Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Blood ; 139(8): 1184-1197, 2022 02 24.
Article in English | MEDLINE | ID: mdl-33908607

ABSTRACT

Cancer cells are in most instances characterized by rapid proliferation and uncontrolled cell division. Hence, they must adapt to proliferation-induced metabolic stress through intrinsic or acquired antimetabolic stress responses to maintain homeostasis and survival. One mechanism to achieve this is reprogramming gene expression in a metabolism-dependent manner. MondoA (also known as Myc-associated factor X-like protein X-interacting protein [MLXIP]), a member of the MYC interactome, has been described as an example of such a metabolic sensor. However, the role of MondoA in malignancy is not fully understood and the underlying mechanism in metabolic responses remains elusive. By assessing patient data sets, we found that MondoA overexpression is associated with worse survival in pediatric common acute lymphoblastic leukemia (ALL; B-precursor ALL [B-ALL]). Using clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) and RNA-interference approaches, we observed that MondoA depletion reduces the transformational capacity of B-ALL cells in vitro and dramatically inhibits malignant potential in an in vivo mouse model. Interestingly, reduced expression of MondoA in patient data sets correlated with enrichment in metabolic pathways. The loss of MondoA correlated with increased tricarboxylic acid cycle activity. Mechanistically, MondoA senses metabolic stress in B-ALL cells by restricting oxidative phosphorylation through reduced pyruvate dehydrogenase activity. Glutamine starvation conditions greatly enhance this effect and highlight the inability to mitigate metabolic stress upon loss of MondoA in B-ALL. Our findings give novel insight into the function of MondoA in pediatric B-ALL and support the notion that MondoA inhibition in this entity offers a therapeutic opportunity and should be further explored.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Neoplasm Proteins/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Stress, Physiological , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Line, Tumor , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Proteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics
2.
Cardiovasc Diagn Ther ; 11(6): 1295-1309, 2021 Dec.
Article in English | MEDLINE | ID: mdl-35070799

ABSTRACT

BACKGROUND: Right ventricular impairment (RVI) secondary to altered hemodynamics contributes to morbidity and mortality in adult patients after tetralogy of Fallot (TOF) repair. The goal of this study was to describe signaling pathways contributing to right ventricular (RV) remodeling by analyzing over lifetime alterations of RV gene expression in affected patients. METHODS: RV tissue was collected at the time of cardiac surgery in 13 patients with a diagnosis of TOF. RNA was isolated and whole transcriptome sequencing was performed. Gene profiles were compared between a group of 6 adults with signs of RVI undergoing right ventricle to pulmonary artery conduit surgery and a group of 7 infants, undergoing TOF correction. Definition of RVI in adult patients was based on clinical symptoms, evidence of RV hypertrophy, dilation, dysfunction or elevated pressure on echocardiographic, cardiovascular magnetic resonance, or catheterization evaluation. RESULTS: Median age was 34 years in RVI patients and 5 months in infants. Based on P adjusted value <0.01, RNA sequencing of RV specimens identified a total of 3,010 differentially expressed genes in adult patients with TOF and RVI as compared to infant patients with TOF. Gene Ontology and Kyoto Encyclopedia of Genes databases highlighted pathways involved in cellular metabolism, cell-cell communication, cell cycling and cellular contractility to be dysregulated in adults with corrected TOF and chronic RVI. CONCLUSIONS: RV transcriptome profiling in adult patients with RVI after TOF repair allows identification of signaling pathways, contributing to pathologic RV remodeling and helps in the discovery of biomarkers for disease progression and of new therapeutic targets.

3.
Redox Biol ; 34: 101536, 2020 07.
Article in English | MEDLINE | ID: mdl-32413743

ABSTRACT

Cardiovascular side effects are frequent problems accompanying systemic glucocorticoid therapy, although the underlying mechanisms are not fully resolved. Reactive oxygen species (ROS) have been shown to promote various cardiovascular diseases although the link between glucocorticoid and ROS signaling has been controversial. As the family of NADPH oxidases has been identified as important source of ROS in the cardiovascular system we investigated the role of NADPH oxidases in response to the synthetic glucocorticoid dexamethasone in the cardiovascular system in vitro and in vivo in mice lacking functional NADPH oxidases due to a mutation in the gene coding for the essential NADPH oxidase subunit p22phox. We show that dexamethasone induced NADPH oxidase-dependent ROS generation, leading to vascular proliferation and angiogenesis due to activation of the transcription factor hypoxia-inducible factor-1 (HIF1). Chronic treatment of mice with low doses of dexamethasone resulted in the development of systemic hypertension, cardiac hypertrophy and left ventricular dysfunction, as well as in pulmonary hypertension and pulmonary vascular remodeling. In contrast, mice deficient in p22phox-dependent NADPH oxidases were protected against these cardiovascular side effects. Mechanistically, dexamethasone failed to upregulate HIF1α levels in these mice, while vascular HIF1α deficiency prevented pulmonary vascular remodeling. Thus, p22phox-dependent NADPH oxidases and activation of the HIF pathway are critical elements in dexamethasone-induced cardiovascular pathologies and might provide interesting targets to limit cardiovascular side effects in patients on chronic glucocorticoid therapy.


Subject(s)
Heart Diseases , Hypertension, Pulmonary , Animals , Glucocorticoids , Humans , Hypertension, Pulmonary/chemically induced , Hypoxia-Inducible Factor 1 , Mice , NADPH Oxidases/genetics , Reactive Oxygen Species
4.
Sci Rep ; 9(1): 8369, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31182733

ABSTRACT

Exposure to chronic hypoxia results in pulmonary hypertension characterized by increased vascular resistance and pulmonary vascular remodeling, changes in functional parameters of the pulmonary vasculature, and right ventricular hypertrophy, which can eventually lead to right heart failure. The underlying mechanisms of hypoxia-induced pulmonary hypertension have still not been fully elucidated while no curative treatment is currently available. Commonly employed pre-clinical analytic methods are largely limited to invasive studies interfering with cardiac tissue or otherwise ex vivo functional studies and histopathology. In this work, we suggest volumetric optoacoustic tomography (VOT) for non-invasive assessment of heart function in response to chronic hypoxia. Mice exposed for 3 consecutive weeks to normoxia or chronic hypoxia were imaged in vivo with heart perfusion tracked by VOT using indocyanide green contrast agent at high temporal (100 Hz) and spatial (200 µm) resolutions in 3D. Unequivocal difference in the pulmonary transit time was revealed between the hypoxic and normoxic conditions concomitant with the presence of pulmonary vascular remodeling within hypoxic models. Furthermore, a beat-to-beat analysis of the volumetric image data enabled identifying and characterizing arrhythmic events in mice exposed to chronic hypoxia. The newly introduced non-invasive methodology for analysis of impaired pulmonary vasculature and heart function under chronic hypoxic exposure provides important inputs into development of early diagnosis and treatment strategies in pulmonary hypertension.


Subject(s)
Cone-Beam Computed Tomography , Heart/diagnostic imaging , Hypertension, Pulmonary/diagnostic imaging , Pulmonary Artery/diagnostic imaging , Animals , Cardiovascular Physiological Phenomena , Disease Models, Animal , Heart/physiopathology , Humans , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/diagnostic imaging , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/diagnostic imaging , Hypoxia/physiopathology , Lung/diagnostic imaging , Lung/pathology , Mice , Muscle, Smooth, Vascular/diagnostic imaging , Muscle, Smooth, Vascular/physiopathology , Photoacoustic Techniques , Pulmonary Artery/pathology , Vascular Remodeling/physiology
5.
Antioxid Redox Signal ; 30(1): 40-55, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30044122

ABSTRACT

BACKGROUND: Cardiovascular diseases have been associated with stress in the endoplasmic reticulum (ER) and accumulation of unfolded proteins leading to the unfolded protein response (UPR). Reactive oxygen species (ROS) such as superoxide and H2O2 derived from NADPH oxidases have been implicated in the pathogenesis of cardiovascular diseases. ROS have also been associated with ER stress. The role NADPH oxidases in the UPR is, however, not completely resolved yet. AIM: In this study, we investigated the role of p22phox, an essential component of most NADPH oxidases, in the UPR of endothelial cells. RESULTS: Induction of ER stress increased p22phox expression at the transcriptional level. p22phox was identified as novel target of the UPR transcription factor ATF4 (activator of transcription factor 4) under ER stress conditions by promoter analyses and ChIP. Depletion of ATF4 and p22phox diminished the levels of superoxide and H2O2 under ER stress conditions. On the contrary, p22phox was instrumental in increasing eIF2α phosphorylation and subsequent ATF4 expression on induction of ER stress by chemicals, oxysterols, or severe hypoxia in vitro and in vivo, leading to increased expression of CHOP and activation of effector caspases. INNOVATION: p22phox is a novel target of ATF4 in response to ER stress, which can promote the PERK-ATF4 branch of the UPR in vitro and in vivo. CONCLUSION: p22phox-dependent NADPH oxidases are important mediators of ER stress driving the UPR.


Subject(s)
Activating Transcription Factor 4/metabolism , Endothelial Cells/metabolism , NADPH Oxidases/metabolism , Unfolded Protein Response , Activating Transcription Factor 4/genetics , Cells, Cultured , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , HeLa Cells , Humans , NADPH Oxidases/genetics
6.
Antioxid Redox Signal ; 30(1): 56-73, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30044141

ABSTRACT

AIMS: Hypoxia and reactive oxygen species (ROS) have been shown to play a role in the pathogenesis of pulmonary hypertension (PH), a potentially fatal disorder characterized by pulmonary vascular remodeling, elevated pulmonary arterial pressure, and right ventricular hypertrophy. However, how they are linked in the context of PH is not completely understood. We, therefore, investigated the role of the NADPH oxidase subunit p22phox in the response to hypoxia both in vitro and in vivo. RESULTS: We found that hypoxia decreased ubiquitinylation and proteasomal degradation of p22phox dependent on prolyl hydroxylases (PHDs) and the E3 ubiquitin ligase protein von Hippel Lindau (pVHL), which resulted in p22phox stabilization and accumulation. p22phox promoted vascular proliferation, migration, and angiogenesis under normoxia and hypoxia. Increased levels of p22phox were also detected in lungs and hearts from mice with hypoxia-induced PH. Mice harboring a point mutation (Y121H) in the p22phox gene, which resulted in decreased p22phox stability and subsequent loss of this protein, were protected against hypoxia-induced PH. Mechanistically, p22phox contributed to ROS generation under normoxia, hypoxia, and hypoxia/reoxygenation. p22phox increased the levels and activity of HIF1α, the major cellular regulator of hypoxia adaptation, under normoxia and hypoxia, possibly by decreasing the levels of the PHD cofactors ascorbate and iron(II), and it contributed to the downregulation of the tumor suppressor miR-140 by hypoxia. INNOVATION: These data identify p22phox as an important regulator of the hypoxia response both in vitro and in vivo. CONCLUSION: p22phox-dependent NADPH oxidases contribute to the pathophysiology of PH induced by hypoxia.


Subject(s)
Cytochrome b Group/metabolism , Hypertension, Pulmonary/metabolism , NADPH Oxidases/metabolism , Animals , Cell Hypoxia , Cells, Cultured , Cytochrome b Group/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , NADPH Oxidases/genetics , Point Mutation , Reactive Oxygen Species/metabolism
7.
Antioxid Redox Signal ; 30(1): 74-94, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29466865

ABSTRACT

SIGNIFICANCE: G protein-coupled receptors (GPCR) are the largest group of cell surface receptors, which link cells to their environment. Reactive oxygen species (ROS) can act as important cellular signaling molecules. The family of NADPH oxidases generates ROS in response to activated cell surface receptors. Recent Advances: Various signaling pathways linking GPCRs and activation of NADPH oxidases have been characterized. CRITICAL ISSUES: Still, a more detailed analysis of G proteins involved in the GPCR-mediated activation of NADPH oxidases is needed. In addition, a more precise discrimination of NADPH oxidase activation due to either upregulation of subunit expression or post-translational subunit modifications is needed. Also, the role of noncanonical modulators of NADPH oxidase activation in the response to GPCRs awaits further analyses. FUTURE DIRECTIONS: As GPCRs are one of the most popular classes of investigational drug targets, further detailing of G protein-coupled mechanisms in the activation mechanism of NADPH oxidases as well as better understanding of the link between newly identified NADPH oxidase interaction partners and GPCR signaling will provide new opportunities for improved efficiency and decreased off target effects of therapies targeting GPCRs.


Subject(s)
NADPH Oxidases/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Humans , Reactive Oxygen Species/metabolism
8.
Sci Rep ; 7(1): 13342, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29042581

ABSTRACT

Management of protein homeostasis by the ubiquitin-proteasome system is critical for atherosclerosis development. Recent studies showed controversial results on the role of immunoproteasome (IP) subunit ß5i/LMP7 in maintenance of protein homeostasis under cytokine induced oxidative stress. The present study aimed to investigate the effect of ß5i/LMP7-deficiency on the initiation and progression of atherosclerosis as a chronic inflammatory, immune cell driven disease. LDLR-/-LMP7-/- and LDLR-/- mice were fed a Western-type diet for either 6 or 24 weeks to induce early and advanced stage atherosclerosis, respectively. Lesion burden was similar between genotypes in both stages. Macrophage content and abundance of polyubiquitin conjugates in aortic root plaques were unaltered by ß5i/LMP7-deficiency. In vitro experiments using bone marrow-derived macrophages (BMDM) showed that ß5i/LMP7-deficiency did not influence macrophage polarization or accumulation of polyubiquitinated proteins and cell survival upon hydrogen peroxide and interferon-γ treatment. Analyses of proteasome core particle composition by Western blot revealed incorporation of standard proteasome subunits in ß5i/LMP7-deficient BMDM and spleen. Chymotrypsin-, trypsin- and caspase-like activities assessed by using short fluorogenic peptides in BMDM whole cell lysates were similar in both genotypes. Taken together, deficiency of IP subunit ß5i/LMP7 does not disturb protein homeostasis and does not aggravate atherogenesis in LDLR-/- mice.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Atherosclerosis/pathology , Disease Models, Animal , Disease Progression , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Proteasome Endopeptidase Complex/deficiency , Proteolysis
9.
Redox Biol ; 13: 94-162, 2017 10.
Article in English | MEDLINE | ID: mdl-28577489

ABSTRACT

The European Cooperation in Science and Technology (COST) provides an ideal framework to establish multi-disciplinary research networks. COST Action BM1203 (EU-ROS) represents a consortium of researchers from different disciplines who are dedicated to providing new insights and tools for better understanding redox biology and medicine and, in the long run, to finding new therapeutic strategies to target dysregulated redox processes in various diseases. This report highlights the major achievements of EU-ROS as well as research updates and new perspectives arising from its members. The EU-ROS consortium comprised more than 140 active members who worked together for four years on the topics briefly described below. The formation of reactive oxygen and nitrogen species (RONS) is an established hallmark of our aerobic environment and metabolism but RONS also act as messengers via redox regulation of essential cellular processes. The fact that many diseases have been found to be associated with oxidative stress established the theory of oxidative stress as a trigger of diseases that can be corrected by antioxidant therapy. However, while experimental studies support this thesis, clinical studies still generate controversial results, due to complex pathophysiology of oxidative stress in humans. For future improvement of antioxidant therapy and better understanding of redox-associated disease progression detailed knowledge on the sources and targets of RONS formation and discrimination of their detrimental or beneficial roles is required. In order to advance this important area of biology and medicine, highly synergistic approaches combining a variety of diverse and contrasting disciplines are needed.


Subject(s)
International Cooperation , Reactive Oxygen Species/metabolism , Animals , European Union , Humans , Molecular Biology/organization & administration , Molecular Biology/trends , Oxidation-Reduction , Reactive Oxygen Species/chemistry , Signal Transduction , Societies, Scientific
10.
J Mol Med (Berl) ; 95(8): 825-837, 2017 08.
Article in English | MEDLINE | ID: mdl-28550361

ABSTRACT

SDF-1/CXCR4 activation facilitates myocardial repair. Therefore, we aimed to activate the HIF-1α target genes SDF-1 and CXCR4 by dimethyloxalylglycine (DMOG)-induced prolyl-hydroxylase (PH) inhibition to augment CXCR4+ cell recruitment and myocardial repair. SDF-1 and CXCR4 expression was analyzed under normoxia and ischemia ± DMOG utilizing SDF-1-EGFP and CXCR4-EGFP reporter mice. In bone marrow and heart, CXCR4-EGFP was predominantly expressed in CD45+/CD11b+ leukocytes which significantly increased after myocardial ischemia. PH inhibition with 500 µM DMOG induced upregulation of SDF-1 mRNA in human microvascular endothelial cells (HMEC-1) and aortic vascular smooth muscle cells (HAVSMC). CXCR4 was highly elevated in HMEC-1 but almost no detectable in HAVSMC. In vivo, systemic administration of the PH inhibitor DMOG without pretreatment upregulated nuclear HIF-1α and SDF-1 in the ischemic mouse heart associated with increased recruitment of CD45+/CXCR4-EGFP+/CD11b+ cell subsets. Enhanced PH inhibition significantly upregulated reparative M2 like CXCR4-EGFP+ CD11b+/CD206+ cells compared to inflammatory M2-like CXCR4-EGFP+ CD11b+/CD86+ cells associated with reduced apoptotic cell death, increased neovascularization, reduced scar size, and an improved heart function after MI. In summary, our data suggest increased PH inhibition as a promising tool for a customized upregulation of SDF-1 and CXCR4 expression to attract CXCR4+/CD11b+ cells to the ischemic heart associated with increased cardiac repair. KEY MESSAGES: DMOG-induced prolyl-hydroxylase inhibition upregulates SDF-1 and CXCR4 in human endothelial cells. Systemic application of DMOG upregulates nuclear HIF-1α and SDF-1 in vivo. Enhanced prolyl-hydroxylase inhibition increases mainly CXCR4+/CD11b+ cells. DMOG increased reparative M2-like CD11b+/CD206+ cells compared to M1-like cells after MI. Enhanced prolyl-hydroxylase inhibition improved cardiac repair and heart function.


Subject(s)
Amino Acids, Dicarboxylic/pharmacology , Chemokine CXCL12/metabolism , Prolyl-Hydroxylase Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Bone Marrow/metabolism , CD11b Antigen/metabolism , Cell Line , Chemokine CXCL12/genetics , Hemodynamics/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice, Inbred C57BL , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Neovascularization, Physiologic/drug effects , RNA, Messenger/metabolism , Receptors, CXCR4/genetics
11.
Br J Pharmacol ; 174(12): 1533-1554, 2017 06.
Article in English | MEDLINE | ID: mdl-28332701

ABSTRACT

Cardiovascular diseases are among the leading causes of death worldwide. Reactive oxygen species (ROS) can act as damaging molecules but also represent central hubs in cellular signalling networks. Increasing evidence indicates that ROS play an important role in the pathogenesis of cardiovascular diseases, although the underlying mechanisms and consequences of pathophysiologically elevated ROS in the cardiovascular system are still not completely resolved. More recently, alterations of the epigenetic landscape, which can affect DNA methylation, post-translational histone modifications, ATP-dependent alterations to chromatin and non-coding RNA transcripts, have been considered to be of increasing importance in the pathogenesis of cardiovascular diseases. While it has long been accepted that epigenetic changes are imprinted during development or even inherited and are not changed after reaching the lineage-specific expression profile, it becomes more and more clear that epigenetic modifications are highly dynamic. Thus, they might provide an important link between the actions of ROS and cardiovascular diseases. This review will provide an overview of the role of ROS in modulating the epigenetic landscape in the context of the cardiovascular system. LINKED ARTICLES: This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.


Subject(s)
Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular System/metabolism , Epigenesis, Genetic/genetics , Reactive Oxygen Species/metabolism , Animals , Humans
12.
J Mol Med (Berl) ; 94(10): 1153-1166, 2016 10.
Article in English | MEDLINE | ID: mdl-27286880

ABSTRACT

Arsenite (As(III)) is widely distributed in nature and can be found in water, food, and air. There is significant evidence that exposure to As(III) is associated with human cancers originated from liver, lung, skin, bladder, kidney, and prostate. Hypoxia plays a role in tumor growth and aggressiveness; adaptation to it is, at least to a large extent, mediated by hypoxia-inducible factor-1α (HIF-1α). In the current study, we investigated As(III) effects on HIF-1α under normoxia and hypoxia in the hepatoma cell line HepG2. We found that As(III) increased HIF-1α protein levels under normoxia while the hypoxia-mediated induction of HIF1α was reduced. Thereby, the As(III) effects on HIF-1α were dependent on both, transcriptional regulation via the transcription factor Nrf2 mediated by NOX4, PI3K/Akt, and ERK1/2 as well as by modulation of HIF-1α protein stability. In line, the different effects of As(III) via participation of HIF-1α and Nrf2 were also seen in tube formation assays with endothelial cells where knockdown of Nrf2 and HIF-1α abolished As(III) effects. Overall, the present study shows that As(III) is a potent inducer of HIF-1α under normoxia but not under hypoxia which may explain, in part, its carcinogenic as well as anti-carcinogenic actions. KEY MESSAGE: As(III) increased HIF-1α under normoxia but reduced its hypoxia-dependent induction. The As(III) effects on HIF-1α were dependent on ROS, NOX4, PI3K/Akt, and ERK1/2. The As(III) effects under normoxia involved transcriptional regulation via Nrf2. Knockdown of Nrf2 and HIF-1α abolished As(III) effects in tube formation assays. The data may partially explain As(III)'s carcinogenic and anti-carcinogenic actions.


Subject(s)
Arsenites/pharmacology , Cell Hypoxia/genetics , Gene Expression Regulation/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Animals , Antineoplastic Agents/pharmacology , Carcinogens/pharmacology , Cell Hypoxia/physiology , Cell Line , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases , Fibroblasts/drug effects , Fibroblasts/metabolism , Heme Oxygenase-1/genetics , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , NADPH Oxidase 4 , NADPH Oxidases , NF-E2-Related Factor 2 , Phosphatidylinositol 3-Kinases , Plasminogen Activator Inhibitor 1/genetics , Proto-Oncogene Proteins c-akt , Reactive Oxygen Species/metabolism
13.
Redox Biol ; 6: 372-385, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26339717

ABSTRACT

Within the last twenty years the view on reactive oxygen species (ROS) has changed; they are no longer only considered to be harmful but also necessary for cellular communication and homeostasis in different organisms ranging from bacteria to mammals. In the latter, ROS were shown to modulate diverse physiological processes including the regulation of growth factor signaling, the hypoxic response, inflammation and the immune response. During the last 60-100 years the life style, at least in the Western world, has changed enormously. This became obvious with an increase in caloric intake, decreased energy expenditure as well as the appearance of alcoholism and smoking; These changes were shown to contribute to generation of ROS which are, at least in part, associated with the occurrence of several chronic diseases like adiposity, atherosclerosis, type II diabetes, and cancer. In this review we discuss aspects and problems on the role of intracellular ROS formation and nutrition with the link to diseases and their problematic therapeutical issues.


Subject(s)
Atherosclerosis/metabolism , Diabetes Mellitus, Type 2/metabolism , Hypoxia/metabolism , Neoplasms/metabolism , Nutritional Requirements/physiology , Obesity/metabolism , Reactive Oxygen Species/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Energy Metabolism/physiology , Gene Expression Regulation , Humans , Hypoxia/genetics , Hypoxia/pathology , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasms/genetics , Neoplasms/pathology , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , Obesity/genetics , Obesity/pathology , Oxidative Stress , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , Signal Transduction
14.
Thromb Haemost ; 108(5): 849-62, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23014943

ABSTRACT

Disruption of endothelial homeostasis results in endothelial dysfunction, characterised by a dysbalance between nitric oxide (NO) and reactive oxygen species (ROS) levels often accompanied by a prothrombotic and proproliferative state. The serine protease thrombin not only is instrumental in formation of the fibrin clot, but also exerts direct effects on the vessel wall by activating proliferative and angiogenic responses. In endothelial cells, thrombin can induce NO as well as ROS levels. However, the relative contribution of these reactive species to the angiogenic response towards thrombin is not completely clear. Since plasminogen activator inhibitor-1 (PAI-1), a direct target of the proangiogenic transcription factors hypoxia-inducible factors (HIFs), exerts prothrombotic and proangiogenic activities we investigated the role of ROS and NO in the regulation of HIF-1α, PAI-1 and capillary formation in response to thrombin. Thrombin enhanced the formation of NO as well as ROS generation involving the GTPase Rac1 in endothelial cells. Rac1-dependent ROS formation promoted induction of HIF-1α, PAI-1 and capillary formation by thrombin, while NO reduced ROS bioavailability and subsequently limited induction of HIF-1α, PAI-1 and the angiogenic response. Importantly, thrombin activation of Rac1 was diminished by NO, but enhanced by ROS. Thus, our findings show that capillary formation induced by thrombin via Rac1-dependent activation of HIF-1 and PAI-1 is limited by the concomitant release of NO which reduced ROS bioavailability. Rac1 activity is sensitive to ROS and NO, thereby playing an essential role in fine tuning the endothelial response to thrombin.


Subject(s)
Capillaries/growth & development , Capillaries/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Nitric Oxide Synthase Type III/antagonists & inhibitors , Plasminogen Activator Inhibitor 1/biosynthesis , rac1 GTP-Binding Protein/metabolism , Capillaries/drug effects , Cell Line , HEK293 Cells , Humans , NG-Nitroarginine Methyl Ester/pharmacology , Neovascularization, Physiologic/drug effects , Nitric Oxide/metabolism , Nitroarginine/pharmacology , Plasminogen Activator Inhibitor 1/genetics , Reactive Oxygen Species/metabolism , Thrombin/metabolism , Thrombin/pharmacology , rac1 GTP-Binding Protein/genetics
15.
J Cell Sci ; 125(Pt 4): 956-64, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22399808

ABSTRACT

Urotensin-II (U-II) has been considered as one of the most potent vasoactive peptides, although its physiological and pathophysiological role is still not finally resolved. Recent evidence suggests that it promotes angiogenic responses in endothelial cells, although the underlying signalling mechanisms are unclear. Reactive oxygen species derived from NADPH oxidases are major signalling molecules in the vasculature. Because NOX2 is functional in endothelial cells, we investigated the role of the NOX2-containing NADPH oxidase in U-II-induced angiogenesis and elucidated a possible contribution of hypoxia-inducible factor-1 (HIF-1), the master regulator of hypoxic angiogenesis, in the response to U-II. We found that U-II increases angiogenesis in vitro and in vivo, and these responses were prevented by antioxidants, NOX2 knockdown and in Nox2(-/-) mice. In addition, U-II-induced angiogenesis was dependent on HIF-1. Interestingly, U-II increased NOX2 transcription involving HIF-1, and chromatin immunoprecipitation confirmed NOX2 as a target gene of HIF-1. In support, NOX2 levels were greatly diminished in U-II-stimulated isolated vessels derived from mice deficient in endothelial HIF-1. Conversely, reactive oxygen species derived from NOX2 were required for U-II activation of HIF and upregulation of HIF-1. In line with this, U-II-induced upregulation of HIF-1 was absent in Nox2(-/-) vessels. Collectively, these findings identified HIF-1 and NOX2 as partners acting in concert to promote angiogenesis in response to U-II. Because U-II has been found to be elevated in cardiovascular disorders and in tumour tissues, this feed-forward mechanism could be an interesting anti-angiogenic therapeutic option in these disorders.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Neovascularization, Physiologic , Urotensins/metabolism , Animals , Feedback, Physiological , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , NADPH Oxidase 2 , NADPH Oxidases/biosynthesis , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Neovascularization, Pathologic , Neovascularization, Physiologic/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Receptors, G-Protein-Coupled/metabolism , Up-Regulation/drug effects , Urotensins/pharmacology
16.
Mol Biol Cell ; 22(22): 4424-34, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21965295

ABSTRACT

The vasoactive peptide urotensin-II (U-II) has been associated with vascular remodeling in different cardiovascular disorders. Although U-II can induce reactive oxygen species (ROS) by the NADPH oxidase NOX4 and stimulate smooth muscle cell (SMC) proliferation, the precise mechanisms linking U-II to vascular remodeling processes remain unclear. Forkhead Box O (FoxO) transcription factors have been associated with redox signaling and control of proliferation and apoptosis. We thus hypothesized that FoxOs are involved in the SMC response toward U-II and NOX4. We found that U-II and NOX4 stimulated FoxO activity and identified matrix metalloproteinase-2 (MMP2) as target gene of FoxO3a. FoxO3a activation by U-II was preceded by NOX4-dependent phosphorylation of c-Jun NH(2)-terminal kinase and 14-3-3 and decreased interaction of FoxO3a with its inhibitor 14-3-3, allowing MMP2 transcription. Functional studies in FoxO3a-depleted SMCs and in FoxO3a(-/-) mice showed that FoxO3a was important for basal and U-II-stimulated proliferation and vascular outgrowth, whereas treatment with an MMP2 inhibitor blocked these responses. Our study identified U-II and NOX4 as new activators of FoxO3a, and MMP2 as a novel target gene of FoxO3a, and showed that activation of FoxO3a by this pathway promotes vascular growth. FoxO3a may thus contribute to progression of cardiovascular diseases associated with vascular remodeling.


Subject(s)
Forkhead Transcription Factors/metabolism , Matrix Metalloproteinase 2/metabolism , Muscle, Smooth, Vascular/metabolism , NADPH Oxidases/metabolism , Urotensins/metabolism , 14-3-3 Proteins/metabolism , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase Inhibitors , Mice , Mice, Knockout , Muscle, Smooth, Vascular/growth & development , Myocytes, Smooth Muscle/metabolism , NADPH Oxidase 4 , Phosphorylation , Reactive Oxygen Species/metabolism , Signal Transduction
17.
Mol Biol Cell ; 21(12): 2087-96, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20427574

ABSTRACT

NADPH oxidases are important sources of reactive oxygen species (ROS), possibly contributing to various disorders associated with enhanced proliferation. NOX4 appears to be involved in vascular signaling and may contribute to the response to hypoxia. However, the exact mechanisms controlling NOX4 levels under hypoxia are not resolved. We found that hypoxia rapidly enhanced NOX4 mRNA and protein levels in pulmonary artery smooth-muscle cells (PASMCs) as well as in pulmonary vessels from mice exposed to hypoxia. This response was dependent on the hypoxia-inducible transcription factor HIF-1alpha because overexpression of HIF-1alpha increased NOX4 expression, whereas HIF-1alpha depletion prevented this response. Mutation of a putative hypoxia-responsive element in the NOX4 promoter abolished hypoxic and HIF-1alpha-induced activation of the NOX4 promoter. Chromatin immunoprecipitation confirmed HIF-1alpha binding to the NOX4 gene. Induction of NOX4 by HIF-1alpha contributed to maintain ROS levels after hypoxia and hypoxia-induced proliferation of PASMCs. These findings show that NOX4 is a new target gene of HIF-1alpha involved in the response to hypoxia. Together with our previous findings that NOX4 mediates HIF-1alpha induction under normoxia, these data suggest an important role of the signaling axis between NOX4 and HIF-1alpha in various cardiovascular disorders under hypoxic and also nonhypoxic conditions.


Subject(s)
NADPH Oxidases/genetics , Protein Subunits/genetics , Animals , Cell Hypoxia , Cell Movement , Cell Proliferation , Enzyme Induction , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/enzymology , NADPH Oxidase 4 , NADPH Oxidases/biosynthesis , Protein Subunits/biosynthesis , Pulmonary Artery/cytology , Reactive Oxygen Species/metabolism , Transcription, Genetic
18.
Antioxid Redox Signal ; 13(4): 467-87, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20001746

ABSTRACT

Reactive oxygen species (ROS) have been implicated in many intra- and intercellular processes. High levels of ROS are generated as part of the innate immunity in the respiratory burst of phagocytic cells. Low levels of ROS, however, are generated in a highly controlled manner by various cell types to act as second messengers in redox-sensitive pathways. A NADPH oxidase has been initially described as the respiratory burst enzyme in neutrophils. Stimulation of this complex enzyme system requires specific signaling cascades linking it to membrane-receptor activation. Subsequently, a family of NADPH oxidases has been identified in various nonphagocytic cells. They mainly differ in containing one out of seven homologous catalytic core proteins termed NOX1 to NOX5 and DUOX1 or 2. NADPH oxidase activity is controlled by regulatory subunits, including the NOX regulators p47phox and p67phox, their homologs NOXO1 and NOXA1, or the DUOX1 or 2 regulators DUOXA1 and 2. In addition, the GTPase Rac modulates activity of several of these enzymes. Recently, additional proteins have been identified that seem to have a regulatory function on NADPH oxidase activity under certain conditions. We will thus summarize molecular pathways linking activation of different membrane-bound receptors with increased ROS production of NADPH oxidases.


Subject(s)
Membrane Proteins/metabolism , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Animals , Humans , Membrane Proteins/genetics , Models, Biological , NADPH Oxidases/genetics , Signal Transduction/genetics , Signal Transduction/physiology
19.
Antioxid Redox Signal ; 13(4): 399-412, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20001745

ABSTRACT

Pulmonary vascular remodeling associated with pulmonary hypertension is characterized by media thickening, disordered proliferation, and in situ thrombosis. The p21-activated kinase-1 (PAK-1) can control growth, migration, and prothrombotic activity, and the hypoxia-inducible transcription factor HIF-1alpha was associated with pulmonary vascular remodeling. Here we studied whether PAK-1 and HIF-1alpha are linked in pulmonary vascular remodeling. PAK-1 was expressed in the media of remodeled pulmonary vessels from patients with pulmonary vasculopathy and was upregulated, together with its upstream regulator Rac1 and HIF-1alpha in lung tissue from lambs with pulmonary vascular remodeling. PAK-1 and Rac1 were activated by thrombin involving calcium, thus resulting in enhanced generation of reactive oxygen species (ROS) in human pulmonary artery smooth muscle cells (PASMCs). Activation of PAK-1 stimulated HIF activity and HIF-1alpha expression involving ROS and NF-kappaB, enhanced the expression of the HIF-1 target gene plasminogen activator inhibitor-1, and stimulated PASMC proliferation. Importantly, HIF-1 itself bound to the Rac1 promoter and enhanced Rac1 and PAK-1 transcription. Thus, PAK-1 and its activator Rac1 are novel HIF-1 targets that may constitute a positive-feedback loop for induction of HIF-1alpha by thrombin and ROS, thus explaining elevated levels of PAK-1, Rac1, and HIF-1alpha in remodeled pulmonary vessels.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Pulmonary Artery/metabolism , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Blotting, Western , Calcium/metabolism , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Chromatin Immunoprecipitation , Enzyme Activation/drug effects , Female , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunohistochemistry , In Vitro Techniques , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Pregnancy , Pulmonary Artery/pathology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sheep , Thrombin/pharmacology , p21-Activated Kinases/genetics , rac1 GTP-Binding Protein/genetics
20.
Circ Res ; 104(10): 1169-77, 2009 May 22.
Article in English | MEDLINE | ID: mdl-19390057

ABSTRACT

Cyclic nucleotide phosphodiesterases (PDEs) control the levels of the second messengers cAMP and cGMP in many cell types including endothelial cells. Although PDE2 has the unique property to be activated by cGMP but to hydrolyze cAMP, its role in endothelial function is only poorly understood. Reactive oxygen species (ROS) have been recognized as signaling molecules controlling many endothelial functions. We thus investigated whether PDE2 would link to ROS generation and proliferative responses in human umbilical vein endothelial cells in response to thrombin. Thrombin stimulated the GTPase Rac1, known to activate NADPH oxidases, and enhanced ROS formation, whereas PDE2 inhibition or depletion by short hairpin (sh)RNA prevented these responses. Similar observations were made with 8-Br-cGMP or atrial natriuretic peptide. In agreement, thrombin elevated cGMP but decreased cAMP levels, whereas db-cAMP or forskolin diminished Rac1 activity and ROS production. Subsequently, PDE2 overexpression activated Rac1, increased ROS generation, and enhanced proliferation and in vitro capillary formation. These responses were not observed in the presence of inactive Rac1 or shRNA against the NADPH oxidase subunit NOX2. Inhibition or depletion of PDE2 also prevented thrombin-induced proliferation and capillary formation. Importantly, downregulation of PDE2 by lentiviral shRNA or PDE2 inhibition prevented vessel sprouting from mouse aortic explants and in vivo angiogenesis in a mouse model, respectively. In summary, PDE2 promotes activation of NADPH oxidase-dependent ROS production and subsequent endothelial proliferation and angiogenesis. Targeting PDE2 may provide a new therapeutic approach in diseases associated with endothelial dysfunction, oxidative stress, vascular proliferation, and angiogenesis.


Subject(s)
Cell Proliferation , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Endothelium, Vascular/metabolism , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Neovascularization, Physiologic/physiology , Thrombin/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Aorta/cytology , Aorta/metabolism , Cells, Cultured , Cyclic GMP/metabolism , Endothelium, Vascular/cytology , Humans , Male , Mice , Mice, Inbred C57BL , NADPH Oxidase 2 , Reactive Oxygen Species/metabolism , Umbilical Veins/cytology , Umbilical Veins/metabolism , p21-Activated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...