironment .
Thrombosis Models:
Ferric chloride-induced arterial thrombosis: Particularly useful for studying sCD40L's role in thrombus formation and stability .
Intravital microscopy of cremaster muscle microcirculation: Allows real-time visualization of platelet-vessel wall interactions under the influence of sCD40L.
Pulmonary microvascular injury models: For studying sCD40L's role in transfusion-related acute lung injury (TRALI) .
Ex Vivo Models:
Isolated Vessel Preparations:
Whole Blood Assays:
Flow chamber systems: For studying platelet adhesion and aggregation under flow conditions.
Thromboelastography: For assessing sCD40L's effects on whole blood coagulation parameters.
When selecting an experimental model, researchers should consider:
Relevance to the specific aspect of sCD40L biology being studied
Species differences in CD40-CD40L interactions
Availability of appropriate controls (e.g., blocking antibodies, knockout models)
Complementary approaches to validate findings across different model systems
The structural organization of sCD40L significantly impacts its biological functions, with important distinctions between monomeric and trimeric forms:
Receptor Binding Properties:
While all forms (monomeric, dimeric, and trimeric) can bind to CD40, the trimeric form demonstrates the most potent biological activity through efficient oligomerization of cell surface CD40 .
Binding kinetics and affinity measurements reveal:
Differential Biological Activities:
Immune Cell Activation:
Trimeric sCD40L is significantly more potent at inducing B-cell proliferation and antibody production
Monomeric sCD40L can act as a partial antagonist in some systems by binding CD40 without efficient receptor clustering
In cancer immunology, recombinant monomeric sCD40L enriches MDSCs and has an inhibitory effect on T-cell proliferation, suggesting distinct functional properties
Platelet Activation:
Signaling Pathway Activation:
Trimeric sCD40L more efficiently recruits TRAFs to CD40's cytoplasmic domain
Differences in MAPK and NF-κB pathway activation kinetics and magnitude exist between monomeric and trimeric forms
Certain downstream effects may require the higher-order clustering of CD40 that only trimers can efficiently induce
Experimental and Therapeutic Implications:
Researchers should carefully consider the oligomeric state of sCD40L preparations in experimental design:
Commercial recombinant proteins may vary in their proportions of monomeric vs. trimeric forms
Expression systems (bacterial vs. mammalian) influence trimerization efficiency
Storage conditions and freeze-thaw cycles can affect oligomeric stability
For therapeutic applications, engineered stabilized trimers may offer superior activity profiles
Understanding these differences is critical for interpreting experimental results and developing CD40L-based therapeutic approaches.
The relationship between platelet-derived sCD40L and transfusion-related acute lung injury (TRALI) represents a critical area of transfusion medicine research:
sCD40L Accumulation in Blood Components:
Soluble CD40L accumulates during storage of blood components, with particularly high levels in platelet concentrates (PCs). Studies have shown that:
All blood components contain higher levels of sCD40L than fresh plasma, with a hierarchy of sCD40L concentration:
The accumulation increases with storage duration, with levels rising significantly after just a few days of storage
PCs implicated in TRALI reactions contain significantly higher sCD40L levels than control PCs that did not elicit transfusion reactions
Mechanistic Links to TRALI:
TRALI is characterized by acute pulmonary endothelial damage and neutrophil infiltration following transfusion. sCD40L contributes to TRALI pathogenesis through several mechanisms:
Neutrophil Priming and Activation:
Two-event Model of TRALI:
In a "two-hit" model of TRALI, sCD40L can serve as either the first or second event
As a first hit, sCD40L primes neutrophils, making them more responsive to subsequent inflammatory signals
As a second hit, sCD40L promotes PMN-mediated cytotoxicity of human pulmonary microvascular endothelial cells (HMVECs) in pre-primed recipients
Endothelial Dysfunction:
Clinical Implications and Risk Mitigation:
Understanding the role of sCD40L in TRALI has important implications for transfusion practice:
Risk Assessment:
Storage Strategies:
Targeted Interventions:
CD40L-CD40 pathway blocking agents could potentially reduce TRALI risk in high-risk transfusions
Antioxidants or inhibitors of neutrophil activation may mitigate sCD40L-mediated effects
Patient-specific risk factors (prior inflammation, sepsis) may identify individuals at heightened risk for sCD40L-mediated TRALI
The identification of sCD40L as a mediator in TRALI represents an important advance in understanding this serious transfusion complication and may lead to improved risk assessment and prevention strategies.
The role of sCD40L in creating an immunosuppressive tumor microenvironment has emerged as a significant area of cancer immunology research with important therapeutic implications:
Mechanisms of sCD40L-Mediated Immunosuppression in Cancer:
Expansion and Activation of Myeloid-Derived Suppressor Cells (MDSCs):
Cancer patients have significantly elevated serum sCD40L compared to healthy donors
PBMCs from cancer patients contain larger populations of MDSCs (CD33+HLA-DR− cells) with higher CD40 expression
These MDSCs express higher levels of CD40 (20% in cancer patients vs. 2% in healthy donors)
sCD40L directly enriches MDSCs and enhances their immunosuppressive function
T-cell proliferation and IFN-γ production decrease when T cells are cocultured with MDSCs, an effect amplified by sCD40L
Regulatory T Cell (Treg) Expansion:
Induction of Inhibitory Checkpoint Expression:
sCD40L induces greater programmed death-1 (PD-1) expression on T cells from cancer patients compared to healthy donors
There is a 5.5-fold increase in PD-1 expression for CD4+ T cells and a 9.7-fold increase for CD8+ T cells in cancer patients
Enhanced PD-1 expression contributes to T cell exhaustion and dysfunction
Cytokine Modulation:
Therapeutic Implications and Strategies:
sCD40L as a Therapeutic Target:
Combination with Checkpoint Inhibitors:
Targeting the MDSC-Treg Axis:
Strategies to deplete or reprogram MDSCs could counteract sCD40L's immunosuppressive effects
Combinatorial approaches targeting both sCD40L signaling and Treg function might overcome resistance to current immunotherapies
The CD40-MDSC-Treg axis represents a potential therapeutic vulnerability in many cancers
Biomarker-Guided Treatment Selection:
Paradoxical CD40 Agonism:
Despite sCD40L's immunosuppressive effects, strong CD40 agonism with antibodies has shown anti-tumor activity
This apparent contradiction might be explained by differential effects of varying CD40 stimulation intensity and context
Understanding this paradox is essential for developing optimal therapeutic strategies
Understanding sCD40L's contributions to tumor immunosuppression provides new avenues for therapeutic intervention in cancer. As our knowledge of this pathway deepens, more targeted and effective immunotherapeutic approaches may emerge to overcome the immunosuppressive tumor microenvironment.
Several cutting-edge technologies are poised to transform our understanding of sCD40L biology and its role in disease:
Single-cell Analysis Technologies:
Single-cell RNA sequencing can identify specific cell populations responsive to sCD40L with unprecedented resolution
Mass cytometry (CyTOF) allows simultaneous detection of dozens of cell surface and intracellular markers to characterize sCD40L-responsive cells
Single-cell proteomics techniques can elucidate how sCD40L alters protein expression patterns at the individual cell level
These approaches can clarify cell-specific responses to sCD40L in heterogeneous samples like tumor microenvironments or blood
Advanced Imaging Techniques:
Intravital microscopy enables visualization of sCD40L-mediated interactions in living tissues
Super-resolution microscopy can reveal nanoscale organization of CD40 receptors upon sCD40L binding
Correlative light and electron microscopy (CLEM) can connect functional responses to ultrastructural changes
These techniques may provide insights into how sCD40L influences cellular behavior in physiologically relevant contexts
Proteomics and Interactomics:
Proximity labeling techniques (BioID, APEX) can identify proteins interacting with CD40 after sCD40L stimulation
Phosphoproteomics can map signaling networks activated by sCD40L with temporal resolution
Secretome analysis can characterize how sCD40L alters cellular secretion profiles
These approaches may reveal new components of sCD40L signaling pathways and identify potential therapeutic targets
CRISPR-based Functional Genomics:
Genome-wide CRISPR screens can identify genes essential for sCD40L responses
CRISPR activation/inhibition screens can map regulatory networks controlling CD40L expression
Base editing and prime editing enable precise modification of CD40L or CD40 to study structure-function relationships
These approaches may uncover new regulators and effectors of sCD40L signaling
Humanized Mouse Models:
Immune system humanized mice can better recapitulate human sCD40L biology
Patient-derived xenograft models can assess sCD40L's role in specific disease contexts
Conditional knockout models with human immune components can provide insights into tissue-specific functions
These models may bridge the translational gap between basic research and clinical applications
Multiplexed Biomarker Profiling:
Multiplex immunoassay panels can simultaneously measure sCD40L alongside dozens of other biomarkers
Machine learning algorithms can identify biomarker signatures incorporating sCD40L for disease diagnosis or prognosis
Point-of-care testing devices may enable rapid sCD40L quantification in clinical settings
These approaches may enhance the clinical utility of sCD40L as a biomarker
Structural Biology Advances:
Cryo-electron microscopy can determine high-resolution structures of sCD40L-CD40 complexes
Hydrogen-deuterium exchange mass spectrometry can map conformational changes upon receptor binding
Molecular dynamics simulations can predict how different forms of sCD40L interact with receptors
These techniques may guide the design of more specific modulators of CD40-CD40L interactions
The integration of these emerging technologies promises to significantly advance our understanding of sCD40L biology and accelerate the development of targeted therapies for diseases involving CD40-CD40L dysregulation.
Despite significant advances in understanding sCD40L biology, several important questions and controversies remain unresolved:
Functional Duality: Friend or Foe?
Controversy: sCD40L demonstrates both pro-inflammatory and immunosuppressive effects depending on the context, creating apparent contradictions in the literature.
Unresolved Questions:
Receptor Specificity and Hierarchy:
Controversy: While CD40 is the canonical receptor for sCD40L, it also binds to other receptors like αIIbβ3 integrin, raising questions about receptor hierarchy and specificity.
Unresolved Questions:
Source-Dependent Functional Differences:
Controversy: sCD40L derived from different cellular sources (platelets vs. T cells) may have distinct functional properties, but evidence is limited.
Unresolved Questions:
Biomarker Utility and Standardization:
Controversy: While numerous studies suggest sCD40L as a biomarker for various diseases, significant methodological variability and inconsistent results limit clinical application.
Unresolved Questions:
Cause vs. Consequence in Disease:
Controversy: Whether elevated sCD40L is a cause or consequence of disease processes remains debated across multiple conditions.
Unresolved Questions:
Therapeutic Targeting Strategies:
Controversy: Whether to block or stimulate CD40-CD40L signaling therapeutically remains context-dependent and controversial.
Unresolved Questions:
In what disease contexts should CD40-CD40L signaling be blocked versus enhanced?
How can therapeutic interventions target specific aspects of CD40-CD40L signaling while preserving beneficial functions?
What are the long-term consequences of modulating this pathway given its fundamental role in immunity?
Regulatory Mechanisms:
Controversy: The mechanisms regulating sCD40L production, release, and clearance remain incompletely understood.
Unresolved Questions:
Interaction with Other Signaling Systems:
Controversy: How sCD40L interacts with other inflammatory and immune signaling pathways remains incompletely mapped.
Unresolved Questions:
Resolving these questions and controversies will require interdisciplinary approaches combining advanced molecular and cellular techniques with carefully designed clinical studies. Progress in addressing these unresolved issues will significantly advance our understanding of sCD40L biology and its therapeutic potential.
Soluble CD40 Ligand (sCD40L), also known as Tumor Necrosis Factor-related Activation Protein (TRAP), is a recombinant protein that plays a crucial role in the immune system. It is a member of the tumor necrosis factor (TNF) superfamily and is primarily expressed on activated T cells . The recombinant form of this protein, tagged with a His tag, is widely used in research to study its biological functions and interactions.
The soluble CD40 Ligand/TRAP is produced as a recombinant protein in various expression systems, including E. coli and HEK 293 cells . The protein is typically fused with a His tag at the N-terminus to facilitate purification. The recombinant protein consists of 169 amino acids and has a molecular mass of approximately 18.3 kDa . The His tag allows for easy purification using nickel affinity chromatography, ensuring high purity and yield.
CD40 Ligand (CD40L) interacts with its receptor CD40, which is expressed on antigen-presenting cells (APCs) such as B cells, dendritic cells, and macrophages . This interaction is critical for various immune responses, including:
CD40L is primarily expressed on activated CD4+ T lymphocytes . However, it is also found on other cell types, including platelets and endothelial cells . The expression of CD40L is tightly regulated and is induced upon T cell activation. Soluble CD40L (sCD40L) is generated through proteolytic cleavage of the membrane-bound form and can be detected in the circulation .
The expression and activity of CD40L are regulated at multiple levels:
Recombinant soluble CD40 Ligand/TRAP with a His tag is widely used in research to study its role in immune responses and its potential therapeutic applications. It is used in various assays, including: