Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.999
Filter
1.
J Cell Mol Med ; 28(13): e18457, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38963011

ABSTRACT

Allogeneic haematopoietic stem cell transplantation (allo-HSCT) can potentially cure malignant blood disorders and benign conditions such as haemoglobinopathies and immunologic diseases. However, allo-HSCT is associated with significant complications. The most common and debilitating among them is graft-versus-host disease (GVHD). In GVHD, donor-derived T cells mount an alloimmune response against the recipient. The alloimmune response involves several steps, including recognition of recipient antigens, activation and proliferation of T cells in secondary lymphoid organs, and homing into GVHD-targeted organs. Adhesion molecules on T cells and endothelial cells mediate homing of T cells into lymphoid and non-lymphoid tissues. In this study, we showed that Von Willebrand factor (VWF), an adhesion molecule secreted by activated endothelial cells, plays an important role in mouse models of GVHD. We investigated the effect of the VWF-cleaving protease ADAMTS13 on GVHD. We found that ADAMTS13 reduced the severity of GVHD after bone marrow transplantation from C57BL6 donor to BALB/C recipient mice. A recombinant VWF-A2 domain peptide also reduced GVHD in mice. We showed that ADAMTS13 and recombinant VWF-A2 reduced the binding of T cells to endothelial cells and VWF in vitro, and reduced the number of T cells in lymph nodes, Peyer's patches and GVHD-targeted organs in vivo. We identified LFA-1 (αLß2) as the binding site of VWF on T cells. Our results showed that blocking T-cell homing by ADAMTS13 or VWF-A2 peptide reduced the severity of the GVHD after allo-HSCT, a potentially novel method for treating and preventing GVHD.


Subject(s)
ADAMTS13 Protein , Graft vs Host Disease , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes , von Willebrand Factor , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Animals , ADAMTS13 Protein/metabolism , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , von Willebrand Factor/metabolism , Humans , Transplantation, Homologous , Hematopoietic Stem Cell Transplantation/adverse effects , Disease Models, Animal , Bone Marrow Transplantation , Endothelial Cells/metabolism
2.
Zhonghua Gan Zang Bing Za Zhi ; 32(6): 484-488, 2024 Jun 20.
Article in Chinese | MEDLINE | ID: mdl-38964888

ABSTRACT

Portal vein thrombosis (PVT) is divided into cirrhotic and non-cirrhotic PVTs. The incidence rate of PVT varies greatly among different clinical stages of cirrhosis, with an overall incidence rate of about 13.92%, and the prevalence of cirrhotic PVT following splenectomy is as high as 60%. The pathogenesis of cirrhotic PVT is still unclear. However, the activation of Janus kinase/signal transduction and activator transcription signaling pathways, the rise in the expression of von Willebrand factor, and the gut microbiota along with its metabolite trimethylamine-N-oxide play an important role in the injury of vascular endothelial cells and the formation of PVT in cirrhosis. Therefore, these could be a new target for cirrhotic PVT prevention and treatment.


Subject(s)
Liver Cirrhosis , Portal Vein , Venous Thrombosis , Humans , Venous Thrombosis/etiology , Venous Thrombosis/prevention & control , Liver Cirrhosis/complications , Signal Transduction , Methylamines/metabolism , Gastrointestinal Microbiome , von Willebrand Factor/metabolism , Janus Kinases/metabolism
3.
BMC Pulm Med ; 24(1): 325, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965511

ABSTRACT

BACKGROUND: SARS-CoV-2 is a systemic disease that affects endothelial function and leads to coagulation disorders, increasing the risk of mortality. Blood levels of endothelial biomarkers such as Von Willebrand Factor (VWF), Thrombomodulin or Blood Dendritic Cell Antigen-3 (BDCA3), and uUokinase (uPA) increase in patients with severe disease and can be prognostic indicators for mortality. Therefore, the aim of this study was to determine the effect of VWF, BDCA3, and uPA levels on mortality. METHODS: From May 2020 to January 2021, we studied a prospective cohort of hospitalized adult patients with polymerase chain reaction (PCR)-confirmed COVID-19 with a SaO2 ≤ 93% and a PaO2/FiO2 ratio < 300. In-hospital survival was evaluated from admission to death or to a maximum of 60 days of follow-up with Kaplan-Meier survival curves and Cox proportional hazard models as independent predictor measures of endothelial dysfunction. RESULTS: We recruited a total of 165 subjects (73% men) with a median age of 57.3 ± 12.9 years. The most common comorbidities were obesity (39.7%), hypertension (35.4%) and diabetes (30.3%). Endothelial biomarkers were increased in non-survivors compared to survivors. According to the multivariate Cox proportional hazard model, those with an elevated VWF concentration ≥ 4870 pg/ml had a hazard ratio (HR) of 4.06 (95% CI: 1.32-12.5) compared to those with a lower VWF concentration adjusted for age, cerebrovascular events, enoxaparin dose, lactate dehydrogenase (LDH) level, and bilirubin level. uPA and BDCA3 also increased mortality in patients with levels ≥ 460 pg/ml and ≥ 3600 pg/ml, respectively. CONCLUSION: The risk of mortality in those with elevated levels of endothelial biomarkers was observable in this study.


Subject(s)
Biomarkers , COVID-19 , Thrombomodulin , Urokinase-Type Plasminogen Activator , von Willebrand Factor , Humans , COVID-19/mortality , COVID-19/blood , Male , von Willebrand Factor/metabolism , von Willebrand Factor/analysis , Middle Aged , Female , Biomarkers/blood , Aged , Urokinase-Type Plasminogen Activator/blood , Thrombomodulin/blood , Prospective Studies , Prognosis , SARS-CoV-2 , Adult , Endothelium, Vascular/physiopathology , Hospital Mortality , Proportional Hazards Models
4.
Blood Coagul Fibrinolysis ; 35(5): 256-264, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38973517

ABSTRACT

Normally, von Willebrand factor (VWF) remains inactive unless its A1A2 domains undergo a shear stress-triggered conformational change. We demonstrated the capacity of a recombinant A2 domain of VWF to bind and to affect fibrin formation, altering the fibrin clot structure. The data indicated that VWF contains an additional binding site for fibrin in the A2 domain that plays a role in the incorporation of VWF to the polymerizing fibrin. This study is to examine the hypothesis that active plasma VWF directly influence fibrin polymerization and the structure of fibrin clots. The study used healthy and type 3 von Willebrand disease (VWD) plasma, purified plasma VWF, fibrin polymerization assays, confocal microscopy and scanning electron microscopy. The exposed A2 domain in active VWF harbors additional binding sites for fibrinogen, and significantly potentiates fibrin formation (P < 0.02). Antibody against the A2 domain of VWF significantly decreased the initial rate of change of fibrin formation (P < 0.002). Clot analyses revealed a significant difference in porosity between normal and type 3 VWD plasma (P < 0.008), further supported by scanning electron microscopy, which demonstrated thicker fibrin fibers in the presence of plasma VWF (P < 0.0003). Confocal immunofluorescence microscopy showed punctate VWF staining along fibrin fibrils, providing visual evidence of the integration of plasma VWF into the fibrin matrix. The study with type 3 VWD plasma supports the hypothesis that plasma VWF directly influences fibrin polymerization and clot structure. In addition, a conformational change in the A1A2 domains exposes a hidden fibrin(ogen) binding site, indicating that plasma VWF determines the fibrin clot structure.


Subject(s)
Fibrin , von Willebrand Factor , von Willebrand Factor/metabolism , Humans , Fibrin/metabolism , Fibrin/ultrastructure , von Willebrand Disease, Type 3/blood , Binding Sites , Microscopy, Electron, Scanning/methods
5.
Mar Drugs ; 22(6)2024 Jun 16.
Article in English | MEDLINE | ID: mdl-38921594

ABSTRACT

Endothelial hyperpermeability is pivotal in sepsis-associated multi-organ dysfunction. Increased von Willebrand factor (vWF) plasma levels, stemming from activated platelets and endothelium injury during sepsis, can bind to integrin αvß3, exacerbating endothelial permeability. Hence, targeting this pathway presents a potential therapeutic avenue for sepsis. Recently, we identified isaridin E (ISE), a marine-derived fungal cyclohexadepsipeptide, as a promising antiplatelet and antithrombotic agent with a low bleeding risk. ISE's influence on septic mortality and sepsis-induced lung injury in a mouse model of sepsis, induced by caecal ligation and puncture, is investigated in this study. ISE dose-dependently improved survival rates, mitigating lung injury, thrombocytopenia, pulmonary endothelial permeability, and vascular inflammation in the mouse model. ISE markedly curtailed vWF release from activated platelets in septic mice by suppressing vesicle-associated membrane protein 8 and soluble N-ethylmaleide-sensitive factor attachment protein 23 overexpression. Moreover, ISE inhibited healthy human platelet adhesion to cultured lipopolysaccharide (LPS)-stimulated human umbilical vein endothelial cells (HUVECs), thereby significantly decreasing vWF secretion and endothelial hyperpermeability. Using cilengitide, a selective integrin αvß3 inhibitor, it was found that ISE can improve endothelial hyperpermeability by inhibiting vWF binding to αvß3. Activation of the integrin αvß3-FAK/Src pathway likely underlies vWF-induced endothelial dysfunction in sepsis. In conclusion, ISE protects against sepsis by inhibiting endothelial hyperpermeability and platelet-endothelium interactions.


Subject(s)
Blood Platelets , Human Umbilical Vein Endothelial Cells , Sepsis , von Willebrand Factor , Animals , Sepsis/drug therapy , von Willebrand Factor/metabolism , Humans , Mice , Human Umbilical Vein Endothelial Cells/drug effects , Male , Blood Platelets/drug effects , Blood Platelets/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Integrin alphaVbeta3/metabolism , Integrin alphaVbeta3/antagonists & inhibitors , Capillary Permeability/drug effects
6.
Sci Rep ; 14(1): 14524, 2024 06 24.
Article in English | MEDLINE | ID: mdl-38914598

ABSTRACT

NLRP3 inflammasome has been implicated in neutrophil polarization and extrusion of neutrophil extracellular traps (NETs) in vitro and facilitates secretion of Il1-beta (IL-1ß). Permanent ligation of the left anterior descending artery was used to induce MI in WT and NLRP3-/- mice as well as in NLRP3-/- recipient mice transfused with either WT or NLRP3-/- neutrophils. NLRP3 deficiency reduced infarct size to roughly a third of WT heart injury and preserved left ventricular (LV) function at 12 h after MI as assessed by echocardiography and triphenyltetrazolium chloride staining of live tissue. Transfusion of WT but not NLRP3-/- neutrophils after MI increased infarct size in NLRP3-/- mice and significantly reduced LV function. The key features of myocardial tissue in WT neutrophil transfused recipients were increased H3Cit-positive deposits with NET-like morphology and increased tissue levels of IL-1ß and plasma levels of von Willebrand Factor (VWF). Flow cytometry analysis also revealed that neutrophil NLRP3 increased the number of labeled and transfused neutrophils in the bone marrow of recipient mice following MI. Our data suggest a key role for neutrophil NLRP3 in the production of IL-1ß and deposition of NETs in cardiac tissue exacerbating injury following MI. We provide evidence for a link between neutrophil NLRP3 and VWF release likely enhancing thromboinflammation in the heart. Neutrophil NLRP3 deficiency conferred similar cardioprotective effects to general NLRP3 deletion in MI rendering anti-neutrophil NLRP3 therapy a promising target for early cardioprotective treatment.


Subject(s)
Extracellular Traps , Interleukin-1beta , Mice, Knockout , Myocardial Infarction , Myocardium , NLR Family, Pyrin Domain-Containing 3 Protein , Neutrophils , von Willebrand Factor , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Neutrophils/metabolism , Interleukin-1beta/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Mice , Extracellular Traps/metabolism , Myocardium/metabolism , Myocardium/pathology , von Willebrand Factor/metabolism , von Willebrand Factor/genetics , Mice, Inbred C57BL , Male , Inflammasomes/metabolism , Disease Models, Animal
7.
Expert Rev Hematol ; 17(7): 391-403, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38889268

ABSTRACT

BACKGROUND: Platelet storage is complicated by deleterious changes, among which reduction of ristocetin-induced platelet aggregation (RIPA) has a poorly understood mechanism. The study elucidates the mechanistic roles of all the possible players in this process. RESEARCH DESIGN AND METHODS: PRP-platelet concentrates were subjected to RIPA, collagen-induced platelet aggregation (CIPA), and flowcytometric analysis of GPIbα and PAC-1 binding from days 0 to 5 of storage. Platelet-poor plasma was subjected to colorimetric assays for glucose/LDH evaluation and automatic analyzer to examine VWF antigen and activity. RESULTS: From day three of platelet storage, reducing CIPA but not RIPA was correlated with the reduction of both metabolic state and integrin activity. RIPA reduction was directly related to the decreased levels of total-content/expression of GPIbα, and inversely related to its shedding levels during storage. Re-suspension of 5-day stored platelet in fresh plasma compensated CIPA, but not RIPA. VWF concentration and its activity did not change during storage while they had no correlation with RIPA. CONCLUSIONS: This study identified the irreversible loss of platelet GPIbα, but not VWF status, as the primary cause of the storage-dependent decrease of RIPA. Unlike CIPA, this observation was not compensated by plasma refreshment, suggesting that some evidence of PSL may not be recovered after transfusion.


Subject(s)
Blood Platelets , Blood Preservation , Platelet Aggregation , Platelet Glycoprotein GPIb-IX Complex , Ristocetin , von Willebrand Factor , Humans , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Aggregation/drug effects , Ristocetin/pharmacology , Blood Platelets/metabolism , Blood Preservation/methods , von Willebrand Factor/metabolism , Hemostasis/drug effects
8.
Thromb Res ; 240: 109038, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38850807

ABSTRACT

BACKGROUND: Shiga toxin (Stx) can activate inflammatory signaling, leading to vascular dysfunction and promotion of a pro-thrombotic tissue microenvironment. Stx can trigger the development of the enterohemorrhagic (childhood) hemolytic uremic syndrome (eHUS), a triad of thrombocytopenia, hemolytic anemia, and acute kidney injury, often requiring dialysis. Additional features may include damage to other organs, including the gastrointestinal tract, pancreas, brain and cardiovascular system; death occurs in 2-5 %. eHUS is a thrombotic microangiopathy; thus, endothelial cell (EC) injury and platelet fibrin thrombus formation in glomerular arterioles and in the arterioles of other affected organs are likely. To elucidate mechanisms of this microangiopathy, we examined in human ECs the regulation of the platelet adhesion proteins P-selectin and von Willebrand factor (VWF), along with the downregulation of erythroblast-transformation-specific transcription factor (ERG) a key regulator of angiogenesis and megakaryocyte development. METHODS: VWF, P-selectin, and ERG levels were determined using immunofluorescence and Western blot in human umbilical endothelial cells (HUVECs). HUVECs were treated with tumor necrosis factor-alpha (TNF-α), Stx-1 or both, versus normal controls. Capillary morphogenesis on Matrigel was performed using HUVECs treated, for 22 h with TNF-α, Stx-1, or both, or treated 4 h with Stx-1 alone or in combination with TNF-α for 22 h. RESULTS: Stx-1 significantly reduced ERG and VWF expression on HUVECs, but upregulated P-selectin expression. ERG levels decreased with Stx-1 alone or in combination with TNF-α, in the nuclear, perinuclear and cytoplasmatic regions. Stx-1 reduced capillary morphogenesis, while Stx-1-TNF-α combined treatment reduced capillary morphogenesis still further. CONCLUSIONS: In the presence of Stx-1 or TNF-α or both treatments, ECs were activated, expressing higher levels of P-selectin and lower levels of VWF. Our findings, further, provide evidence that Stx-1 downregulates ERG, repressing angiogenesis in vitro.


Subject(s)
Down-Regulation , Human Umbilical Vein Endothelial Cells , Humans , Down-Regulation/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Transcriptional Regulator ERG/metabolism , Shiga Toxin/metabolism , Shiga Toxin/pharmacology , Endothelial Cells/metabolism , Endothelial Cells/drug effects , von Willebrand Factor/metabolism , Angiogenesis
9.
Thromb Res ; 240: 109061, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38870771

ABSTRACT

BACKGROUND: Von Willebrand Disease (VWD) is the most common inherited bleeding disorder. VWD is characterized by an abnormal quantity or quality of von Willebrand Factor (VWF). Anemia is often found at presentation for a bleeding disorder evaluation due to chronic blood loss. OBJECTIVES/HYPOTHESIS: We hypothesized that anemia is associated with elevations in both VWF and factor VIII (FVIII) over baseline. We also hypothesized that obesity would be associated with increased levels of VWF. METHODS: We conducted a single-center review of the electronic health record for patients that had proximal von Willebrand profiles and Hb data. RESULTS: We identified 4552 unique subjects with VWF studies and a CBC within 24 h. We found that decreasing hemoglobin inversely correlated with VWF antigen, VWF ristocetin cofactor activity, and FVIII activity. We also found that obesity and Black race were independently associated with increased VWF antigen, activity, and FVIII activity. Hb, race, and body mass index (BMI) continued to be determinants of VWF and FVIII levels in multivariable analysis. CONCLUSION: Our study demonstrates that anemia, race, and BMI were found to be associated with elevation of VWF antigen, VWF activity, and FVIII levels. As many individuals with anemia present for evaluation for a bleeding disorder, these variables need to be considered. KEY POINTS: - Anemia was found to be associated with elevation of VWF antigen, VWF activity and FVIII levels. - Testing von Willebrand factor at times of anemia may mask a diagnosis of von Willebrand Disease.


Subject(s)
Body Mass Index , Factor VIII , Hemoglobins , von Willebrand Factor , Humans , von Willebrand Factor/analysis , von Willebrand Factor/metabolism , Factor VIII/analysis , Factor VIII/metabolism , Male , Female , Middle Aged , Adult , Hemoglobins/analysis , von Willebrand Diseases/blood , Anemia/blood , Aged , Obesity/blood , Obesity/complications
10.
Front Endocrinol (Lausanne) ; 15: 1397062, 2024.
Article in English | MEDLINE | ID: mdl-38836224

ABSTRACT

Background and aims: A prothrombotic state was demonstrated in patients with Cushing's syndrome and is involved in the development and progression of cardiovascular and renal damage in hypertensive patients. This study was designed to examine the relationships between cortisol secretion and the hemostatic and fibrinolytic systems in hypertension. Methods: In 149 middle-aged, nondiabetic, essential hypertensive patients free of cardiovascular and renal complications, we measured hemostatic markers that express the spontaneous activation of the coagulation and fibrinolytic systems and assessed daily cortisol levels (8 AM, 3 PM, 12 AM; area under the curve, AUC-cortisol) together with the cortisol response to dexamethasone overnight suppression (DST-cortisol). Results: Plasma levels of D-dimer (D-dim), prothrombin fragment 1 + 2 (F1 + 2), and von Willebrand factor (vWF) were progressively and significantly higher across tertiles of AUC-cortisol and DST-cortisol, whereas no differences were observed in fibrinogen, tissue plasminogen activator, plasminogen activator inhibitor-1, antithrombin III, protein C, and protein S. D-dim, F1 + 2, and vWF were significantly and directly correlated with age and both AUC-cortisol and DST-cortisol. Multivariate regression analysis showed that both AUC-cortisol and DST-cortisol were related to plasma D-dim, F1 + 2, and vWF independently of age, body mass index, blood pressure, and renal function. Conclusion: Greater daily cortisol profile and cortisol response to overnight suppression are independently associated with a prothrombotic state in hypertensive patients and might contribute to the development of organ damage and higher risk of cardiovascular complications.


Subject(s)
Dexamethasone , Hydrocortisone , Hypertension , Humans , Male , Middle Aged , Female , Hydrocortisone/blood , Hypertension/blood , Hypertension/complications , Adult , Thrombosis/blood , Thrombosis/etiology , von Willebrand Factor/metabolism , von Willebrand Factor/analysis , Circadian Rhythm/physiology , Aged , Biomarkers/blood
11.
Immun Inflamm Dis ; 12(6): e1267, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38888384

ABSTRACT

OBJECTIVES: Acute pancreatitis (AP) is an inflammatory disease of the pancreas, and the prognosis of severe AP (SAP) is poor. The study aimed to identify promising biomarkers for predicting the occurrence and survival outcome of SAP patients. MATERIALS AND METHODS: Two hundred and forty AP patients were retrospectively recruited, in which 72 cases with SAP. Blood test was done for collection of laboratory indicators. After treatment, the mortality of patients was recorded. RESULTS: Patients in the SAP group had higher intensive care unit admissions and longer hospital stays (p < .001). Among laboratory parameters, significantly high values of C-reactive protein (CRP), triglycerides and glucose (TyG) index, Von willebrand factor antigen (vWF:Ag) and D-dimer were found in SAP groups relative to non-SAP ones. Receiver operating characteristic curve indicated the good performance of CRP, TyG index, vWF:Ag and D-dimer in SAP diagnosis. Among all SAP cases, 51 survived while 21 died. TyG index (odds ratio [OR] = 6.914, 95% confidence interval [CI] = 1.193-40.068, p = .028), vWF:Ag (OR = 7.441, 95% CI = 1.236-244.815, p = .028), and D-dimer (OR = 7.987, 95% CI = 1.251-50.997, p = .028) were significantly related to survival outcome of SAP patients by multiple logistic regression analysis. Both TyG index and vWF showed favorable efficiency in predicting overall prognosis. The area under the curve for the multivariate model (PRE = -35.908 + 2.764 × TyG + 0.021 × vWF:Ag) was 0.909 which was greater than 0.9, indicating its excellent performance in prognosis prediction. CONCLUSION: CRP, TyG index, vWF:Ag, and D-dimer values on admission may be potential clinical predictors of the development of SAP. Moreover, TyG index and vWF:Ag may be helpful to predict survival outcome.


Subject(s)
Biomarkers , C-Reactive Protein , Fibrin Fibrinogen Degradation Products , Pancreatitis , Humans , Male , Female , Middle Aged , Pancreatitis/mortality , Pancreatitis/blood , Pancreatitis/diagnosis , Retrospective Studies , Prognosis , Adult , Fibrin Fibrinogen Degradation Products/analysis , Fibrin Fibrinogen Degradation Products/metabolism , C-Reactive Protein/analysis , C-Reactive Protein/metabolism , Biomarkers/blood , Aged , von Willebrand Factor/metabolism , von Willebrand Factor/analysis , ROC Curve , Acute Disease , Triglycerides/blood , Blood Glucose/metabolism , Blood Glucose/analysis , Severity of Illness Index
12.
J Thromb Haemost ; 22(7): 2052-2058, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38704122

ABSTRACT

BACKGROUND: The activity of von Willebrand factor (VWF) in facilitating platelet adhesion and aggregation correlates with its multimer size. Traditional ristocetin-dependent functional assays lack sensitivity to multimer sizes. Recently, nanobodies targeting the autoinhibitory module and activating VWF were identified. OBJECTIVES: To develop an assay that can differentiate the platelet-binding activity of VWF multimers. METHODS: A novel enzyme-linked immunosorbent assay (nanobody-triggered glycoprotein Ib binding assay [VWF:GPIbNab]) utilizing a VWF-activating nanobody was developed. Recombinant VWF, plasma-derived VWF (pdVWF), and selected gel-filtrated fractions of pdVWF were evaluated for VWF antigen and activity levels. A linear regression model was developed to estimate the specific activity of VWF multimers. RESULTS: Of the 3 activating nanobodies tested, 6C11 with the lowest activation effect exhibited the highest sensitivity for high-molecular-weight multimers (HMWMs) of VWF. VWF:GPIbNab utilizing 6C11 (VWF:GPIbNab6C11) produced significantly higher activity/antigen ratios for recombinant VWF (>2.0) and HMWM-enriched pdVWF fractions (>2.0) than for pdVWF (∼1.0) or fractions enriched with shorter multimers (<1.0). The differences were much larger than those produced by VWF:GPIbNab utilizing other nanobodies, VWF:GPIbM, VWF:GPIbR, or VWF:CB assays. Linear regression analysis of 5 pdVWF fractions of various multimer sizes produced an estimated specific activity of 2.7 for HMWMs. The analysis attributed >90% of the VWF activity measured by VWF:GPIbNab6C11 to that of HMWMs, which is significantly higher than all other activity assays tested. CONCLUSION: The VWF:GPIbNab6C11 assay exhibits higher sensitivity to HMWMs than ristocetin-based and collagen-binding assays. Future studies examining the application of this assay in clinical settings and any associated therapeutic benefit of doing so are warranted.


Subject(s)
Enzyme-Linked Immunosorbent Assay , Protein Multimerization , Single-Domain Antibodies , von Willebrand Factor , von Willebrand Factor/metabolism , von Willebrand Factor/analysis , Humans , Single-Domain Antibodies/immunology , Single-Domain Antibodies/chemistry , Linear Models , Recombinant Proteins , Blood Platelets/metabolism , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Protein Binding , Platelet Adhesiveness , Molecular Weight
13.
Blood Adv ; 8(13): 3388-3401, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38701351

ABSTRACT

ABSTRACT: Glycoprotein Ibα (GPIbα), the ligand-binding subunit of platelet GPIb-IX complex, interacts with von Willebrand factor (VWF) exposed at the injured vessel wall, initiating platelet adhesion, activation, hemostasis, and thrombus formation. The cytoplasmic tail of GPIbα interacts with 14-3-3ζ, regulating the VWF-GPIbα-elicited signal transduction and VWF binding function of GPIbα. However, we unexpectedly found that the GPIbα-14-3-3ζ association, beyond VWF-dependent function, is essential for general platelet activation. We found that the myristoylated peptide of GPIbα C-terminus MPαC, a potential GPIbα inhibitor, by itself induced platelet aggregation, integrin αIIbß3 activation, granule secretion, and phosphatidylserine (PS) exposure. Conversely, the deletion of the cytoplasmic tail of GPIbα in mouse platelets (10aa-/-) decreased platelet aggregation, integrin αIIbß3 activation, granule secretion, and PS exposure induced by various physiological agonists. Phosphoproteome-based kinase activity profiling revealed significantly upregulated protein kinase C (PKC) activity in MPαC-treated platelets. MPαC-induced platelet activation was abolished by the pan-PKC inhibitor and PKCα deletion. Decreased PKC activity was observed in both resting and agonist-stimulated 10aa-/- platelets. GPIbα regulates PKCα activity by sequestering 14-3-3ζ from PKCα. In vivo, the deletion of the GPIbα cytoplasmic tail impaired mouse hemostasis and thrombus formation and protected against platelet-dependent pulmonary thromboembolism. Therefore, our findings demonstrate an essential role for the GPIbα cytoplasmic tail in regulating platelet general activation and thrombus formation beyond the VWF-GPIbα axis.


Subject(s)
Blood Platelets , Platelet Activation , Platelet Glycoprotein GPIb-IX Complex , Platelet Glycoprotein GPIb-IX Complex/metabolism , Animals , Mice , Humans , Blood Platelets/metabolism , 14-3-3 Proteins/metabolism , von Willebrand Factor/metabolism , Thrombosis/metabolism , Signal Transduction , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Mice, Knockout , Platelet Aggregation
14.
J Transl Med ; 22(1): 412, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693516

ABSTRACT

BACKGROUND: Thromboinflammation involving platelet adhesion to endothelial surface-associated von Willebrand factor (VWF) has been implicated in the accelerated progression of non-culprit plaques after MI. The aim of this study was to use arterial endothelial molecular imaging to mechanistically evaluate endothelial-associated VWF as a therapeutic target for reducing remote plaque activation after myocardial infarction (MI). METHODS: Hyperlipidemic mice deficient for the low-density lipoprotein receptor and Apobec-1 underwent closed-chest MI and were treated chronically with either: (i) recombinant ADAMTS13 which is responsible for proteolytic removal of VWF from the endothelial surface, (ii) N-acetylcysteine (NAC) which removes VWF by disulfide bond reduction, (iii) function-blocking anti-factor XI (FXI) antibody, or (iv) no therapy. Non-ischemic controls were also studied. At day 3 and 21, ultrasound molecular imaging was performed with probes targeted to endothelial-associated VWF A1-domain, platelet GPIbα, P-selectin and vascular cell adhesion molecule-1 (VCAM-1) at lesion-prone sites of the aorta. Histology was performed at day 21. RESULTS: Aortic signal for P-selectin, VCAM-1, VWF, and platelet-GPIbα were all increased several-fold (p < 0.01) in post-MI mice versus sham-treated animals at day 3 and 21. Treatment with NAC and ADAMTS13 significantly attenuated the post-MI increase for all four molecular targets by > 50% (p < 0.05 vs. non-treated at day 3 and 21). On aortic root histology, mice undergoing MI versus controls had 2-4 fold greater plaque size and macrophage content (p < 0.05), approximately 20-fold greater platelet adhesion (p < 0.05), and increased staining for markers of platelet transforming growth factor-ß1 signaling. Accelerated plaque growth and inflammatory activation was almost entirely prevented by ADAMTS13 and NAC. Inhibition of FXI had no significant effect on molecular imaging signal or plaque morphology. CONCLUSIONS: Plaque inflammatory activation in remote arteries after MI is strongly influenced by VWF-mediated platelet adhesion to the endothelium. These findings support investigation into new secondary preventive therapies for reducing non-culprit artery events after MI.


Subject(s)
ADAMTS13 Protein , Myocardial Infarction , von Willebrand Factor , Animals , von Willebrand Factor/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/complications , ADAMTS13 Protein/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Mice , Plaque, Atherosclerotic/pathology , P-Selectin/metabolism , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Male , Molecular Imaging , Aorta/pathology , Aorta/drug effects , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Mice, Inbred C57BL
15.
J Chem Inf Model ; 64(10): 4158-4167, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38751042

ABSTRACT

The cyclic peptide OS1 (amino acid sequence: CTERMALHNLC), which has a disulfide bond between both termini cysteine residues, inhibits complex formation between the platelet glycoprotein Ibα (GPIbα) and the von Willebrand factor (vWF) by forming a complex with GPIbα. To study the binding mechanism between GPIbα and OS1 and, therefore, the inhibition mechanism of the protein-protein GPIbα-vWF complex, we have applied our multicanonical molecular dynamics (McMD)-based dynamic docking protocol starting from the unbound state of the peptide. Our simulations have reproduced the experimental complex structure, although the top-ranking structure was an intermediary one, where the peptide was bound in the same location as in the experimental structure; however, the ß-switch of GPIbα attained a different conformation. Our analysis showed that subsequent refolding of the ß-switch results in a more stable binding configuration, although the transition to the native configuration appears to take some time, during which OS1 could dissociate. Our results show that conformational changes in the ß-switch are crucial for successful binding of OS1. Furthermore, we identified several allosteric binding sites of GPIbα that might also interfere with vWF binding, and optimization of the peptide to target these allosteric sites might lead to a more effective inhibitor, as these are not dependent on the ß-switch conformation.


Subject(s)
Molecular Docking Simulation , Molecular Dynamics Simulation , Peptides, Cyclic , Platelet Glycoprotein GPIb-IX Complex , Protein Binding , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Peptides, Cyclic/metabolism , Platelet Glycoprotein GPIb-IX Complex/chemistry , Platelet Glycoprotein GPIb-IX Complex/metabolism , Protein Conformation , von Willebrand Factor/chemistry , von Willebrand Factor/metabolism , Humans , Binding Sites
16.
Ann Clin Lab Sci ; 54(2): 246-250, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38802159

ABSTRACT

Von Willebrand disease (VWD) is the most common inherited bleeding disorder. It is primarily attributed to malfunctioning or deficient Von Willebrand factor (VWF). Thromboelastography (TEG) has emerged as a valuable tool for assessing coagulation dynamics and guiding transfusion therapy in bleeding patients. Given this, we present a case study of a 23-year-old pregnant female with a past medical history of type 2B VWD, wherein TEG was employed to optimize disease screening and therapy monitoring while minimizing costs and preventing complications associated with low platelet counts. This case underscores the potential utility of TEG in enhancing the care of VWD patients, particularly in unique critical settings such as pregnancy.


Subject(s)
Thrombelastography , von Willebrand Diseases , Humans , Thrombelastography/methods , Female , von Willebrand Diseases/diagnosis , von Willebrand Diseases/blood , Pregnancy , Young Adult , von Willebrand Factor/metabolism , von Willebrand Factor/analysis , Adult
17.
Expert Rev Hematol ; 17(7): 341-351, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38752747

ABSTRACT

INTRODUCTION: Hereditary thrombotic thrombocytopenic purpura (hTTP) is caused by deficiency of plasma ADAMTS13 activity, resulting from ADAMTS13 mutations. ADAMTS13 cleaves ultra large von Willebrand factor (VWF), thus reducing its multimer sizes. Hereditary deficiency of plasma ADAMTS13 activity leads to the formation of excessive platelet-VWF aggregates in small arterioles and capillaries, resulting in hTTP. AREAS COVERED: PubMed search from 1956 to 2024 using thrombotic thrombocytopenic purpura and therapy identified 3,675 articles. Only the articles relevant to the topic were selected for discussion, which focuses on pathophysiology, clinical presentations, and mechanisms of action of emerging therapeutics for hTTP. Current therapies include infusion of plasma, or coagulation factor VIII, or recombinant ADAMTS13. Emerging therapies include anti-VWF A1 aptamers or nanobody and gene therapies with adeno-associated viral vector or self-inactivated lentiviral vector or a sleeping beauty transposon system for a long-term expression of a functional ADAMTS13 enzyme. EXPERT OPINION: Frequent plasma infusion remains to be the standard of care in most parts of the world, while recombinant ADAMTS13 has become the treatment of choice for hTTP in some of the Western countries. The success of gene therapies in preclinical models may hold a promise for future development of these novel approaches for a cure of hTTP.


Subject(s)
ADAMTS13 Protein , Genetic Therapy , Purpura, Thrombotic Thrombocytopenic , Humans , Purpura, Thrombotic Thrombocytopenic/therapy , Purpura, Thrombotic Thrombocytopenic/genetics , ADAMTS13 Protein/genetics , ADAMTS13 Protein/metabolism , ADAMTS13 Protein/deficiency , von Willebrand Factor/metabolism , von Willebrand Factor/genetics , Single-Domain Antibodies/therapeutic use , Disease Management , Factor VIII/genetics , Factor VIII/therapeutic use , Factor VIII/metabolism , Mutation , Animals
18.
J Thromb Haemost ; 22(7): 2018-2026, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38670315

ABSTRACT

BACKGROUND: Free labile hemin acts as a damage-associated molecular pattern during acute and chronic hemolysis and muscle injury, supporting platelet activation and thrombosis. OBJECTIVES: To investigate the anti-thrombotic potential of hydroxychloroquine on hemolysis-induced platelet activation and arterial thrombosis. METHODS: The effect of hydroxychloroquine on hemin-induced platelet activation and hemolysis-induced platelet recruitment and aggregation was measured in washed platelets and hemolyzed blood, respectively. Its effect on ferric-chloride (FeCl3)-induced arterial thrombosis and lung perfusion following hemin injection was assessed in wild-type mice. RESULTS: Erythrocyte lysis and endothelial cell activation cooperatively supported platelet aggregation and thrombosis at arterial shear stress. This thrombotic effect was reversed by hydroxychloroquine. In a purified system, hydroxychloroquine inhibited platelet build-up on immobilized von Willebrand factor in hemolyzed blood without altering initial platelet recruitment. Hydroxychloroquine inhibited hemin-induced platelet activation and phosphatidylserine exposure independently of reactive oxygen species generation. In the presence of hemin, hydroxychloroquine did not alter glycoprotein VI shedding but reduced C-type-lectin-like-2 expression on platelets. In vivo, hydroxychloroquine reversed pulmonary perfusion decline induced by exogenous administration of hemin. In arterial thrombosis models, hydroxychloroquine inhibited ferric-chloride-induced thrombosis in the carotid artery and reduced von Willebrand factor accumulation in the thrombi. CONCLUSION: Hydroxychloroquine inhibited hemolysis-induced arterial thrombosis ex vivo and improved pulmonary perfusion in hemin-treated mice, supporting a potential benefit of its use as an adjuvant therapy in hemolytic diseases to limit arterial thrombosis and to improve organ perfusion.


Subject(s)
Hemin , Hemolysis , Hydroxychloroquine , Lung , Platelet Activation , Thrombosis , Animals , Hydroxychloroquine/pharmacology , Hemolysis/drug effects , Hemin/pharmacology , Thrombosis/drug therapy , Thrombosis/blood , Lung/drug effects , Lung/blood supply , Platelet Activation/drug effects , Mice , Mice, Inbred C57BL , Platelet Aggregation/drug effects , Ferric Compounds , Humans , Male , Chlorides , Disease Models, Animal , Blood Platelets/drug effects , Blood Platelets/metabolism , Erythrocytes/drug effects , Erythrocytes/metabolism , von Willebrand Factor/metabolism
19.
J Thromb Haemost ; 22(7): 1844-1846, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38679336

ABSTRACT

A remarkable step forward in the treatment of hemophilia A has recently been achieved with the development of an Ultra-Long modified factor (F)VIII. Leveraging expertise gained with fusion to immunoglobulin Fc fragments, disconnecting FVIII from endogenous von Willebrand factor (via a D'-D3 fragment), and benefiting from the pharmacokinetic prolongation provided by the addition of hydrophilic polypeptides, efanesoctocog alfa opens a new era in the treatment of hemophilia A. The term Ultra-Long FVIII has been proposed to designate it and differentiate it from extended half-life FVIII. The level of FVIII correction within the normal range for several days provided by this molecule should allow an increasing number of patients to free themselves from the physical and psychological constraints of hemophilia A. Certainly, the burden of weekly intravenous infusions persists but is compensated by a correction of hemostasis whose amplitude and duration remain unmatched by other therapeutic options currently available.


Subject(s)
Factor VIII , Hemophilia A , Humans , Hemophilia A/blood , Hemophilia A/drug therapy , Factor VIII/pharmacokinetics , Factor VIII/administration & dosage , Factor VIII/therapeutic use , von Willebrand Factor/metabolism , Half-Life , Coagulants/pharmacokinetics , Coagulants/therapeutic use , Coagulants/administration & dosage , Immunoglobulin Fc Fragments , Treatment Outcome , Hemostasis/drug effects , Infusions, Intravenous , Blood Coagulation/drug effects , Animals , Recombinant Fusion Proteins/therapeutic use , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/administration & dosage
20.
J Thromb Haemost ; 22(7): 1894-1908, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38679335

ABSTRACT

BACKGROUND: von Willebrand disease (VWD) is the most common inherited bleeding disorder, characterized by either partial or complete von Willebrand factor (VWF) deficiency or by the occurrence of VWF proteoforms of altered functionality. The gene encoding VWF is highly polymorphic, giving rise to a variety of proteoforms with varying plasma concentrations and clinical significance. OBJECTIVES: To address this complexity, we translated genomic variation in VWF to corresponding VWF proteoforms circulating in blood. METHODS: VWF was characterized in VWD patients (n = 64) participating in the Willebrand in the Netherlands study by conventional laboratory testing, DNA sequencing and complementary discovery, and targeted mass spectrometry-based plasma proteomic strategies. RESULTS: Unbiased plasma profiling combined with immune enrichment of VWF verified VWF and its binding partner factor VIII as key determinants of VWD and revealed a remarkable heterogeneity in VWF amino acid sequence coverage among patients. Subsequent VWF proteotyping enabled identification of both polymorphisms (eg, p.Thr789Ala, p.Gln852Arg, and p.Thr1381Ala), as well as pathogenic variants (n = 16) along with their corresponding canonical sequences. Targeted proteomics using stable isotope-labeled peptides confirmed unbiased proteotyping for 5 selected variants and suggested differential proteoform quantities in plasma. The variant-to-wild-type peptide ratio was determined in 6 type 2B patients heterozygous for p.Arg1306Trp, confirming the relatively low proteoform concentration of the pathogenic variant. The elevated VWF propeptide/VWF ratio indicated increased clearance of specific VWF proteoforms. CONCLUSION: This study highlights how VWF proteotyping from plasma could be the first step to bridge the gap between genotyping and functional testing in VWD.


Subject(s)
Proteomics , von Willebrand Diseases , von Willebrand Factor , Humans , von Willebrand Factor/genetics , von Willebrand Factor/analysis , von Willebrand Factor/metabolism , von Willebrand Diseases/diagnosis , von Willebrand Diseases/blood , von Willebrand Diseases/genetics , Proteomics/methods , Netherlands , Phenotype , Female , Factor VIII/genetics , Factor VIII/analysis , Factor VIII/metabolism , Mass Spectrometry , Male , Predictive Value of Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...