Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Acta Physiol (Oxf) ; 240(3): e14099, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38230889

ABSTRACT

AIM: Heart Failure with preserved Ejection Fraction (HFpEF) is characterized by diastolic dysfunction and reduced cardiac output, but its pathophysiology remains poorly understood. Animal models of HFpEF are challenging due to difficulties in assessing the degree of heart failure in small animals. This study aimed at inducing HFpEF in a mouse model to probe preload-dependency. METHODS: Increased body mass and arterial hypertension were induced in mice using a Western diet and NO synthase inhibition. Preload dependence was tested ex vivo. RESULTS: Mice with obesity and hypertension exhibited reduced cardiac output, indicating a failing heart. Increased left ventricular filling pressure during diastole suggested reduced compliance. Notably, the ejection fraction was preserved, suggesting the development of HFpEF. Spontaneous physical activity at night was reduced in HFpEF mice, indicating exercise intolerance; however, the cardiac connective tissue content was comparable between HFpEF and control mice. The HFpEF mice showed increased vulnerability to reduced preload ex vivo, indicating that elevated left ventricular filling pressure compensated for the rigid left ventricle, preventing a critical decrease in cardiac output. CONCLUSION: This animal model successfully developed mild HFpEF with a reduced pump function that was dependent on a high preload. A model of mild HFpEF may serve as a valuable tool for studying disease progression and interventions aimed at delaying or reversing symptom advancement, considering the slow development of HFpEF in patients.


Subject(s)
Cardiomyopathies , Heart Failure , Hypertension , Humans , Mice , Animals , Stroke Volume , Disease Models, Animal , Ventricular Function, Left
2.
Thromb Haemost ; 2023 Oct 16.
Article in English | MEDLINE | ID: mdl-37846465

ABSTRACT

BACKGROUND: Deep vein thrombosis (DVT) is a common condition associated with significant mortality due to pulmonary embolism. Despite advanced prevention and anticoagulation therapy, the incidence of venous thromboembolism remains unchanged. Individuals with elevated hematocrit and/or excessively high erythropoietin (EPO) serum levels are particularly susceptible to DVT formation. We investigated the influence of short-term EPO administration compared to chronic EPO overproduction on DVT development. Additionally, we examined the role of the spleen in this context and assessed its impact on thrombus composition. METHODS: We induced ligation of the caudal vena cava (VCC) in EPO-overproducing Tg(EPO) mice as well as wildtype mice treated with EPO for two weeks, both with and without splenectomy. The effect on platelet circulation time was evaluated through FACS analysis, and thrombus composition was analyzed using immunohistology. RESULTS: We present evidence for an elevated thrombogenic phenotype resulting from chronic EPO overproduction, achieved by combining an EPO-overexpressing mouse model with experimental DVT induction. This increased thrombotic state is largely independent of traditional contributors to DVT, such as neutrophils and platelets. Notably, the pronounced prothrombotic effect of red blood cells (RBCs) only manifests during chronic EPO overproduction and is not influenced by splenic RBC clearance, as demonstrated by splenectomy. In contrast, short-term EPO treatment does not induce thrombogenesis in mice. Consequently, our findings support the existence of a differential thrombogenic effect between chronic enhanced erythropoiesis and exogenous EPO administration. CONCLUSION: Chronic EPO overproduction significantly increases the risk of DVT, while short-term EPO treatment does not. These findings underscore the importance of considering EPO-related factors in DVT risk assessment and potential therapeutic strategies.

3.
Int J Sports Med ; 42(14): 1297-1304, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33975368

ABSTRACT

An athlete's skin has to cope with various stressors that influence skin integrity. This study investigated the effect of intense sporting activity at a competitive level on skin health, independent of the type of sport. The prevalence of dermatoses in elite athletes who exercise 8 or more hours per week was compared to recreational athletes. By applying a questionnaire, we acquired data from n=492 recruited athletes and recreational athletes regarding the prevalence of dermatoses, the extent of physical activity, and sports discipline practiced. Compared to the reference group, elite athletes showed less inflammatory, traumatic, infectious, and sebaceous skin diseases and especially neurodermitis, pruritus, bullae, tinea pedis, acne, and herpes were less common. Women suffered from skin diseases more often than men. With advanced age, the incidence of dermatoses increased, but less so among elite athletes. The discipline practiced and the duration of training, especially when performed outdoors, strongly influenced the development of dermatoses. Even though the skin of athletes is exposed to higher stress levels and physical strain, we can state that intensive physical activity seems to act as a protecting factor against skin diseases and significantly promotes skin health.


Subject(s)
Athletes , Skin Diseases , Sports , Exercise , Humans , Skin Diseases/epidemiology
4.
Skin Res Technol ; 27(5): 723-729, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33511695

ABSTRACT

BACKGROUND: Keratinocyte cancers, also referred to as non-melanoma skin cancers (NMSCs), are one of the most common malignant skin tumors. We performed a retrospective analysis of lesions from patients of a private dermatology practice to evaluate the use of electrical impedance spectroscopy (EIS) in detecting keratinocyte malignancies. The aim of the study is to assess the accuracy of the technique and to rate its use as supportive tool in NMSC diagnosis. MATERIAL AND METHODS: The period evaluated ranges from September 2015 to November 2019. In total, 1712 lesions from 951 patients were included. All lesions suspicious for malignancy were gauged with the Nevisense device. Excised lesions were sent in for histopathological classification, and the results were compared to the Nevisense score. RESULTS: A total of 767 lesions (44.8%) received a negative score (0-3) from the Nevisense system and 945 lesions (55.2%) a positive score (4-10). The combination of the dermatologist's visual assessment plus the technical determined Neviscore resulted in the excision of 52.5% of all 1712 suspicious lesions whereof 15% were found to be malignant. The sensitivity of Nevisense was 98.4% for NMSC detection. CONCLUSION: Electrical impedance spectroscopy was found to be a valuable adjunct support tool in clinical decisions for cases with suspicion for NMSC.


Subject(s)
Melanoma , Skin Neoplasms , Dielectric Spectroscopy , Electric Impedance , Humans , Keratinocytes , Melanoma/diagnosis , Retrospective Studies , Sensitivity and Specificity , Skin Neoplasms/diagnosis
6.
Arterioscler Thromb Vasc Biol ; 38(4): 772-786, 2018 04.
Article in English | MEDLINE | ID: mdl-29419408

ABSTRACT

OBJECTIVE: Cancer patients are at high risk of developing deep venous thrombosis (DVT) and venous thromboembolism, a leading cause of mortality in this population. However, it is largely unclear how malignant tumors drive the prothrombotic cascade culminating in DVT. APPROACH AND RESULTS: Here, we addressed the pathophysiology of malignant DVT compared with nonmalignant DVT and focused on the role of tumor microvesicles as potential targets to prevent cancer-associated DVT. We show that microvesicles released by pancreatic adenocarcinoma cells (pancreatic tumor-derived microvesicles [pcMV]) boost thrombus formation in a model of flow restriction of the mouse vena cava. This depends on the synergistic activation of coagulation by pcMV and host tissue factor. Unlike nonmalignant DVT, which is initiated and propagated by innate immune cells, thrombosis triggered by pcMV was largely independent of myeloid leukocytes or platelets. Instead, we identified externalization of the phospholipid phosphatidylethanolamine as a major mechanism controlling the prothrombotic activity of pcMV. Disrupting phosphatidylethanolamine-dependent activation of factor X suppressed pcMV-induced DVT without causing changes in hemostasis. CONCLUSIONS: Together, we show here that the pathophysiology of pcMV-associated experimental DVT differs markedly from innate immune cell-promoted nonmalignant DVT and is therefore amenable to distinct antithrombotic strategies. Targeting phosphatidylethanolamine on tumor microvesicles could be a new strategy for prevention of cancer-associated DVT without causing bleeding complications.


Subject(s)
Adenocarcinoma/complications , Blood Coagulation , Cell-Derived Microparticles/metabolism , Pancreatic Neoplasms/complications , Vena Cava, Inferior/metabolism , Venous Thrombosis/etiology , Adenocarcinoma/blood , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Bacteriocins/pharmacology , Blood Coagulation/drug effects , Cell Line, Tumor , Cell-Derived Microparticles/drug effects , Cell-Derived Microparticles/pathology , Disease Models, Animal , Drug Design , Factor Xa/metabolism , Fibrinolytic Agents/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Peptides/pharmacology , Phosphatidylethanolamines/antagonists & inhibitors , Phosphatidylethanolamines/blood , Signal Transduction , Thromboplastin/metabolism , Vena Cava, Inferior/drug effects , Vena Cava, Inferior/pathology , Venous Thrombosis/blood , Venous Thrombosis/pathology , Venous Thrombosis/prevention & control
7.
PLoS One ; 13(1): e0190728, 2018.
Article in English | MEDLINE | ID: mdl-29293656

ABSTRACT

AIMS: Medical treatment of arterial thrombosis is mainly directed against platelets and coagulation factors, and can lead to bleeding complications. Novel antithrombotic therapies targeting immune cells and neutrophil extracellular traps (NETs) are currently being investigated in animals. We addressed whether immune cell composition of arterial thrombi induced in mouse models of thrombosis resemble those of human patients with acute myocardial infarction (AMI). METHODS AND RESULTS: In a prospective cohort study of patients suffering from AMI, 81 human arterial thrombi were harvested during percutaneous coronary intervention and subjected to detailed histological analysis. In mice, arterial thrombi were induced using two distinct experimental models, ferric chloride (FeCl3) and wire injury of the carotid artery. We found that murine arterial thrombi induced by FeCl3 were highly concordant with human coronary thrombi regarding their immune cell composition, with neutrophils being the most abundant cell type, as well as the presence of NETs and coagulation factors. Pharmacological treatment of mice with the protein arginine deiminase (PAD)-inhibitor Cl-amidine abrogated NET formation, reduced arterial thrombosis and limited injury in a model of myocardial infarction. CONCLUSIONS: Neutrophils are a hallmark of arterial thrombi in patients suffering from acute myocardial infarction and in mouse models of arterial thrombosis. Inhibition of PAD could represent an interesting strategy for the treatment of arterial thrombosis to reduce neutrophil-associated tissue damage and improve functional outcome.


Subject(s)
Disease Models, Animal , Myocardial Infarction/pathology , Ornithine/analogs & derivatives , Thrombosis/pathology , Aged , Animals , Chlorides/administration & dosage , Coronary Vessels/pathology , Female , Ferric Compounds/administration & dosage , Humans , Male , Mice , Middle Aged , Ornithine/pharmacology , Prospective Studies
8.
Cell ; 171(6): 1368-1382.e23, 2017 Nov 30.
Article in English | MEDLINE | ID: mdl-29195076

ABSTRACT

Blood platelets are critical for hemostasis and thrombosis and play diverse roles during immune responses. Despite these versatile tasks in mammalian biology, their skills on a cellular level are deemed limited, mainly consisting in rolling, adhesion, and aggregate formation. Here, we identify an unappreciated asset of platelets and show that adherent platelets use adhesion receptors to mechanically probe the adhesive substrate in their local microenvironment. When actomyosin-dependent traction forces overcome substrate resistance, platelets migrate and pile up the adhesive substrate together with any bound particulate material. They use this ability to act as cellular scavengers, scanning the vascular surface for potential invaders and collecting deposited bacteria. Microbe collection by migrating platelets boosts the activity of professional phagocytes, exacerbating inflammatory tissue injury in sepsis. This assigns platelets a central role in innate immune responses and identifies them as potential targets to dampen inflammatory tissue damage in clinical scenarios of severe systemic infection.


Subject(s)
Bacterial Infections/immunology , Blood Platelets/immunology , Animals , Bacteria/classification , Blood Platelets/cytology , Blood Vessels/injuries , Blood Vessels/pathology , Calcium/metabolism , Cell Movement , Cell Polarity , Humans , Inflammation/immunology , Integrins/metabolism , Mice , Myosins/metabolism , Neutrophils/cytology
9.
J Exp Med ; 214(7): 2121-2138, 2017 Jul 03.
Article in English | MEDLINE | ID: mdl-28566277

ABSTRACT

Blood coagulation is essential for physiological hemostasis but simultaneously contributes to thrombotic disease. However, molecular and cellular events controlling initiation and propagation of coagulation are still incompletely understood. In this study, we demonstrate an unexpected role of eosinophils during plasmatic coagulation, hemostasis, and thrombosis. Using a large-scale epidemiological approach, we identified eosinophil cationic protein as an independent and predictive risk factor for thrombotic events in humans. Concurrent experiments showed that eosinophils contributed to intravascular thrombosis by exhibiting a strong endogenous thrombin-generation capacity that relied on the enzymatic generation and active provision of a procoagulant phospholipid surface enriched in 12/15-lipoxygenase-derived hydroxyeicosatetraenoic acid-phosphatidylethanolamines. Our findings reveal a previously unrecognized role of eosinophils and enzymatic lipid oxidation as regulatory elements that facilitate both hemostasis and thrombosis in response to vascular injury, thus identifying promising new targets for the treatment of thrombotic disease.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Blood Coagulation , Eosinophils/metabolism , Hemostasis , Lipids/analysis , Thrombosis/metabolism , Adult , Aged , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Atherosclerosis/diagnosis , Atherosclerosis/metabolism , Blotting, Western , Cells, Cultured , Eosinophil Cationic Protein/metabolism , Humans , Hydroxyeicosatetraenoic Acids/metabolism , Logistic Models , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Oxidation-Reduction , Phosphatidylethanolamines/metabolism , Prospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors , Thrombin/metabolism
10.
Sci Transl Med ; 8(367): 367ra168, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27903864

ABSTRACT

In patients with atrial fibrillation, oral anticoagulation with oral thrombin inhibitors (OTIs), in contrast to vitamin K antagonists (VKAs), associates with a modest increase in acute coronary syndromes (ACSs). Whether this observation is causatively linked to OTI treatment and, if so, whether OTI action is the result of a lower antithrombotic efficacy of OTI compared to VKA or reflects a yet undefined prothrombotic activity of OTI remain unclear. We analyzed platelet function in patients receiving OTI or dose-adapted VKA under static and flow conditions. In vivo, we studied arterial thrombosis in OTI-, VKA-, and vehicle-treated mice using carotid ligation and wire injury models. Further, we examined thrombus formation on human atherosclerotic plaque homogenates under arterial shear to address the relevance to human pathology. Under static conditions, aggregation in the presence of ristocetin was increased in OTI-treated blood, whereas platelet reactivity and aggregation to other agonists were only marginally affected. Under flow conditions, firm platelet adhesion and thrombus formation on von Willebrand factor, collagen, and human atherosclerotic plaque were increased in the presence of OTI in comparison to VKA. OTI treatment was associated with increased thrombus formation in injured carotid arteries of mice. Inhibition or ablation of GPIbα-thrombin interactions abolished the effect of OTI on thrombus formation, suggesting a mechanistic role of the platelet receptor GPIbα and its thrombin-binding site. The effect of OTI was also abrogated in the presence of aspirin. In summary, OTI treatment has prothrombotic activity that might contribute to the increase in ACS observed clinically in patients.


Subject(s)
Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Thrombin/antagonists & inhibitors , Thrombosis/pathology , Acute Coronary Syndrome/pathology , Administration, Oral , Animals , Anticoagulants/pharmacology , Arteries/pathology , Aspirin/pharmacology , Atherosclerosis/pathology , Binding Sites , Blood Platelets/drug effects , Fibrinolytic Agents/pharmacology , Humans , Mice , Platelet Aggregation Inhibitors/pharmacology , Randomized Controlled Trials as Topic , Vitamin K/antagonists & inhibitors
11.
Blood ; 128(20): 2435-2449, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27574188

ABSTRACT

Deep venous thrombosis (DVT) is one of the most common cardiovascular diseases, but its pathophysiology remains incompletely understood. Although sterile inflammation has recently been shown to boost coagulation during DVT, the underlying molecular mechanisms are not fully resolved, which could potentially identify new anti-inflammatory approaches to prophylaxis and therapy of DVT. Using a mouse model of venous thrombosis induced by flow reduction in the vena cava inferior, we identified blood-derived high-mobility group box 1 protein (HMGB1), a prototypical mediator of sterile inflammation, to be a master regulator of the prothrombotic cascade involving platelets and myeloid leukocytes fostering occlusive DVT formation. Transfer of platelets into Hmgb1-/- chimeras showed that this cell type is the major source of HMGB1, exposing reduced HMGB1 on their surface upon activation thereby enhancing the recruitment of monocytes. Activated leukocytes in turn support oxidation of HMGB1 unleashing its prothrombotic activity and promoting platelet aggregation. This potentiates the amount of HMGB1 and further nurtures the accumulation and activation of monocytes through receptor for advanced glycation end products (RAGE) and Toll-like receptor 2, leading to local delivery of monocyte-derived tissue factor and cytokines. Moreover, disulfide HMGB1 facilitates formation of prothrombotic neutrophil extracellular traps (NETs) mediated by RAGE, exposing additional HMGB1 on their extracellular DNA strands. Eventually, a vicious circle of coagulation and inflammation is set in motion leading to obstructive DVT formation. Therefore, platelet-derived disulfide HMGB1 is a central mediator of the sterile inflammatory process in venous thrombosis and could be an attractive target for an anti-inflammatory approach for DVT prophylaxis.


Subject(s)
Blood Platelets/metabolism , HMGB1 Protein/physiology , Venous Thrombosis/genetics , Animals , Blood Platelets/pathology , Disulfides/chemistry , Disulfides/metabolism , HMGB1 Protein/chemistry , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Receptor for Advanced Glycation End Products/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Venous Thrombosis/metabolism , Venous Thrombosis/pathology
12.
Circ Res ; 117(4): 376-87, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26129975

ABSTRACT

RATIONALE: Platelets are known to play a crucial role in hemostasis. Sphingosine kinases (Sphk) 1 and 2 catalyze the conversion of sphingosine to the bioactive metabolite sphingosine 1-phosphate (S1P). Although platelets are able to secrete S1P on activation, little is known about a potential intrinsic effect of S1P on platelet function. OBJECTIVE: To investigate the role of Sphk1- and Sphk2-derived S1P in the regulation of platelet function. METHODS AND RESULTS: We found a 100-fold reduction in intracellular S1P levels in platelets derived from Sphk2(-/-) mutants compared with Sphk1(-/-) or wild-type mice, as analyzed by mass spectrometry. Sphk2(-/-) platelets also failed to secrete S1P on stimulation. Blood from Sphk2-deficient mice showed decreased aggregation after protease-activated receptor 4-peptide and adenosine diphosphate stimulation in vitro, as assessed by whole blood impedance aggregometry. We revealed that S1P controls platelet aggregation via the sphingosine 1-phosphate receptor 1 through modulation of protease-activated receptor 4-peptide and adenosine diphosphate-induced platelet activation. Finally, we show by intravital microscopy that defective platelet aggregation in Sphk2-deficient mice translates into reduced arterial thrombus stability in vivo. CONCLUSIONS: We demonstrate that Sphk2 is the major Sphk isoform responsible for the generation of S1P in platelets and plays a pivotal intrinsic role in the control of platelet activation. Correspondingly, Sphk2-deficient mice are protected from arterial thrombosis after vascular injury, but have normal bleeding times. Targeting this pathway could therefore present a new therapeutic strategy to prevent thrombosis.


Subject(s)
Blood Platelets/enzymology , Lysophospholipids/blood , Phosphotransferases (Alcohol Group Acceptor)/blood , Platelet Aggregation , Sphingosine/analogs & derivatives , Animals , Arachidonic Acid/blood , Blood Coagulation , Blood Coagulation Tests , Carotid Artery Injuries/blood , Carotid Artery Injuries/enzymology , Disease Models, Animal , Erythrocytes/enzymology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Platelet Adhesiveness , Platelet Function Tests , Receptors, Lysosphingolipid/blood , Signal Transduction , Sphingosine/blood , Sphingosine-1-Phosphate Receptors , Thrombosis/blood , Thrombosis/enzymology , Thrombosis/prevention & control , Thromboxane A2/blood , Vascular System Injuries/blood , Vascular System Injuries/enzymology
13.
J Leukoc Biol ; 95(2): 265-74, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24072879

ABSTRACT

Inflammatory conditions induce redistribution of junctional adhesion receptors toward the apical regions of endothelial cells promoting lymphocyte TEM. Much of the molecular structures of TEM have been revealed; however, the biophysical mechanisms underlying this process remain to be fully elucidated. Here, we used immunofluorescence microscopy and AFM to study endothelial distribution of adhesion molecules upon lymphocyte activation and transmigration. Our immunofluorescence results revealed redistribution of JAM-A and PECAM-1 but not ICAM-1 or VCAM-1 toward the apical junctional regions of HUVECs following a 6-h stimulation with TNF-α and IFN-γ. Consistently, our SCFS studies revealed that Jurkat cell adhesion to stimulated HUVEC monolayers was significantly greater in junctional regions. Enhanced adhesion was mediated mostly by JAM-A receptors. Further AFM adhesion mapping of the homophilic JAM-A/JAM-A interaction on the surfaces of HUVECs revealed a greater number of JAM-A receptors available for binding along junctional regions after TNF-α and IFN-γ stimulation. Our data reveal for the first time that adhesion "hot spots" of JAM-A receptors are involved in initiating lymphocyte TEM under inflammatory conditions.


Subject(s)
Human Umbilical Vein Endothelial Cells/cytology , Intercellular Junctions/metabolism , Interferon-gamma/pharmacology , Lymphocytes/cytology , Transendothelial and Transepithelial Migration/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cell Communication/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intercellular Junctions/drug effects , Jurkat Cells , Lymphocytes/drug effects , Lymphocytes/metabolism , Microscopy, Atomic Force , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptors, Cell Surface/metabolism , Spectrum Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...