Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Curr Mol Med ; 14(10): 1247-51, 2014.
Article in English | MEDLINE | ID: mdl-25470293

ABSTRACT

Over the past few years it has become clear that mitochondria are not merely the powerhouses of cells. Proteome-analyses of mitochondria from different organisms and organs revealed that more than 1000 proteins are localized in and/or on mitochondria. This by far exceeds the number of proteins required for classical mitochondrial functions, e.g. the respiratory chain, the tricarboxylic acid cycle, fatty acid oxidation and apoptosis. This suggests that many of these proteins have other, as yet unknown functions. Several proteins with well-described nuclear functions, like the transcription factor FoxO3A or the Telomerase Reverse Transcriptase, have recently been shown to be localized also within the mitochondria. This mini-review will focus on the description of the functions of these two proteins in the nucleus and in the mitochondria - as two examples of many more proteins, which are yet to be uncovered. It will give insights into the role of these proteins within different organelles of the cell and will reveal that the functions of the proteins are probably not the same in the nucleus and the mitochondria. Therefore, these differences have to be considered when targeting proteins for therapeutic approaches.


Subject(s)
Aging/genetics , Forkhead Transcription Factors/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Telomerase/genetics , Aging/metabolism , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Humans , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protein Binding , Protein Transport , Reactive Oxygen Species/metabolism , Signal Transduction , Telomerase/metabolism
2.
Z Gerontol Geriatr ; 46(7): 635-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23958997

ABSTRACT

The endothelium is located in a strategic anatomical position within the blood vessel wall and thus constitutes a barrier between the blood and all tissues. The integrity of the endothelial cells, which line the entire circulatory system like wallpaper, is essential to prevent the onset of cardiovascular disorders. Aging is one of the major risk factors for the development of heart and vascular diseases. However, over the past years it has become clear that the functional capacity of endothelial cells declines with age and that physiological aging occurs independently of pathological changes. One important mechanism contributing to the onset of the aging. process is the disturbance of the cellular redox homeostasis. Two key molecules involved in maintaining the delicate balance between oxidative and antioxidative systems are NADPH oxidase 4, an enzyme whose sole function is to produce reactive oxygen species and the oxidoreductase thioredoxin-1, which reduces oxidized proteins. Therefore, this review will focus on the role of these two proteins in cardiovascular aging.


Subject(s)
Aging/physiology , Cellular Senescence/physiology , Endothelial Cells/physiology , Models, Cardiovascular , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Thioredoxins/metabolism , Animals , Endothelial Cells/cytology , Humans , NADPH Oxidase 4 , Oxidation-Reduction , Oxidative Stress/physiology
4.
Z Gerontol Geriatr ; 45(2): 90-4, 2012 Feb.
Article in German | MEDLINE | ID: mdl-22262415

ABSTRACT

Reactive oxygen species (ROS) are important signaling molecules in human cells. At physiological concentrations, they can for instance protect against apoptosis and act as secondary messengers in many different signaling pathways. Disturbance of redox homeostasis, i.e., the physiological balance between ROS generation and degradation, leads to not only increased ROS levels, so-called oxidative stress, but also results in damage to macromolecules and promotes the development of diseases and accelerates the aging process. The organism has various enzyme systems at hand to eliminate excess ROS. Their inactivation or degradation under conditions of oxidative stress is tightly linked to endothelial dysfunction due to endothelial cell apoptosis, a loss of telomerase activity, and telomere shortening. Restricted endothelial function causes cardiovascular diseases and diabetes type 2.


Subject(s)
Cardiovascular Diseases/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Endothelial Cells/metabolism , Models, Cardiovascular , Oxidative Stress , Reactive Oxygen Species/metabolism , Animals , Cardiovascular Diseases/etiology , Diabetes Mellitus, Type 2/complications , Humans
5.
Z Gerontol Geriatr ; 40(5): 334-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17943236

ABSTRACT

Aging is one major risk factor for the incidence of coronary artery disease and the development of atherosclerosis. The functional integrity of the endothelial cell monolayer is essential to prevent lesion formation. Endothelial cells show profound changes with age. However, the molecular mechanisms are not well understood. Important players in the process of endothelial cell aging are reactive oxygen species, nitric oxide bioavailability, mitochondrial integrity and the activity of telomerase reverse transcriptase. This review will demonstrate the evidence that these processes are involved in endothelial cell aging and linked to each other. The future goal of understanding endothelial cell aging would allow for an anti-aging therapy to reduce the influence of aging in the development of atherosclerosis and coronary artery disease.


Subject(s)
Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cellular Senescence , Endothelial Cells/metabolism , Endothelial Cells/pathology , Models, Cardiovascular , Telomerase/metabolism , Animals , Humans , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism
6.
Circ Res ; 89(8): 709-15, 2001 Oct 12.
Article in English | MEDLINE | ID: mdl-11597994

ABSTRACT

Advanced aging leads to impaired endothelial NO synthesis and enhanced endothelial cell apoptosis; therefore, we investigated the sensitivity of aged endothelial cells toward apoptotic stimuli and determined the role of NO. Human umbilical vein endothelial cells (HUVECs) were cultured until 14th passage. In aged cells, oxLDL and tumor necrosis factor-alpha-induced apoptosis and caspase-3-like activity were significantly enhanced more than 3-fold compared with young cells (passage 3). Because NO contributes to protection against endothelial cell death via S-nitrosylation of caspases, we determined endothelial NO synthase (eNOS) protein expression and the content of S-nitrosylated proteins. Aged HUVECs showed significantly reduced eNOS expression (35+/-10%) and a decrease in the overall S-NO content (33+/-3%), suggesting that eNOS downregulation may be involved in age-dependent increase of apoptosis sensitivity. Indeed, eNOS knockout endothelial cells showed a significantly enhanced apoptosis induction. Exogenous NO donors abolished increased apoptosis and caspase-3-like activity. In contrast, the application of shear stress, which exerts a profound apoptosis inhibitory effect via upregulation of NO synthesis in young cells, failed to inhibit apoptosis in aged cells. Moreover, no upregulation of eNOS protein expression and S-NO content in response to shear stress was detected in aged cells. Overexpression of wild-type eNOS completely restored the antiapoptotic effect of shear stress, whereas only a partial inhibitory effect was detected under steady conditions. Strikingly, transfection of constitutively active phosphomimetic eNOS (S1177D) further abrogated apoptosis in aged HUVECs. Thus, aging of endothelial cells is associated with decreased NO synthesis and concomitantly increased sensitivity of apoptosis, which may contribute to functional impairment of the endothelial monolayer.


Subject(s)
Aging/metabolism , Apoptosis/physiology , Endothelium, Vascular/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Protein Serine-Threonine Kinases , Aging/drug effects , Amino Acid Substitution , Animals , Aorta , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Enzyme Activation/genetics , Humans , Lipoproteins, LDL/pharmacology , Male , Mice , Mice, Knockout , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Nitroso Compounds/metabolism , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Stress, Mechanical , Sulfhydryl Compounds/metabolism , Transfection , Tumor Necrosis Factor-alpha/pharmacology
7.
Mol Cell Biol ; 21(19): 6387-94, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11533228

ABSTRACT

Previous studies showed that the epidermal growth factor receptor (EGFR) can be transactivated by platelet-derived growth factor (PDGF) stimulation and that EGFR transactivation is required for PDGF-stimulated cell migration. To investigate the mechanism for cross talk between the PDGF beta receptor (PDGFbetaR) and the EGFR, we stimulated rat aortic vascular smooth muscle cells (VSMC) with 20 ng of PDGF/ml. Transactivation of the EGFR, defined by receptor tyrosine phosphorylation, occurred with the same time course as PDGFbetaR activation. Basal formation of PDGFbetaR-EGFR heterodimers was shown by coimmunoprecipitation studies, and interestingly, disruption of this receptor heterodimer abolished EGFR transactivation. Breakdown of the heterodimer was observed when VSMC were pretreated with antioxidants or with a Src family kinase inhibitor. Disruption of heterodimers decreased ERK1 and ERK2 activation by PDGF. Although PDGF-induced PDGFbetaR activation was abolished after pretreatment with 1 microM AG1295 (a specific PDGF receptor kinase inhibitor), EGFR transactivation was still observed, indicating that PDGFbetaR kinase activity is not required. In conclusion, our data demonstrate that the PDGFbetaR and the EGFR form PDGFbetaR-EGFR heterodimers basally, and we suggest that heterodimers represent a novel signaling complex which plays an important role in PDGF signal transduction.


Subject(s)
ErbB Receptors/metabolism , Platelet-Derived Growth Factor/pharmacology , Receptor Cross-Talk , 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt/pharmacology , Acetylcysteine/pharmacology , Animals , Cells, Cultured , Dimerization , Free Radical Scavengers/pharmacology , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Protein Tyrosine Phosphatases/physiology , Rats , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor beta/metabolism , Transcriptional Activation , src-Family Kinases/physiology
8.
J Biol Chem ; 276(44): 41383-7, 2001 Nov 02.
Article in English | MEDLINE | ID: mdl-11524431

ABSTRACT

Nitric oxide (NO) plays an important role in the regulation of the functional integrity of the endothelium. The intracellular reaction of NO with reactive cysteine groups leads to the formation of S-nitrosothiols. To investigate the regulation of S-nitrosothiols in endothelial cells, we first analyzed the composition of the S-nitrosylated molecules in endothelial cells. Gel filtration revealed that more than 95% of the detected S-nitrosothiols had a molecular mass of more than 5000 Da. Moreover, inhibition of de novo synthesis of glutathione using N-butyl-sulfoximine did not diminish the overall cellular S-NO content suggesting that S-nitrosylated glutathione quantitatively plays only a minor role in endothelial cells. Having demonstrated that most of the S-nitrosothiols are proteins, we determined the regulation of the S-nitrosylation by pro-inflammatory and pro-atherogenic factors, such as TNFalpha and mildly oxidized low density lipoprotein (oxLDL). TNFalpha and oxLDL induced denitrosylation of various proteins as assessed by Saville-Griess assay, by immunostaining with an anti-S-nitrosocysteine antibody, and by a Western blot approach. Furthermore, the caspase-3 p17 subunit, which has previously been shown to be S-nitrosylated and thereby inhibited, was denitrosylated by TNFalpha treatment suggesting that S-nitrosylation and denitrosylation are important regulatory mechanisms in endothelial cells contributing to the integrity of the endothelial cell monolayer.


Subject(s)
Endothelium, Vascular/metabolism , Lipoproteins, LDL/metabolism , S-Nitrosothiols/metabolism , Tumor Necrosis Factor-alpha/metabolism , Blotting, Western , Chromatography, Gel , Endothelium, Vascular/cytology , Humans , Molecular Weight , S-Nitrosothiols/chemistry
9.
J Am Coll Cardiol ; 36(7): 2081-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11127444

ABSTRACT

OBJECTIVES: The purposes of this study were to determine whether the serum of patients with congestive heart failure (CHF) can induce apoptosis of endothelial cells and to elucidate the underlying mechanisms. Moreover, the effect of the beta-blocker carvedilol was investigated. BACKGROUND: Congestive heart failure is associated with impaired endothelial function in the peripheral systemic vasculature and with systemic release of inflammatory cytokines. Pro-inflammatory cytokines have been shown to induce endothelial cell apoptosis in vitro. Therefore, we hypothesized that CHF is associated with enhanced apoptosis of endothelial cells. METHODS: Human umbilical vein endothelial cells were exposed to the serum of patients with CHF (n = 15) or healthy volunteers (n = 11), and apoptosis was determined by fluorescence staining of the nuclei and demonstration of deoxyribonucleic acid laddering. Moreover, apoptotic membrane particles were detected in plasma samples of patients with CHF. RESULTS: The serum of patients with CHF revealed a significantly enhanced pro-apoptotic activity as compared with age- and gender-matched healthy volunteers (p < 0.001). Furthermore, patients with CHF revealed significantly elevated plasma concentrations of apoptotic membrane particles. Apoptosis of endothelial cells correlated with elevated tumor necrosis factor-alpha (TNF-alpha) (r = 0.585, p = 0.002) and soluble TNF receptor serum levels (r = 0.517, p = 0.007). Carvedilol completely suppressed the increase in apoptosis induced by the serum of patients with CHF. Moreover, carvedilol dose-dependently inhibited TNF-alpha-induced apoptosis. The antiapoptotic activity of carvedilol was mediated by reduced activation of the caspase cascade through inhibition of mitochondrial cytochrome c release. The suppression of apoptosis by carvedilol was due to its antioxidative rather than beta-blocking effects, as the analogue BM91.0228, which has no beta-blocking activity, exerted similar effects. CONCLUSIONS: These findings indicate that endothelial cell apoptosis may play a role in the pathophysiology of heart failure. Inhibition of endothelial cell apoptosis by carvedilol may contribute to the beneficial effects of carvedilol in patients with heart failure.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Antioxidants/pharmacology , Apoptosis/drug effects , Carbazoles/pharmacology , Heart Failure/physiopathology , Propanolamines/pharmacology , Aged , Antioxidants/therapeutic use , Carvedilol , Cells, Cultured , Cytochrome c Group/metabolism , Epithelial Cells/physiology , Female , Heart Failure/blood , Humans , Male , Middle Aged
10.
Circ Res ; 87(10): 881-7, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-11073883

ABSTRACT

Retinoids exert antiproliferative and prodifferentiating effects in vascular smooth muscle cells (SMCs) and reduce neointimal mass in balloon-injured blood vessels. The mechanisms through which retinoids carry out these effects are unknown but likely involve retinoid receptor-mediated changes in gene expression. Here we report the cloning, chromosomal mapping, and biological activity of the retinoid-response gene rat tissue transglutaminase (tTG). Northern blotting studies showed that tTG is rapidly and dose-dependently induced in a protein synthesis-independent manner after stimulation with the natural retinoid all-trans retinoic acid (atRA). The induction of tTG was selective for atRA and its stereoisomers 9-cis and 13-cis RA, because little or no elevation in mRNA expression was observed with a panel of growth factors. Western blotting and immunofluorescence confocal microscopy showed an accumulation of cytosolic tTG protein after atRA stimulation. Radiolabeled cross-linking studies revealed a corresponding elevation in in vitro tTG activity. The increase in tTG activity was reduced in the presence of 2 distinct inhibitors of tTG (monodansylcadaverine and cystamine). atRA-induced tTG mRNA and protein expression were followed by a significant elevation in SMC apoptosis. Such retinoid-induced programmed cell death could be partially inhibited with each tTG inhibitor and was completely blocked when both inhibitors were used simultaneously. These results establish a role for atRA in the sequential stimulation of tTG and apoptosis in cultured SMCs. atRA-mediated apoptosis in SMCs seems to require the participation of active tTG, suggesting a potential mechanistic link between this retinoid-inducible gene and programmed cell death.


Subject(s)
Apoptosis , Cadaverine/analogs & derivatives , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Transglutaminases/genetics , Transglutaminases/metabolism , Tretinoin/metabolism , Animals , Blotting, Northern , Blotting, Western , Cadaverine/pharmacology , Cells, Cultured , Chromosome Mapping , Cloning, Molecular , Cystamine/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , Growth Substances/metabolism , Growth Substances/pharmacology , Male , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Protein Glutamine gamma Glutamyltransferase 2 , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Transcription, Genetic/drug effects , Transglutaminases/antagonists & inhibitors , Tretinoin/pharmacology
11.
Regul Pept ; 95(1-3): 1-7, 2000 Nov 24.
Article in English | MEDLINE | ID: mdl-11062326

ABSTRACT

It has been 100 years since the discovery of renin by Bergman and Tigerstedt. Since then, numerous studies have advanced our understanding of the renin-angiotensin system. A remarkable aspect was the discovery that angiotensin II (AngII) is the central product of the renin-angiotensin system and that this octapeptide induces multiple physiological responses in different cell types. In addition to its well known vasoconstrictive effects, growing evidence supports the notion that AngII may play a central role not only in hypertension, but also in cardiovascular and renal diseases. Binding of AngII to the seven-transmembrane angiotensin II type 1 receptor is responsible for nearly all of the physiological actions of AngII. Recent studies underscore the new concept that activation of intracellular second messengers by AngII requires tyrosine phosphorylation. An increasing number of tyrosine kinases have been shown to be activated by AngII, including the Src kinase family, the focal adhesion kinase family, the Janus kinases and receptor tyrosine kinases. These actions of AngII contribute to the pathophysiology of cardiac hypertrophy and remodeling, vascular thickening, heart failure and atherosclerosis. In this review, we discuss the important role of tyrosine kinases in AngII-mediated signal transduction. Understanding the importance of tyrosine phosphorylation in AngII-stimulated signaling events may contribute to new therapies for cardiovascular and renal diseases.


Subject(s)
Angiotensin II/physiology , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Animals , Cardiovascular Diseases/physiopathology , Humans , Hypertension/physiopathology , Kidney Diseases/physiopathology , Receptor Protein-Tyrosine Kinases/metabolism , Renin-Angiotensin System/physiology
12.
Cardiovasc Res ; 48(1): 148-57, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11033117

ABSTRACT

OBJECTIVE: The signaling pathways mediating proliferation and apoptosis in vascular smooth muscle cells (VSMC) are not well established. It has previously been shown that activation of the phosphoinositide 3-OH kinase (PI3K)/Akt pathway or the ERK 1/2 pathway can mediate anti-apoptotic function in different cell types. This study determined the specific contribution of the PI3K/Akt and ERK pathway in the regulation of apoptosis and proliferation of VSMC. METHODS AND RESULTS: Incubation of rat VSMC with FCS, insulin or IGF-1 time-dependently stimulated the phosphorylation of Akt, however FCS but not insulin or IGF-1 activated the MAP-kinase ERK 1/2. Moreover, insulin inhibited H(2)O(2)-induced apoptosis via the Akt pathway as demonstrated by pharmacological inhibition of the PI3K or overexpression of a dominant negative Akt mutant. In contrast, FCS inhibited H(2)O(2)-induced apoptosis via the Akt and also the ERK pathway. FCS, but not insulin or IGF-1 induced VSMC proliferation, suggesting that Akt activation is necessary but not sufficient for VSMC proliferation. FCS-induced proliferation of VSMC was only mediated via the Akt pathway and not the ERK pathway. CONCLUSIONS: These results define a link between cell proliferation and programmed cell death in VSMC via the same signal transduction pathway, namely activation of the serine/threonine kinase Akt, which may have significant implication for the development of vascular diseases or remodeling.


Subject(s)
Growth Substances/pharmacology , Muscle, Smooth, Vascular/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins , Signal Transduction/drug effects , Analysis of Variance , Animals , Aorta , Apoptosis/drug effects , Cell Division/drug effects , Cells, Cultured , Flow Cytometry , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt , Rats , Stimulation, Chemical
13.
Circ Res ; 87(9): 789-96, 2000 Oct 27.
Article in English | MEDLINE | ID: mdl-11055983

ABSTRACT

Reactive oxygen species have been implicated in the pathogenesis of atherosclerosis, hypertension, and restenosis, in part by promoting vascular smooth muscle cell (VSMC) growth. Many VSMC growth factors are secreted by VSMC and act in an autocrine manner. Here we demonstrate that cyclophilin A (CyPA), a member of the immunophilin family, is secreted by VSMCs in response to oxidative stress and mediates extracellular signal-regulated kinase (ERK1/2) activation and VSMC growth by reactive oxygen species. Human recombinant CyPA can mimic the effects of secreted CyPA to stimulate ERK1/2 and cell growth. The peptidyl-prolyl isomerase activity is required for ERK1/2 activation by CyPA. In vivo, CyPA expression and secretion are increased by oxidative stress and vascular injury. These findings are the first to identify CyPA as a secreted redox-sensitive mediator, establish CyPA as a VSMC growth factor, and suggest an important role for CyPA and enzymes with peptidyl-prolyl isomerase activity in the pathogenesis of vascular diseases.


Subject(s)
Cyclophilin A/metabolism , Muscle, Smooth, Vascular/metabolism , NADPH Oxidases , Oxidative Stress , 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt/pharmacology , Aminoquinolines/pharmacology , Animals , Apoptosis/drug effects , Carotid Artery Injuries/metabolism , Cell Division/drug effects , Cyclophilin A/analysis , Enzyme Activation , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Male , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , Nitroprusside , Onium Compounds/pharmacology , Peptidylprolyl Isomerase , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism , Transfection , Vascular Diseases/etiology
14.
J Biol Chem ; 275(33): 25502-7, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10846176

ABSTRACT

MAP kinase-dependent phosphorylation processes have been shown to interfere with the degradation of the antiapoptotic protein Bcl-2. The cytosolic MAP kinase phosphatase MAP kinase phosphatase-3 (MKP-3) induces apoptosis of endothelial cells in response to tumor necrosis factor alpha (TNFalpha) via dephosphorylation of the MAP kinase ERK1/2, leading to Bcl-2 proteolysis. Here we report that the endothelial cell survival factor nitric oxide (NO) down-regulated MKP-3 by destabilization of MKP-3 mRNA. This effect of NO was paralleled by a decrease in MKP-3 protein levels. Moreover, ERK1/2 was found to be protected against TNFalpha-induced dephosphorylation by coincubation of endothelial cells with the NO donor. Subsequently, both the decrease in Bcl-2 protein levels and the mitochondrial release of cytochrome c in response to TNFalpha were largely prevented by exogenous NO. In cells overexpressing MKP-3, no differences in phosphatase activity in the presence or absence of NO were found, excluding potential posttranslational modifications of MKP-3 protein by NO. These data demonstrate that upstream of the S-nitrosylation of caspase-3, NO exerts additional antiapoptotic effects in endothelial cells, which rely on the down-regulation of MKP-3 mRNA.


Subject(s)
Down-Regulation , Nitric Oxide/physiology , Protein Tyrosine Phosphatases/metabolism , RNA, Messenger/metabolism , Animals , Apoptosis , Blotting, Western , COS Cells , Cells, Cultured , Cytochrome c Group/metabolism , Dual Specificity Phosphatase 6 , Endothelium, Vascular/metabolism , Enzyme Activation , Humans , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Nitric Oxide/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Plasmids/metabolism , Protein Biosynthesis , Proto-Oncogene Proteins c-bcl-2/metabolism , Time Factors , Transcription, Genetic , Transfection , Tumor Necrosis Factor-alpha/metabolism , Umbilical Veins/metabolism
16.
J Biol Chem ; 275(16): 11706-12, 2000 Apr 21.
Article in English | MEDLINE | ID: mdl-10766791

ABSTRACT

c-Jun NH(2)-terminal kinase (JNK) is activated by a number of cellular stimuli such as inflammatory cytokines and environmental stresses. Reactive oxygen species also cause activation of JNK; however, the signaling cascade that leads to JNK activation remains to be elucidated. Because recent reports showed that expression of Cas, a putative Src substrate, stimulates JNK activation, we hypothesized that the Src kinase family and Cas would be involved in JNK activation by reactive oxygen species. An essential role for both Src and Cas was demonstrated. First, the specific Src family tyrosine kinase inhibitor, PP2, inhibited JNK activation by H(2)O(2) in a concentration-dependent manner but had no effect on extracellular signal-regulated kinases 1 and 2 and p38 activation. Second, JNK activation in response to H(2)O(2) was completely inhibited in cells derived from transgenic mice deficient in Src but not Fyn. Third, expression of a dominant negative mutant of Cas prevented H(2)O(2)-mediated JNK activation but had no effect on extracellular signal-regulated kinases 1 and 2 and p38 activation. Finally, the importance of Src was further supported by the inhibition of both H(2)O(2)-mediated Cas tyrosine phosphorylation and Cas.Crk complex formation in Src-/- but not Fyn-/- cells. These results demonstrate an essential role for Src and Cas in H(2)O(2)-mediated activation of JNK and suggest a new redox-sensitive pathway for JNK activation mediated by Src.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Proteins , Proto-Oncogene Proteins/metabolism , Reactive Oxygen Species/metabolism , Ubiquitin-Protein Ligases , src-Family Kinases/metabolism , Animals , Crk-Associated Substrate Protein , Dose-Response Relationship, Drug , Enzyme Activation , Fibroblasts/metabolism , Hydrogen Peroxide/administration & dosage , Hydrogen Peroxide/metabolism , JNK Mitogen-Activated Protein Kinases , Male , Mice , Mitogen-Activated Protein Kinase 3 , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-cbl , Rats , Rats, Sprague-Dawley , Retinoblastoma-Like Protein p130 , Signal Transduction , p38 Mitogen-Activated Protein Kinases
17.
Circ Res ; 86(7): 729-36, 2000 Apr 14.
Article in English | MEDLINE | ID: mdl-10764405

ABSTRACT

The third cytoplasmic loop of the angiotensin (Ang) II type 1 receptor (AT(1)) is important for receptor coupling to G proteins and activation of downstream events. Therefore, we determined whether specific AT(1) sequences were required for kinase activation and inhibition of apoptosis by transfecting wild-type (AT1Rwt) and mutated AT(1) into 293 cells. Ang II stimulated a 19.4-fold increase in extracellular signal-regulated kinase (ERK1/ERK2) activity in 293 cells transfected with AT1Rwt. However, in 293 cells that expressed a receptor in which amino acids 221 and 222 were deleted (AT1R[Del221/222]), Ang II-mediated ERK1/ERK2 activation was inhibited by >85%. In contrast, c-Jun NH(2)-terminal protein kinase (JNK) activation was similar in AT1Rwt- and AT1R(Del221/222)-transfected cells. Activation of ERK1/ERK2 by AT1Rwt was independent of Ca(2+), whereas the low level of ERK1/ERK2 activation by AT1R(Del221/222) was completely Ca(2+) dependent. Activation of ERK1/ERK2 in AT1Rwt required Ras, whereas AT1R(Del221/222) required Rap1. These results demonstrate the presence of 2 different pathways for ERK1/ERK2 activation by Ang II, which differ in their requirements for Ca(2+) and small G proteins (Ras versus Rap1). Furthermore, Ang II prevented serum deprivation-induced apoptosis in cells transfected with AT1Rwt but not AT1R(Del221/222). AKT was only phosphorylated by Ang II in AT1Rwt-transfected cells. Overexpression of constitutively active AKT significantly reduced serum deprivation-induced apoptosis in cells transfected with AT1R(Del221/222). This study shows for the first time a direct link between kinase activation and inhibition of apoptosis dependent on amino acids 221 and 222 in the third cytoplasmic loop of the AT(1).


Subject(s)
Receptors, Angiotensin/chemistry , Receptors, Angiotensin/physiology , Calcium/physiology , Cell Death , Cell Line , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Protein Structure, Secondary , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Deletion , Signal Transduction , Transfection , rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism
18.
J Biol Chem ; 275(21): 15926-32, 2000 May 26.
Article in English | MEDLINE | ID: mdl-10748142

ABSTRACT

Several signal transduction events induced by angiotensin II (AngII) binding to the angiotensin II type 1 receptor resemble those evoked by platelet-derived growth factor (PDGF) binding to the PDGF-beta receptor (PDGFbeta-R). We report here, in agreement with previous data, that AngII and PDGF-B-chain homodimer (PDGF-BB) stimulate tyrosine phosphorylation of the PDGFbeta-R. Both AngII and PDGF-BB stimulated the phosphorylation of PDGFbeta-R via the binding of tyrosine-phosphorylated Shc to PDGFbeta-R. Both PDGF-BB- and AngII-induced phosphorylation of the Shc.PDGFbeta-R complex was inhibited by antioxidants such as N-acetylcysteine and Tiron, but not by calcium chelation. However, transactivation of PDGFbeta-R by AngII (measured by PDGFbeta-R tyrosine phosphorylation) differed significantly from PDGF-BB. Evidence to support different mechanisms of PDGFbeta-R phosphorylation includes differences in the time course of PDGFbeta-R phosphorylation, differing effects of inhibitors of the endogenous PDGFbeta-R tyrosine kinase and Src family tyrosine kinases, differing results when the PDGFbeta-R was directly immunoprecipitated (PDGFbeta-R-antibody) versus coimmunoprecipitated (Shc-antibody), and cell fractionation studies that suggested that the Shc.PDGFbeta-R complexes phosphorylated by AngII and PDGF-BB were located in separate subcellular compartments. These studies are the first to suggest that transactivation of tyrosine kinase receptors by G protein-coupled receptors involves a unique pathway that regulates a population of tyrosine kinase receptors different from the endogenous tyrosine kinase ligand.


Subject(s)
Angiotensin II/pharmacology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Transcriptional Activation/drug effects , Animals , Antioxidants/pharmacology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Male , Muscle, Smooth, Vascular/drug effects , Phosphorylation , Phosphotyrosine/analysis , Platelet-Derived Growth Factor/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Tyrphostins/pharmacology , src Homology Domains
19.
Cardiovasc Res ; 45(3): 671-8, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10728388

ABSTRACT

OBJECTIVE: Apoptosis of cardiomyocytes may contribute to ischemia-reperfusion injury. The role of nitric oxide (NO) in apoptosis is controversial. Therefore, we investigated the effect of NO synthase inhibition on apoptosis of cardiomyocytes during ischemia and reperfusion and elucidated the underlying mechanisms. METHODS AND RESULTS: Isolated perfused rat hearts (n = 6/group) were subjected to ischemia (30 min) and reperfusion (30 min) in the presence or absence of the NO synthase inhibitor NG-mono-methyl-L-arginine. Reperfusion induced cardiomyocyte apoptosis as assessed by immunohistochemistry (TUNEL-staining) and the demonstration of the typical DNA laddering. Apoptosis during reperfusion was associated with the cleavage of caspase-3, the final down-stream executioner caspase, whereas the protein levels of the anti-apoptotic protein Bcl-2 and the pro-apoptotic protein Bax were unchanged. Inhibition of the NO synthase drastically increased ischemia and reperfusion-induced apoptosis of cardiomyocytes. Moreover, the NO synthase inhibitor enhanced the activation of caspase-3, suggesting that NO interferes with the activation of caspases in ischemia-reperfusion. CONCLUSION: The results of the present study demonstrate that inhibition of endogenous NO synthesis during ischemia and reperfusion leads to an enhanced induction of apoptosis, suggesting that the endogenous NO synthesis protects against apoptotic cell death. Inhibition of NO synthesis thereby activates the caspase cascade, whereas the Bcl-2/Bax protein levels remained unchanged.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Myocardial Reperfusion Injury/enzymology , Myocardium/enzymology , Nitric Oxide Synthase/antagonists & inhibitors , Signal Transduction , omega-N-Methylarginine/pharmacology , Analysis of Variance , Animals , Blotting, Western , Caspase 3 , Cells, Cultured , Enzyme Activation , In Situ Nick-End Labeling , Male , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , bcl-2-Associated X Protein
20.
Mol Cell Biol ; 20(5): 1886-96, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10669763

ABSTRACT

The ratio of proapoptotic versus antiapoptotic Bcl-2 members is a critical determinant that plays a significant role in altering susceptibility to apoptosis. Therefore, a reduction of antiapoptotic protein levels in response to proximal signal transduction events may switch on the apoptotic pathway. In endothelial cells, tumor necrosis factor alpha (TNF-alpha) induces dephosphorylation and subsequent ubiquitin-dependent degradation of the antiapoptotic protein Bcl-2. Here, we investigate the role of different putative phosphorylation sites to facilitate Bcl-2 degradation. Mutation of the consensus protein kinase B/Akt site or of potential protein kinase C or cyclic AMP-dependent protein kinase sites does not affect Bcl-2 stability. In contrast, inactivation of the three consensus mitogen-activated protein (MAP) kinase sites leads to a Bcl-2 protein that is ubiquitinated and subsequently degraded by the 26S proteasome. Inactivation of these sites within Bcl-2 revealed that dephosphorylation of Ser87 appears to play a major role. A Ser-to-Ala substitution at this position results in 50% degradation, whereas replacement of Thr74 with Ala leads to 25% degradation, as assessed by pulse-chase studies. We further demonstrated that incubation with TNF-alpha induces dephosphorylation of Ser87 of Bcl-2 in intact cells. Furthermore, MAP kinase triggers phosphorylation of Bcl-2, whereas a reduction in Bcl-2 phosphorylation was observed in the presence of MAP kinase-specific phosphatases or the MAP kinase-specific inhibitor PD98059. Moreover, we show that oxidative stress mediates TNF-alpha-stimulated proteolytic degradation of Bcl-2 by reducing MAP kinase activity. Taken together, these results demonstrate a direct protective role for Bcl-2 phosphorylation by MAP kinase against apoptotic challenges to endothelial cells and other cells.


Subject(s)
Cysteine Endopeptidases/metabolism , Multienzyme Complexes/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Apoptosis , Cells, Cultured , Cysteine Endopeptidases/genetics , Humans , MAP Kinase Signaling System , Multienzyme Complexes/genetics , Mutation , Phosphorylation , Proteasome Endopeptidase Complex , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...