Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cardiovasc Res ; 120(4): 417-432, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-37976180

ABSTRACT

AIMS: Abdominal aortic aneurysm (AAA) is a highly lethal disease with progressive dilatation of the abdominal aorta accompanied by degradation and remodelling of the vessel wall due to chronic inflammation. Platelets play an important role in cardiovascular diseases, but their role in AAA is poorly understood. METHODS AND RESULTS: The present study revealed that platelets play a crucial role in promoting AAA through modulation of inflammation and degradation of the extracellular matrix (ECM). They are responsible for the up-regulation of SPP1 (osteopontin, OPN) gene expression in macrophages and aortic tissue, which triggers inflammation and remodelling and also platelet adhesion and migration into the abdominal aortic wall and the intraluminal thrombus (ILT). Further, enhanced platelet activation and pro-coagulant activity result in elevated gene expression of various cytokines, Mmp9 and Col1a1 in macrophages and Il-6 and Mmp9 in fibroblasts. Enhanced platelet activation and pro-coagulant activity were also detected in AAA patients. Further, we detected platelets and OPN in the vessel wall and in the ILT of patients who underwent open repair of AAA. Platelet depletion in experimental murine AAA reduced inflammation and ECM remodelling, with reduced elastin fragmentation and aortic diameter expansion. Of note, OPN co-localized with platelets, suggesting a potential role of OPN for the recruitment of platelets into the ILT and the aortic wall. CONCLUSION: In conclusion, our data strongly support the potential relevance of anti-platelet therapy to reduce AAA progression and rupture in AAA patients.


Subject(s)
Aortic Aneurysm, Abdominal , Matrix Metalloproteinase 9 , Humans , Animals , Mice , Matrix Metalloproteinase 9/metabolism , Osteopontin/genetics , Osteopontin/metabolism , Aortic Aneurysm, Abdominal/metabolism , Aorta, Abdominal/metabolism , Inflammation/metabolism , Macrophages/metabolism , Fibroblasts/metabolism
2.
Diabetologia ; 64(8): 1834-1849, 2021 08.
Article in English | MEDLINE | ID: mdl-34131781

ABSTRACT

AIMS/HYPOTHESIS: People with diabetes have an increased cardiovascular risk with an accelerated development of atherosclerosis and an elevated mortality rate after myocardial infarction. Therefore, cardioprotective effects of glucose-lowering therapies are of major importance for the pharmacotherapy of individuals with type 2 diabetes. For sodium-glucose cotransporter 2 inhibitors (SGLT2is), in addition to a reduction in blood glucose, beneficial effects on atherosclerosis, obesity, renal function and blood pressure have been observed. Recent results showed a reduced risk of worsening heart failure and cardiovascular deaths under dapagliflozin treatment irrespective of the diabetic state. However, the underlying mechanisms are yet unknown. Platelets are known drivers of atherosclerosis and atherothrombosis and disturbed platelet activation has also been suggested to occur in type 2 diabetes. Therefore, the present study investigates the impact of the SGLT2i dapagliflozin on the interplay between platelets and inflammation in atherogenesis. METHODS: Male, 8-week-old LDL-receptor-deficient (Ldlr-/-) mice received a high-fat, high-sucrose diabetogenic diet supplemented without (control) or with dapagliflozin (5 mg/kg body weight per day) for two time periods: 8 and 25 weeks. In a first translational approach, eight healthy volunteers received 10 mg dapagliflozin/day for 4 weeks. RESULTS: Dapagliflozin treatment ameliorated atherosclerotic lesion development, reduced circulating platelet-leucocyte aggregates (glycoprotein [GP]Ib+CD45+: 29.40 ± 5.94 vs 17.00 ± 5.69 cells, p < 0.01; GPIb+lymphocyte antigen 6 complex, locus G+ (Ly6G): 8.00 ± 2.45 vs 4.33 ± 1.75 cells, p < 0.05) and decreased aortic macrophage infiltration (1.31 ± 0.62 vs 0.70 ± 0.58 ×103 cells/aorta, p < 0.01). Deeper analysis revealed that dapagliflozin decreased activated CD62P-positive platelets in Ldlr-/- mice fed a diabetogenic diet (3.78 ± 1.20% vs 2.83 ± 1.06%, p < 0.01) without affecting bleeding time (85.29 ± 37.27 vs 89.25 ± 16.26 s, p = 0.78). While blood glucose was only moderately affected, dapagliflozin further reduced endogenous thrombin generation (581.4 ± 194.6 nmol/l × min) × 10-9 thrombin vs 254.1 ± 106.4 (nmol/l × min) × 10-9 thrombin), thereby decreasing one of the most important platelet activators. We observed a direct inhibitory effect of dapagliflozin on isolated platelets. In addition, dapagliflozin increased HDL-cholesterol levels. Importantly, higher HDL-cholesterol levels (1.70 ± 0.58 vs 3.15 ± 1.67 mmol/l, p < 0.01) likely contribute to dapagliflozin-mediated inhibition of platelet activation and thrombin generation. Accordingly, in line with the results in mice, treatment with dapagliflozin lowered CD62P-positive platelet counts in humans after stimulation by collagen-related peptide (CRP; 88.13 ± 5.37% of platelets vs 77.59 ± 10.70%, p < 0.05) or thrombin receptor activator peptide-6 (TRAP-6; 44.23 ± 15.54% vs 28.96 ± 11.41%, p < 0.01) without affecting haemostasis. CONCLUSIONS/INTERPRETATION: We demonstrate that dapagliflozin-mediated atheroprotection in mice is driven by elevated HDL-cholesterol and ameliorated thrombin-platelet-mediated inflammation without interfering with haemostasis. This glucose-independent mechanism likely contributes to dapagliflozin's beneficial cardiovascular risk profile.


Subject(s)
Benzhydryl Compounds/therapeutic use , Coronary Artery Disease/prevention & control , Diabetes Mellitus, Type 2/drug therapy , Glucosides/therapeutic use , Platelet Activation/drug effects , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Thrombin/metabolism , Adult , Animals , Blood Glucose/metabolism , Blood Platelets/drug effects , Blood Platelets/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/prevention & control , Cholesterol, HDL/blood , Coronary Artery Disease/metabolism , Diabetes Mellitus, Type 2/metabolism , Female , Flow Cytometry , Healthy Volunteers , Humans , Immunohistochemistry , Male , Mice, Inbred C57BL , Middle Aged , P-Selectin/metabolism , Platelet Count , Real-Time Polymerase Chain Reaction , Risk Reduction Behavior
3.
Int J Mol Sci ; 22(7)2021 Apr 02.
Article in English | MEDLINE | ID: mdl-33918229

ABSTRACT

BACKGROUND: The use of knock-out mouse models is crucial to understand platelet activation and aggregation. METHODS: Analysis of the global double fluorescent Cre reporter mouse mT/mG that has been crossbred with the megakaryocyte/platelet specific PF4-Cre mouse. RESULTS: Platelets show bright mT (PF4-Cre negative) and mG (PF4-Cre positive) fluorescence. However, a small proportion of leukocytes was positive for mG fluorescence in PF4-Cre positive mice. In mT/mG;PF4-Cre mice, platelets, and megakaryocytes can be tracked by their specific fluorescence in blood smear, hematopoietic organs and upon thrombus formation. No differences in platelet activation and thrombus formation was observed between mT/mG;PF4-Cre positive and negative mice. Furthermore, hemostasis and in vivo thrombus formation was comparable between genotypes as analyzed by intravital microscopy. Transplantation studies revealed that bone marrow of mT/mG;PF4-Cre mice can be transferred to C57BL/6 mice. CONCLUSIONS: The mT/mG Cre reporter mouse is an appropriate model for real-time visualization of platelets, the analysis of cell morphology and the identification of non-recombined platelets. Thus, mT/mG;PF4-Cre mice are important for the analysis of platelet-specific knockout mice. However, a small proportion of leukocytes exhibit mG fluorescence. Therefore, the analysis of platelets beyond hemostasis and thrombosis should be critically evaluated when recombination of immune cells is increased.


Subject(s)
Blood Platelets , Fluorescent Dyes , Megakaryocytes , Animals , Integrases , Mice , Mice, Transgenic
4.
Hepatology ; 74(1): 411-427, 2021 07.
Article in English | MEDLINE | ID: mdl-33369745

ABSTRACT

BACKGROUND AND AIMS: Thrombocytopenia has been described in most patients with acute and chronic liver failure. Decreased platelet production and decreased half-life of platelets might be a consequence of low levels of thrombopoietin (TPO) in these patients. Platelet production is tightly regulated to avoid bleeding complications after vessel injury and can be enhanced under elevated platelet destruction as observed in liver disease. Thrombopoietin (TPO) is the primary regulator of platelet biogenesis and supports proliferation and differentiation of megakaryocytes. APPROACH AND RESULTS: Recent work provided evidence for the control of TPO mRNA expression in liver and bone marrow (BM) by scanning circulating platelets. The Ashwell-Morell receptor (AMR) was identified to bind desialylated platelets to regulate hepatic thrombopoietin (TPO) production by Janus kinase (JAK2)/signal transducer and activator of transcription (STAT3) activation. Two-thirds partial hepatectomy (PHx) was performed in mice. Platelet activation and clearance by AMR/JAK2/STAT3 signaling and TPO production were analyzed at different time points after PHx. Here, we demonstrate that PHx in mice led to thrombocytopenia and platelet activation defects leading to bleeding complications, but unaltered arterial thrombosis, in these mice. Platelet counts were rapidly restored by up-regulation and crosstalk of the AMR and the IL-6 receptor (IL-6R) to induce JAK2-STAT3-TPO activation in the liver, accompanied by an increased number of megakaryocytes in spleen and BM before liver was completely regenerated. CONCLUSIONS: The AMR/IL-6R-STAT3-TPO signaling pathway is an acute-phase response to liver injury to reconstitute hemostasis. Bleeding complications were attributable to thrombocytopenia and platelet defects induced by elevated PGI2 , NO, and bile acid plasma levels early after PHx that might also be causative for the high mortality in patients with liver disease.


Subject(s)
Hepatectomy/adverse effects , Thrombocytopenia/blood , Thrombopoietin/biosynthesis , Animals , Asialoglycoprotein Receptor/genetics , Asialoglycoprotein Receptor/metabolism , Disease Models, Animal , Humans , Janus Kinase 2/metabolism , Mice , Mice, Knockout , Platelet Count , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Specific Pathogen-Free Organisms , Thrombocytopenia/etiology , Thrombopoietin/blood
5.
Sci Signal ; 13(643)2020 08 04.
Article in English | MEDLINE | ID: mdl-32753479

ABSTRACT

Cerebral amyloid angiopathy (CAA) and ß-amyloid (Aß) deposition in the brain parenchyma are hallmarks of Alzheimer's disease (AD). We previously reported that platelets contribute to Aß aggregation in cerebral vessels by secreting the factor clusterin upon binding of Aß40 to the fibrinogen receptor integrin αIIbß3 Here, we investigated the contribution of the collagen receptor GPVI (glycoprotein VI) in platelet-induced amyloid aggregation. Using platelets isolated from GPVI-wild type and GPVI-deficient human donors and mice, we found that Aß40 bound to GPVI, which induced the release of ATP and fibrinogen, resulting in platelet aggregation. Binding of Aß40 to integrin αIIbß3, fibrinogen, and GPVI collectively contributed to the formation of amyloid clusters at the platelet surface. Consequently, blockade of αIIbß3 or genetic loss of GPVI reduced amyloid fibril formation in cultured platelets and decreased the adhesion of Aß-activated platelets to injured carotid arteries in mice. Application of losartan to inhibit collagen binding to GPVI resulted in decreased Aß40-stimulated platelet activation, factor secretion, and platelet aggregation. Furthermore, the application of GPVI- or integrin-blocking antibodies reduced the formation of platelet-associated amyloid aggregates. Our findings indicate that Aß40 promotes platelet-mediated amyloid aggregation by binding to both GPVI and integrin αIIbß3 Blocking these pathways may therapeutically reduce amyloid plaque formation in cerebral vessels and the brain parenchyma of patients.


Subject(s)
Amyloid beta-Peptides/metabolism , Blood Platelets/metabolism , Peptide Fragments/metabolism , Platelet Membrane Glycoproteins/metabolism , Protein Aggregation, Pathological/metabolism , Receptors, Collagen/metabolism , Adult , Alzheimer Disease/metabolism , Animals , Blood Platelets/cytology , Cells, Cultured , Fibrinogen/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/genetics , Protein Binding , Receptors, Collagen/genetics , Signal Transduction
6.
Sci Rep ; 8(1): 10006, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29968773

ABSTRACT

Sepsis is a systemic inflammatory disorder with organ dysfunction and represents the leading cause of mortality in non-coronary intensive care units. A key player in septic shock is Tumor Necrosis Factor-alpha (TNF-α). Phospholipase (PL)D1 is involved in the regulation of TNF-α upon ischemia/reperfusion injury in mice. In this study we analyzed the impact of PLD1 in the regulation of TNF-α, inflammation and organ damage in experimental sepsis. PLD1 deficiency increased survival of mice and decreased vital organ damage after LPS injections. Decreased TNF-α plasma levels and reduced migration of leukocytes and platelets into lungs was associated with reduced apoptosis in lung and liver tissue of PLD1 deficient mice. PLD1 deficient platelets contribute to preserved outcome after LPS-induced sepsis because platelets exhibit an integrin activation defect suggesting reduced platelet activation in PLD1 deficient mice. Furthermore, reduced thrombin generation of PLD1 deficient platelets might be responsible for reduced fibrin formation in lungs suggesting reduced disseminated intravascular coagulation (DIC). The analysis of Pld1fl/fl-PF4-Cre mice revealed that migration of neutrophils and cell apoptosis in septic animals is not due to platelet-mediated processes. The present study has identified PLD1 as a regulator of innate immunity that may be a new target to modulate sepsis.


Subject(s)
Lipopolysaccharides/toxicity , Phospholipase D/metabolism , Shock, Septic/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/immunology , Blood Platelets/metabolism , Cell Movement/genetics , Cells, Cultured , Disease Models, Animal , Fibrin/metabolism , Immunity, Innate/immunology , Inflammation/pathology , Leukocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Phospholipase D/deficiency , Phospholipase D/genetics , Platelet Activation/genetics
7.
J Clin Invest ; 128(9): 3906-3925, 2018 08 31.
Article in English | MEDLINE | ID: mdl-29952767

ABSTRACT

Red blood cells (RBCs) influence rheology, and release ADP, ATP, and nitric oxide, suggesting a role for RBCs in hemostasis and thrombosis. Here, we provide evidence for a significant contribution of RBCs to thrombus formation. Anemic mice showed enhanced occlusion times upon injury of the carotid artery. A small population of RBCs was located to platelet thrombi and enhanced platelet activation by a direct cell contact via the FasL/FasR (CD95) pathway known to induce apoptosis. Activation of platelets in the presence of RBCs led to platelet FasL exposure that activated FasR on RBCs responsible for externalization of phosphatidylserine (PS) on the RBC membrane. Inhibition or genetic deletion of either FasL or FasR resulted in reduced PS exposure of RBCs and platelets, decreased thrombin generation, and reduced thrombus formation in vitro and protection against arterial thrombosis in vivo. Direct cell contacts between platelets and RBCs via FasL/FasR were shown after ligation of the inferior vena cava (IVC) and in surgical specimens of patients after thrombectomy. In a flow restriction model of the IVC, reduced thrombus formation was observed in FasL-/- mice. Taken together, our data reveal a significant contribution of RBCs to thrombosis by the FasL/FasR pathway.


Subject(s)
Blood Platelets/physiology , Erythrocytes/physiology , Fas Ligand Protein/blood , Thrombosis/blood , fas Receptor/blood , Aged , Aged, 80 and over , Anemia/blood , Animals , Blood Platelets/ultrastructure , Cell Communication/physiology , Disease Models, Animal , Erythrocytes/ultrastructure , Fas Ligand Protein/deficiency , Fas Ligand Protein/genetics , Female , Hemorheology/physiology , Hemostasis/physiology , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Middle Aged , Phosphatidylserines/blood , Platelet Activation/physiology , Thrombosis/etiology , Thrombosis/pathology
8.
Cell Signal ; 40: 210-221, 2017 12.
Article in English | MEDLINE | ID: mdl-28943410

ABSTRACT

Reelin is a secreted glycoprotein and essential for brain development and plasticity. Recent studies provide evidence that Reelin modifies platelet actin cytoskeletal dynamics. In this study we sought to dissect the contribution of Reelin in arterial thrombus formation. Here we analyzed the impact of Reelin in arterial thrombosis ex vivo and in vivo using Reelin deficient (reeler) and wildtype mice. We found that Reelin is secreted upon platelet activation and mediates signaling via glycoprotein (GP)Ib, the amyloid precursor protein (APP) and apolipoprotein E receptor 2 (ApoER2) to induce activation of Akt, extracellular signal-regulated kinase (Erk), SYK and Phospholipase Cγ2. Moreover, our data identifies Reelin as first physiological ligand for platelet APP. Platelets from reeler mice displayed attenuated platelet adhesion and significantly reduced thrombus formation under high shear conditions indicating an important role for Reelin in GPIb-dependent integrin αIIbß3 activation. Accordingly, adhesion to immobilized vWF as well as integrin activation and the phosphorylation of Erk and Akt after GPIb engagement was reduced in Reelin deficient platelets. Defective Reelin signaling translated into protection from arterial thrombosis and cerebral ischemia/reperfusion injury beside normal hemostasis. Furthermore, treatment with an antagonistic antibody specific for Reelin protects wildtype mice from occlusive thrombus formation. Mechanistically, GPIb co-localizes to the major Reelin receptor APP in platelets suggesting that Reelin-induced effects on GPIb signaling are mediated by APP-GPIb interaction. These results indicate that Reelin is an important regulator of GPIb-mediated platelet activation and may represent a new therapeutic target for the prevention and treatment of cardio- and cerebrovascular diseases.


Subject(s)
Blood Platelets/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Nerve Tissue Proteins/genetics , Platelet Aggregation/genetics , Platelet Glycoprotein GPIb-IX Complex/genetics , Serine Endopeptidases/genetics , Thrombosis/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Arteries/physiopathology , Blood Platelets/pathology , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Humans , LDL-Receptor Related Proteins/genetics , Mice , Nerve Tissue Proteins/metabolism , Phosphorylation , Platelet Adhesiveness/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIb-IX Complex/metabolism , Receptors, Cell Surface/genetics , Reelin Protein , Serine Endopeptidases/metabolism , Signal Transduction , Thrombosis/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...