Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 57(1): 68-85.e11, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38141610

ABSTRACT

Tissue factor (TF), which is a member of the cytokine receptor family, promotes coagulation and coagulation-dependent inflammation. TF also exerts protective effects through unknown mechanisms. Here, we showed that TF bound to interferon-α receptor 1 (IFNAR1) and antagonized its signaling, preventing spontaneous sterile inflammation and maintaining immune homeostasis. Structural modeling and direct binding studies revealed binding of the TF C-terminal fibronectin III domain to IFNAR1, which restricted the expression of interferon-stimulated genes (ISGs). Podocyte-specific loss of TF in mice (PodΔF3) resulted in sterile renal inflammation, characterized by JAK/STAT signaling, proinflammatory cytokine expression, disrupted immune homeostasis, and glomerulopathy. Inhibiting IFNAR1 signaling or loss of Ifnar1 expression in podocytes attenuated these effects in PodΔF3 mice. As a heteromer, TF and IFNAR1 were both inactive, while dissociation of the TF-IFNAR1 heteromer promoted TF activity and IFNAR1 signaling. These data suggest that the TF-IFNAR1 heteromer is a molecular switch that controls thrombo-inflammation.


Subject(s)
Signal Transduction , Thromboplastin , Animals , Mice , Inflammation , Interferon-alpha , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Thromboplastin/genetics
2.
Psychol Med ; : 1-12, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38450444

ABSTRACT

BACKGROUND: Physical sequelae of anorexia nervosa (AN) include a marked reduction in whole brain volume and subcortical structures such as the hippocampus. Previous research has indicated aberrant levels of inflammatory markers and growth factors in AN, which in other populations have been shown to influence hippocampal integrity. METHODS: Here we investigated the influence of concentrations of two pro-inflammatory cytokines (tumor necrosis factor-alpha [TNF-α] and interleukin-6 [IL-6]) and brain-derived neurotrophic factor (BDNF) on the whole hippocampal volume, as well as the volumes of three regions (the hippocampal body, head, and tail) and 18 subfields bilaterally. Investigations occurred both cross-sectionally between acutely underweight adolescent/young adult females with AN (acAN; n = 82) and people recovered from AN (recAN; n = 20), each independently pairwise age-matched with healthy controls (HC), and longitudinally in acAN after partial renourishment (n = 58). Hippocampal subfield volumes were quantified using FreeSurfer. Concentrations of molecular factors were analyzed in linear models with hippocampal (subfield) volumes as the dependent variable. RESULTS: Cross-sectionally, there was no evidence for an association between IL-6, TNF-α, or BDNF and between-group differences in hippocampal subfield volumes. Longitudinally, increasing concentrations of BDNF were positively associated with longitudinal increases in bilateral global hippocampal volumes after controlling for age, age2, estimated total intracranial volume, and increases in body mass index (BMI). CONCLUSIONS: These findings suggest that increases in BDNF may contribute to global hippocampal recovery over and above increases in BMI during renourishment. Investigations into treatments targeted toward increasing BDNF in AN may be warranted.

3.
J Cardiovasc Pharmacol ; 77(3): 378-385, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33662980

ABSTRACT

ABSTRACT: The calcium sensitizer levosimendan is indicated for the hemodynamic stabilization of patients with acutely decompensated heart failure and has been shown to be protective against reperfusion injury after myocardial infarction. However, affected forms of cell death and underlying signaling pathways remain controversial. Therefore, the aim of this study was to examine the influence of levosimendan preconditioning and postconditioning on anoxia/reoxygenation-induced apoptosis, necrosis, and autophagy in H9c2 myoblasts. To mimic conditions of myocardial ischemia/reperfusion, rat cardiac H9c2 myoblasts were exposed to anoxia/starvation, followed by reoxygenation/refeeding. Apoptosis, necrosis, autophagy, cell viability, survival signaling, and mitochondrial permeability transition pore (mPTP) opening were measured. Both, pharmacological preconditioning and postconditioning with levosimendan were capable to reduce apoptosis as well as necrosis in stressed H9c2 cells. However, preconditioning showed to have the stronger impact compared with postconditioning. Moreover, levosimendan preconditioning increased autophagy, suggesting enhanced repair processes initiated by the early presence of the drug. Underlying mechanisms differ between both interventions: Although both are associated with PI3/Akt activation and reduced mPTP opening, only postconditioning but not preconditioning depended on mKATP activation. This variation might indicate that a pharmacological treatment after the onset of reoxygenation at least in part directly addresses mitochondrial structures for protection. In conclusion, we demonstrate that both pharmacological preconditioning and postconditioning with levosimendan protect anoxia/reoxygenation-stressed cells but differ in the underlying mechanisms. These results are decisive to obtain more insights into the beneficial effects of levosimendan in the treatment of reperfusion-mediated damage.


Subject(s)
Cardiovascular Agents/pharmacology , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Simendan/pharmacology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Hypoxia , Cell Line , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondrial Permeability Transition Pore/metabolism , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Necrosis , Potassium Channels/metabolism , Rats , Signal Transduction
4.
Arterioscler Thromb Vasc Biol ; 39(6): 1137-1148, 2019 06.
Article in English | MEDLINE | ID: mdl-31070476

ABSTRACT

Objective- Pathological angiogenesis, such as exuberant retinal neovascularization during proliferative retinopathies, involves endothelial responses to ischemia/hypoxia and oxidative stress. Autophagy is a clearance system enabling bulk degradation of intracellular components and is implicated in cellular adaptation to stressful conditions. Here, we addressed the role of the ATG5 (autophagy-related protein 5) in endothelial cells in the context of pathological ischemia-related neovascularization in the murine model of retinopathy of prematurity. Approach and Results- Autophagic vesicles accumulated in neovascular tufts of the retina of retinopathy of prematurity mice. Endothelium-specific Atg5 deletion reduced pathological neovascularization in the retinopathy of prematurity model. In contrast, no alterations in physiological retina vascularization were observed in endothelial-specific ATG5 deficiency, suggesting a specific role of endothelial ATG5 in pathological hypoxia/reoxygenation-related angiogenesis. Consistently, in an aortic ring angiogenesis assay, endothelial ATG5 deficiency resulted in impaired angiogenesis under hypoxia/reoxygenation conditions. As compared to ATG5-sufficient endothelial cells, ATG5-deficient cells displayed impaired mitochondrial respiratory activity, diminished production of mitochondrial reactive oxygen species and decreased phosphorylation of the VEGFR2 (vascular endothelial growth factor receptor 2). Consistently, ATG5-deficient endothelial cells displayed decreased oxidative inactivation of PTPs (phospho-tyrosine phosphatases), likely due to the reduced reactive oxygen species levels resulting from ATG5 deficiency. Conclusions- Our data suggest that endothelial ATG5 supports mitochondrial function and proangiogenic signaling in endothelial cells in the context of pathological hypoxia/reoxygenation-related neovascularization. Endothelial ATG5, therefore, represents a potential target for the treatment of pathological neovascularization-associated diseases, such as retinopathies.


Subject(s)
Autophagy-Related Protein 5/deficiency , Endothelial Cells/metabolism , Neovascularization, Pathologic , Retinal Vessels/metabolism , Retinopathy of Prematurity/metabolism , Animals , Autophagy-Related Protein 5/genetics , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Humans , Mice, Knockout , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Stress , Phosphorylation , Reactive Oxygen Species/metabolism , Retinal Vessels/pathology , Retinopathy of Prematurity/genetics , Retinopathy of Prematurity/pathology , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism
5.
Euro Surveill ; 25(25)2020 06.
Article in English | MEDLINE | ID: mdl-32613938

ABSTRACT

We investigated why a clinical meticillin-resistant Staphylococcus aureus (MRSA) isolate yielded false-negative results with some commercial PCR tests for MRSA detection. We found that an epidemic European CC1-MRSA-IV clone generally exhibits this behaviour. The failure of the assays was attributable to a large insertion in the orfX/SCCmec integration site. To ensure the reliability of molecular MRSA tests, it is vital to monitor emergence of new SCCmec types and junction sites.


Subject(s)
Methicillin-Resistant Staphylococcus aureus/isolation & purification , Staphylococcal Infections/diagnosis , Austria/epidemiology , False Negative Reactions , Female , Germany/epidemiology , Humans , Ireland/epidemiology , Methicillin-Resistant Staphylococcus aureus/genetics , Middle Aged , Staphylococcal Infections/epidemiology
6.
Biochim Biophys Acta Mol Basis Dis ; 1864(8): 2526-2537, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29777903

ABSTRACT

Vascular smooth muscle cells (VSMC) exhibit a dual role in progression and maintenance of arteriosclerosis. They are fundamental for plaque stability but also can drive plaque progression. During pathogenic vascular remodeling, VSMC transdifferentiate into a phenotype with enhanced proliferation and migration. Moreover, they exert an increased capacity to generate extracellular matrix proteins. A special lineage of transdifferentiated VSMC expresses Sox9, a multi-functional transcription factor. The aim of the study was to examine the role of Sox9 in phenotypic alterations leading to arteriosclerosis. Using mouse models for arterial stenosis, Sox9 induction in diseased vessels was verified. The phenotypic switch of VSMC from contractile to proliferative nature caused a significant increase of Sox9 expression. Various factors known to be involved in the progression of arteriosclerosis were examined for their ability to modulate Sox9 expression in VSMC. While PDGF-BB resulted in a strong transient upregulation of Sox9, TGF-ß1 appeared to be responsible for a moderate, but prolonged increase of Sox9 expression. Beside the regulation, functional studies focused on knockout and overexpression of Sox9. A Sox9-dependent alteration of extracellular matrix could be revealed and was associated with an upregulated calcium deposition. Taken together, Sox9 is identified as important factor of VSMC function by modulation the extracellular matrix composition and calcium deposition, which are important processes in plaque development.


Subject(s)
Arteriosclerosis/metabolism , Extracellular Matrix/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic/metabolism , SOX9 Transcription Factor/metabolism , Vascular Calcification/metabolism , Animals , Arteriosclerosis/genetics , Arteriosclerosis/pathology , Disease Models, Animal , Extracellular Matrix/genetics , Mice , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology , SOX9 Transcription Factor/genetics , Vascular Calcification/genetics , Vascular Calcification/pathology
7.
Cell Tissue Res ; 374(3): 473-485, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30159755

ABSTRACT

Pheochromocytomas and paragangliomas (PGLs) due to mutations of succinate dehydrogenase (SDH) B, a subunit of the SDH complex with a role in the Krebs cycle and the respiratory chain, tend to be larger at diagnosis and more prone to metastatic disease than other tumors. This presentation contrasts with the behavior of some cell line models of SDHB impairment, which show reduced growth compared to wild type. We hypothesize that reduced growth of SDHB-impaired monolayer culture models might reflect lack of support from sources within the tumor microenvironment. The present study therefore investigates how the microenvironment, modeled here by fibroblast co-culture, modulates cell metabolism, growth and invasion in an Sdhb-impaired mouse pheochromocytoma cell line. We employed two different constructs of short hairpin RNA to knockdown Sdhb and compared growth in a monolayer with and without fibroblast co-culture. Sdhb-silenced cells showed functional impairment of SDH with elevated succinate to fumarate ratio and decreased oxidative capacity. Cell growth was delayed with an increase in doubling time of 2 h or 20 h. Clonogenic cell survival and viability, on the other hand, were either unchanged or increased compared to control. In standard monolayer culture, no differences in pro-metastatic features were present. Co-culture with primary mouse fibroblast reversed the difference of proliferation between control and Sdhb knockdown but was unable to significantly influence invasiveness under these culture conditions. Metabolic studies identified that lactate secreted by fibroblasts was taken up preferentially by Sdhb-silenced cells. In summary, the present study identified a potential role for the tumor microenvironment in influencing phenotypic features of SDHB-mutated PGLs, providing a basis for the use of therapies targeted towards the tumor microenvironment.


Subject(s)
Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Fibroblasts/metabolism , Pheochromocytoma/metabolism , Pheochromocytoma/pathology , Succinate Dehydrogenase/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Coculture Techniques , Gene Knockdown Techniques , Gene Silencing , Mice , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Succinate Dehydrogenase/genetics
8.
Biomarkers ; 23(1): 78-87, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29098879

ABSTRACT

BACKGROUND: Chromogranin B (CGB) regulates B-type natriuretic peptide (BNP) production. Circulating CGB levels are elevated in heart failure (HF) animal models and HF patients, but also increase in healthy individuals in response to physical activity. Therefore, CGB seems to integrate information from myocardial stress and systemic neuro-endocrine activation. Substantial gaps remain in our understanding of CGB regulation in HF. METHODS AND RESULTS: We conducted a retrospective registry study including 372 patients. CGB and N-terminal pro-BNP (NT-proBNP) plasma levels were assessed in acute HF and chronic valvular HF patients and controls. CGB levels were significantly increased in acute HF and chronic valvular HF, but significantly higher in the latter. Patients in chronic valvular HF with severe mitral regurgitation (cHF-MR) showed significantly higher CGB levels than patients in chronic valvular HF with severe aortic stenosis. CGB levels progressively increased with worsening NYHA functional status and were moderately correlated to NT-proBNP, but independent of left ventricular (LV) ejection fraction (LVEF), LV mass, age and body weight. Finally, cHF-MR patients showed significant reductions of CGB levels after interventional mitral valve repair. CONCLUSION: CGB is a promising emerging biomarker in HF patients with unique potential to integrate information from myocardial stress and neuro-endocrine activation.


Subject(s)
Biomarkers/blood , Chromogranin B/blood , Heart Failure/blood , Mitral Valve Insufficiency/blood , Aged , Aged, 80 and over , Chronic Disease , Female , Heart Failure/complications , Heart Failure/diagnosis , Humans , Male , Middle Aged , Mitral Valve Insufficiency/complications , Mitral Valve Insufficiency/diagnosis , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Retrospective Studies , Sensitivity and Specificity
9.
Europace ; 20(3): 459-465, 2018 03 01.
Article in English | MEDLINE | ID: mdl-28073885

ABSTRACT

Aims: It is hypothesized that inflammation could promote structural and electrical remodelling processes in atrial fibrillation (AF). Atrial infiltration of monocytes and granulocytes has been shown to be dependent on CD11b expression. The aim of this study was to investigate whether treatment of AF by pulmonary vein isolation (PVI) may lead to reduced inflammation, as indicated by a decrease of CD11b expression on monocytes and granulocytes. Methods and results: Flow-cytometric quantification analysis and determination of systemic inflammatory markers of peripheral blood were performed in 75 patients undergoing PVI 1 day before and 6 months after PVI. The extent of activation of monocytes and granulocytes was measured by quantifying the cell adhesion molecule CD11b. The mean expression of CD11b on monocytes (20.9 ± 2.5 vs. 10.2 ± 1.4; P < 0.001) and granulocytes (13.9 ± 1.6 vs. 6.8 ± 0.5; P < 0.001), as well as the relative count of CD11b-positive monocytes (P < 0.05) and CD11b-positive granulocytes (P < 0.01) were significantly reduced when comparing the identical patients before and 6 months after PVI. Systemic inflammatory parameters showed only a declining tendency after 6 months. Patients with unsuccessful PVI and ongoing AF on the day of follow-up showed no decrease in CD11b expression. Conclusions: A significant reduction of CD11b expression on monocytes and granulocytes, as a sign of reduced cellular inflammation, was achieved by treatment of AF using PVI. These data strongly support that AF is not only a consequence of but also a cause for inflammatory processes, which, in turn, may contribute to atrial remodelling.


Subject(s)
Atrial Fibrillation/surgery , CD11b Antigen/metabolism , Catheter Ablation , Granulocytes/metabolism , Inflammation Mediators/metabolism , Monocytes/metabolism , Pulmonary Veins/surgery , Action Potentials , Aged , Atrial Fibrillation/immunology , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Atrial Remodeling , CD11b Antigen/immunology , Catheter Ablation/adverse effects , Down-Regulation , Female , Granulocytes/immunology , Heart Rate , Humans , Inflammation Mediators/immunology , Male , Middle Aged , Monocytes/immunology , Pulmonary Veins/physiopathology , Risk Factors , Time Factors , Treatment Outcome
10.
Platelets ; 29(1): 41-47, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28287031

ABSTRACT

Atrial fibrillation (AF) is well known for being a major risk factor of thromboembolic stroke. We could recently demonstrate an association of monocyte-platelet aggregates (MPAs) with the degree of thrombogenicity in patients with AF. This study investigated platelet activation markers, as potential biomarkers for the presence of left atrial (LA) thrombus in patients with AF. One hundred and eight patients with symptomatic AF underwent transesophageal echocardiography (TEE) before scheduled cardioversion or pulmonary vein isolation. In order to determine the content of MPAs by flow-cytometric quantification analyses, blood was drawn on the day of TEE. The soluble CD40 Ligand (sCD40L) and soluble P-selectin (sP-selectin) were obtained by Cytometric Bead Arrays (CBA). D-dimer levels were detected by quantitative immunological determination of fibrin degradation products. Clinical, laboratory, and echocardiographic standard parameters were obtained from all patients, including the determination of the flow in the left atrial appendage (LAA). Patients with detected LA thrombus (n = 28) compared with patients without thrombus (n = 80) showed an increased number of common risk factors, such as age, diabetes, heart failure, and coronary artery disease (CAD). The presence of LA thrombus was associated with significantly increased levels of MPAs (147 ± 12 vs. 304 ± 29 per µl; p < 0.00), sCD40L (106.3 ± 31.0 vs. 33.5 ± 2.1 pg/ml, p = 0.027), and D-dimer (0.13 ± 0.02 vs. 0.69 ± 0.21 mg FEU/l, p = 0.015). In contrast, sP-selectin showed no association with LA thrombus. A multivariate regression analysis showed that MPAs, sCD40L as well as D-dimers were independent indicators for the existence of LA thrombus. MPAs above 170 cells/µl indicated LA thrombus with a high sensitivity of 93% and a specificity of 73% (OR 62, 95% CI. 6.9-557.2, p < 0.001) in patients with AF, whereas the D-dimer lost their quality as independent indicator by using the conventional cut-off of 0.5 mg/l within the regression analysis. MPAs, as well as the D-dimer, correlated significantly negatively with the flow in the LAA measured during TEE. The content of MPAs, sCD40L, and D-dimer, but not sP-selectin showed an increased dependence on LA thrombus in patients with AF. In our study group, MPAs showed the best diagnostic test accuracy of the compared platelet markers. The different results of the examined platelet activation markers could be an indication of diverse mechanisms of LA thrombus in AF. Further studies should evaluate whether determination of MPAs in clinical routine may suffice to indicate the presence of LA thrombus in patients with AF.


Subject(s)
Atrial Fibrillation/complications , Blood Platelets/metabolism , Heart Diseases/diagnosis , Heart Diseases/etiology , Platelet Activation , Thrombosis/diagnosis , Thrombosis/etiology , Aged , Aged, 80 and over , Atrial Fibrillation/etiology , Biomarkers , CD40 Ligand/metabolism , Echocardiography, Transesophageal , Female , Fibrin Fibrinogen Degradation Products , Heart Atria/pathology , Heart Atria/physiopathology , Humans , Male , Middle Aged , P-Selectin/blood , Platelet Aggregation , ROC Curve
11.
Biochim Biophys Acta Mol Basis Dis ; 1863(2): 529-536, 2017 02.
Article in English | MEDLINE | ID: mdl-27915034

ABSTRACT

Endothelial dysfunction is crucial in the initiation of atherosclerosis, which is associated with a lack of nitric oxide. The endothelial NO synthase (eNOS) is responsible for constitutive synthesis of NO and inhibited by caveolin-1 (Cav1). In the current study, we examined the influence on intima formation through single and combined deletion of eNOS and Cav1 with a focus on differentiation of local and systemic effects. A sex-mismatch transplantation of denudated aortae from female C57BL/6n (WT), Cav1-/-, eNOS-/- and Cav1-/-/eNOS-/- (C/e--/--) mice in common carotid artery of male WT mice was performed. After six weeks on Western-type diet, the aortae were explanted and intimal lesions were quantified by determining the intima-media-ratio (IMR). Significantly larger plaques were observed in all knockout mice compared to WT. The highest IMR was detected in Cav1-/- arteries associated with an increased expression of α-smooth muscle actin (αSMA) and the proliferating cell nuclear antigen (PCNA). Both were reduced in aortae from C/e--/--. Galectin-3 (Gal3) immunostaining revealed only small infiltrations of macrophages. Systemic cell invasion was detected by Y chromosome fluorescence in situ hybridization (Y-FISH), which showed only small numbers of systemic cells and no differences between the genotypes. Loss of Cav1 increased vascular lesion by enhancing neointimal proliferation. The combined loss of Cav1 and eNOS, compared to Cav1-/-, lowered intima formation, suggesting an increasing effect of eNOS in the absence of Cav1 on vascular lesion. Furthermore, these effects seem to be mediated by local cells rather than by systemically invaded ones.


Subject(s)
Aorta/transplantation , Carotid Artery, Common/surgery , Carotid Stenosis/etiology , Caveolin 1/genetics , Neointima/etiology , Nitric Oxide Synthase Type III/genetics , Vascular Grafting/adverse effects , Animals , Aorta/pathology , Carotid Artery, Common/pathology , Carotid Stenosis/genetics , Carotid Stenosis/pathology , Female , Gene Knockout Techniques , Male , Mice , Mice, Inbred C57BL , Neointima/genetics , Neointima/pathology , Tunica Intima/pathology , Vascular Grafting/methods
12.
Platelets ; 28(4): 394-399, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27736274

ABSTRACT

Atrial fibrillation (AF) is known to cause platelet activation. AF and its degree of thrombogenesis could be associated with monocyte-platelet aggregates (MPAs). We investigated on whether the content of MPAs or other platelet activation markers is associated with the recurrence of AF after pulmonary vein isolation (PVI). A total of 73 patients with symptomatic AF underwent PVI. After 6 months, all patients were evaluated for episodes of AF recurrence. At the same time, flow-cytometric quantification analyses were performed to determine the content of MPAs. Further platelet activation parameters were detected by using either cytometric bead arrays or quantitative immunological determination. Patients with recurrent AF (n = 20) compared to individuals without AF relapse (n = 53) were associated with an increased content of MPAs (43 ± 3% vs. 33 ± 2%, p = 0.004), as well as an increased CD41 expression on monocytes (191 ± 20 vs. 113 ± 6, p = 0.001). The level of the soluble platelet activation markers such as D-dimer, sCD40L, and sP-selectin did not differ between these groups. The content of MPAs correlated weakly with the level of sCD40L (r = 0.26, p = 0.03), but not with sP-selectin and D-dimer, whereas sP-selectin and sCD40L correlated with each other (r = 0.38, p = 0.001). Only the cellular marker of platelet activation, the content of MPAs, was increased in patients with recurrent AF after PVI. In contrast, soluble markers remained unaltered. These data indicate a distinct mechanism and level of platelet activation in AF. The clinical relevance of MPAs in identifying AF recurrence or in guiding the therapy with anticoagulants remains to be elucidated.


Subject(s)
Atrial Fibrillation/etiology , Platelet Activation/physiology , Pulmonary Veins/physiopathology , Aged , Female , Humans , Male , Middle Aged , Recurrence
13.
Blood ; 121(8): 1436-45, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23264599

ABSTRACT

Erythropoiesis must be tightly balanced to guarantee adequate oxygen delivery to all tissues in the body. This process relies predominantly on the hormone erythropoietin (EPO) and its transcription factor hypoxia inducible factor (HIF). Accumulating evidence suggests that oxygen-sensitive prolyl hydroxylases (PHDs) are important regulators of this entire system. Here, we describe a novel mouse line with conditional PHD2 inactivation (cKO P2) in renal EPO producing cells, neurons, and astrocytes that displayed excessive erythrocytosis because of severe overproduction of EPO, exclusively driven by HIF-2α. In contrast, HIF-1α served as a protective factor, ensuring survival of cKO P2 mice with HCT values up to 86%. Using different genetic approaches, we show that simultaneous inactivation of PHD2 and HIF-1α resulted in a drastic PHD3 reduction with consequent overexpression of HIF-2α-related genes, neurodegeneration, and lethality. Taken together, our results demonstrate for the first time that conditional loss of PHD2 in mice leads to HIF-2α-dependent erythrocytosis, whereas HIF-1α protects these mice, providing a platform for developing new treatments of EPO-related disorders, such as anemia.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hematopoiesis, Extramedullary/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Polycythemia/genetics , Procollagen-Proline Dioxygenase/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain/physiology , Cells, Cultured , Erythropoietin/genetics , Erythropoietin/metabolism , Female , Fibroblasts/cytology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases , Keratinocytes/cytology , Kidney/cytology , Kidney/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Polycythemia/metabolism , Polycythemia/pathology , Procollagen-Proline Dioxygenase/metabolism , Severity of Illness Index , Thrombocytopenia/genetics , Thrombocytopenia/metabolism , Thrombocytopenia/pathology
14.
Scand Cardiovasc J ; 49(6): 361-6, 2015.
Article in English | MEDLINE | ID: mdl-26287645

ABSTRACT

OBJECTIVE: The soluble form of ST2 (sST2) is a novel laboratory parameter for cardiac risk prediction, and over the past years, several studies have tried to evaluate its utility, especially in the management of heart failure. We investigated whether increased serum levels of sST2 show a characteristic pathomorphologic pattern in 3-Tesla cardiac magnetic resonance imaging (CMRI). METHODS: One hundred and fifty-six patients referred to 3T CMRI due to suspected coronary artery disease (CAD) or myocarditis were prospectively enrolled in the study. Ninety patients were diagnosed with CAD, 22 patients with myocarditis, and 44 patients, who constituted the reference group, showed no pathologic CMRI pattern. RESULTS: There was no significant difference between the sST2 values for patients in the reference group and patients with CAD or myocarditis. The sST2 concentration showed a weak correlation with the NYHA functional class (P = 0.002, r = 0.22), but correlation of sST2 and LGE, left ventricular parameters, and LVEF could not be seen. In contrast NT-proBNP was positively correlated to left ventricular parameters, LGE, and NYHA class function (P < 0.05). Additionally, it showed an inverse relationship to LVEF (P < 0.001, r = - 0.42). CONCLUSIONS: Soluble ST2 is not able to detect myocardial scar and should not be used alone as a parameter for detection of inflammation and myocardial scar formation.


Subject(s)
Coronary Artery Disease/blood , Coronary Artery Disease/pathology , Magnetic Resonance Imaging , Myocarditis/blood , Myocarditis/pathology , Myocardium/metabolism , Myocardium/pathology , Receptors, Cell Surface/blood , Adult , Aged , Biomarkers/blood , Case-Control Studies , Cicatrix/blood , Cicatrix/pathology , Coronary Artery Disease/physiopathology , Female , Fibrosis , Humans , Interleukin-1 Receptor-Like 1 Protein , Male , Middle Aged , Myocarditis/physiopathology , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Predictive Value of Tests , Prospective Studies , Stroke Volume , Up-Regulation , Ventricular Function, Left
15.
J Mol Cell Cardiol ; 77: 125-35, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25451169

ABSTRACT

The ligand ephrin A1 is more often discussed to play a role in the development of the atherosclerotic plaque and in this context especially in the monocyte adhesion to endothelial cells. As tumor necrosis factor-α (TNF-α) is known to induce monocyte adhesion to endothelium and ephrin A1 expression, the present study focuses on the involvement of ephrin A1 in TNF-α-mediated monocyte adhesion. The analysis of different members of the Eph/ephrin system in TNF-α-treated human umbilical vein endothelial cells (HUVEC) revealed that especially ephrinA1 was found to be highly regulated by TNF-α compared to other members of the Eph family. This effect is also present in arterial endothelial cells from the umbilical artery and from the coronary artery. This regulation is dependent on NFκB-activation as shown by the expression of a constitutive-active IκB-mutant. By using siRNA-mediated silencing and adenoviral overexpression of ephrinA1 in HUVEC, the involvement of ephrinA1 in the TNF-α triggered monocyte adhesion to endothelial cells could be demonstrated. In addition, these results could be verified by quantitative adhesion measurement using atomic force microscopy-based single-cell force spectroscopy and under flow conditions. Furthermore, this effect is mediated via the EphA4 receptor. EphrinA1 does not influence the mRNA or protein expression of the adhesion receptors VCAM-1 and ICAM-1 in endothelial cells. However, the surface presentation of these adhesion receptors is modulated in an ephrinA1-dependent manner. In conclusion, these data demonstrate that ephrinA1 plays an important role in the TNF-α-mediated adhesion of monocytes to endothelial cells, which might be of great importance in the context of atherosclerosis.


Subject(s)
Ephrin-A1/physiology , Human Umbilical Vein Endothelial Cells/physiology , Monocytes/physiology , Tumor Necrosis Factor-alpha/physiology , Cell Adhesion , Cell Line , Endothelium, Vascular/pathology , Humans , Intercellular Adhesion Molecule-1/metabolism , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
16.
Biochim Biophys Acta ; 1833(10): 2201-11, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23707953

ABSTRACT

The Eph receptors represent the largest family of receptor tyrosine kinases. Both Eph receptors and their ephrin ligands are cell-surface proteins, and they typically mediate cell-to-cell communication by interacting at sites of intercellular contact. The major aim of the present study was to investigate the involvement of EphA4-ephrin-A1 interaction in monocyte adhesion to endothelial cells, as this process is a crucial step during the initiation and progression of the atherosclerotic plaque. Immunohistochemical analysis of human atherosclerotic plaques revealed expression of EphA4 receptor and ephrin-A1 ligand in major cell types within the plaque. Short-time stimulation of endothelial cells with the soluble ligand ephrin-A1 leads to a fourfold increase in adhesion of human monocytes to endothelial cells. In addition, ephrin-A1 further increases monocyte adhesion to already inflamed endothelial cells. EphrinA1 mediates its effect on monocyte adhesion via the activated receptor EphA4. This ephrinA1/EphA4 induced process involves the activation of the Rho signaling pathway and does not require active transcription. Rho activation downstream of EphA4 leads to increased polymerization of actin filaments in endothelial cells. This process was shown to be crucial for the proadhesive effect of ephrin-A1. The results of the present study show that ephrin-A1-induced EphA4 forward signaling promotes monocyte adhesion to endothelial cells via activation of RhoA and subsequent stress-fiber formation by a non-transcriptional mechanism.


Subject(s)
Atherosclerosis/metabolism , Cell Adhesion , Endothelium, Vascular/metabolism , Ephrin-A1/metabolism , Ephrin-A4/metabolism , Monocytes/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Blotting, Western , Cell Proliferation , Cells, Cultured , Endothelium, Vascular/cytology , Ephrin-A1/antagonists & inhibitors , Ephrin-A1/genetics , Ephrin-A4/antagonists & inhibitors , Ephrin-A4/genetics , Flow Cytometry , Humans , Immunoenzyme Techniques , Immunoprecipitation , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
17.
Biochim Biophys Acta ; 1833(12): 3396-3404, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24135056

ABSTRACT

The composition of the hematopoietic stem cell (HSC) niche within the bone marrow is highly dynamic, tightly regulated, and of importance for various HSC properties. Integrins are important molecules within this niche that influence those properties through the interactions of HSCs and mesenchymal stem cells (MSCs). Here we investigated the function of miR-134 in integrin regulation in MSCs. In MSCs, miR-134 post-transcriptionally regulated ß1 integrin expression. This negative regulation of ß1 integrin was mediated by the binding of miR-134 to its 3' untranslated region, which contains two conserved binding sites for miR-134. The miR-134-mediated silencing of ß1 integrin in MSCs was shown by atomic force microscopy to decrease the adhesion of 32D cells to MSCs transfected with miR-134. Furthermore, the adhesion of MSCs to fibronectin was reduced after transfection with miR-134. MSCs from patients with myelodysplastic syndrome (MDS) revealed highly significant miR-134 overexpression compared with MSCs from healthy bone marrow donors. MSCs from MDS patients showed lower ß1 integrin protein, but not lower mRNA, expression, suggesting post-transcriptional regulation. The present study demonstrates miR-134-mediated negative regulation of ß1 integrin that influences cell adhesion to and of MSCs. These results further contribute to our understanding of the complexity of MDS.


Subject(s)
Integrin beta1/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions/genetics , Adult , Aged , Aged, 80 and over , Base Sequence , Binding Sites , Cell Adhesion/genetics , Gene Expression Regulation , HeLa Cells , Humans , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , Middle Aged , Molecular Sequence Data , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Protein Binding/genetics , Transfection , Young Adult
18.
Haematologica ; 99(6): 997-1005, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24584347

ABSTRACT

The chemokine CXCL12 regulates the interaction between hematopoietic stem and progenitor cells and bone marrow stromal cells. Although its relevance in the bone marrow niche is well recognized, the regulation of CXCL12 by microRNA is not completely understood. We transfected a library of 486 microRNA in the bone marrow stromal cell line SCP-1 and studied the expression of CXCL12. Twenty-seven microRNA were shown to downregulate expression of CXCL12. Eight microRNA (miR-23a, 130b, 135, 200b, 200c, 216, 222, and 602) interacted directly with the 3'UTR of CXCL12. Next, we determined that only miR-23a is predicted to bind to the 3'UTR and is strongly expressed in primary bone marrow stromal cells. Modulation of miR-23a changes the migratory potential of hematopoietic progenitor cells in co-culture experiments. We discovered that TGFB1 mediates its inhibitory effect on CXCL12 levels by upregulation of miR-23a. This process was partly reversed by miR-23a molecules. Finally, we determined an inverse expression of CXCL12 and miR-23a in stromal cells from patients with myelodys-plastic syndrome indicating that the interaction has a pathophysiological role. Here, we show for the first time that CXCL12-targeting miR23a regulates the functional properties of the hematopoietic niche.


Subject(s)
Chemokine CXCL12/genetics , Gene Expression Regulation , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , RNA Interference , RNA Processing, Post-Transcriptional , Cell Line , Gene Expression , Humans , Myelodysplastic Syndromes/genetics , RNA, Messenger/genetics , Reproducibility of Results , Transfection
19.
Article in English | MEDLINE | ID: mdl-38461330

ABSTRACT

Previous studies of brain structure in anorexia nervosa (AN) have reported reduced gray matter in underweight patients, which largely normalizes upon weight gain. One underlying biological mechanism may be glial cell alterations related to low-grade inflammation. Here, we investigated relationships between brain structure as measured by magnetic resonance imaging and serum concentrations of two pro-inflammatory cytokines (interleukin-6 and tumor necrosis factor alpha) cross-sectionally in 82 underweight adolescent and young adult female patients (mean age 16.8 years; 59 of whom were observed longitudinally after short-term weight restoration; mean duration 2.8 months), 20 individuals long-term weight-recovered from AN (mean age 22.7 years) and 105 healthy control (HC) participants (mean age 17.2 years). We measured cortical thickness, subcortical volumes and local gyrification index, a measure of cortical folding. In contrast to most previous studies of cytokine concentrations in AN, we found no cross-sectional group differences (interleukin-6: p = 0.193, tumor necrosis factor alpha: p = 0.057) or longitudinal changes following weight restoration (interleukin-6: p = 0.201, tumor necrosis factor alpha: p = 0.772). As expected, widespread gray matter reductions (cortical thickness, subcortical volumes, cortical folding) were observed in underweight patients with AN compared to HC. However, we found no evidence of associations between cytokine concentrations and structural brain measures in any participant group. Furthermore, longitudinal changes in cytokine concentrations were unrelated to changes in gray matter. In conclusion, we did not identify any association between (sub-)inflammatory processes and structural brain changes in AN. Future studies are needed to elucidate which other factors besides nutritional status may contribute to brain morphological alterations.

20.
Basic Res Cardiol ; 108(6): 390, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24121466

ABSTRACT

Availability of adiponectin plays a crucial role in cardiovascular function. The present study was conducted to evaluate the presence, alterations and impact of the various adiponectin isoforms in vascular tissue under clinically relevant in vitro conditions (cold storage). Presence of various adiponectin isoforms in vascular smooth muscle cells and their regulation during cold storage was evaluated by PCR, western blot, ELISA and immunohistochemistry. The impact of the various isoforms for vessel preservation was assessed using isometric force measurement as an in vitro assay for vascular function. Adiponectin is expressed in smooth muscle cells from murine aortae and human saphenous veins. Following 2 days of cold storage adiponectin mRNA expression in mouse aorta is reduced, which appears to be regulated indirectly by miR-292-3p. Despite the reduced mRNA expression, adiponectin accumulated in cold storage supernatant over 2 days indicating a net release of adiponectin. Two days of cold storage resulted in an impairment of endothelium-dependent relaxation which was prevented by addition of full-length adiponectin in concentrations similar to normal plasma levels during storage. In contrast, addition of recombinant adiponectin which is unable to form high order multimers failed to improve vessel function. High concentrations (20 µg/mL) of this trimeric isoform even reduced the vasorelaxation response and facilitated uncoupling of endothelial nitric oxide synthase. Endothelial injury by cold storage may partly be prevented by addition of high-molecular-weight adiponectin. This effect may support graft patency to avoid coagulation- and atherosclerosis-associated impairment of perfusion.


Subject(s)
Adiponectin/metabolism , Aorta/metabolism , Cryopreservation , Muscle, Smooth, Vascular/metabolism , Animals , Aorta/transplantation , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Protein Isoforms , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Transplants , Vasodilation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL