Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(15)2022 Jul 29.
Article in English | MEDLINE | ID: mdl-35955568

ABSTRACT

There is evidence for increased angiogenesis in the (ectopic) endometrium of adenomyosis patients under the influence of vascular endothelial growth factor (VEGF). VEGF stimulates both angiogenesis and lymph-angiogenesis. However, information on lymph vessels in the (ectopic) endometrium of adenomyosis patients is lacking. In this retrospective matched case-control study, multiplex immunohistochemistry was performed on thirty-eight paraffin embedded specimens from premenopausal women who had undergone a hysterectomy at the Amsterdam UMC between 2001 and 2018 to investigate the evidence for (lymph) angiogenesis in the (ectopic) endometrium or myometrium of patients with adenomyosis versus controls with unrelated pathologies. Baseline characteristics of both groups were comparable. In the proliferative phase, the blood and lymph vessel densities were, respectively, higher in the ectopic and eutopic endometrium of patients with adenomyosis than in the endometrium of controls. The relative number of blood vessels without α-smooth muscle actinin (α SMA) was higher in the eutopic and ectopic endometrium of adenomyosis patients versus controls. The level of VEGF staining intensity was highest in the myometrium but did not differ between patients with adenomyosis or controls. The results indicate increased angiogenesis and lymphangiogenesis in the (ectopic) endometrium affected by adenomyosis. The clinical relevance of our findings should be confirmed in prospective clinical studies.


Subject(s)
Adenomyosis , Endometriosis , Adenomyosis/metabolism , Adenomyosis/pathology , Case-Control Studies , Endometriosis/pathology , Endometrium/metabolism , Female , Humans , Immunohistochemistry , Lymphangiogenesis , Neovascularization, Pathologic/metabolism , Prospective Studies , Retrospective Studies , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factors/metabolism
4.
Cytotherapy ; 20(9): 1143-1154, 2018 09.
Article in English | MEDLINE | ID: mdl-30107976

ABSTRACT

BACKGROUND AIMS: After a myocardial infarction (MI) atherosclerosis is accelerated leading to destabilization of the atherosclerotic plaque. mesenchymal stromal cells are a promising therapeutic option for atherosclerosis. Previously, we demonstrated a novel stem cell delivery technique, with adipose stem cells coupled to microbubbles (i.e., StemBells) as therapy after MI. In this study, we aim to investigate the effect of StemBell therapy on atherosclerotic plaques in an atherosclerotic mouse model after MI. METHODS: MI was induced in atherosclerotic Apolipoprotein E-deficient mice that were fed a high-fat Western diet. Six days post-MI, the mice received either 5 × 105/100 µL StemBells or vehicle intravenously. The effects of StemBell treatment on the size and stability of aortic root atherosclerotic plaques and the infarcted heart were determined 28 days post-MI via (immuno)histological analyses. Moreover, monocyte subtypes and lipids in the blood were studied. RESULTS: StemBell treatment resulted in significantly increased cap thickness, decreased intra-plaque macrophage density and increased percentage of intra-plaque anti-inflammatory macrophages and chemokines, without affecting plaque size and serum cholesterol/triglycerides. Furthermore, StemBell treatment significantly increased the percentage of anti-inflammatory macrophages within the infarcted myocardium but did not affect cardiac function nor infarct size. Finally, also the average percentage of anti-inflammatory monocytes in the circulation was increased after StemBell therapy. DISCUSSION: StemBell therapy increased cap thickness and decreased intra-plaque inflammation after MI, indicative of stabilized atherosclerotic plaque. It also induced a shift of circulating monocytes and intra-plaque and intra-cardiac macrophages towards anti-inflammatory phenotypes. Hence, StemBell therapy may be a therapeutic option to prevent atherosclerosis acceleration after MI.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Myocardial Infarction/complications , Plaque, Atherosclerotic/therapy , Animals , Aorta/pathology , Apolipoproteins E/genetics , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Lipids/blood , Macrophages/pathology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice, Inbred C57BL , Mice, Knockout , Microbubbles , Monocytes/pathology , Myocardial Infarction/pathology , Plaque, Atherosclerotic/etiology
5.
Ultrasound Med Biol ; 44(8): 1901-1909, 2018 08.
Article in English | MEDLINE | ID: mdl-29735316

ABSTRACT

Contrast-enhanced ultrasound (CEUS) is an innovative ultrasound technique capable of visualizing both the macro- and microvasculature of tissues. In this prospective pilot study, we evaluated the feasibility of using CEUS to visualize the microvasculature of uterine fibroids and compared CEUS with conventional ultrasound. Four women with fibroids underwent gray-scale ultrasound, sonoelastography and power/color Doppler scans followed by CEUS examination. Analysis of CEUS images revealed initial perfusion of the peripheral rim, that is, a pseudo-capsule, followed by enhancement of the entire lesion through vessels traveling from the exterior to the interior of the fibroid. The pseudo-capsules exhibited slight hyper-enhancement, making a clear delineation of the fibroids possible. The centers of three fibroids exhibited areas lacking vascularization, information not obtainable with the other imaging techniques. CEUS is a feasible technique for imaging and quantifying the microvasculature of fibroids. In comparison with conventional ultrasound imaging modalities, CEUS can provide additional diagnostic information based on the microvasculature.


Subject(s)
Contrast Media , Image Enhancement/methods , Leiomyoma/diagnostic imaging , Ultrasonography/methods , Uterine Neoplasms/diagnostic imaging , Adult , Feasibility Studies , Female , Humans , Leiomyoma/blood supply , Microvessels , Middle Aged , Pilot Projects , Prospective Studies , Uterine Neoplasms/blood supply , Uterus/blood supply , Uterus/diagnostic imaging
6.
Heart Fail Rev ; 23(4): 583-595, 2018 07.
Article in English | MEDLINE | ID: mdl-29536322

ABSTRACT

Infectious myocarditis is the result of an immune response to a microbial infection of the heart. The blood vessels of the heart, both the intramyocardial microvasculature and the large epicardial coronary arteries, play an important role in the pathogenesis of infectious myocarditis. First of all, in addition to cardiomyocytes, endothelial cells of the cardiac (micro)vasculature are direct targets for infection. Moreover, through the expression of adhesion molecules and antigen presenting Major Histocompatibility Complex molecules, the blood vessels assist in shaping the cellular immune response in infectious myocarditis. In addition, damage and dysfunction of the cardiac (micro)vasculature are associated with thrombus formation as well as aberrant regulation of vascular tone including coronary vasospasm. These in turn can cause cardiac perfusion abnormalities and even myocardial infarction. In this review, we will discuss the role of the cardiac (micro)vasculature in the pathogenesis of infectious myocarditis.


Subject(s)
Coronary Vessels/pathology , Infections/complications , Myocarditis/pathology , Myocytes, Cardiac/pathology , Endothelium, Vascular/pathology , Humans , Infections/pathology , Myocarditis/etiology
8.
Cytotherapy ; 19(1): 131-140, 2017 01.
Article in English | MEDLINE | ID: mdl-27856230

ABSTRACT

BACKGROUND: Adipose-derived stromal cells (ASCs) are a promising new therapeutic option for patients with acute myocardial infarction (AMI). Previously, we found that ASCs coupled to antibody-targeted microbubbles (StemBells [StBs]) improved cardiac function when administered intravenously 7 days post-AMI in rats. In this study, we compared the efficacy of intravenous StB administration at different administration time points following AMI in rats. METHODS: AMI, followed by reperfusion, was induced in four groups of male Wistar rats, which subsequently received an intravenous 1 × 106 StB bolus 1 day post-AMI (StB1; n = 8), 7 days post-AMI (StB7; n = 9), at both time points (StB1+7; n = 7) or neither (Control; n = 7). The effect onrdiac function was determined using echocardiography prior to AMI, 7 days post-AMI and 42 days post-AMI. The effect on infarct size and macrophages in the infarct core were determined (immuno)histochemically 42 days post-AMI. RESULTS: At 42 days post-AMI, all three StB groups had a significantly improved fractional shortening compared with the control group. Between the StB-treated groups, the effects did not differ significantly at 42 days post-AMI. At 7 days post-AMI, the StB1 group had a significantly improved fractional shortening compared with the control and StB7 groups. No significant changes in infarct size or macrophage numbers were found compared with the control group for any StB group. CONCLUSIONS: StB administration resulted in long-term improvement of cardiac function, independent of the time point of administration. When administered at 1 day post-AMI, this improvement was already evident at 7 days post-AMI.


Subject(s)
Adipose Tissue/cytology , Myocardial Infarction/therapy , Administration, Intravenous , Animals , Cells, Cultured , Echocardiography , Male , Microbubbles , Myocardial Infarction/diagnostic imaging , Rats, Wistar , Stromal Cells/transplantation , Time Factors
9.
Int J Cardiol ; 216: 58-65, 2016 Aug 01.
Article in English | MEDLINE | ID: mdl-27140338

ABSTRACT

BACKGROUND: There is a clinical need for immunosuppressive therapy that can treat myocarditis patients in the presence of an active viral infection. In this study we therefore investigated the effects of colchicine, an immunosuppressive drug which has been used successfully as treatment for pericarditis patients, in a mouse model of coxsackievirus B3(CVB3)-induced myocarditis. METHODS: Four groups of C3H mice were included: control mice (n=8), mice infected with CVB3 (1×10(5) PFU, n=10), mice with colchicine administration (2mg/kg i.p, n=5) and mice with combined CVB3 infection and colchicine administration (n=10). After three days, the heart, pancreas and spleen were harvested and evaluated using (immuno)histochemical analysis and CVB3 qPCR. RESULTS: Mice were terminated at day 3 post-virus infection as colchicine treatment rapidly resulted in severe illness and mortality in CVB3-infected mice. Colchicine significantly decreased the number of macrophages in the heart in CVB3-infected mice (p<0.01) but significantly increased the number of neutrophils (p<0.01). In the pancreas, colchicine caused complete destruction of the acini in the CVB3-infected mice and also significantly decreased macrophage (p<0.01) and increased neutrophil numbers (p<0.01). In the spleen, colchicine treatment of CVB3-infected mice induced massive apoptosis in the white pulp and significantly inhibited the virus-induced increase of megakaryocytes in the spleen (p<0.001). Finally, we observed that colchicine significantly increased CVB3 levels in both the pancreas and the heart. CONCLUSIONS: Colchicine treatment in CVB3-induced myocarditis has a detrimental effect as it causes complete destruction of the exocrine pancreas and enhances viral load in both heart and pancreas.


Subject(s)
Colchicine/administration & dosage , Coxsackievirus Infections/drug therapy , Myocarditis/virology , Pancreas/pathology , Spleen/pathology , Animals , Colchicine/adverse effects , Colchicine/pharmacology , Coxsackievirus Infections/mortality , Disease Models, Animal , Enterovirus B, Human/physiology , Heart/drug effects , Heart/virology , Humans , Male , Mice , Mice, Inbred C3H , Myocarditis/drug therapy , Myocarditis/mortality , Pancreas/drug effects , Pancreas/virology , Spleen/drug effects , Spleen/virology , Treatment Outcome , Viral Load/drug effects
10.
Ultrasound Med Biol ; 42(8): 1919-28, 2016 08.
Article in English | MEDLINE | ID: mdl-27160847

ABSTRACT

High-mechanical-index ultrasound and intravenous microbubbles might prove beneficial in treating microvascular obstruction caused by microthrombi after primary percutaneous coronary intervention for ST-segment elevation myocardial infarction (STEMI). Experiments in animals have revealed that longer-pulse-duration ultrasound is associated with an improvement in microvascular recovery. This trial tested long-pulse-duration, high-mechanical-index ultrasound in STEMI patients. Non-randomly assigned, non-blinded patients were included in this phase 2 trial. The primary endpoint was any side effect possibly related to the ultrasound treatment. The study was aborted after six patients were included; three patients experienced coronary vasoconstriction of the culprit artery, unresponsive to nitroglycerin. Therefore, coronary artery diameter was measured in five pigs. Coronary artery diameters distal to the injury site decreased after application of ultrasound, after balloon injury plus thrombus injection (from 1.89 ± 0.24 mm before to 1.78 ± 0.17 after ultrasound, p = 0.05). Long-pulse-duration ultrasound might cause coronary vasoconstriction distal to the culprit vessel location.


Subject(s)
Coronary Vessels/physiopathology , Microvessels/physiopathology , Myocardial Infarction/therapy , Ultrasonic Therapy/adverse effects , Ultrasonic Therapy/methods , Vasoconstriction/physiology , Adult , Animals , Disease Models, Animal , Female , Humans , Incidence , Male , Middle Aged , Swine , Treatment Outcome , Young Adult
11.
Cardiovasc Pathol ; 25(1): 33-9, 2016.
Article in English | MEDLINE | ID: mdl-26476955

ABSTRACT

BACKGROUND: Complement activation contributes significantly to inflammation-related damage in the heart after acute myocardial infarction. Knowledge on factors that regulate postinfraction complement activation is incomplete however. In this study, we investigated whether endogenous C1-inhibitor, a well-known inhibitor of complement activation, is expressed in the heart after acute myocardial infarction. MATERIALS AND METHODS: C1-inhibitor and complement activation products C3d and C4d were analyzed immunohistochemically in the hearts of patients who died at different time intervals after acute myocardial infarction (n=28) and of control patients (n=8). To determine putative local C1-inhibitor production, cardiac transcript levels of the C1-inhibitor-encoding gene serping1 were determined in rats after induction of acute myocardial infarction (microarray). Additionally, C1-inhibitor expression was analyzed (fluorescence microscopy) in human endothelial cells and rat cardiomyoblasts in vitro. RESULTS: C1-inhibitor was found predominantly in and on jeopardized cardiomyocytes in necrotic infarct cores between 12h and 5days old. C1-inhibitor protein expression coincided in time and colocalized with C3d and C4d. In the rat heart, serping1 transcript levels were increased from 2h up until 7days after acute myocardial infarction. Both endothelial cells and cardiomyoblasts showed increased intracellular expression of C1-inhibitor in response to ischemia in vitro (n=4). CONCLUSIONS: These observations suggest that endogenous C1-inhibitor is likely involved in the regulation of complement activity in the myocardium following acute myocardial infarction. Observations in rat and in vitro suggest that C1-inhibitor is produced locally in the heart after acute myocardial infarction.


Subject(s)
Complement C1 Inactivator Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , RNA, Messenger/metabolism , Animals , Cell Line , Complement C1 Inhibitor Protein , Complement C3d/metabolism , Complement C4b/metabolism , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/pathology , Humans , Male , Myoblasts, Cardiac/metabolism , Myoblasts, Cardiac/pathology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/pathology , Necrosis , Peptide Fragments/metabolism , RNA, Messenger/genetics , Rats , Rats, Wistar , Retrospective Studies , Time Factors , Up-Regulation
12.
Ultrasound Med Biol ; 40(3): 532-40, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24361223

ABSTRACT

Ultrasound and microbubble-targeted delivery (UMTD) is a promising non-viral technique for genetic-based therapy. We found that UMTD of small interfering RNA (siRNA) is more effective than delivery of plasmid DNA (pDNA). UMTD (1 MHz, 0.22 MPa) of fluorescently labeled siRNA resulted in 97.9 ± 1.5% transfected cells, with siRNA localized homogenously in the cytoplasm directly after ultrasound exposure. UMTD of fluorescently labeled pDNA resulted in only 43.0 ± 4.2% transfected cells, with localization mainly in vesicular structures, co-localizing with endocytosis markers clathrin and caveolin. Delivery of siRNA against GAPDH (glyceraldehyde-3-phosphate dehydrogenase) effectively decreased protein levels to 24.3 ± 7.9% of non-treated controls (p < 0.01). In contrast, 24 h after delivery of pDNA encoding GAPDH, no increase in protein levels was detected. Transfection efficiency, verified with red fluorescently labeled pDNA encoding enhanced green fluorescent protein, revealed that of the transfected cells, only 2.0 ± 0.7% expressed the transgene. In conclusion, the difference in localization between siRNA and pDNA after UMTD is an important determinant of the effectiveness of these genetic-based technologies.


Subject(s)
Endothelial Cells/physiology , Endothelial Cells/radiation effects , Phospholipids/radiation effects , Plasmids/genetics , RNA, Small Interfering/genetics , Sonication/methods , Sulfur Hexafluoride/radiation effects , Transfection/methods , Animals , Cells, Cultured , Electroporation/methods , Endothelial Cells/cytology , High-Energy Shock Waves , Microbubbles , Plasmids/administration & dosage , RNA, Small Interfering/administration & dosage , Swine
13.
Ultrasound Med Biol ; 39(3): 490-506, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23347643

ABSTRACT

In this study, we investigated the effect of secondary Bjerknes forces on targeted microbubbles using high-speed optical imaging. We observed that targeted microbubbles attached to an underlying surface and subject to secondary Bjerknes forces deform in the direction of their neighboring bubble, thereby tending toward a prolate shape. The deformation induces an elastic restoring force, causing the bubbles to recoil back to their equilibrium position; typically within 100 µs after low-intensity ultrasound application. The temporal dynamics of the recoil was modeled as a simple mass-spring system, from which a value for the effective spring constant k of the order 10(-3) Nm(-1) was obtained. Moreover, the translational dynamics of interacting targeted microbubbles was predicted by a hydrodynamic point particle model, including a value of the spring stiffness k of the very same order as derived experimentally from the recoiling curves. For higher acoustic pressures, secondary Bjerknes forces rupture the molecular adhesion of the bubbles to the surface. We used this mutual attraction to quantify the binding force between a single biotinylated microbubble and an avidin-coated surface, which was found to be between 0.9 and 2 nanonewtons (nN). The observation of patches of lipids left at the initial binding site suggests that lipid anchors are pulled out of the microbubble shell, rather than biotin molecules unbinding from avidin. Understanding the effect of ultrasound application on targeted microbubbles is crucial for further advances in the realm of molecular imaging.


Subject(s)
Contrast Media , Microbubbles , Ultrasonics/methods , Avidin/chemistry , Biotin/chemistry , Elasticity , Image Processing, Computer-Assisted/methods , Lipids/chemistry , Models, Theoretical
14.
Cell Biochem Biophys ; 67(2): 341-52, 2013 Nov.
Article in English | MEDLINE | ID: mdl-22038300

ABSTRACT

Apoptosis of endothelial cells related to homocysteine (Hcy) has been reported in several studies. In this study, we evaluated whether reactive oxygen species (ROS)-producing signaling pathways contribute to Hcy-induced apoptosis induction, with specific emphasis on NADPH oxidases. Human umbilical vein endothelial cells were incubated with 0.01-2.5 mM Hcy. We determined the effect of Hcy on caspase-3 activity, annexin V positivity, intracellular NOX1, NOX2, NOX4, and p47(phox) expression and localization, nuclear nitrotyrosine accumulation, and mitochondrial membrane potential (ΔΨ m). Hcy induced caspase-3 activity and apoptosis; this effect was concentration dependent and maximal after 6-h exposure to 2.5 mM Hcy. It was accompanied by a significant increase in ΔΨ m. Cysteine was inactive on these parameters excluding a reactive thiol group effect. Hcy induced an increase in cellular NOX2, p47(phox), and NOX4, but not that of NOX1. 3D digital imaging microscopy followed by image deconvolution analysis showed nuclear accumulation of NOX2 and p47(phox) in endothelial cells exposed to Hcy, but not in control cells, which coincided with accumulation of nuclear nitrotyrosine residues. Furthermore, Hcy enhanced peri-nuclear localization of NOX4 coinciding with accumulation of peri-nuclear nitrotyrosine residues, a reflection of local ROS production. p47(phox) was also increased in the peri-nuclear region. The Hcy-induced increase in caspase-3 activity was prevented by DPI and apocynin, suggesting involvement of NOX activity. The data presented in this article reveal accumulation of nuclear NOX2 and peri-nuclear NOX4 accumulation as potential source of ROS production in Hcy-induced apoptosis in endothelial cells.


Subject(s)
Apoptosis/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Homocysteine/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrogen Peroxide/metabolism , Membrane Potential, Mitochondrial/drug effects , NADPH Oxidase 2 , NADPH Oxidase 4 , Nitric Oxide/metabolism , Protein Transport/drug effects
15.
Eur J Clin Invest ; 40(10): 893-902, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20854280

ABSTRACT

BACKGROUND: Clusterin (Apolipoprotein J), a plasma protein with cytoprotective and complement-inhibiting activities, localizes in the infarcted heart during myocardial infarction (MI). Recently, we have shown a protective effect of exogenous clusterin in vitro on ischaemically challenged cardiomyocytes independent of complement. We therefore hypothesized that intravenous clusterin administration would reduce myocardial infarction damage. METHODS: Wistar rats undergoing experimental MI, induced by 40 min ligation of a coronary vessel, were treated with either clusterin (n=15) or vehicle (n=13) intravenously, for 3 days post-MI. After 4 weeks, hearts were analysed. The putative role of megalin, a clusterin receptor, was also studied. RESULTS: Administration of human clusterin significantly reduced both infarct size (with 75 ± 5%) and death of animals (23% vehicle group vs. 0% clusterin group). Importantly, histochemical analysis showed no signs of impaired wound healing in the clusterin group. In addition, significantly increased numbers of macrophages were found in the clusterin group. We also found that the clusterin receptor megalin was present on cardiomyocytes in vitro which, however, was not influenced by ischaemia. Human clusterin co-localized with this receptor in vitro, but not in the human heart. In addition, using a megalin inhibitor, we found that clusterin did not exert its protective effect on cardiomyocytes through megalin. CONCLUSIONS: Our results thus show that clusterin has a protective effect on cardiomyocytes after acute myocardial infarction in vivo, independent of its receptor megalin. This indicates that clusterin, or a clusterin derivate, is a potential therapeutic agent in the treatment of MI.


Subject(s)
Clusterin/therapeutic use , Myocardial Infarction/therapy , Myocardium/metabolism , Animals , Immunohistochemistry , Low Density Lipoprotein Receptor-Related Protein-2/therapeutic use , Myocardial Infarction/physiopathology , Rats , Rats, Wistar , Treatment Outcome
16.
Ultrasound Med Biol ; 35(11): 1917-27, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19766381

ABSTRACT

Recent developments in the field of ultrasound (US) contrast agents have demonstrated that these encapsulated microbubbles can not only be used for diagnostic imaging but may also be employed as therapeutic carriers for localized, targeted drug or gene delivery. The exact mechanisms behind increased uptake of therapeutic compounds by US-exposed microbubbles are still not fully understood. Therefore, we studied the effects of stably oscillating SonoVue microbubbles on relevant parameters of cellular and intercellular permeability, i.e., reactive oxygen species (ROS) homeostasis, calcium permeability, F-actin cytoskeleton, monolayer integrity and cell viability using live-cell fluorescence microscopy. US was applied at 1-MHz, 0.1MPa peak-negative pressure, 0.2% duty cycle and 20Hz pulse repetition frequency to primary endothelial cells. We demonstrated increased membrane permeability for calcium ions, with an important role for H(2)O(2). Catalase, an extracellular H(2)O(2) scavenger, significantly blocked the influx of calcium ions. Further changes in ROS homeostasis involved an increase in intracellular H(2)O(2) levels, protein nitrosylation and a decrease in total endogenous glutathione levels. In addition, an increase in the number of F-actin stress fibers and F-actin cytoskeletal rearrangement were observed. Furthermore, US-exposed microbubbles significantly affected endothelial monolayer integrity, but importantly, disrupted cell-cell interactions were restored within 30min. Finally, cell viability was not affected. In conclusion, these data provide more insight in the interactions between US, microbubbles and endothelial cells, which is important for understanding the mechanisms behind US and microbubble-enhanced uptake of drugs or genes.


Subject(s)
Contrast Media/pharmacology , Endothelial Cells/cytology , Endothelial Cells/diagnostic imaging , Endothelium, Vascular/cytology , Endothelium, Vascular/diagnostic imaging , Phospholipids/pharmacology , Sulfur Hexafluoride/pharmacology , Actins/metabolism , Calcium/metabolism , Cell Membrane Permeability/drug effects , Cell Survival , Cells, Cultured , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Homeostasis , Humans , Infant, Newborn , Microbubbles , Microscopy, Fluorescence/methods , Reactive Oxygen Species/metabolism , Ultrasonography
17.
Circ Res ; 104(5): 679-87, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19168443

ABSTRACT

Contrast microbubbles in combination with ultrasound (US) are promising vehicles for local drug and gene delivery. However, the exact mechanisms behind intracellular delivery of therapeutic compounds remain to be resolved. We hypothesized that endocytosis and pore formation are involved during US and microbubble targeted delivery (UMTD) of therapeutic compounds. Therefore, primary endothelial cells were subjected to UMTD of fluorescent dextrans (4.4 to 500 kDa) using 1 MHz pulsed US with 0.22-MPa peak-negative pressure, during 30 seconds. Fluorescence microscopy showed homogeneous distribution of 4.4- and 70-kDa dextrans through the cytosol, and localization of 155- and 500-kDa dextrans in distinct vesicles after UMTD. After ATP depletion, reduced uptake of 4.4-kDa dextran and no uptake of 500-kDa dextran was observed after UMTD. Independently inhibiting clathrin- and caveolae-mediated endocytosis, as well as macropinocytosis significantly decreased intracellular delivery of 4.4- to 500-kDa dextrans. Furthermore, 3D fluorescence microscopy demonstrated dextran vesicles (500 kDa) to colocalize with caveolin-1 and especially clathrin. Finally, after UMTD of dextran (500 kDa) into rat femoral artery endothelium in vivo, dextran molecules were again localized in vesicles that partially colocalized with caveolin-1 and clathrin. Together, these data indicated uptake of molecules via endocytosis after UMTD. In addition to triggering endocytosis, UMTD also evoked transient pore formation, as demonstrated by the influx of calcium ions and cellular release of preloaded dextrans after US and microbubble exposure. In conclusion, these data demonstrate that endocytosis is a key mechanism in UMTD besides transient pore formation, with the contribution of endocytosis being dependent on molecular size.


Subject(s)
Caveolae/metabolism , Dextrans/metabolism , Drug Delivery Systems , Endocytosis , Endothelial Cells/metabolism , Fluorescent Dyes/metabolism , Microbubbles , Ultrasonics , Adenosine Triphosphate/metabolism , Androstadienes/pharmacology , Animals , Biological Transport , Cattle , Caveolin 1/metabolism , Cells, Cultured , Chlorpromazine/pharmacology , Clathrin/metabolism , Contrast Media/administration & dosage , Cytosol/metabolism , Dextrans/administration & dosage , Dextrans/chemistry , Endocytosis/drug effects , Endothelial Cells/drug effects , Femoral Artery/metabolism , Filipin/pharmacology , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/chemistry , Imaging, Three-Dimensional , Infusions, Intravenous , Microscopy, Fluorescence , Molecular Weight , Phospholipids/administration & dosage , Pinocytosis , Pressure , Rats , Rats, Wistar , Sulfur Hexafluoride/administration & dosage , Time Factors , Transport Vesicles/metabolism , Wortmannin
18.
Ultrasound Med Biol ; 34(3): 502-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17993242

ABSTRACT

Ultrasound (US) contrast agents have gained wide interest in gene therapy as many researchers reported increased membrane permeability and transfection efficiency by sonoporation in the presence of US contrast agents. We recently demonstrated an increase in cell membrane permeability for Ca2+ in rat cardiomyoblast (H9c2) cells insonified in the presence of microbubbles. In the present study, we specifically investigated whether US-exposed microbubbles have an effect on the cell membrane potential and whether Ca2+-dependent potassium (BK(Ca)) channels are involved. We particularly focused on local events where the microbubble was in contact with the cell membrane. H9c2 cells were cultured on US transparent membranes. US exposure consisted of bursts with a frequency of 1 MHz with a peak-to-peak pressure of 0.1 or 0.5 MPa. Pulse repetition frequency was set to 20 Hz, with a duty cycle of 0.2%. Cells were insonified during 30 s in the presence of Sonovue(trade mark) microbubbles. The membrane potential was monitored during US exposure using the fluorescent dye di-4-aminonaphtylethenylpyridinium (di-4-ANEPPS). The experiments were repeated in the presence of iberiotoxin (100 nM), a specific inhibitor of BK(Ca) channels. Surprisingly, despite the previously reported Ca(2+) influx, we found patches of hyperpolarization of the cell membrane, as reflected by local increases in di-4-ANEPPS mean intensity of fluorescence (MIF) to 118.6 +/- 2.5% (p < 0.001, n = 267) at 0.1 MPa and 125.7 +/- 5.9% (p < 0.001, n = 161) at 0.5 MPa at t = 74 s, respectively, compared with "no US" (100.3 +/- 3.4%, n = 52). This hyperpolarization was caused by the activation of BK(Ca) channels, as iberiotoxin completely prevented hyperpolarization. (MIF(t74) = 100.6 +/- 1.4%; p < 0.001, n = 267) and 0.5 MPa (MIF(t74) = 88.8 +/- 2.0%; p< 0.001, n = 193), compared with 0.1 and 0.5 MPa microbubbles without iberiotoxin. In conclusion, US-exposed microbubbles elicit a Ca2+ influx, which leads to activation of BK(Ca) channels and a subsequent, local hyperpolarization of the cell membrane. This local hyperpolarization of the cell membrane may facilitate uptake of macromolecules through endocytosis and macropinocytosis. (E-mail: ljm.juffermans@vumc.nl).


Subject(s)
Myocytes, Cardiac/diagnostic imaging , Ultrasonics , Animals , Cell Line , Cell Membrane Permeability/drug effects , Hydrogen Peroxide/metabolism , Membrane Potentials/drug effects , Microbubbles , Microscopy, Fluorescence , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phospholipids/pharmacology , Potassium Channels, Calcium-Activated/drug effects , Rats , Sulfur Hexafluoride/pharmacology , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL
...