Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(12): e0278895, 2022.
Article in English | MEDLINE | ID: mdl-36520838

ABSTRACT

During tissue regeneration, mesenchymal stem cells can support endothelial cells in the process of new vessel formation. For a functional interaction of endothelial cells with mesenchymal stem cells a vascular inductive microenvironment is required. Using a cellular model for neo-vessel formation, we could show that newly formed vascular structures emanated from the embedded aggregates, consisting of mesenchymal stem cells co-cultured with autologous human umbilical vein endothelial cells, into avascular human platelet lysate-based matrices, bridging distances up to 5 mm to join with adjacent aggregates with the same morphology forming an interconnected network. These newly formed vascular sprouts showed branch points and generated a lumen, as sign of mature vascular development. In two-dimensional culture, we detected binding of mesenchymal stem cells to laser-damaged endothelial cells under flow conditions, mimicking the dynamics in blood vessels. In conclusion, we observed that mesenchymal stem cells can support human umbilical vein endothelial cells in their vitality and functionality. In xeno-free human platelet lysate-based matrices, endothelial cells form complex vascular networks in a primarily avascular scaffold with the aid of mesenchymal stem cells, when co-cultured in three-dimensional spherical aggregates. Under dynamic conditions, representing the flow rate of venous vessel, mesenchymal stem cells preferably bind to damaged endothelial cells presumably assisting in the healing process.


Subject(s)
Mesenchymal Stem Cells , Neovascularization, Physiologic , Humans , Coculture Techniques , Human Umbilical Vein Endothelial Cells/metabolism , Cells, Cultured
2.
Proc Natl Acad Sci U S A ; 119(29): e2207020119, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858345

ABSTRACT

Changes in Ca2+ influx during proinflammatory stimulation modulates cellular responses, including the subsequent activation of inflammation. Whereas the involvement of Ca2+ has been widely acknowledged, little is known about the role of Na+. Ranolazine, a piperazine derivative and established antianginal drug, is known to reduce intracellular Na+ as well as Ca2+ levels. In stable coronary artery disease patients (n = 51) we observed reduced levels of high-sensitive C-reactive protein (CRP) 3 mo after the start of ranolazine treatment (n = 25) as compared to the control group. Furthermore, we found that in 3,808 acute coronary syndrome patients of the MERLIN-TIMI 36 trial, individuals treated with ranolazine (1,934 patients) showed reduced CRP values compared to placebo-treated patients. The antiinflammatory effects of sodium modulation were further confirmed in an atherosclerotic mouse model. LDL-/- mice on a high-fat diet were treated with ranolazine, resulting in a reduced atherosclerotic plaque burden, increased plaque stability, and reduced activation of the immune system. Pharmacological Na+ inhibition by ranolazine led to reduced express of adhesion molecules and proinflammatory cytokines and reduced adhesion of leukocytes to activated endothelium both in vitro and in vivo. We demonstrate that functional Na+ shuttling is required for a full cellular response to inflammation and that inhibition of Na+ influx results in an attenuated inflammatory reaction. In conclusion, we demonstrate that inhibition of Na+-Ca2+ exchange during inflammation reduces the inflammatory response in human endothelial cells in vitro, in a mouse atherosclerotic disease model, and in human patients.


Subject(s)
Acute Coronary Syndrome , C-Reactive Protein , Cardiovascular Agents , Coronary Artery Disease , Ranolazine , Sodium Channel Blockers , Sodium , Acute Coronary Syndrome/drug therapy , Animals , C-Reactive Protein/analysis , C-Reactive Protein/metabolism , Cardiovascular Agents/pharmacology , Cardiovascular Agents/therapeutic use , Coronary Artery Disease/drug therapy , Endothelial Cells/metabolism , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Mice , Ranolazine/pharmacology , Ranolazine/therapeutic use , Sodium/metabolism , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use
3.
J Mol Med (Berl) ; 99(8): 1151-1171, 2021 08.
Article in English | MEDLINE | ID: mdl-34018016

ABSTRACT

Obesity and body fat distribution are important risk factors for the development of type 2 diabetes and metabolic syndrome. Evidence has accumulated that this risk is related to intrinsic differences in behavior of adipocytes in different fat depots. We recently identified LIM domain only 3 (LMO3) in human mature visceral adipocytes; however, its function in these cells is currently unknown. The aim of this study was to determine the potential involvement of LMO3-dependent pathways in the modulation of key functions of mature adipocytes during obesity. Based on a recently engineered hybrid rAAV serotype Rec2 shown to efficiently transduce both brown adipose tissue (BAT) and white adipose tissue (WAT), we delivered YFP or Lmo3 to epididymal WAT (eWAT) of C57Bl6/J mice on a high-fat diet (HFD). The effects of eWAT transduction on metabolic parameters were evaluated 10 weeks later. To further define the role of LMO3 in insulin-stimulated glucose uptake, insulin signaling, adipocyte bioenergetics, as well as endocrine function, experiments were conducted in 3T3-L1 adipocytes and newly differentiated human primary mature adipocytes, engineered for transient gain or loss of LMO3 expression, respectively. AAV transduction of eWAT results in strong and stable Lmo3 expression specifically in the adipocyte fraction over a course of 10 weeks with HFD feeding. LMO3 expression in eWAT significantly improved insulin sensitivity and healthy visceral adipose tissue expansion in diet-induced obesity, paralleled by increased serum adiponectin. In vitro, LMO3 expression in 3T3-L1 adipocytes increased PPARγ transcriptional activity, insulin-stimulated GLUT4 translocation and glucose uptake, as well as mitochondrial oxidative capacity in addition to fatty acid oxidation. Mechanistically, LMO3 induced the PPARγ coregulator Ncoa1, which was required for LMO3 to enhance glucose uptake and mitochondrial oxidative gene expression. In human mature adipocytes, LMO3 overexpression promoted, while silencing of LMO3 suppressed mitochondrial oxidative capacity. LMO3 expression in visceral adipose tissue regulates multiple genes that preserve adipose tissue functionality during obesity, such as glucose metabolism, insulin sensitivity, mitochondrial function, and adiponectin secretion. Together with increased PPARγ activity and Ncoa1 expression, these gene expression changes promote insulin-induced GLUT4 translocation, glucose uptake in addition to increased mitochondrial oxidative capacity, limiting HFD-induced adipose dysfunction. These data add LMO3 as a novel regulator improving visceral adipose tissue function during obesity. KEY MESSAGES: LMO3 increases beneficial visceral adipose tissue expansion and insulin sensitivity in vivo. LMO3 increases glucose uptake and oxidative mitochondrial activity in adipocytes. LMO3 increases nuclear coactivator 1 (Ncoa1). LMO3-enhanced glucose uptake and mitochondrial gene expression requires Ncoa1.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adipocytes/metabolism , Energy Metabolism , Intra-Abdominal Fat/metabolism , LIM Domain Proteins/genetics , Obesity/metabolism , 3T3-L1 Cells , Adaptor Proteins, Signal Transducing/metabolism , Animals , Biomarkers , Disease Models, Animal , Disease Susceptibility , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Glucose/metabolism , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Humans , Insulin/metabolism , Intra-Abdominal Fat/cytology , LIM Domain Proteins/metabolism , Mice , Mitochondria/genetics , Mitochondria/metabolism , Models, Biological , Obesity/etiology , Oxidation-Reduction , Oxidative Phosphorylation , PPAR gamma/metabolism , Protein Binding
4.
J Stroke Cerebrovasc Dis ; 28(6): 1540-1545, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30952532

ABSTRACT

BACKGROUND: Stroke is one of the leading causes of morbidity and mortality. Thromboembolism, as a major cause of carotid artery-related stroke, can be caused by plaque rupture which is associated with neoangiogenesis within the carotid plaque. AIM: We sought to investigate a possible correlation between angiogenesis-related factors and preoperative neurological manifestations in patients with internal carotid artery stenosis, for a better understanding of thromboembolism in internal carotid artery stenosis-related stroke. METHODS: This study included 54 patients (asymptomatic, n = 20 and symptomatic, n = 34) undergoing carotid endarterectomy for high-grade internal carotid artery stenosis. In the retrieved carotid plaques, angiogenesis-related factors (vascular endothelial growth factor [VEGF], hypoxia inducible factor-1 alpha [HIF-1α], and Clusterin) were measured by immunohistochemistry and quantified by real-time polymerase chain reaction. RESULTS: We demonstrated the expression of VEGF, HIF-1α, and Clusterin by endothelial cells and smooth muscle cells in the carotid plaques. Noteworthy, mRNA VEGF levels were .7-fold higher in symptomatic patients (P = .017) compared to asymptomatic patients. In contrast, mRNA Clusterin levels were 1.8-fold lower (P = .021). Levels of mRNA HIF-1α were 1.5-fold higher in asymptomatic patients, but no statistical significance was reached between the 2 groups. CONCLUSIONS: Our results show an association between VEGF and Clusterin and neurological symptoms of patients with high-grade carotid artery stenosis.


Subject(s)
Carotid Artery, Internal/chemistry , Carotid Stenosis/metabolism , Clusterin/analysis , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Plaque, Atherosclerotic , Vascular Endothelial Growth Factor A/analysis , Aged , Aged, 80 and over , Asymptomatic Diseases , Carotid Artery, Internal/pathology , Carotid Artery, Internal/surgery , Carotid Stenosis/complications , Carotid Stenosis/pathology , Carotid Stenosis/surgery , Clusterin/genetics , Female , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Middle Aged , Neovascularization, Pathologic , Prospective Studies , RNA, Messenger/genetics , Rupture, Spontaneous , Severity of Illness Index , Stroke/etiology , Vascular Endothelial Growth Factor A/genetics
5.
JAMA Dermatol ; 155(2): 188-195, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30566178

ABSTRACT

Importance: Information on risk factors of subsequent melanomas would be helpful to identify patients at risk after the diagnosis of their first melanomas. Objective: To determine risk factors of subsequent melanomas. Design, Setting, and Participants: In this retrospective case-control study, 1648 participants with histologically verified cutaneous melanoma diagnosed from January 1, 1968, though March 16, 2015, were recruited from a tertiary referral center as part of the Molecular Markers of Melanoma study. CDKN2A was sequenced in 514 and MC1R in 953 participants. Data were analyzed from March 7, 2008, through March 25, 2015. Main Outcomes and Measures: Phenotypic traits and internal and external risk factors for the development of a second, third, or fourth melanoma. Results: In total, 1648 patients (53.6% men; mean [SD] age, 54 [15] years) were enrolled, including 1349 with single and 299 with multiple primary melanoma. Mean (SD) age at recruitment was 57 (15) years for the single-melanoma and 62 (14) years for the multiple-melanoma groups. From the internal risk factors, family history (odds ratio [OR], 1.76; 95% CI, 1.22-2.55; P = .006), CDKN2A high-risk mutations (OR, 4.03; 95% CI, 1.28-12.70; P = .02), and high numbers of nevi as a phenotypic risk factor (ORs, 2.23 [95% CI, 1.56-3.28, P < .001] for 20-30 smaller nevi and 2.56 [95% CI, 1.50-4.36; P = .003] for 20-30 larger nevi) were significantly associated with the risk of developing a subsequent primary melanoma using multivariate logistic regression analysis. Nonmelanoma skin cancer (OR, 2.57; 95% CI, 1.84-3.58; P < .001) and signs of actinic skin damage, particularly on the back (ORs, 1.91 [95% CI, 1.12-3.25; P = .04] for freckling and 1.92 [95% CI, 1.29-3.08; P = .007] for solar lentigines), additionally increased risk of a subsequent melanoma. All those factors were also associated with an earlier development of the second melanoma. Patients with 3 melanomas developed their second melanoma earlier than patients with only 2 melanomas (mean [SD] age, 55 [15] years for those with 2 primary melanomas; 52 [15] years for those with 3 primary melanomas). Time spent outdoors, solarium use, outdoor occupation, and hair color had no significant associations in these models. Conclusions and Relevance: According to the results of this study, internal factors (family history and genetic variants), number of nevi, and actinic damage on the back are more relevant for the development of subsequent melanomas than skin phototype or hair color. Patients with many nevi were younger at the time of the diagnosis of their first melanoma. This finding could help to identify persons at increased risk of developing multiple primary melanomas.


Subject(s)
Genetic Predisposition to Disease/epidemiology , Melanoma/epidemiology , Melanoma/genetics , Neoplasm Recurrence, Local/epidemiology , Skin Neoplasms/epidemiology , Skin Neoplasms/genetics , Age Distribution , Analysis of Variance , Austria , Case-Control Studies , Female , Humans , Logistic Models , Male , Melanoma/pathology , Neoplasm Invasiveness/pathology , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prevalence , Retrospective Studies , Risk Factors , Sex Distribution , Skin Neoplasms/pathology , Survival Analysis , Melanoma, Cutaneous Malignant
6.
Thromb Haemost ; 118(12): 2074-2085, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30419595

ABSTRACT

Thrombospondin-1 (TSP-1) is primarily expressed by platelets and endothelial cells (ECs) and rapidly released upon their activation. It functions in haemostasis as a bridging molecule in platelet aggregation, by promoting platelet adhesion to collagen and by protecting von Willebrand factor strings from degradation. In blood of patients undergoing surgery and in co-cultures of neutrophils with platelets or ECs, we observed proteolysis of the 185 kDa full-length TSP-1 to a 160-kDa isoform. We hypothesized that TSP-1 processing may alter its haemostatic properties. Selective enzyme inhibitors in co-cultures revealed that neutrophil proteases elastase and cathepsin G mediate TSP-1 processing. The cut site of cathepsin G was mapped to TSP-1 amino acids R237/T238 by Edman sequencing. Formation of neutrophil extracellular traps protected TSP-1 from complete degradation and promoted controlled processing to the 160-kDa isoform. Haemostatic properties were tested by platelet aggregation, adhesion, coagulation and string formation under flow. Platelets from TSP-1 deficient mice did not differ from wild-type in platelet aggregation but showed severe impairment of platelet adhesion to collagen and string formation under flow. Reconstitution experiments revealed that the 160-kDa TSP-1 isoform was markedly more potent than the 185-kDa full-length molecule in restoring function. Thus, TSP-1 processing by neutrophil proteases yields a 160-kDa isoform which shows enhanced potency to promote platelet adhesion and string formation. This finding reveals a novel mechanism of neutrophil-mediated thrombus formation and provides first evidence for the impact of TSP-1 proteolysis on its haemostatic properties.


Subject(s)
Blood Platelets/physiology , Endothelium, Vascular/physiology , Neutrophils/physiology , Thrombospondin 1/metabolism , Animals , Cells, Cultured , Coculture Techniques , Hemostasis , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Adhesiveness , Platelet Aggregation , Protein Multimerization , Proteolysis , Thrombospondin 1/genetics , Thrombospondin 1/immunology , von Willebrand Factor/metabolism
7.
JAMA Dermatol ; 154(7): 789-795, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29898205

ABSTRACT

Importance: Recently, the red hair variants of MC1R were found to contribute differently to pigmentation phenotype in males and females. Objective: To investigate the role of these variants in melanoma risk in males and females separately because carriers of the red hair variants of MC1R are at increased risk of melanoma. Design, Setting, and Participants: In this hospital-based, case-control study, we evaluated the effect of MC1R and melanoma risk for males and females separately by performing multivariate logistic regression analyses. Main Outcomes and Measures: Association of MC1R variants and melanoma risk in males and females. Results: A total of 905 females (473 melanoma cases, 432 controls) and 886 males (518 melanoma cases, 368 controls) were included in the analyses. The mean (SD) age of the study population was 59.2 (15.6). In females, carrying any MC1R red hair variants remained an independent risk factor of melanoma in a multivariable analysis (adjusted odds ratio [OR], 2.19 [95% CI, 1.60-2.99]), whereas in males, only signs of actinic skin damage (lentigines on the back [OR, 2.56; 95% CI, 1.47-4.45; P = .001] and the hands [OR, 2.31; 95% CI, 1.24-4.29; P = .008] and wrinkling on the neck [OR, 2.17; 95% CI, 1.23-3.82; P = .007]) and sunburns (OR, 1.65; 95% CI, 1.12-2.42; P = .01) remained significant risk factors. Conclusions and Relevance: MC1R variants contribute differently to melanoma risk in males and females. This could be helpful to better classify melanoma risk factors between the sexes.


Subject(s)
Melanoma/epidemiology , Melanoma/genetics , Receptor, Melanocortin, Type 1/genetics , Skin Neoplasms/epidemiology , Skin Neoplasms/genetics , Adult , Aged , Austria/epidemiology , Case-Control Studies , Female , Genetic Variation , Hair Color/genetics , Humans , Lentigo/epidemiology , Male , Middle Aged , Neck , Phenotype , Risk Factors , Sex Factors , Skin Aging , Skin Pigmentation/genetics , Sunburn/epidemiology
8.
Sci Rep ; 8(1): 6598, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29700367

ABSTRACT

Secretion and exchange of biomolecules via extracellular vesicles (EVs) are crucial mechanisms in intercellular communication, and the roles of EVs in infection, inflammation, or thrombosis have been increasingly recognized. EVs have emerged as central players in immune regulation and can enhance or suppress the immune response, depending on the state of donor and recipient cells. We investigated the interaction of blood cell-derived EVs with leukocyte subpopulations (monocytes and their subsets, granulocytes, B cells, T cells, and NK cells) directly in whole blood using a combination of flow cytometry, imaging flow cytometry, cell sorting, and high resolution confocal microscopy. Platelet-derived EVs constituted the majority of circulating EVs and were preferentially associated with granulocytes and monocytes, while they scarcely interacted with lymphocytes. Further flow cytometric differentiation of monocyte subsets provided clear indications for a preferential association of platelet-derived EVs with intermediate (CD14++CD16+) monocytes in whole blood.


Subject(s)
Blood Platelets/metabolism , Cell-Derived Microparticles/metabolism , Extracellular Vesicles/metabolism , Leukocytes/metabolism , Biomarkers , Blood Platelets/immunology , Cell Separation/methods , Granulocytes/immunology , Granulocytes/metabolism , Humans , Immunophenotyping , Leukocytes/immunology , Monocytes/immunology , Monocytes/metabolism
9.
Thromb Haemost ; 117(7): 1379-1390, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28492698

ABSTRACT

Monocytes and monocyte-derived microvesicles (MVs) are the main source of circulating tissue factor (TF). Increased monocyte TF expression and increased circulating levels of procoagulant MVs contribute to the formation of a prothrombotic state in patients with cardiovascular disease. Interleukin (IL)-33 is a pro-inflammatory cytokine involved in atherosclerosis and other inflammatory diseases, but its role in regulating thrombosis is still unclear. The aim of the present study was to investigate the effects of IL-33 on the procoagulant properties of human monocytes and monocyte-derived MVs. IL-33 induced a time- and concentration-dependent increase of monocyte TF mRNA and protein levels via binding to the ST2-receptor and activation of the NF-κB-pathway. The IL-33 treated monocytes also released CD14+TF+ MVs and IL-33 was found to increase the TF activity of both the isolated monocytes and monocyte-derived MVs. The monocytes were classified into subsets according to their CD14 and CD16 expression. Intermediate monocytes (IM) showed the highest ST2 receptor expression, followed by non-classical monocytes (NCM), and classical monocytes (CM). IL-33 induced a significant increase of TF only in the IM (p<0.01), with a tendency in NCM (p=0.06), but no increase was observed in CM. Finally, plasma levels of IL-33 were positively correlated with CD14+TF+ MVs in patients undergoing carotid endarterectomy (r=0.480; p=0.032; n=20). We hereby provide novel evidence that the proinflammatory cytokine IL-33 induces differential TF expression and activity in monocyte subsets, as well as the release of procoagulant MVs. In this manner, IL-33 may contribute to the formation of a prothrombotic state characteristic for cardiovascular disease.


Subject(s)
Cell-Derived Microparticles/metabolism , Interleukin-33/physiology , Monocytes/physiology , Thromboplastin/physiology , Aged , Carotid Stenosis/blood , Carotid Stenosis/immunology , Cells, Cultured , Female , Humans , Interleukin-1 Receptor-Like 1 Protein/blood , Interleukin-33/pharmacology , Lipopolysaccharide Receptors/blood , Male , Middle Aged , Monocytes/classification , Monocytes/immunology , NF-kappa B/blood , RNA, Messenger/blood , RNA, Messenger/genetics , Recombinant Proteins/pharmacology , Thromboplastin/genetics , Thrombosis/etiology
10.
Vascul Pharmacol ; 90: 44-50, 2017 03.
Article in English | MEDLINE | ID: mdl-28192257

ABSTRACT

AIMS: Levosimendan is an inodilator for the treatment of acute decompensated heart failure (HF). Data from clinical studies suggest that levosimendan is particularly effective in HF due to myocardial infarction. After acute revascularization, no reflow-phenomenon is a common complication that may lead to pump failure and cardiogenic shock. Our aim was to examine whether levosimendan interferes with the pro-thrombotic phenotype of activated endothelial cells in vitro. METHODS: Human heart microvascular endothelial cells (HHMEC) and human umbilical vein endothelial cells (HUVEC) were treated with interleukin-1ß (IL-1ß) (200U/mL) or thrombin (5U/mL) and co-treated with or without levosimendan (0.1-10µM) for 2-24h. In addition, flow experiments were performed. Effects on plasminogen activator inhibitor-1 (PAI-1) and tissue factor (TF) expression and activity were measured by rt-PCR, specific ELISA and flow cytometry. RESULTS: Treatment with IL-1ß or thrombin significantly increased the expression of PAI-1 and TF in endothelial cells. Co-treatment with levosimendan strongly attenuated the effects of IL-1ß and thrombin on PAI-1 and TF mRNA by up to 50% and 45%, in a dose- and time-dependent manner. Similar results were obtained under flow conditions. Furthermore, co-treatment with levosimendan dampened the antigen production of PAI-1 and the surface expression of TF by 35% and 45%, respectively. Additionally, levosimendan diminished both TF and PAI-1 activity. CONCLUSION: Levosimendan down-regulates the expression of the pro-thrombotic and anti-fibrinolytic biomolecules TF and PAI-1 in activated human endothelial cells. Our findings may, at least in part, explain some of the beneficial effects of levosimendan after myocardial reperfusion.


Subject(s)
Fibrinolysis/drug effects , Fibrinolytic Agents/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Hydrazones/pharmacology , Pyridazines/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-1beta/pharmacology , Plasminogen Activator Inhibitor 1/metabolism , Simendan , Thrombin/pharmacology , Thromboplastin/metabolism , Time Factors
11.
Sci Rep ; 7: 40881, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28102348

ABSTRACT

Excessive accumulation of white adipose tissue (WAT) is a hallmark of obesity. The expansion of WAT in obesity involves proliferation and differentiation of adipose precursors, however, the underlying molecular mechanisms remain unclear. Here, we used an unbiased transcriptomics approach to identify the earliest molecular underpinnings occuring in adipose precursors following a brief HFD in mice. Our analysis identifies Heme Oxygenase-1 (HO-1) as strongly and selectively being upregulated in the adipose precursor fraction of WAT, upon high-fat diet (HFD) feeding. Specific deletion of HO-1 in adipose precursors of Hmox1fl/flPdgfraCre mice enhanced HFD-dependent visceral adipose precursor proliferation and differentiation. Mechanistically, HO-1 reduces HFD-induced AKT2 phosphorylation via ROS thresholding in mitochondria to reduce visceral adipose precursor proliferation. HO-1 influences adipogenesis in a cell-autonomous way by regulating events early in adipogenesis, during the process of mitotic clonal expansion, upstream of Cebpα and PPARγ. Similar effects on human preadipocyte proliferation and differentiation in vitro were observed upon modulation of HO-1 expression. This collectively renders HO-1 as an essential factor linking extrinsic factors (HFD) with inhibition of specific downstream molecular mediators (ROS &AKT2), resulting in diminished adipogenesis that may contribute to hyperplastic adipose tissue expansion.


Subject(s)
Cell Differentiation , Cell Proliferation , Heme Oxygenase-1/metabolism , Obesity/pathology , Proto-Oncogene Proteins c-akt/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Diet, High-Fat , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , PPAR gamma/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism
12.
Inflammation ; 39(5): 1737-46, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27503310

ABSTRACT

Endothelial activation with excessive recruitment and adhesion of immune cells plays a central role in the progression of sepsis. We established a microfluidic system to study the activation of human umbilical vein endothelial cells by conditioned medium containing plasma from lipopolysaccharide-stimulated whole blood or from septic blood and to investigate the effect of adsorption of inflammatory mediators on endothelial activation. Treatment of stimulated whole blood with polystyrene-divinylbenzene-based cytokine adsorbents (average pore sizes 15 or 30 nm) prior to passage over the endothelial layer resulted in significantly reduced endothelial cytokine and chemokine release, plasminogen activator inhibitor-1 secretion, adhesion molecule expression, and in diminished monocyte adhesion. Plasma samples from sepsis patients differed substantially in their potential to induce endothelial activation and monocyte adhesion despite their almost identical interleukin-6 and tumor necrosis factor-alpha levels. Pre-incubation of the plasma samples with a polystyrene-divinylbenzene-based adsorbent (30 nm average pore size) reduced endothelial intercellular adhesion molecule-1 expression to baseline levels, resulting in significantly diminished monocyte adhesion. Our data support the potential of porous polystyrene-divinylbenzene-based adsorbents to reduce endothelial activation under septic conditions by depletion of a broad range of inflammatory mediators.


Subject(s)
Cell Adhesion , Endothelium, Vascular/immunology , Monocytes/cytology , Polystyrenes/therapeutic use , Sepsis/immunology , Vinyl Compounds/therapeutic use , Adsorption , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Porosity , Sepsis/therapy
13.
Biomed Opt Express ; 7(4): 1479-95, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27446670

ABSTRACT

We present a high resolution polarization sensitive optical coherence tomography (PS-OCT) system for ocular imaging in rodents. The system operates at 840 nm and uses a broadband superluminescent diode providing an axial resolution of 5.1 µm in air. PS-OCT data was acquired at 83 kHz A-scan rate by two identical custom-made spectrometers for orthogonal polarization states. Pigmented (Brown Norway, Long Evans) and non-pigmented (Sprague Dawley) rats as well as pigmented mice (C57BL/6) were imaged. Melanin pigment related depolarization was analyzed in the retinal pigment epithelium (RPE) and choroid of these animals using the degree of polarization uniformity (DOPU). For all rat strains, significant differences between RPE and choroidal depolarization were observed. In contrast, DOPU characteristics of RPE and choroid were similar for C57BL/6 mice. Moreover, the depolarization within the same tissue type varied significantly between different rodent strains. Retinal nerve fiber layer thickness, phase retardation, and birefringence were mapped and quantitatively measured in Long Evans rats in vivo for the first time. In a circumpapillary annulus, retinal nerve fiber layer birefringence amounted to 0.16°/µm ± 0.02°/µm and 0.17°/µm ± 0.01°/µm for the left and right eyes, respectively.

14.
Sci Rep ; 6: 25171, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27142573

ABSTRACT

Tissue factor (TF) is the primary trigger of coagulation. Elevated levels of TF are found in atherosclerotic plaques, and TF leads to thrombus formation when released upon plaque rupture. Interleukin (IL)-33 was previously shown to induce angiogenesis and inflammatory activation of endothelial cells (ECs). Here, we investigated the impact of IL-33 on TF in human ECs, as a possible new link between inflammation and coagulation. IL-33 induced TF mRNA and protein in human umbilical vein ECs and coronary artery ECs. IL-33-induced TF expression was ST2- and NF-κB-dependent, but IL-1-independent. IL-33 also increased cell surface TF activity in ECs and TF activity in ECs-derived microparticles. IL-33-treated ECs reduced coagulation time of whole blood and plasma but not of factor VII-deficient plasma. In human carotid atherosclerotic plaques (n = 57), TF mRNA positively correlated with IL-33 mRNA expression (r = 0.691, p < 0.001). In this tissue, IL-33 and TF protein was detected in ECs and smooth muscle cells by immunofluorescence. Furthermore, IL-33 and TF protein co-localized at the site of clot formation within microvessels in plaques of patients with symptomatic carotid stenosis. Through induction of TF in ECs, IL-33 could enhance their thrombotic capacity and thereby might impact on thrombus formation in the setting of atherosclerosis.


Subject(s)
Blood Coagulation , Endothelial Cells/metabolism , Inflammation/pathology , Interleukin-33/metabolism , Thromboplastin/biosynthesis , Cells, Cultured , Humans , RNA, Messenger/biosynthesis
15.
Thromb Haemost ; 116(2): 317-27, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27173404

ABSTRACT

Interleukin (IL)-33, a member of the IL-1 family of cytokines, is involved in various inflammatory conditions targeting amongst other cells the endothelium. Besides regulating the maturation and functions of myeloid cells, granulocyte macrophage-colony stimulating factor (GM-CSF) and macrophage-CSF (M-CSF) have been shown to play a role in such pathologies too. It was the aim of our study to investigate a possible influence of IL-33 on GM-CSF and M-CSF production by human endothelial cells. IL-33, but not IL-18 or IL-37, stimulated GM-CSF and M-CSF mRNA expression and protein production by human umbilical vein endothelial cells (HUVECs) and human coronary artery ECs (HCAECs) through the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway in an IL-1-independent way. This effect was inhibited by the soluble form of ST2 (sST2), which is known to act as a decoy receptor for IL-33. The 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitor fluvastatin could also be shown to moderately reduce the IL-33-mediated effect on M-CSF, but not on GM-CSF expression. In addition, IL-33, IL-1ß, GM-CSF and M-CSF were detected in endothelial cells of human carotid atherosclerotic plaques using immunofluorescence. Upregulation of GM-CSF and M-CSF production by human endothelial cells, an effect that appears to be mediated by NF-κB and to be independent of IL-1, may be an additional mechanism through which IL-33 contributes to inflammatory activation of the vessel wall.


Subject(s)
Endothelial Cells/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Interleukin-33/metabolism , Macrophage Colony-Stimulating Factor/biosynthesis , Carotid Stenosis/immunology , Carotid Stenosis/metabolism , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/immunology , Fatty Acids, Monounsaturated/pharmacology , Fluvastatin , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Human Umbilical Vein Endothelial Cells , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Indoles/pharmacology , Inflammation Mediators/metabolism , Inflammation Mediators/pharmacology , Interleukin-1/metabolism , Interleukin-1/pharmacology , Interleukin-18/metabolism , Interleukin-18/pharmacology , Interleukin-1beta/metabolism , Interleukin-33/pharmacology , Macrophage Colony-Stimulating Factor/genetics , NF-kappa B/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Up-Regulation
16.
JAMA Dermatol ; 152(7): 776-82, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27050141

ABSTRACT

IMPORTANCE: Despite the unquestioned relationship of UV radiation (UVR) exposure and melanoma development, UVR-independent development of melanoma has only recently been described in mice. These findings in mice highlight the importance of the genetic background of the host and could be relevant for preventive measures in humans. OBJECTIVE: To study the role of the melanocortin-1 receptor (MC1R) and melanoma risk independently from UVR in a clinical setting. DESIGN, SETTING, AND PARTICIPANTS: Hospital-based case-control study, including genetic testing, questionnaires, and physical data (Molecular Markers of Melanoma Study data set) including 991 melanoma patients (cases) and 800 controls. MAIN OUTCOMES AND MEASURES: Association of MC1R variants and melanoma risk independent from sun exposure variables. RESULTS: The 1791 participants included 991 with a diagnosis of melanoma and 800 control patients (mean [SD] age, 59.2 [15.6] years; 50.5% male). Compared with wild-type carriers, carriers of MC1R variants were at higher melanoma risk after statistically adjusting for previous UVR exposure (represented by prior sunburns and signs of actinic skin damage identified by dermatologists), age, and sex compared with wild-type carriers (≥2 variants, OR, 2.13 [95% CI, 1.66-2.75], P < .001; P for trend <.001). After adjustment for sex, age, sunburns in the past, and signs of actinic skin damage, the associations remained significant (OR, 1.65 [95% CI, 1.02-2.67] for R/R, OR, 2.63 [95% CI, 1.82-3.81] for R/r; OR, 1.83 [95% CI, 1.36-2.48] for R/0; and OR, 1.50 [95% CI, 1.01-2.21] for r/r, with P values ranging from <.001 to .04 when adjusted for facial actinic skin damage; OR, 2.36 [95% CI, 1.62-3.43] for R/r; and OR, 1.47 [95% CI, 1.08-1.99] for R/0 with P values ranging from <.001 to .01 when adjusted for dorsal actinic skin damage; and OR, 2.54 [95% CI, 1.76-3.67] for R/r, OR, 1.75 [95% CI, 1.30-2.36] for R/0; and OR, 1.50 [95% CI, 1.02-2.20] for r/r with P values ranging from <.001 to .04 when adjusted for actinic skin damage on the hands). CONCLUSIONS AND RELEVANCE: Carriers of MC1R variants were at increased melanoma risk independent of their sun exposure. Further studies are required to elucidate the causes of melanoma development in these individuals.


Subject(s)
Melanoma/genetics , Receptor, Melanocortin, Type 1/genetics , Skin Neoplasms/genetics , Ultraviolet Rays/adverse effects , Adult , Aged , Back , Case-Control Studies , Face , Female , Genetic Testing , Genetic Variation , Hand , Humans , Keratosis, Actinic/etiology , Male , Middle Aged , Risk Factors , Sunburn/etiology , Surveys and Questionnaires
17.
Arterioscler Thromb Vasc Biol ; 36(4): 647-54, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26821949

ABSTRACT

OBJECTIVE: Cingulin is a cytoplasmic component of tight junctions. Although modulation of cingulin levels in cultured epithelial model systems has no significant effect on barrier function, evidence from cingulin knockout mice suggests that cingulin may be involved in the regulation of the behavior of epithelial or endothelial cells. Here, we investigate the role of cingulin in the barrier function of endothelial cells. APPROACH AND RESULTS: We show that cingulin is expressed in human endothelial cells of the skin, brain, and lung in vivo and in vitro. Endothelial cingulin colocalizes and coimmunoprecipitates with the tight junction proteins zonula occludens-1 and guanine nucleotide exchange factor-H1. Cingulin overexpression in human umbilical vein endothelial cell induces tight junction formation, increases transendothelial electric resistance, and strengthens barrier function for low and high molecular weight tracers. In contrast, cultured endothelial cells lacking cingulin are more permeable for low molecular weight tracers. In cingulin knockout mice, neurons of the area postrema and Purkinje cells show an increased uptake of small molecular weight tracers indicating decreased barrier function at these sites. CONCLUSIONS: We demonstrate that cingulin participates in the modulation of endothelial barrier function both in human cultured cells in vitro and in mouse brains in vivo. Understanding the role of cingulin in maintaining tight barriers in endothelia may allow developing new strategies for the treatment of vascular leak syndromes.


Subject(s)
Blood-Brain Barrier/metabolism , Capillary Permeability , Endothelial Cells/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Animals , Area Postrema/metabolism , Cell Proliferation , Cells, Cultured , Claudin-5/metabolism , Electric Impedance , Genotype , Humans , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Phenotype , Purkinje Cells/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , Tight Junctions/metabolism , Time Factors , Transfection , Zonula Occludens-1 Protein/metabolism
18.
J Mod Opt ; 62(21): 1758-1763, 2015.
Article in English | MEDLINE | ID: mdl-26689829

ABSTRACT

Depolarization of light can be measured by polarization-sensitive optical coherence tomography (PS-OCT) and has been used to improve tissue discrimination as well as segmentation of pigmented structures. Most approaches to depolarization assessment for PS-OCT - such as the degree of polarization uniformity (DOPU) - rely on measuring the uniformity of polarization states using spatial evaluation kernels. In this article, we present a different approach which exploits the spectral dimension. We introduce the spectral DOPU for the pixelwise analysis of polarization state variations between sub-bands of the broadband light source spectrum. Alongside a comparison with conventional spatial and temporal DOPU algorithms, we demonstrate imaging in the healthy human retina, and apply the technique for contrasting hard exudates in diabetic retinopathy and investigating the pigment epithelium of the rat iris.

19.
Invest Ophthalmol Vis Sci ; 56(12): 7462-72, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26595606

ABSTRACT

PURPOSE: The purpose of this study was to demonstrate polarization-sensitive optical coherence tomography (PS-OCT) for imaging pigmented structures in the posterior eye segments of albino and pigmented rats and to correlate depolarization contrast of the retinal pigment epithelium (RPE) and choroid in in vivo PS-OCT to melanin pigmentation detected in postmortem histologic serial sections. METHODS: In vivo three-dimensional PS-OCT imaging was performed in adult albino and pigmented rat eyes at 70-kHz A-line rate. Degree of polarization uniformity (DOPU) fundus maps and radial DOPU profiles were generated. Postmortem histomorphologic analysis was performed in order to investigate melanin pigmentation of the RPE and choroid. Fundus pigmentation maps were extracted from histologic serial sections. Pigmentation profiles were correlated to DOPU profiles of the same eyes. RESULTS: Strong depolarization was found in the RPE/choroid complex of pigmented rats, whereas the same structures exhibited uniform polarization in albino rats. The difference between the depolarization characteristics between albino and pigmented animals was statistically significant. In the fundus pigmentation maps, optical pigment density was zero in albino rat eyes. In pigmented rat eyes, a strong negative correlation between optical pigment density and DOPU was observed. CONCLUSIONS: This in vivo and ex vivo investigation of posterior rat eyes indicates that melanin is the cause of depolarization in retinal PS-OCT images. It further demonstrates that melanin pigmentation in the RPE and choroid can be quantified via depolarization imaging and therefore suggests that PS-OCT is a useful tool for the noninvasive quantitative assessment of pigmentary changes in vision-threatening diseases such as age-related macular degeneration.


Subject(s)
Albinism/pathology , Melanins/metabolism , Retinal Pigment Epithelium/pathology , Tomography, Optical Coherence/methods , Animals , Disease Models, Animal , Fluorescein Angiography , Fundus Oculi , Male , Rats , Rats, Inbred BN , Rats, Long-Evans , Rats, Sprague-Dawley , Retinal Pigment Epithelium/metabolism , Visual Acuity
20.
PLoS One ; 10(11): e0142115, 2015.
Article in English | MEDLINE | ID: mdl-26536466

ABSTRACT

BACKGROUND: Despite significant advances in organ preservation, surgical techniques and perioperative care, primary graft dysfunction is a serious medical problem in transplantation medicine in general and a specific problem in patients undergoing lung transplantation. As a result, patients develop lung edema, causing reduced tissue oxygenation capacity, reduced lung compliance and increased requirements for mechanical ventilatory support. Yet, there is no effective strategy available to protect the grafted organ from stress reactions induced by ischemia/reperfusion and by the surgical procedure itself. METHODS: We assessed the effect of a cingulin-derived peptide, XIB13 or a random peptide in an established rat model of allogeneic lung transplantation. Donor lungs and recipients received therapeutic peptide at the time of transplantation and outcome was analyzed 100min and 28 days post grafting. RESULTS: XIB13 improved blood oxygenation and reduced vascular leak 100min post grafting. Even after 28 days, lung edema was significantly reduced by XIB13 and lungs had reduced fibrotic or necrotic zones. Moreover, the induction of an allogeneic T cell response was delayed indicating a reduced antigen exchange between the donor and the host. CONCLUSIONS: In summary, we provide a new tool to strengthen endothelial barrier function thereby improving outcomes in lung transplantation.


Subject(s)
Disease Models, Animal , Lung Transplantation/adverse effects , Membrane Proteins , Microfilament Proteins , Peptide Fragments/pharmacology , Primary Graft Dysfunction/prevention & control , Pulmonary Edema/prevention & control , Vascular Resistance/drug effects , Animals , Male , Organ Preservation , Primary Graft Dysfunction/etiology , Pulmonary Edema/etiology , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Ventilation
SELECTION OF CITATIONS
SEARCH DETAIL
...