Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Ann Vasc Surg ; 42: 293-298, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28300679

ABSTRACT

BACKGROUND: Arterial blood pressure-induced shear stress causes endothelial cell apoptosis and inflammation in vein grafts after coronary artery bypass grafting. As the inflammatory protein type IIA secretory phospholipase A2 (sPLA2-IIA) has been shown to progress atherosclerosis, we hypothesized a role for sPLA2-IIA herein. METHODS: The effects of PX-18, an inhibitor of both sPLA2-IIA and apoptosis, on residual endothelium and the presence of sPLA2-IIA were studied in human saphenous vein segments (n = 6) perfused at arterial blood pressure with autologous blood for 6 hrs. RESULTS: The presence of PX-18 in the perfusion blood induced a significant 20% reduction in endothelial cell loss compared to veins perfused without PX18, coinciding with significantly reduced sPLA2-IIA levels in the media of the vein graft wall. In addition, PX-18 significantly attenuated caspase-3 activation in human umbilical vein endothelial cells subjected to shear stress via mechanical stretch independent of sPLA2-IIA. CONCLUSIONS: In conclusion, PX-18 protects saphenous vein endothelial cells from arterial blood pressure-induced death, possibly also independent of sPLA2-IIA inhibition.


Subject(s)
Alkanesulfonic Acids/pharmacology , Arterial Pressure , Endothelial Cells/drug effects , Group II Phospholipases A2/antagonists & inhibitors , Mechanotransduction, Cellular/drug effects , Oleic Acids/pharmacology , Phospholipase A2 Inhibitors/pharmacology , Saphenous Vein/drug effects , Apoptosis/drug effects , Cells, Cultured , Endothelial Cells/enzymology , Endothelial Cells/pathology , Group II Phospholipases A2/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Saphenous Vein/enzymology , Saphenous Vein/pathology , Time Factors
2.
PLoS One ; 9(9): e107494, 2014.
Article in English | MEDLINE | ID: mdl-25244123

ABSTRACT

Duchenne muscular dystrophy (DMD) is characterized by the absence or reduced levels of dystrophin expression on the inner surface of the sarcolemmal membrane of muscle fibers. Clinical development of therapeutic approaches aiming to increase dystrophin levels requires sensitive and reproducible measurement of differences in dystrophin expression in muscle biopsies of treated patients with DMD. This, however, poses a technical challenge due to intra- and inter-donor variance in the occurrence of revertant fibers and low trace dystrophin expression throughout the biopsies. We have developed an immunofluorescence and semi-automated image analysis method that measures the sarcolemmal dystrophin intensity per individual fiber for the entire fiber population in a muscle biopsy. Cross-sections of muscle co-stained for dystrophin and spectrin have been imaged by confocal microscopy, and image analysis was performed using Definiens software. Dystrophin intensity has been measured in the sarcolemmal mask of spectrin for each individual muscle fiber and multiple membrane intensity parameters (mean, maximum, quantiles per fiber) were calculated. A histogram can depict the distribution of dystrophin intensities for the fiber population in the biopsy. This method was tested by measuring dystrophin in DMD, Becker muscular dystrophy, and healthy muscle samples. Analysis of duplicate or quadruplicate sections of DMD biopsies on the same or multiple days, by different operators, or using different antibodies, was shown to be objective and reproducible (inter-assay precision, CV 2-17% and intra-assay precision, CV 2-10%). Moreover, the method was sufficiently sensitive to detect consistently small differences in dystrophin between two biopsies from a patient with DMD before and after treatment with an investigational compound.


Subject(s)
Dystrophin/metabolism , Fluorescent Antibody Technique/methods , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Biopsy , Humans , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Duchenne/pathology , Reproducibility of Results , Sensitivity and Specificity
3.
Mol Ther Nucleic Acids ; 3: e148, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24549299

ABSTRACT

Antisense-mediated exon skipping is currently in clinical development for Duchenne muscular dystrophy (DMD) to amend the consequences of the underlying genetic defect and restore dystrophin expression. Due to turnover of compound, transcript, and protein, chronic treatment with effector molecules (antisense oligonucleotides) will be required. To investigate the dynamics and persistence of antisense 2'-O-methyl phosphorothioate oligonucleotides, exon skipping, and dystrophin expression after dosing was concluded, mdx mice were treated subcutaneously for 8 weeks with 100 mg/kg oligonucleotides twice weekly. Thereafter, mice were sacrificed at different time points after the final injection (36 hours-24 weeks). Oligonucleotide half-life was longer in heart (~65 days) compared with that in skeletal muscle, liver, and kidney (~35 days). Exon skipping half-lives varied between 33 and 53 days, whereas dystrophin protein showed a long half-life (>100 days). Oligonucleotide and exon-skipping levels peaked in the first week and declined thereafter. By contrast, dystrophin expression peaked after 3-8 weeks and then slowly declined, remaining detectable after 24 weeks. Concordance between levels of oligonucleotides, exon skipping, and proteins was observed, except in heart, wherein high oligonucleotide levels but low exon skipping and dystrophin expression were seen. Overall, these results enhance our understanding of the pharmacokinetics and pharmacodynamics of 2'-O-methyl phosphorothioate oligos used for the treatment of DMD.Molecular Therapy-Nucleic Acids (2014) 3, e148; doi:10.1038/mtna.2014.1; published online 18 February 2014.

4.
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
5.
Atherosclerosis ; 221(1): 48-54, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22204864

ABSTRACT

OBJECTIVE: We have previously shown that homocysteine (Hcy) induces phosphatidylserine (PS) exposure, apoptosis and necrosis in human endothelial cells. Since it has been suggested that S-adenosylhomocysteine (SAH) is the main causative factor in Hcy-induced pathogenesis of cardiovascular disease, we evaluate here whether the cytotoxic Hcy effect in endothelial cells is also SAH dependent. METHODS AND RESULTS: Human umbilical vein endothelial cells (HUVECs) were exposed to the following conditions: (1) non-treated control (resulting in 2.8 nM intracellular SAH and 3.1 µM extracellular l-Hcy); and incubation with (2) 50 µM adenosine-2,3-dialdehyde (ADA; resulting in 17.7 nM intracellular SAH and 3.1 µM extracellular l-Hcy), (3) 2.5 mM Hcy (resulting in 20.9 nM intracellular SAH and 1.8 mM extracellular l-Hcy), and (4) 1, 10 and 100 µM SAH. We then determined the effect of treatment on annexin V-positivity, caspase-3 activity, cytochrome c release (sub)cellular expression of NOX2, NOX4, p47(phox) and nitrotyrosine, and H(2)O(2). Both Hcy and ADA significantly increased PS exposure (n=5), caspase-3 activity (n=6) and cytochrome c release (n=3). Incubation with extracellular SAH alone did not affect cell viability. Both Hcy and ADA also induced similar increases in nuclear NOX2 and (peri)nuclear NOX4, coinciding with (peri)nuclear p47(phox) expression and local reactive oxygen species (ROS) (n=3). Inhibition of NOX-mediated ROS by the flavoenzyme inhibitor diphenylene iodonium (DPI) significantly decreased apoptosis induction (n=3) and ROS production (n=3). CONCLUSION: SAH induces PS exposure and apoptosis in endothelial cells independently of Hcy. Our study therefore shows that Hcy-mediated endothelial dysfunction, as determined in the cell model used, is mainly due to SAH accumulation.


Subject(s)
Apoptosis , Endothelial Cells/metabolism , Homocysteine/metabolism , Phosphatidylserines/metabolism , S-Adenosylhomocysteine/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Cell Survival , Cells, Cultured , Cytochromes c/metabolism , Endothelial Cells/drug effects , Endothelial Cells/pathology , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Peroxide/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Onium Compounds/pharmacology , Tyrosine/analogs & derivatives , Tyrosine/metabolism
6.
Basic Res Cardiol ; 107(1): 233, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22167343

ABSTRACT

Pro-coagulant and pro-inflammatory intramyocardial (micro)vasculature plays an important role in acute myocardial infarction (AMI). Currently, inhibition of serine protease dipeptidyl peptidase 4 (DPP4) receives a lot of interest as an anti-hyperglycemic therapy in type 2 diabetes patients. However, DPP4 also possesses anti-thrombotic properties and may behave as an immobilized anti-coagulant on endothelial cells. Here, we studied the expression and activity of endothelial DPP4 in human myocardial infarction in relation to a prothrombogenic endothelial phenotype. Using (immuno)histochemistry, DPP4 expression and activity were found on the endothelium of intramyocardial blood vessels in autopsied control hearts (n = 9). Within the infarction area of AMI patients (n = 73), this DPP4 expression and activity were significantly decreased, coinciding with an increase in Tissue Factor expression. In primary human umbilical vein endothelial cells (HUVECs), Western blot analysis and digital imaging fluorescence microscopy revealed that DPP4 expression was strongly decreased after metabolic inhibition, also coinciding with Tissue Factor upregulation. Interestingly, inhibition of DPP4 activity with diprotin A also enhanced the amount of Tissue Factor encountered and induced the adherence of platelets under flow conditions. Ischemia induces loss of coronary microvascular endothelial DPP4 expression and increased Tissue Factor expression in AMI as well as in vitro in HUVECs. Our data suggest that the loss of DPP4 activity affects the anti-thrombogenic nature of the endothelium.


Subject(s)
Coronary Thrombosis/enzymology , Coronary Vessels/enzymology , Dipeptidyl Peptidase 4/metabolism , Microvessels/enzymology , Myocardial Infarction/enzymology , Adult , Aged , Aged, 80 and over , Female , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Male , Middle Aged , Myocardium/enzymology , Platelet Adhesiveness , Thromboplastin/metabolism
7.
Cell Physiol Biochem ; 28(1): 53-62, 2011.
Article in English | MEDLINE | ID: mdl-21865848

ABSTRACT

AIMS: Increased levels of homocysteine (Hcy) form an independent risk factor for cardiovascular disease. In a previous study we have shown that Hcy induced phosphatidylserine (PS) exposure to the outer leaflet of the plasma membrane in cardiomyocytes, inducing a pro-inflammatory phenotype. In the present study the mechanism(s) involved in Hcy-induced PS exposure were analyzed. METHODS: H9c2 rat cardiomyoblasts were subjected to 2.5 mM D,L-Hcy and analyzed for RhoA translocation and activity, Rho Kinase (ROCK) activity and expression and flippase activity. In addition, the effect of ROCK inhibition with Y27632 on Hcy-induced PS exposure and flippase activity was analyzed. Furthermore, GTP and ATP levels were determined. RESULTS: Incubation of H9c2 cells with 2.5 mM D,L-Hcy did not inhibit RhoA translocation to the plasma membrane. Neither did it inhibit activation of RhoA, even though GTP levels were significantly decreased. Hcy did significantly inhibit ROCK activation, but not its expression, and did inhibit flippase activity, in advance of a significant decrease in ATP levels. ROCK inhibition via Y27632 did not have significant added effects on this. CONCLUSION: Hcy induced PS exposure in the outer leaflet of the plasma membrane in cardiomyocytes via inhibition of ROCK and flippase activity. As such Hcy may induce cardiomyocytes vulnerable to inflammation in vivo in hyperhomocysteinaemia patients.


Subject(s)
Homocysteine/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Phosphatidylserines/metabolism , Phospholipid Transfer Proteins/antagonists & inhibitors , rho-Associated Kinases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Amides/pharmacology , Animals , Cells, Cultured , Guanosine Triphosphate/metabolism , Phospholipid Transfer Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Rats , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
8.
Mol Cell Biochem ; 358(1-2): 229-39, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21739151

ABSTRACT

We previously found that homocysteine (Hcy) induced plasma membrane flip-flop, apoptosis, and necrosis in cardiomyocytes. Inactivation of flippase by Hcy induced membrane flip-flop, while apoptosis was induced via a NOX2-dependent mechanism. It has been suggested that S-adenosylhomocysteine (SAH) is the main causative factor in hyperhomocysteinemia (HHC)-induced pathogenesis of cardiovascular disease. Therefore, we evaluated whether the observed cytotoxic effect of Hcy in cardiomyocytes is SAH dependent. Rat cardiomyoblasts (H9c2 cells) were treated under different conditions: (1) non-treated control (1.5 nM intracellular SAH with 2.8 µM extracellular L -Hcy), (2) incubation with 50 µM adenosine-2,3-dialdehyde (ADA resulting in 83.5 nM intracellular SAH, and 1.6 µM extracellular L -Hcy), (3) incubation with 2.5 mM D, L -Hcy (resulting in 68 nM intracellular SAH and 1513 µM extracellular L -Hcy) with or without 10 µM reactive oxygen species (ROS)-inhibitor apocynin, and (4) incubation with 100 nM, 10 µM, and 100 µM SAH. We then determined the effect on annexin V/propodium iodide positivity, flippase activity, caspase-3 activity, intracellular NOX2 and p47(phox) expression and localization, and nuclear ROS production. In contrast to Hcy, ADA did not induce apoptosis, necrosis, or membrane flip-flop. Remarkably, both ADA and Hcy induced a significant increase in nuclear NOX2 expression. However, in contrast to ADA, Hcy additionally induced nuclear p47(phox) expression, increased nuclear ROS production, and inactivated flippase. Incubation with SAH did not have an effect on cell viability, nor on flippase activity, nor on nuclear NOX2-, p47phox expression or nuclear ROS production. HHC-induced membrane flip-flop and apoptosis in cardiomyocytes is due to increased Hcy levels and not primarily related to increased intracellular SAH, which plays a crucial role in nuclear p47(phox) translocation and subsequent ROS production.


Subject(s)
Apoptosis/drug effects , Cell Membrane/metabolism , Cell Nucleus/enzymology , Homocysteine/pharmacology , Myocytes, Cardiac/cytology , NADPH Oxidases/metabolism , S-Adenosylhomocysteine/pharmacology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine Triphosphate/metabolism , Animals , Caspase 3/metabolism , Cell Membrane/drug effects , Cell Nucleus/drug effects , Cell Survival/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Homocysteine/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Membrane Glycoproteins/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , NADPH Oxidase 2 , Phospholipid Transfer Proteins/metabolism , Rats , Reactive Oxygen Species/metabolism , S-Adenosylmethionine/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
9.
N Engl J Med ; 364(16): 1513-22, 2011 Apr 21.
Article in English | MEDLINE | ID: mdl-21428760

ABSTRACT

BACKGROUND: Local intramuscular administration of the antisense oligonucleotide PRO051 in patients with Duchenne's muscular dystrophy with relevant mutations was previously reported to induce the skipping of exon 51 during pre-messenger RNA splicing of the dystrophin gene and to facilitate new dystrophin expression in muscle-fiber membranes. The present phase 1-2a study aimed to assess the safety, pharmacokinetics, and molecular and clinical effects of systemically administered PRO051. METHODS: We administered weekly abdominal subcutaneous injections of PRO051 for 5 weeks in 12 patients, with each of four possible doses (0.5, 2.0, 4.0, and 6.0 mg per kilogram of body weight) given to 3 patients. Changes in RNA splicing and protein levels in the tibialis anterior muscle were assessed at two time points. All patients subsequently entered a 12-week open-label extension phase, during which they all received PRO051 at a dose of 6.0 mg per kilogram per week. Safety, pharmacokinetics, serum creatine kinase levels, and muscle strength and function were assessed. RESULTS: The most common adverse events were irritation at the administration site and, during the extension phase, mild and variable proteinuria and increased urinary α(1)-microglobulin levels; there were no serious adverse events. The mean terminal half-life of PRO051 in the circulation was 29 days. PRO051 induced detectable, specific exon-51 skipping at doses of 2.0 mg or more per kilogram. New dystrophin expression was observed between approximately 60% and 100% of muscle fibers in 10 of the 12 patients, as measured on post-treatment biopsy, which increased in a dose-dependent manner to up to 15.6% of the expression in healthy muscle. After the 12-week extension phase, there was a mean (±SD) improvement of 35.2±28.7 m (from the baseline of 384±121 m) on the 6-minute walk test. CONCLUSIONS: Systemically administered PRO051 showed dose-dependent molecular efficacy in patients with Duchenne's muscular dystrophy, with a modest improvement in the 6-minute walk test after 12 weeks of extended treatment. (Funded by Prosensa Therapeutics; Netherlands National Trial Register number, NTR1241.).


Subject(s)
Alternative Splicing , Muscular Dystrophy, Duchenne/drug therapy , Oligonucleotides/therapeutic use , Adolescent , Child , Child, Preschool , Creatine Kinase/urine , Dose-Response Relationship, Drug , Dystrophin/genetics , Dystrophin/metabolism , Exercise Test , Exons , Humans , Injections, Subcutaneous , Male , Muscle Strength/drug effects , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Mutation , Oligonucleotides/administration & dosage , Oligonucleotides/adverse effects , Oligonucleotides/blood , RNA/analysis
10.
J Mol Cell Cardiol ; 49(5): 781-90, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20691698

ABSTRACT

Subsequent to myocardial infarction, cardiomyocytes within the infarcted areas and border zones expose phosphatidylserine (PS) in the outer plasma membrane leaflet (flip-flop). We showed earlier that in addition to apoptosis, this flip-flop can be reversible in cardiomyocytes. We now investigated a possible role for Rho and downstream effector Rho-associated kinase (ROCK) in the process of (reversible) PS exposure and apoptosis in cardiomyocytes. In rat cardiomyoblasts (H9c2 cells) and isolated adult ventricular rat cardiomyocytes Clostridium difficile Toxin B (TcdB), a Rho GTPase family inhibitor, C3 transferase (C3), a Rho(A,B,C) inhibitor and the ROCK inhibitors Y27632 and H1152 were used to inhibit Rho-ROCK signaling. PS exposure was assessed via flow cytometry and fluorescent digital imaging microscopy using annexin V. Akt expression and phosphorylation were analyzed via Western blot, and Akt activity was inhibited by wortmannin. The cellular concentration activated caspase 3 was determined as a measure of apoptosis, and flippase activity was assessed via flow cytometry using NBD-labeled PS. TcdB, C3, Y27632 and H1152 all significantly increased PS exposure. TcdB, Y27632 and H1152 all significantly inhibited phosphorylation of the anti-apoptotic protein Akt and Akt inhibition by wortmannin lead to increased PS exposure. However, only TcdB and C3, but not ROCK- or Akt inhibition led to caspase 3 activation and thus apoptosis. Notably, pancaspase inhibitor zVAD only partially inhibited TcdB-induced PS exposure indicating the existence of apoptotic and non-apoptotic PS exposure. The induced PS exposure coincided with decreased flippase activity as measured with NBD-labeled PS flip-flop. In this study, we show a regulatory role for a novel signaling route, Rho-ROCK-flippase signaling, in maintaining asymmetrical membrane phospholipid distribution in cardiomyocytes.


Subject(s)
Apoptosis , Myocytes, Cardiac/enzymology , Phosphatidylserines/metabolism , Phospholipid Transfer Proteins/metabolism , Signal Transduction , rho GTP-Binding Proteins/antagonists & inhibitors , rho-Associated Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , ADP Ribose Transferases/pharmacology , Adenosine Triphosphate/metabolism , Amides/pharmacology , Animals , Apoptosis/drug effects , Bacterial Proteins/pharmacology , Bacterial Toxins/pharmacology , Botulinum Toxins/pharmacology , Caspase 3/metabolism , Caspase Inhibitors , Cell Line , Cell Separation , Enzyme Activation/drug effects , Humans , Myocytes, Cardiac/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/pharmacology , Rats , Signal Transduction/drug effects , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
11.
Apoptosis ; 12(8): 1407-18, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17440815

ABSTRACT

BACKGROUND: Hyperhomocysteinaemia (HHC) is thought to be a risk factor for cardiovascular disease including heart failure. While numerous studies have analyzed the role of homocysteine (Hcy) in the vasculature, only a few studies investigated the role of Hcy in the heart. Therefore we have analyzed the effects of Hcy on isolated cardiomyocytes. METHODS: H9c2 cells (rat cardiomyoblast cells) and adult rat cardiomyocytes were incubated with Hcy and were analyzed for cell viability. Furthermore, we determined the effects of Hcy on intracellular mediators related to cell viability in cardiomyocytes, namely NOX2, reactive oxygen species (ROS), mitochondrial membrane potential (DeltaPsi (m)) and ATP concentrations. RESULTS: We found that incubation of H9c2 cells with 0.1 mM D,L-Hcy (= 60 microM L-Hcy) resulted in an increase of DeltaPsi (m) as well as ATP concentrations. 1.1 mM D,L-Hcy (= 460 microM L-Hcy) induced reversible flip-flop of the plasma membrane phospholipids, but not apoptosis. Incubation with 2.73 mM D,L-Hcy (= 1.18 mM L-Hcy) induced apoptosis and necrosis. This loss of cell viability was accompanied by a thread-to-grain transition of the mitochondrial reticulum, ATP depletion and nuclear NOX2 expression coinciding with ROS production as evident from the presence of nitrotyrosin residues. Notably, only at this concentration we found a significant increase in S-adenosylhomocysteine which is considered the primary culprit in HHC. CONCLUSION: We found concentration-dependent effects of Hcy in cardiomyocytes, varying from induction of reversible flip-flop of the plasma membrane phospholipids, to apoptosis and necrosis.


Subject(s)
Apoptosis/drug effects , Cell Membrane/drug effects , Homocysteine/pharmacology , Membrane Fluidity/drug effects , Myocytes, Cardiac/drug effects , Adenosine Triphosphate/metabolism , Animals , Caspase 3/metabolism , Cell Membrane/metabolism , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Homocysteine/analysis , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/drug effects , Models, Biological , Myocytes, Cardiac/cytology , Myocytes, Cardiac/pathology , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Necrosis/chemically induced , Phospholipids/metabolism , Protein Processing, Post-Translational , Rats , S-Adenosylhomocysteine/analysis , S-Adenosylmethionine/analysis
SELECTION OF CITATIONS
SEARCH DETAIL