Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
J Immunol ; 210(11): 1641-1652, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37058108

ABSTRACT

IL-6 family members contribute to host defense through the stimulation of acute-phase signaling, hematopoiesis, immune reactions, and regenerative processes. To investigate essential mechanisms that are linked toward a constitutively activated gp130 signaling, we generated and characterized a mouse model that reflects a constitutive and cytokine-independent activation of JAK/STAT3 signaling by Lgp130 in CD4- and CD8-positive T cells. Lgp130 is an engineered form of gp130 in which dimerization and activation are forced by a leucine zipper. T cell-specific Lgp130 activation resulted in massive phenotypical abnormalities, including splenomegaly, lymphadenopathy, and an upregulation of innate immune system components shown by hyperinflammatory signatures in several organs. Moreover, T cell-restricted expression of Lgp130 resulted in increased numbers of cytotoxic and regulatory T cells, especially in lymph nodes. Consistent with this, we found an elevated platelet production and increase in megakaryocytes in the spleen and bone marrow that are causative for an acute thrombocytosis accompanied by anemia. Due to a shortened life span of T cell-specific Lgp130 mice, we could also show that next to an overall increase in regulatory cell-cycle genes, an activation of p53 and increased expression of p21 provide evidence for a senescence-like phenotype. Together, these data suggest that T cell-restricted gp130 activation is not only involved in autoimmune processes but also in senescence-associated aging. Therefore, Lgp130 expression in T cells might be a suitable model to study inflammation and disease.


Subject(s)
Aging, Premature , Animals , Mice , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Hematopoiesis , Spleen/metabolism , STAT3 Transcription Factor/metabolism
2.
Platelets ; 35(1): 2322733, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38968449

ABSTRACT

Liver failure and cirrhosis are characterized by abnormal hemostasis with aberrant platelet activation. In particular, the consequences of cholestatic liver disease and molecular mechanisms, including the role of bile acids leading to impaired platelet responses, are not well understood. Here, we demonstrate that bile acids inhibit human and murine platelet activation, adhesion and spreading, leading to reduced thrombus formation under flow conditions. We identified the G-protein coupled receptor TGR5 in platelets and provide support for its role as mediator of bile acid-induced impairment of platelet activation. In the liver, TGR5 couples to Gαs proteins, activates the adenylate cyclase to induce a transient cAMP rise and stimulates the MAPK signaling pathway to regulate cholangiocyte proliferation, hepatocyte survival and inflammation. In this report, we demonstrate that the genetic deficiency of TGR5 in mice led to enhanced platelet activation and thrombus formation, suggesting that TGR5 plays an important role in hemostasis. Mechanistically, platelet inhibition is achieved by TGR5 mediated PKA activation and modulation of AKT and ERK1/2 phosphorylation. Thus, this report provides evidence for the ability of TGR5 ligands to reduce platelet activation and identifies TGR5 agonism as a new target for the prevention of cardiovascular diseases.


What is the context? Liver failure or cirrhosis are related to impaired hemostasis and a role of bile acids in impaired platelet responses is known but only less understood.Platelets express the bile acid receptor FXR. Ligand binding to the FXR on platelets causes a shift in platelet reactivity and is atheroprotective suggesting that the FXR is a potential target for the prevention of atherothrombotic diseases.What is new? Treatment of murine and human blood with bile acids in low molecular quantity led to reduced platelet activation, adhesion and thrombus formation.The bile acid receptor TGR5 was identified on human and murine platelets.TGR5 plays an important role in hemostasis because TGR5 deficient mice showed elevated platelet reactivity and enhanced thrombus formation.Loss of TGR5 led to enhanced PKA activation and modulated the phosphorylation of MAPK such as AKT and ERK1/2.What is the impact? Impairment of platelet activation by bile acids is mediated by TGR5 via the protein kinase A signaling pathway.Our findings provide evidence for the modulation of TGR5 activation as a potential new target of both, anti-platelet therapy in cardiovascular diseases and the restoration of hemostasis upon liver injury.


Subject(s)
Platelet Activation , Receptors, G-Protein-Coupled , Thrombosis , Receptors, G-Protein-Coupled/metabolism , Animals , Mice , Humans , Platelet Activation/drug effects , Thrombosis/metabolism , Blood Platelets/metabolism , Bile Acids and Salts/metabolism , Mice, Knockout , Signal Transduction
3.
Blood ; 137(16): 2152-2160, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33649757

ABSTRACT

Thrombotic, vascular, and bleeding complications are the most common causes of morbidity and mortality in the Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs). In these disorders, circulating red cells, leukocytes, and platelets, as well as some vascular endothelial cells, each have abnormalities that are cell-intrinsic to the MPN driver mutations they harbor (eg, JAK2 V617F). When these cells are activated in the MPNs, their interactions with each other create a highly proadhesive and prothrombotic milieu in the circulation that predisposes patients with MPN to venous, arterial, and microvascular thrombosis and occlusive disease. Bleeding problems in the MPNs are caused by the MPN blood cell-initiated development of acquired von Willebrand disease. The inflammatory state created by MPN stem cells in their microenvironment extends systemically to amplify the clinical thrombotic tendency and, at the same time, preferentially promote further MPN stem cell clonal expansion, thereby generating a vicious cycle that favors a prothrombotic state in these diseases.


Subject(s)
Hemorrhage/pathology , Microvessels/pathology , Myeloproliferative Disorders/pathology , Thrombosis/pathology , Vascular Diseases/pathology , Animals , Hemorrhage/etiology , Humans , Inflammation/etiology , Inflammation/pathology , Myeloproliferative Disorders/complications , Neoplasms/complications , Neoplasms/pathology , Thrombosis/etiology , Tumor Microenvironment , Vascular Diseases/etiology
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.
Int J Mol Sci ; 23(9)2022 May 02.
Article in English | MEDLINE | ID: mdl-35563450

ABSTRACT

Pannexin-1 (PANX1) is a transmembrane protein that forms ion channels as hexamers on the plasma membrane. Electrophysiological studies prove that PANX1 has a high conductance for adenosine triphosphate (ATP), which plays an important role as a signal molecule in platelet activation. Recently, it was shown that PANX1 channels modulate platelet functions. To date, it remains unclear how PANX1 channels are activated and which signaling mechanisms are responsible for impaired hemostasis and thrombosis. Analysis of PANX1 phosphorylation at Tyr198 and Tyr308, and the impact on platelet activation and thrombus formation using genetically modified platelets or pharmacological inhibitors. Platelet activation via immunoreceptor tyrosine-based activation motif (ITAM) coupled, G Protein-Coupled Receptors (GPCR) and thromboxane receptor (TP)-mediated signaling pathways led to increased PANX1 phosphorylation at Tyr198 and Tyr308. We identified the Src-GPVI signaling axes as the main pathway inducing PANX1 activation, while PKC and Akt play a minor role. PANX1 channels function as ATP release channels in platelets to support arterial thrombus formation. PANX1 activation is regulated by phosphorylation at Tyr198 and Tyr308 following platelet activation. These results suggest an important role of PANX1 in hemostasis and thrombosis by releasing extracellular ATP to support thrombus formation.


Subject(s)
Connexins , Platelet Aggregation , Thrombosis , Adenosine Triphosphate/metabolism , Animals , Blood Platelets/metabolism , Connexins/metabolism , Humans , Mice , Nerve Tissue Proteins/metabolism , Phosphorylation , Platelet Activation , Platelet Aggregation/physiology , Thrombosis/metabolism
6.
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
7.
Arterioscler Thromb Vasc Biol ; 40(10): 2391-2403, 2020 10.
Article in English | MEDLINE | ID: mdl-32787521

ABSTRACT

OBJECTIVE: Reelin, a secreted glycoprotein, was originally identified in the central nervous system, where it plays an important role in brain development and maintenance. In the cardiovascular system, reelin plays a role in atherosclerosis by enhancing vascular inflammation and in arterial thrombosis by promoting platelet adhesion, activation, and thrombus formation via APP (amyloid precursor protein) and GP (glycoprotein) Ib. However, the role of reelin in hemostasis and arterial thrombosis is not fully understood to date. Approach and Results: In the present study, we analyzed the importance of reelin for cytoskeletal reorganization of platelets and thrombus formation in more detail. Platelets release reelin to amplify alphaIIb beta3 integrin outside-in signaling by promoting platelet adhesion, cytoskeletal reorganization, and clot retraction via activation of Rho GTPases RAC1 (Ras-related C3 botulinum toxin substrate) and RhoA (Ras homolog family member A). Reelin interacts with the collagen receptor GP (glycoprotein) VI with subnanomolar affinity, induces tyrosine phosphorylation in a GPVI-dependent manner, and supports platelet binding to collagen and GPVI-dependent RAC1 activation, PLC gamma 2 (1-phosphatidylinositol-4,5-bisphosphate phosphodiesterase gamma-2) phosphorylation, platelet activation, and aggregation. When GPVI was deleted from the platelet surface by antibody treatment in reelin-deficient mice, thrombus formation was completely abolished after injury of the carotid artery while being only reduced in either GPVI-depleted or reelin-deficient mice. CONCLUSIONS: Our study identified a novel signaling pathway that involves reelin-induced GPVI activation and alphaIIb beta3 integrin outside-in signaling in platelets. Loss of both, GPVI and reelin, completely prevents stable arterial thrombus formation in vivo suggesting that inhibiting reelin-platelet-interaction might represent a novel strategy to avoid arterial thrombosis in cardiovascular disease.


Subject(s)
Blood Platelets/enzymology , Carotid Artery Injuries/enzymology , Cell Adhesion Molecules, Neuronal/blood , Extracellular Matrix Proteins/blood , Nerve Tissue Proteins/blood , Neuropeptides/blood , Phospholipase C gamma/blood , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/metabolism , Serine Endopeptidases/blood , Thrombosis/enzymology , rac1 GTP-Binding Protein/blood , rhoA GTP-Binding Protein/blood , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Blood Coagulation , Carotid Artery Injuries/blood , Carotid Artery Injuries/etiology , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/genetics , Clot Retraction , Cytoskeleton/enzymology , Disease Models, Animal , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Mice, 129 Strain , Mice, Inbred C3H , Mice, Inbred C57BL , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Platelet Activation , Reelin Protein , Serine Endopeptidases/deficiency , Serine Endopeptidases/genetics , Signal Transduction , Thrombosis/blood , Thrombosis/etiology
8.
Int J Mol Sci ; 22(17)2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34502546

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is characterized by an accumulation of amyloid ß (Aß) peptides in the brain and mitochondrial dysfunction. Platelet activation is enhanced in AD and platelets contribute to AD pathology by their ability to facilitate soluble Aß to form Aß aggregates. Thus, anti-platelet therapy reduces the formation of cerebral amyloid angiopathy in AD transgenic mice. Platelet mitochondrial dysfunction plays a regulatory role in thrombotic response, but its significance in AD is unknown and explored herein. METHODS: The effects of Aß-mediated mitochondrial dysfunction in platelets were investigated in vitro. RESULTS: Aß40 stimulation of human platelets led to elevated reactive oxygen species (ROS) and superoxide production, while reduced mitochondrial membrane potential and oxygen consumption rate. Enhanced mitochondrial dysfunction triggered platelet-mediated Aß40 aggregate formation through GPVI-mediated ROS production, leading to enhanced integrin αIIbß3 activation during synergistic stimulation from ADP and Aß40. Aß40 aggregate formation of human and murine (APP23) platelets were comparable to controls and could be reduced by the antioxidant vitamin C. CONCLUSIONS: Mitochondrial dysfunction contributes to platelet-mediated Aß aggregate formation and might be a promising target to limit platelet activation exaggerated pathological manifestations in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Blood Platelets/metabolism , Mitochondria/metabolism , Protein Aggregation, Pathological/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/pharmacology , Animals , Blood Platelets/cytology , Blood Platelets/drug effects , Cells, Cultured , Humans , Integrins/metabolism , Membrane Potential, Mitochondrial/genetics , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Oxygen Consumption/drug effects , Platelet Activation/drug effects , Platelet Function Tests/methods , Reactive Oxygen Species/metabolism
9.
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
10.
Int J Mol Sci ; 22(22)2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34830059

ABSTRACT

BACKGROUND: Vascular injury induces the exposure of subendothelial extracellular matrix (ECM) important to serve as substrate for platelets to adhere to the injured vessel wall to avoid massive blood loss. Different ECM proteins are known to initiate platelet adhesion and activation. In atherosclerotic mice, the small, leucine-rich proteoglycan biglycan is important for the regulation of thrombin activity via heparin cofactor II. However, nothing is known about the role of biglycan for hemostasis and thrombosis under nonatherosclerotic conditions. METHODS: The role of biglycan for platelet adhesion and thrombus formation was investigated using a recombinant protein and biglycan knockout mice. RESULTS: The present study identified biglycan as important ECM protein for the adhesion and activation of platelets, and the formation of three-dimensional thrombi under flow conditions. Platelet adhesion to immobilized biglycan induces the reorganization of the platelet cytoskeleton. Mechanistically, biglycan binds and activates the major collagen receptor glycoprotein (GP)VI, because reduced platelet adhesion to recombinant biglycan was observed when GPVI was blocked and enhanced tyrosine phosphorylation in a GPVI-dependent manner was observed when platelets were stimulated with biglycan. In vivo, the deficiency of biglycan resulted in reduced platelet adhesion to the injured carotid artery and prolonged bleeding times. CONCLUSIONS: Loss of biglycan in the vessel wall of mice but not in platelets led to reduced platelet adhesion at the injured carotid artery and prolonged bleeding times, suggesting a crucial role for biglycan as ECM protein that binds and activates platelets via GPVI upon vessel injury.


Subject(s)
Biglycan/genetics , Biglycan/metabolism , Platelet Adhesiveness/physiology , Platelet Membrane Glycoproteins/metabolism , Thrombosis/metabolism , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , Carotid Arteries/metabolism , Carotid Artery Injuries/metabolism , Collagen/metabolism , Cytoskeleton/metabolism , Extracellular Matrix Proteins/metabolism , Healthy Volunteers , Hemorrhage/genetics , Hemorrhage/metabolism , Humans , Integrins/metabolism , Male , Mice, Inbred C57BL , Platelet Activation/physiology , Platelet Adhesiveness/genetics
11.
Int J Mol Sci ; 21(9)2020 May 01.
Article in English | MEDLINE | ID: mdl-32370031

ABSTRACT

BACKGROUND: Phospholipase (PL)D1 is crucial for integrin αIIbß3 activation of platelets in arterial thrombosis and TNF-α-mediated inflammation and TGF-ß-mediated collagen scar formation after myocardial infarction (MI) in mice. Enzymatic activity of PLD is not responsible for PLD-mediated TNF-α signaling and myocardial healing. The impact of PLD2 in ischemia reperfusion injury is unknown. METHODS: PLD2-deficient mice underwent myocardial ischemia and reperfusion (I/R). RESULTS: Enhanced integrin αIIbß3 activation of platelets resulted in elevated interleukin (IL)-6 release from endothelial cells in vitro and enhanced IL-6 plasma levels after MI in PLD2-deficient mice. This was accompanied by enhanced migration of inflammatory cells into the infarct border zone and reduced TGF-ß plasma levels after 72 h that might account for enhanced inflammation in PLD2-deficient mice. In contrast to PLD1, TNF-α signaling, infarct size and cardiac function 24 h after I/R were not altered when PLD2 was deleted. Furthermore, TGF-ß plasma levels, scar formation and heart function were comparable between PLD2-deficient and control mice 21 days post MI. CONCLUSIONS: The present study contributes to our understanding about the role of PLD isoforms and altered platelet signaling in the process of myocardial I/R injury.


Subject(s)
Blood Platelets/metabolism , Integrins/metabolism , Myocardial Infarction/complications , Myocarditis/etiology , Myocarditis/metabolism , Phospholipase D/deficiency , Animals , Biomarkers , Cell Survival , Cytokines/metabolism , Disease Susceptibility , Endothelial Cells/metabolism , Gene Expression , Integrins/chemistry , Male , Mice , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocarditis/pathology
12.
Int J Mol Sci ; 21(21)2020 Oct 24.
Article in English | MEDLINE | ID: mdl-33114406

ABSTRACT

Cardiovascular diseases (CVDs) are the leading cause of death globally-partly a consequence of increased population size and ageing-and are major contributors to reduced quality of life. Platelets play a major role in hemostasis and thrombosis. While platelet activation and aggregation are essential for hemostasis at sites of vascular injury, uncontrolled platelet activation leads to pathological thrombus formation and provokes thrombosis leading to myocardial infarction or stroke. Platelet activation and thrombus formation is a multistage process with different signaling pathways involved to trigger platelet shape change, integrin activation, stable platelet adhesion, aggregation, and degranulation. Apart from thrombotic events, thrombo-inflammation contributes to organ damage and dysfunction in CVDs and is mediated by platelets and inflammatory cells. Therefore, in the past, many efforts have been made to investigate specific signaling pathways in platelets to identify innovative and promising approaches for novel antithrombotic and anti-thrombo-inflammatory strategies that do not interfere with hemostasis. In this review, we focus on some of the most recent data reported on different platelet receptors, including GPIb-vWF interactions, GPVI activation, platelet chemokine receptors, regulation of integrin signaling, and channel homeostasis of NMDAR and PANX1.


Subject(s)
Blood Platelets/metabolism , Cardiovascular Diseases/metabolism , Gene Regulatory Networks , Anti-Inflammatory Agents/pharmacology , Cardiovascular Diseases/blood , Cardiovascular Diseases/drug therapy , Fibrinolytic Agents/pharmacology , Gene Regulatory Networks/drug effects , Humans , Molecular Targeted Therapy , Platelet Activation , Platelet Glycoprotein GPIb-IX Complex/metabolism , Quality of Life , Reelin Protein , von Willebrand Factor/metabolism
13.
Cell Physiol Biochem ; 41(6): 2133-2149, 2017.
Article in English | MEDLINE | ID: mdl-28441661

ABSTRACT

BACKGROUND/AIMS: Platelets are essential mediators of hemostasis to avoid excessive blood loss. Cirrhosis and chronic liver diseases are characterized by alterations in hemostasis. Alterations in the secondary hemostasis have been well studied, while defects in primary hemostasis, especially the consequences of cholestatic liver disease on platelet function are not well defined. METHODS: After bile duct ligation (BDL) platelet activation and thrombus formation were analyzed in mice. RESULTS: BDL in mice had a moderate effect on platelet counts; however, intrinsic platelet activation was strongly reduced upon activation of the collagen receptor GPVI at early time points. 7 days after bile duct ligation, platelets displayed an almost complete loss of activation with reduced agonist-triggered release of alpha and dense granules and expression of integrin αIIbß3 on the platelet surface. This activation defects resulted in strongly reduced thrombus formation under flow, reduced platelet adhesion to fibrinogen and bleeding complications in BDL mice as measured by tail bleeding experiments. Mechanistically, elevated nitric oxide and prostacyclin levels induced phosphorylation of Vasodilator-stimulated phosphoprotein (VASP), an established inhibitor of platelet activation. Furthermore increased tissue plasminogen activator in plasma of BDL mice led to enhanced plasmin levels that might be responsible for reduced glycoprotein expression of BDL platelets. Besides, high amounts of bile acids contribute to defective signal transduction as shown in platelets from mice fed with a cholic acid diet. CONCLUSIONS: Cholestatic liver disease induces multiple platelet activation defects and impairs thrombus formation responsible for bleeding complications at least in mice.


Subject(s)
Blood Platelets/metabolism , Cholestasis/pathology , Animals , Blood Platelets/cytology , Cell Adhesion Molecules , Cholestasis/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Epoprostenol/analysis , Hemorrhage/etiology , Liver/pathology , Mice , Mice, Inbred C57BL , Microfilament Proteins , Nitric Oxide/metabolism , Phosphoproteins , Phosphorylation , Platelet Activation , Platelet Count , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/metabolism , Spleen/pathology , Thrombosis/metabolism , Thrombosis/pathology , Tissue Plasminogen Activator/blood
14.
Arterioscler Thromb Vasc Biol ; 36(5): e41-50, 2016 05.
Article in English | MEDLINE | ID: mdl-27034473

ABSTRACT

OBJECTIVE: Thrombin signaling promotes atherosclerosis by initiating inflammatory events indirectly through platelet activation and directly via protease-activated receptors. Therefore, endogenous thrombin inhibitors may be relevant modulators of atheroprogression and cardiovascular risk. In addition, endogenous thrombin inhibitors may affect the response to non-vitamin K-dependent oral anticoagulants. Here, the question was addressed whether the small leucine-rich proteoglycan biglycan acts as an endogenous thrombin inhibitor in atherosclerosis through activation of heparin cofactor II. APPROACH AND RESULTS: Biglycan concentrations were elevated in the plasma of patients with acute coronary syndrome and in male Apolipoprotein E-deficient (ApoE(-/-)) mice. Biglycan was detected in the glycocalyx of capillaries and the subendothelial matrix of arterioles of ApoE(-/-) mice and in atherosclerotic plaques. Thereby a vascular compartment is provided that may mediate the endothelial and subendothelial activation of heparin cofactor II through biglycan. ApoE and Bgn double-deficient (ApoE(-/-)/Bgn(-/0)) mice showed higher activity of circulating thrombin, increased platelet activation and platelet adhesion in vivo, supporting a role of biglycan in balancing thrombin activity. Furthermore, concentrations of circulating cytokines and aortic macrophage content were elevated in ApoE(-/-)/Bgn(-/0) mice, suggesting a proinflammatory phenotype. Elevated platelet activation and macrophage accumulation were reversed by treating ApoE(-/-)/Bgn(-/0) mice with the thrombin inhibitor argatroban. Ultimately, ApoE(-/-)/Bgn(-/0) mice developed aggravated atherosclerosis. CONCLUSIONS: The present results indicate that biglycan plays a previously unappreciated protective role during the progression of atherosclerosis by inhibiting thrombin activity, platelet activation, and finally macrophage-mediated plaque inflammation.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Biglycan/deficiency , Inflammation/metabolism , Thrombin/metabolism , Acute Coronary Syndrome/blood , Animals , Antithrombins/pharmacology , Aorta/drug effects , Aorta/pathology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/prevention & control , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Biglycan/blood , Biglycan/genetics , Cytokines/blood , Disease Models, Animal , Genotype , Heparin Cofactor II/metabolism , Humans , Inflammation/blood , Inflammation/genetics , Inflammation/prevention & control , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Plaque, Atherosclerotic , Platelet Activation , Time Factors
15.
Am J Pathol ; 184(9): 2450-64, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25046692

ABSTRACT

Myocardial inflammation is critical for ventricular remodeling after ischemia. Phospholipid mediators play an important role in inflammatory processes. In the plasma membrane they are degraded by phospholipase D1 (PLD1). PLD1 was shown to be critically involved in ischemic cardiovascular events. Moreover, PLD1 is coupled to tumor necrosis factor-α signaling and inflammatory processes. However, the impact of PLD1 in inflammatory cardiovascular disease remains elusive. Here, we analyzed the impact of PLD1 in tumor necrosis factor-α-mediated activation of monocytes after myocardial ischemia and reperfusion using a mouse model of myocardial infarction. PLD1 expression was highly up-regulated in the myocardium after ischemia/reperfusion. Genetic ablation of PLD1 led to defective cell adhesion and migration of inflammatory cells into the infarct border zone 24 hours after ischemia/reperfusion injury, likely owing to reduced tumor necrosis factor-α expression and release, followed by impaired nuclear factor-κB activation and interleukin-1 release. Moreover, PLD1 was found to be important for transforming growth factor-ß secretion and smooth muscle α-actin expression of cardiac fibroblasts because myofibroblast differentiation and interstitial collagen deposition were altered in Pld1(-/-) mice. Consequently, infarct size was increased and left ventricular function was impaired 28 days after myocardial infarction in Pld1(-/-) mice. Our results indicate that PLD1 is crucial for tumor necrosis factor-α-mediated inflammation and transforming growth factor-ß-mediated collagen scar formation, thereby augmenting cardiac left ventricular function after ischemia/reperfusion.


Subject(s)
Inflammation/pathology , Myocardial Reperfusion Injury/pathology , Phospholipase D/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Movement/physiology , Cicatrix/metabolism , Cicatrix/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/enzymology , Myocardial Reperfusion Injury/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
16.
FASEB J ; 28(5): 2108-19, 2014 May.
Article in English | MEDLINE | ID: mdl-24522202

ABSTRACT

Platelets are activated by increased cytosolic Ca(2+) concentration ([Ca(2+)]i) following store-operated calcium entry (SOCE) accomplished by calcium-release-activated calcium (CRAC) channel moiety Orai1 and its regulator STIM1. In other cells, Ca(2+) transport is regulated by 1,25(OH)2 vitamin D3 [1,25(OH)2D3]. 1,25(OH)2D3 formation is inhibited by klotho and excessive in klotho-deficient mice (kl/kl). The present study explored the effect of klotho deficiency on platelet Ca(2+) signaling and activation. Platelets and megakaryocytes isolated from WT and kl/kl-mice were analyzed by RT-PCR, Western blotting, confocal microscopy, Fura-2-fluorescence, patch clamp, flow cytometry, aggregometry, and flow chamber. STIM1/Orai1 transcript and protein levels, SOCE, agonist-induced [Ca(2+)]i increase, activation-dependent degranulation, integrin αIIbß3 activation and aggregation, and thrombus formation were significantly blunted in kl/kl platelets (by 27-90%). STIM1/Orai1 transcript and protein levels, as well as CRAC currents, were significantly reduced in kl/kl megakaryocytes (by 38-73%) and 1,25(OH)2D3-treated WT megakaryocytes. Nuclear NF-κB subunit p50/p65 abundance was significantly reduced in kl/kl-megakaryocytes (by 51-76%). Transfection with p50/p65 significantly increased STIM1/Orai1 transcript and protein levels in megakaryocytic MEG-01 cells (by 46-97%). Low-vitamin D diet (LVD) of kl/kl mice normalized plasma 1,25(OH)2D3 concentration and function of platelets and megakaryocytes. Klotho deficiency inhibits platelet Ca(2+) signaling and activation, an effect at least partially due to 1,25(OH)2D3-dependent down-regulation of NF-κB activity and STIM1/Orai1 expression in megakaryocytes.


Subject(s)
Blood Platelets/metabolism , Calcitriol/metabolism , Calcium Signaling , Calcium/metabolism , Glucuronidase/genetics , Thrombosis/metabolism , Animals , Calcium Channels/metabolism , Cell Nucleus/metabolism , Cytosol/metabolism , Down-Regulation , Klotho Proteins , Megakaryocytes/cytology , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , NF-kappa B/metabolism , ORAI1 Protein , Patch-Clamp Techniques , Platelet Aggregation , Signal Transduction , Stromal Interaction Molecule 1 , Transfection
17.
Arterioscler Thromb Vasc Biol ; 34(1): 61-71, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24233488

ABSTRACT

OBJECTIVE: Platelet activation is essential for primary hemostasis and acute thrombotic vascular occlusions. On activation, platelets release their prothrombotic granules and expose phosphatidylserine, thus fostering thrombin generation and thrombus formation. In other cell types, both degranulation and phosphatidylserine exposure are modified by sphingomyelinase-dependent formation of ceramide. The present study thus explored whether acid sphingomyelinase participates in the regulation of platelet secretion, phosphatidylserine exposure, and thrombus formation. APPROACH AND RESULTS: Collagen-related peptide-induced or thrombin-induced ATP release and P-selectin exposure were significantly blunted in platelets from Asm-deficient mice (Smpd1(-/-)) when compared with platelets from wild-type mice (Smpd1(+/+)). Moreover, phosphatidylserine exposure and thrombin generation were significantly less pronounced in Smpd1(-/-) platelets than in Smpd1(+/+) platelets. In contrast, platelet integrin αIIbß3 activation and aggregation, as well as activation-dependent Ca(2+) flux, were not significantly different between Smpd1(-/-) and Smpd1(+/+) platelets. In vitro thrombus formation at shear rates of 1700 s(-1) and in vivo thrombus formation after FeCl3 injury were significantly blunted in Smpd1(-/-) mice while bleeding time was unaffected. Asm-deficient platelets showed significantly reduced activation-dependent ceramide formation, whereas exogenous ceramide rescued diminished platelet secretion and thrombus formation caused by Asm deficiency. Treatment of Smpd1(+/+) platelets with bacterial sphingomyelinase (0.01 U/mL) increased, whereas treatment with functional acid sphingomyelinase-inhibitors, amitriptyline or fluoxetine (5 µmol/L), blunted activation-dependent platelet degranulation, phosphatidylserine exposure, and thrombus formation. Impaired degranulation and thrombus formation of Smpd1(-/-) platelets were again overcome by exogenous bacterial sphingomyelinase. CONCLUSIONS: Acid sphingomyelinase is a completely novel element in the regulation of platelet plasma membrane properties, secretion, and thrombus formation.


Subject(s)
Blood Platelets/enzymology , Cell Degranulation , Cell Membrane/enzymology , Platelet Activation , Sphingomyelin Phosphodiesterase/blood , Thrombosis/enzymology , Adenosine Triphosphate/blood , Animals , Blood Platelets/drug effects , Calcium/blood , Cell Degranulation/drug effects , Cell Membrane/drug effects , Ceramides/blood , Chlorides , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Ferric Compounds , Fibrinolytic Agents/pharmacology , Male , Mice , Mice, Knockout , P-Selectin/blood , Phosphatidylserines/blood , Platelet Activation/drug effects , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics , Thrombin/metabolism , Thrombosis/blood , Thrombosis/chemically induced , Thrombosis/genetics , Thrombosis/prevention & control , Time Factors
18.
Blood ; 120(6): 1317-26, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22740452

ABSTRACT

Platelet adhesion and aggregation play a critical role in primary hemostasis. Uncontrolled platelet activation leads to pathologic thrombus formation and organ failure. The decisive central step for different processes of platelet activation is the increase in cytosolic Ca(2+) activity ([Ca(2+)](i)). Activation-dependent depletion of intracellular Ca(2+) stores triggers Ca(2+) entry from the extracellular space. Stromal interaction molecule 1 (STIM1) has been identified as a Ca(2+) sensor that regulates store-operated Ca(2+) entry through activation of the pore-forming subunit Orai1, the major store-operated Ca(2+) entry channel in platelets. In the present study, we show for the first time that the chaperone protein cyclophilin A (CyPA) acts as a Ca(2+) modulator in platelets. CyPA deficiency strongly blunted activation-induced Ca(2+) mobilization from intracellular stores and Ca(2+) influx from the extracellular compartment and thus impaired platelet activation substantially. Furthermore, the phosphorylation of the Ca(2+) sensor STIM1 was abrogated upon CyPA deficiency, as shown by immunoprecipitation studies. In a mouse model of arterial thrombosis, CyPA-deficient mice were protected against arterial thrombosis, whereas bleeding time was not affected. The results of the present study identified CyPA as an important Ca(2+) regulator in platelets, a critical mechanism for arterial thrombosis.


Subject(s)
Blood Platelets/metabolism , Calcium/metabolism , Cyclophilin A/physiology , Thrombosis/genetics , Animals , CHO Cells , Calcium Signaling/genetics , Cell Degranulation/genetics , Cell Degranulation/physiology , Cricetinae , Cricetulus , Cyclophilin A/genetics , Cyclophilin A/metabolism , Integrin beta3/metabolism , Intracellular Space/metabolism , Mice , Mice, Knockout , Models, Biological , Peripheral Arterial Disease/genetics , Peripheral Arterial Disease/metabolism , Platelet Activation/genetics , Thrombosis/metabolism
19.
Blood ; 119(1): 251-61, 2012 Jan 05.
Article in English | MEDLINE | ID: mdl-22031864

ABSTRACT

Platelets are activated on increase of cytosolic Ca2+ activity ([Ca2+](i)), accomplished by store-operated Ca2+ entry (SOCE) involving the pore-forming ion channel subunit Orai1. Here, we show, for the first time, that the serum- and glucocorticoid-inducible kinase 1 (SGK1) is expressed in platelets and megakaryocytes. SOCE and agonist-induced [Ca2+](i) increase are significantly blunted in platelets from SGK1 knockout mice (sgk1(-/-)). Similarly, Ca2+ -dependent degranulation, integrin α(IIb)ß3 activation, phosphatidylserine exposure, aggregation, and in vitro thrombus formation were significantly impaired in sgk1(-/-) platelets, whereas tail bleeding time was not significantly enhanced. Platelet and megakaryocyte Orai1 transcript levels and membrane protein abundance were significantly reduced in sgk1(-/-) mice. In human megakaryoblastic cells (MEG-01), transfection with constitutively active (S422D)SGK1 but not with inactive (K127N)SGK1 significantly enhanced Orai1 expression and SOCE, while effects reversed by the SGK1 inhibitor GSK650394 (1µM). Transfection of MEG-01 cells with (S422D)SGK1 significantly increased phosphorylation of IκB kinase α/ß and IκBα resulting in nuclear translocation of NF-κB subunit p65. Treatment of (S422D)SGK1-transfected MEG-01 cells with the IκB kinase inhibitor BMS-345541 (10µM) abolished SGK1-induced increase of Orai1 expression and SOCE. The present observations unravel SGK1 as novel regulator of platelet function, effective at least in part by NF-κB-dependent transcriptional up-regulation of Orai1 in megakaryocytes and increasing platelet SOCE.


Subject(s)
Blood Platelets/metabolism , Calcium Channels/metabolism , Calcium Signaling , Calcium/metabolism , Immediate-Early Proteins/physiology , Megakaryocytes/metabolism , Protein Serine-Threonine Kinases/physiology , Animals , Bleeding Time , Blotting, Western , Calcium Channels/genetics , Cells, Cultured , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Leukemia, Megakaryoblastic, Acute/metabolism , Leukemia, Megakaryoblastic, Acute/pathology , Male , Megakaryocytes/cytology , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , ORAI1 Protein , Phosphorylation , Platelet Aggregation , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thrombosis/etiology , Thrombosis/metabolism , Thrombosis/pathology
20.
Biomedicines ; 12(5)2024 May 13.
Article in English | MEDLINE | ID: mdl-38791039

ABSTRACT

Acute myocardial infarction (AMI) is one of the leading causes of death worldwide. Cell apoptosis in the myocardium plays an important role in ischemia and reperfusion (I/R) injury, leading to cardiac damage and dysfunction. Platelets are major players in hemostasis and play a crucial role in vessel occlusion, inflammation, and cardiac remodeling after I/R. Here, we studied the impact of platelets on cell apoptosis in the myocardium using a close-chest mouse model of AMI. We found caspase-3-positive resident cardiac cells, while leukocytes were negative for caspase-3. Using two different mouse models of thrombocytopenia, we detected a significant reduction in caspase-3 positive cells in the infarct border zone after I/R injury. Further, we identified platelet FasL to induce cell apoptosis via the extrinsic pathway of Fas receptor activation of target cells. Mechanistically, hypoxia triggers platelet adhesion to FasR, suggesting that platelet-induced apoptosis is elevated after I/R. Platelet-specific FasL knock-out mice showed reduced Bax and Bcl2 expression, suggesting that platelets modulate the intrinsic and extrinsic pathways of apoptosis, leading to reduced infarct size after myocardial I/R injury. Thus, a new mechanism for how platelets contribute to tissue homeostasis after AMI was identified that should be validated in patients soon.

SELECTION OF CITATIONS
SEARCH DETAIL