THBD binds thrombin to form a 1:1 complex, converting thrombin from a procoagulant to an anticoagulant enzyme .
This complex activates protein C, which degrades clotting factors Va and VIIIa, reducing thrombin generation .
Simultaneously inhibits fibrinolysis by activating thrombin-activatable fibrinolysis inhibitor (TAFI) .
Modulates angiogenesis and vascular remodeling via endothelial nitric oxide (NO) signaling and VEGF expression .
Regulates complement activity by aiding factor I-mediated C3b inactivation .
Study Design: Brain endothelial-specific Thbd knockout mice subjected to middle cerebral artery occlusion (MCAO) .
Key Results:
Parameter | Wild-Type vs. Thbd Knockout |
---|---|
Infarct Size | Smaller in wild-type |
Vessel Diameter | Reduced in knockout |
Proliferating Endothelial Cells | Fewer in knockout |
Mutations: Linked to atypical hemolytic-uremic syndrome (aHUS) and thrombophilia .
Variants: Rare THBD variants (e.g., rs1042579) show no significant association with venous thromboembolism in large cohorts .
Predominantly expressed in endothelial cells, with high levels in placental trophoblasts, lung, and kidney .
Detected in dendritic cell subsets (BDCA-3+) and monocytes .
Reduced THBD levels correlate with atherosclerosis, thrombosis, and diabetic complications .
Elevated soluble THBD serves as a biomarker for endothelial damage in sepsis and liver cirrhosis .
Recombinant THBD is investigated for treating coagulopathies and ischemic injuries .
Preclinical studies highlight its potential in mitigating stroke-induced vascular dysfunction .
Thrombomodulin (THBD) is a transmembrane glycoprotein primarily found on the surface of endothelial cells. It plays a crucial role in regulating blood coagulation. THBD binds to thrombin, a key enzyme in the coagulation cascade, and this interaction enhances the activation of protein C, an anticoagulant protein. The THBD-thrombin complex promotes the conversion of protein C to its active form, which in turn inhibits excessive clot formation. THBD also influences fibrinolysis, the process of clot breakdown, by activating thrombin-activatable fibrinolysis inhibitor (TAFI). Dysregulation of THBD levels has been implicated in cardiovascular diseases such as atherosclerosis and thrombosis. Additionally, altered THBD levels are associated with conditions like diabetes mellitus, liver cirrhosis, cerebral and myocardial infarction, and multiple sclerosis.
Recombinant human THBD, expressed in Sf9 insect cells using a baculovirus expression system, is a single, glycosylated polypeptide chain. This protein consists of 500 amino acids (residues 22-515) with an approximate molecular weight of 52.6 kDa. For purification purposes, a 6-amino acid Histidine tag is fused to the C-terminus. The protein is purified using proprietary chromatographic techniques.
The THBD protein is supplied as a solution at a concentration of 0.5 mg/ml. The solution is buffered with Phosphate-Buffered Saline (PBS) at a pH of 7.4 and contains 10% glycerol as a stabilizing agent.
For short-term storage (up to 2-4 weeks), the THBD solution should be stored at 4°C. For long-term storage, it is recommended to store the protein at -20°C. To prevent protein degradation during long-term storage, adding a carrier protein like HSA or BSA to a final concentration of 0.1% is advised. To maintain protein integrity, it is crucial to avoid repeated freeze-thaw cycles.
The purity of the THBD protein is determined using SDS-PAGE analysis and is consistently greater than 90%.
THBD, Thrombomodulin, TM, Fetomodulin, CD141, CD141 antigen, THRM, BDCA-3, BDCA3, blood dendritic cell antigen 3, AHUS6, THPH12.
Sf9, Baculovirus cells.
EPQPGGSQCV EHDCFALYPG PATFLNASQI CDGLRGHLMT VRSSVAADVI SLLLNGDGGV GRRRLWIGLQ LPPGCGDPKR LGPLRGFQWV TGDNNTSYSR WARLDLNGAP LCGPLCVAVS AAEATVPSEP IWEEQQCEVK ADGFLCEFHF PATCRPLAVE PGAAAAAVSI TYGTPFAARG ADFQALPVGS SAAVAPLGLQ LMCTAPPGAV QGHWAREAPG AWDCSVENGG CEHACNAIPG
APRCQCPAGA ALQADGRSCT ASATQSCNDL CEHFCVPNPD QPGSYSCMCE TGYRLAADQH RCEDVDDCIL EPSPCPQRCV NTQGGFECHC YPNYDLVDGE CVEPVDPCFR ANCEYQCQPL NQTSYLCVCA EGFAPIPHEP HRCQMFCNQT ACPADCDPNT QASCECPEGY ILDDGFICTD IDECENGGFC SGVCHNLPGT FECICGPDSA LARHIGTDCD SGKVDGGDSG SGEPPPSPTP
GSTLTPPAVG LVHSHHHHHH
Thrombomodulin (THBD, also known as CD141, BDCA-3, and Fetomodulin) is an endothelial cell-expressed transmembrane glycoprotein that plays a critical role in the regulation of coagulation. The human THBD gene encodes a protein with a signal peptide (aa 1-18) and a mature chain (aa 19-575) consisting of multiple functional domains .
The primary functions of THBD include:
Formation of a 1:1 stoichiometric complex with thrombin
Activation of protein C in the anticoagulant pathway
Inhibition of fibrinolysis by facilitating thrombin-activatable fibrinolysis inhibitor (TAFI) activation
Involvement in inflammation and cellular processes beyond coagulation
Researchers investigating THBD should employ multiple methodological approaches to characterize its function, including protein-protein interaction assays, functional coagulation tests, and cellular models of endothelial function.
The THBD protein consists of several distinct functional domains that contribute to its diverse roles:
Signal peptide (aa 1-18)
C-type lectin domain (aa 31-169)
Six EGF-like domains (aa 241-481):
EGF1: aa 241-281
EGF2: aa 284-324
EGF3: aa 325-363
EGF4: aa 365-405
EGF5: aa 404-440
EGF6: aa 441-481
Transmembrane domain (aa 516-539)
For recombinant protein production, the extracellular portion (aa 19-515) is typically used, which contains the domains responsible for thrombin binding and protein C activation . When designing experiments targeting specific THBD functions, researchers should consider which domains are necessary for the activity of interest.
THBD is predominantly expressed by vascular endothelial cells, but its expression has been detected in other cell types including dendritic cells (specifically BDCA-3+ dendritic cells), monocytes, and certain tumor cells .
Regulation of THBD expression involves:
Transcriptional control through promoter elements responsive to inflammatory mediators
Downregulation by inflammatory cytokines (TNF-α, IL-1β)
Upregulation by thrombin, statins, and antioxidants
Post-translational shedding from the cell surface, resulting in soluble THBD in circulation
When analyzing THBD expression, researchers should consider:
Cell-specific expression patterns
Tissue-specific vascular bed differences
Environmental factors affecting expression (flow conditions, hypoxia)
Disease states that may alter expression patterns
The interaction between THBD and thrombin involves:
Initial binding between EGF-like domains 5-6 of THBD and the anion-binding exosite I of thrombin
Conformational changes in thrombin's active site
This interaction alters thrombin's substrate specificity from pro-coagulant to anticoagulant by:
Blocking thrombin's interaction with fibrinogen
Creating a binding site for protein C
Accelerating protein C activation by >1000-fold
To study this interaction experimentally, researchers can use:
Surface plasmon resonance for binding kinetics
Isothermal titration calorimetry for thermodynamic parameters
Functional assays measuring protein C activation rates
Structural analysis techniques (X-ray crystallography, cryo-EM)
Altered soluble THBD levels in circulation are associated with numerous conditions:
Pathological Condition | Change in Soluble THBD | Potential Mechanism |
---|---|---|
Smoking | Increased | Endothelial damage |
Cardiac surgery | Increased | Vascular injury |
Atherosclerosis | Increased | Chronic endothelial dysfunction |
Liver cirrhosis | Increased | Altered clearance |
Diabetes mellitus | Increased | Microvascular damage |
Cerebral infarction | Increased | Acute vascular injury |
Myocardial infarction | Increased | Cardiac endothelial damage |
Multiple sclerosis | Increased | Inflammatory activation |
When measuring soluble THBD levels for research purposes:
Standardize sample collection and processing
Account for confounding factors (renal function, age, medications)
Use appropriate reference ranges for specific populations
Consider longitudinal measurements in dynamic conditions
THBD (as CD141/BDCA-3) is expressed on a specialized subset of dendritic cells with unique functions:
CD141+/BDCA-3+ dendritic cells are efficient at cross-presentation of antigens to CD8+ T cells
These cells play important roles in anti-tumor immunity and viral responses
They can be identified by flow cytometry using specific antibodies against THBD/BDCA-3
For researchers studying THBD in dendritic cells:
Flow cytometry detection typically employs anti-THBD/BDCA-3 antibodies (such as clone #501733)
Co-staining with HLA-DR helps identify the specific dendritic cell population
Setting quadrant markers based on control antibody staining is essential for accurate identification
The study of THBD+ dendritic cells in cancer immunotherapy has shown promising results, with research demonstrating that human induced pluripotent stem cell-derived CD141+XCR1+ dendritic cells can effectively cross-present tumor antigens .
Multiple complementary approaches can be used to detect THBD expression:
Flow cytometry:
Optimal for cell surface detection on intact cells
Allows multi-parameter analysis with other markers
Example protocol: Human PBMCs can be stained with Mouse Anti-Human Thrombomodulin/BDCA-3 PE-conjugated Monoclonal Antibody (Clone #501733)
Co-staining with HLA-DR antibodies helps identify specific dendritic cell populations
Immunohistochemistry/Immunofluorescence:
Provides spatial context in tissue sections
Allows assessment of vascular THBD expression patterns
Requires optimization of fixation and antigen retrieval protocols
ELISA for soluble THBD:
Quantifies circulating THBD in plasma/serum
Reflects endothelial damage/activation
Reference ranges should be established for specific clinical contexts
qRT-PCR for THBD mRNA:
Assesses transcriptional regulation
Requires careful primer design and appropriate controls
Should be normalized to stable reference genes
Production of recombinant THBD typically involves:
Expression systems:
Purification strategy:
Formulation and storage:
Quality control should include functional assays to confirm the activity of the recombinant protein in thrombin binding and protein C activation.
When designing flow cytometry experiments for THBD/BDCA-3 detection:
Antibody selection:
Sample preparation:
Peripheral blood mononuclear cells (PBMCs) can be isolated using standard density gradient methods
Fresh samples are preferred as cryopreservation may affect surface expression
Staining protocol:
Analysis considerations:
Gating strategy should account for potential auto-fluorescence
Assess compensation when using multiple fluorophores
Quantify both percentage of positive cells and mean fluorescence intensity
An example protocol has been validated for detecting Thrombomodulin/BDCA-3 in human PBMCs, demonstrating successful identification of THBD+ cells using PE-conjugated antibodies in combination with HLA-DR staining .
THBD gene polymorphisms have been associated with various pathological conditions:
Types of polymorphisms:
Single nucleotide polymorphisms in coding regions affecting protein function
Promoter variants affecting expression levels
Rare variants linked to specific disorders
Disease associations:
Methodological approaches for studying polymorphisms:
PCR-RFLP analysis for known variants
Targeted sequencing of THBD gene
Case-control association studies
Functional characterization of variants using cellular models
Researchers should consider population-specific allele frequencies and potential gene-environment interactions when designing genetic association studies involving THBD.
Several experimental models can be employed to study THBD function:
Cell culture models:
Primary human endothelial cells (HUVECs, HAECs, HMVECs)
Cell lines expressing recombinant THBD
CRISPR/Cas9-edited cells with THBD mutations or knockout
In vitro coagulation assays:
Protein C activation assays using purified components
Thrombin generation tests with/without soluble THBD
Clot formation and lysis assays
Animal models:
THBD knockout mice (complete knockout is embryonic lethal)
Tissue-specific or inducible knockout systems
Humanized THBD mouse models
Models with mutant THBD mimicking human polymorphisms
Ex vivo systems:
Perfused human vessel segments
Microfluidic devices with endothelialized channels
Platelet adhesion assays under flow conditions
Each model has strengths and limitations that should be considered based on the specific research question being addressed.
Research into THBD-based therapeutics is advancing in several directions:
Recombinant soluble THBD:
Development as anticoagulant therapy
Potential applications in disseminated intravascular coagulation
Advantages over direct anticoagulants in being localized to sites of thrombin generation
THBD in cancer immunotherapy:
Targeting THBD in vascular diseases:
Modulation of THBD expression to prevent thrombosis
Protection against ischemia-reperfusion injury
Development of more stable THBD variants with enhanced activity
Diagnostic applications:
Soluble THBD as a biomarker for endothelial dysfunction
Prognostic indicator in various cardiovascular conditions
Monitoring therapeutic response
For researchers developing THBD-based therapeutics, considerations include pharmacokinetics, immunogenicity, target specificity, and potential off-target effects on hemostasis.
Variability in THBD expression and activity presents methodological challenges:
Sources of variability:
Genetic polymorphisms affecting expression or function
Environmental factors (inflammation, shear stress, hypoxia)
Post-translational modifications (glycosylation, proteolytic processing)
Assay-specific technical factors
Strategies to address variability:
Careful phenotyping and genotyping of study subjects
Standardization of experimental conditions
Use of internal controls and reference standards
Measurement of multiple parameters (expression, activity, soluble levels)
Statistical approaches to account for covariates
Normalization approaches:
Ratio methods comparing THBD to housekeeping genes/proteins
Adjustment for endothelial cell content in tissue samples
Paired experimental designs when possible
Longitudinal measurements to account for temporal variations
When conducting THBD functional assays, critical quality control measures include:
Protein quality assessment:
Assay validation parameters:
Linearity within the relevant concentration range
Precision (intra- and inter-assay variability)
Accuracy (recovery of known additions)
Specificity (absence of interference from related proteins)
Sensitivity (lower limit of detection and quantification)
Experimental controls:
Positive and negative controls for each assay run
Calibration curves with recombinant standards
Inclusion of reference samples across experimental batches
Testing of inhibitors to confirm specificity of measured activities
Technical considerations:
Calcium concentration (critical for THBD-thrombin-protein C interactions)
pH optimization
Temperature control during reactions
Appropriate handling of samples to preserve activity
Conflicting results in THBD research can arise from multiple sources and require careful evaluation:
Methodological differences:
Different antibody clones or detection systems
Variations in recombinant protein sources
Assay-specific biases in measurement
Cell or tissue sources (venous vs. arterial endothelium)
Biological complexities:
Context-dependent functions of THBD
Interactions with other coagulation and inflammatory pathways
Feedback mechanisms affecting THBD regulation
Compensatory mechanisms in genetic models
Approaches to reconciliation:
Direct comparison of methodologies in the same laboratory
Collaborative studies with standardized protocols
Meta-analyses of published data
Comprehensive models incorporating multiple experimental systems
Consideration of biological context and species differences
Reporting recommendations:
Detailed methodological documentation
Transparent reporting of negative results
Acknowledgment of limitations and potential confounders
Discussion of results in context of existing literature
By systematically addressing methodological differences and biological complexities, researchers can develop more integrated models of THBD function that account for apparently conflicting observations.
Thrombomodulin is composed of several domains, each contributing to its function:
When thrombin binds to thrombomodulin, its procoagulant activity is inhibited, and it promotes the activation of protein C. Activated protein C, in the presence of protein S, inactivates factors Va and VIIIa, thus inhibiting further thrombin generation and clot formation .
Recombinant human thrombomodulin (rhTM), also known as thrombomodulin alfa or ART-123, is a soluble form of thrombomodulin comprising all extracellular domains of the protein . It is developed for therapeutic use, particularly in conditions involving excessive coagulation and inflammation, such as disseminated intravascular coagulation (DIC) and sepsis .
rhTM has shown promise in various clinical settings:
rhTM enhances the activation of pro-carboxypeptidase B2 (pro-CPB2) by thrombin. Activated pro-CPB2 (CPB2) exerts anti-inflammatory and anti-fibrinolytic activities . By binding to thrombin, rhTM inhibits its procoagulant activity and promotes the activation of protein C, which in turn inactivates factor Va in the presence of protein S . This mechanism helps to attenuate the extension of clots while other anticoagulants inhibit the initiation of clot formation .