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

Country/Region as subject
Publication year range
1.
Pharmacol Res ; 159: 104999, 2020 09.
Article in English | MEDLINE | ID: mdl-32535224

ABSTRACT

Rimeporide, a first-in-class sodium/proton exchanger Type 1 inhibitor (NHE-1 inhibitor) is repositioned by EspeRare for patients with Duchenne Muscular Dystrophy (DMD). Historically, NHE-1 inhibitors were developed for cardiac therapeutic interventions. There is considerable overlap in the pathophysiological mechanisms in Congestive Heart Failure (CHF) and in cardiomyopathy in DMD, therefore NHE-1 inhibition could be a promising pharmacological approach to the cardiac dysfunctions observed in DMD. Extensive preclinical data was collected in various animal models including dystrophin-deficient (mdx) mice to characterise Rimeporide's anti-fibrotic and anti-inflammatory properties and there is evidence that NHE-1 inhibitors could play a significant role in modifying DMD cardiac and also skeletal pathologies, as the NHE-1 isoform is ubiquitous. We report here the first study with Rimeporide in DMD patients. This 4-week treatment, open label phase Ib, multiple oral ascending dose study, enrolled 20 ambulant boys with DMD (6-11 years), with outcomes including safety, pharmacokinetic (PK) and pharmacodynamic (PD) biomarkers. Rimeporide was safe and well-tolerated at all doses. PK evaluations showed that Rimeporide was well absorbed orally reaching pharmacological concentrations from the lowest dose, with exposure increasing linearly with dose and with no evidence of accumulation upon repeated dosing. Exploratory PD biomarkers showed positive effect upon a 4-week treatment, supporting its therapeutic potential in patients with DMD, primarily as a cardioprotective treatment, and provide rationale for further efficacy studies.


Subject(s)
Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne/drug therapy , Neuromuscular Agents/administration & dosage , Sodium-Hydrogen Exchanger 1/antagonists & inhibitors , Administration, Oral , Child , Drug Administration Schedule , Europe , Humans , Male , Models, Biological , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/physiopathology , Neuromuscular Agents/adverse effects , Neuromuscular Agents/pharmacokinetics , Sodium-Hydrogen Exchanger 1/metabolism , Treatment Outcome
4.
J Pathol ; 235(1): 65-78, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25116588

ABSTRACT

Reactive oxygen species (ROS) participate in the pathogenesis of emphysema. Among ROS-producing enzymes, NOX NADPH oxidases are thought to be responsible for tissue injury associated with several lung pathologies. To determine whether NOX2 and/or NOX1 participate in the development of emphysema, their expression patterns were first studied by immunohistochemistry in the lungs of emphysematous patients. Subsequently, we investigated their contribution to elastase-induced emphysema using NOX2- and NOX1-deficient mice. In human lung, NOX2 was mainly detected in macrophages of control and emphysematous lungs, while NOX1 was expressed in alveolar epithelium and bronchial cells. We observed an elevated number of NOX2-positive cells in human emphysematous lungs, as well as increased NOX2 and NOX1 mRNA expression in mouse lungs following elastase exposure. Elastase-induced alveolar airspace enlargement and elastin degradation were prevented in NOX2-deficient mice, but not in NOX1-deficient mice. This protection was independent of inflammation and correlated with reduced ROS production. Concomitantly, an elevation of sirtuin 1 (SIRT1) level and a decrease of matrix metalloproteinase-9 (MMP-9) expression and activity were observed in alveolar macrophages and neutrophils. We addressed the specific role of macrophage-restricted functional NOX2 in elastase-induced lung emphysema using Ncf1 mutant mice and Ncf1 macrophage rescue mice (Ncf1 mutant mice with transgenic expression of Ncf1 only in CD68-positive mononuclear phagocytes; the MN mouse). Compared to WT mice, the lack of functional NOX2 led to decreased elastase-induced ROS production and protected against emphysema. In contrast, ROS production was restored specifically in macrophages from Ncf1 rescue mice and contributes to emphysema. Taken together, our results demonstrate that NOX2 is involved in the pathogenesis of human emphysema and macrophage-specific NOX2 participates in elastase-induced emphysema through the involvement of SIRT1/MMP-9 pathways in mice.


Subject(s)
Macrophages/metabolism , Matrix Metalloproteinase 9/metabolism , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Pulmonary Emphysema/metabolism , Sirtuin 1/metabolism , Animals , Humans , Inflammation/pathology , Lung/pathology , Macrophages/pathology , Mice , NADPH Oxidase 2 , Neutrophils/pathology , Pulmonary Emphysema/pathology , Reactive Oxygen Species/metabolism
5.
J Immunol ; 193(4): 1690-700, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25009204

ABSTRACT

Arginine, a semiessential amino acid implicated in diverse cellular processes, is a substrate for two arginases-Arg1 and Arg2-having different expression patterns and functions. Although appropriately regulated Arg1 expression is critical for immune responses, this has not been documented for Arg2. We show that Arg2 is the dominant enzyme in dendritic cells (DCs) and is repressed by microRNA-155 (miR155) during their maturation. miR155 is known to be strongly induced in various mouse and human DC subsets in response to diverse maturation signals, and miR155-deficient DCs exhibit an impaired ability to induce Ag-specific T cell responses. By means of expression profiling studies, we identified Arg2 mRNA as a novel miR155 target in mouse DCs. Abnormally elevated levels of Arg2 expression and activity were observed in activated miR155-deficient DCs. Conversely, overexpression of miR155 inhibited Arg2 expression. Bioinformatic and functional analyses confirmed that Arg2 mRNA is a direct target of miR155. Finally, in vitro and in vivo functional assays using DCs exhibiting deregulated Arg2 expression indicated that Arg2-mediated arginine depletion in the extracellular milieu impairs T cell proliferation. These results indicate that miR155-induced repression of Arg2 expression is critical for the ability of DCs to drive T cell activation by controlling arginine availability in the extracellular environment.


Subject(s)
Arginase/biosynthesis , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/enzymology , Lymphocyte Activation/immunology , MicroRNAs/genetics , Animals , Arginase/antagonists & inhibitors , Arginase/genetics , Arginine/metabolism , Cell Proliferation , Dendritic Cells/immunology , Gene Expression Profiling , Gene Expression Regulation/immunology , HEK293 Cells , Humans , Lymphocyte Activation/genetics , Mice , Mice, Knockout , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics
7.
Respir Res ; 16: 118, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26415510

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a rapid progressive fibro-proliferative disorder with poor prognosis similar to lung cancer. The pathogenesis of IPF is uncertain, but loss of epithelial cells and fibroblast proliferation are thought to be central processes. Previous reports have shown that BARD1 expression is upregulated in response to hypoxia and associated with TGF-ß signaling, both recognized factors driving lung fibrosis. Differentially spliced BARD1 isoforms, in particular BARD1ß, are oncogenic drivers of proliferation in cancers of various origins. We therefore hypothesized that BARD1 and/or its isoforms might play a role in lung fibrosis. METHODS: We investigated BARD1 expression as a function of TGF-ß in cultured cells, in mice with experimentally induced lung fibrosis, and in lung biopsies from pulmonary fibrosis patients. RESULTS: FL BARD1 and BARD1ß were upregulated in response to TGF-ß in epithelial cells and fibroblasts in vitro and in vivo. Protein and mRNA expression studies showed very low expression in healthy lung tissues, but upregulated expression of full length (FL) BARD1 and BARD1ß in fibrotic tissues. CONCLUSION: Our data suggest that FL BARD1 and BARD1ß might be mediators of pleiotropic effects of TGF-ß. In particular BARD1ß might be a driver of proliferation and of pulmonary fibrosis pathogenesis and progression and represent a target for treatment.


Subject(s)
Idiopathic Pulmonary Fibrosis/metabolism , Lung/drug effects , Transforming Growth Factor beta1/pharmacology , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis/drug effects , Bleomycin , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Lung/metabolism , Lung/pathology , Male , Mice, Inbred C57BL , Protein Isoforms , Signal Transduction/drug effects , Time Factors , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics
9.
J Pathol ; 228(3): 341-50, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22685019

ABSTRACT

Chronic granulomatous disease (CGD), caused by a lack of reactive oxygen species (ROS) generation by the phagocyte NADPH oxidase NOX2, leads to massively increased inflammatory responses. In order to identify the type of phagocyte which requires NOX2 activity to limit inflammation, we investigated mice with a loss of function mutation in the Ncf1 gene coding for the p$47^{\rm{phox}}$ subunit of NOX2 and mice with transgenic rescue of Ncf1 under control of the CD68 promoter. To induce CGD hyperinflammation, different mouse genotypes were injected intradermally with ß-glucan. Ncf1 mutant mice showed massive and prolonged hyperinflammation. Hyperinflammatory lesions were characterized by persistent neutrophilic infiltration, along with ulceration and necrosis. In contrast, in CD68 promoter rescue mice inflammation resolved within days, as seen in wild-type animals. Measurements of ROS in rescue mice demonstrated functional NOX2 in mononuclear phagocytes (macrophages and dendritic cells) but not in neutrophils. This absence of NOX2 function was also confirmed in inflammatory tissue neutrophils. Lack of functional NOX2 in mononuclear phagocytes increased the secretion of IL-1ß at early time points and of IL-6 and TNFα at later time points. Thus, CGD hyperinflammation is a redox dysregulation in mononuclear phagocytes, demonstrating a cell type-specific anti-inflammatory function of NOX2.


Subject(s)
Dendritic Cells/metabolism , Granulomatous Disease, Chronic/prevention & control , Inflammation/prevention & control , Macrophages/metabolism , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cytokines/metabolism , Dendritic Cells/pathology , Disease Models, Animal , Granulomatous Disease, Chronic/metabolism , Granulomatous Disease, Chronic/pathology , Inflammation/chemically induced , Inflammation/pathology , Macrophages/pathology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Mutant Strains , Mice, Transgenic , NADPH Oxidase 2 , NADPH Oxidases/genetics , Neutrophils/metabolism , Neutrophils/pathology , Proteoglycans/adverse effects , Reactive Oxygen Species/metabolism , Receptors, Transforming Growth Factor beta
10.
Anesth Analg ; 116(6): 1257-64, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23592610

ABSTRACT

BACKGROUND: Volatile inhaled anesthetics exert a differential protective effect against bronchospasm development after cholinergic stimulation. However, their ability to inhibit the adverse respiratory consequences of an anaphylactic reaction after exposure to an allergen has not been characterized. We therefore compared the abilities of isoflurane, sevoflurane, and desflurane to prevent the lung constriction induced by an allergic reaction in a pediatric model of an anaphylactic reaction. METHODS: Low-frequency respiratory input impedance (Zrs) was measured in 4 groups of ovalbumin (OVA)-sensitized 5-week-old rabbit pups anesthetized with midazolam (group IV) and with inhaled isoflurane (group ISO), sevoflurane (group SEVO), or desflurane (group DES) at 1 minimum alveolar concentration. Zrs was measured under baseline conditions and after an anaphylactic lung response provoked by IV allergen injection (OVA 1 mg), during which the changes in airway resistance (Raw), tissue damping (G), and elastance obtained from Zrs were followed for 15 minutes. RESULTS: Allergen provocation generated immediate severe bronchoconstriction, with no statistically significant difference in Raw increase among the groups in the first 3 minutes. Conversely, the inhalation of volatile anesthetics accelerated the recovery from the allergen-induced bronchoconstriction, particularly in group SEVO where the Raw was significantly lower than that in group IV 4 minutes after the allergen challenge. These changes were paralleled by significant elevations in G in all groups, with a significantly more pronounced deterioration in the animals in group DES. The anesthetic regimen did not statistically significantly affect the sustained increases in elastance after OVA injections. CONCLUSIONS: Our results reveal the lack of potential of the commonly used volatile anesthetics to inhibit the most severe acute phase of the constrictor response to allergen after anaphylaxis in both the central airway and peripheral lung compartments. Inhalation of volatile anesthetics, particularly sevoflurane, promotes an earlier easing of the bronchospasm; this beneficial profile may be advantageous in children with atopic lung diseases.


Subject(s)
Anesthetics, Inhalation/pharmacology , Bronchoconstriction/drug effects , Hypersensitivity/physiopathology , Animals , Blood Pressure/drug effects , Female , Male , Ovalbumin/immunology , Rabbits
11.
Cell Mol Life Sci ; 69(14): 2373-85, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22581364

ABSTRACT

Acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS), is characterized by acute inflammation, disruption of the alveolar-capillary barrier, and in the organizing stage by alveolar pneumocytes hyperplasia and extensive lung fibrosis. The cellular and molecular mechanisms leading to the development of ALI/ARDS are not completely understood, but there is evidence that reactive oxygen species (ROS) generated by inflammatory cells as well as epithelial and endothelial cells are responsible for inflammatory response, lung damage, and abnormal repair. Among all ROS-producing enzymes, the members of NADPH oxidases (NOXs), which are widely expressed in different lung cell types, have been shown to participate in cellular processes involved in the maintenance of lung integrity. It is not surprising that change in NOXs' expression and function is involved in the development of ALI/ARDS. In this context, the use of NOX inhibitors could be a possible therapeutic perspective in the management of this syndrome. In this article, we summarize the current knowledge concerning some cellular aspects of NOXs localization and function in the lungs, consider their contribution in the development of ALI/ARDS and discuss the place of NOX inhibitors as potential therapeutical target.


Subject(s)
Acute Lung Injury/therapy , NADPH Oxidases/antagonists & inhibitors , Acute Lung Injury/enzymology , Acute Lung Injury/pathology , Cytokines/metabolism , Enzyme Inhibitors/therapeutic use , Humans , NADPH Oxidases/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Respiratory Distress Syndrome/enzymology , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/therapy
12.
Cells ; 11(3)2022 01 31.
Article in English | MEDLINE | ID: mdl-35159311

ABSTRACT

Cell therapies based on pluripotent stem cells (PSC), have opened new therapeutic strategies for neurodegenerative diseases. However, insufficiently differentiated PSC can lead to tumor formation. Ideally, safety switch therapies should selectively kill proliferative transplant cells while preserving post-mitotic neurons. In this study, we evaluated the potential of nucleoside analogs and thymidine kinase-based suicide genes. Among tested thymidine kinase variants, the humanized SR39 (SR39h) variant rendered cells most sensitive to suicide induction. Unexpectedly, post-mitotic neurons with ubiquitous SR39h expression were killed by ganciclovir, but were spared when SR39h was expressed under the control of the cell cycle-dependent Ki67 promoter. The efficacy of six different nucleoside analogs to induce cell death was then evaluated. Penciclovir (PCV) showed the most interesting properties with an efficiency comparable to ganciclovir (GCV), but low toxicity. We tested three nucleoside analogs in vivo: at concentrations of 40 mg/kg/day, PCV and GCV prevented tumor formation, while acyclovir (ACV) did not. In summary, SR39h under the control of a cell cycle-dependent promoter appears most efficient and selective as safety switch for neural transplants. In this setting, PCV and GCV are efficient inducers of cell death. Because of its low toxicity, PCV might become a preferred alternative to GCV.


Subject(s)
Nucleosides , Thymidine Kinase , Cell- and Tissue-Based Therapy , Ganciclovir/pharmacology , Humans , Neurons/metabolism , Thymidine Kinase/genetics , Thymidine Kinase/metabolism
13.
Am J Physiol Lung Cell Mol Physiol ; 299(3): L312-22, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20382751

ABSTRACT

Bcl-2 is an anti-apoptotic molecule preventing oxidative stress damage and cell death. We have previously shown that Bcl-2 is able to prevent hyperoxia-induced cell death when overexpressed in a murine fibrosarcoma cell line L929. We hypothesized that its specific overexpression in pulmonary epithelial type II cells could prevent hyperoxia-induced lung injury by protecting the epithelial side of the alveolo-capillary barrier. In the present work, we first showed that in vitro Bcl-2 can rescue murine pulmonary epithelial cells (MLE12) from oxygen-induced cell apoptosis, as shown by analysis of LDH release, annexin V/propidium staining, and caspase-3 activity. We then generated transgenic mice overexpressing specifically Bcl-2 in lung epithelial type II cells under surfactant protein C (SP-C) promoter (Tg-Bcl-2) and exposed them to hyperoxia. Bcl-2 did not hinder hyperoxia-induced mitochondria and DNA oxidative damage of type II cell in vivo. Accordingly, lung damage was identical in both Tg-Bcl-2 and littermate mice strains, as measured by lung weight, bronchoalveolar lavage, and protein content. Nevertheless, we observed a significant lower number of TUNEL-positive cells in type II cells isolated from Tg-Bcl-2 mice exposed to hyperoxia compared with cells isolated from littermate mice. In summary, these results show that although Bcl-2 overexpression is able to prevent hyperoxia-induced cell death at single cell level in vitro and ex vivo, it is not sufficient to prevent cell death of parenchymal cells and to protect the lung from acute damage in mice.


Subject(s)
Acute Lung Injury/prevention & control , Epithelial Cells/metabolism , Hyperoxia/complications , Hyperoxia/metabolism , Lung/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Acute Lung Injury/etiology , Animals , Apoptosis , Cell Death , Cells, Cultured , DNA Damage , Epithelial Cells/classification , Hyperoxia/pathology , Hyperoxia/physiopathology , Lung/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/pathology , Oxidative Stress , Proto-Oncogene Proteins c-bcl-2/genetics , Up-Regulation
14.
Am J Respir Crit Care Med ; 180(10): 972-81, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19661248

ABSTRACT

RATIONALE: Hyperoxia-induced acute lung injury has been used for many years as a model of oxidative stress mimicking clinical acute lung injury and the acute respiratory distress syndrome. Excess quantities of reactive oxygen species (ROS) are responsible for oxidative stress-induced lung injury. ROS are produced by mitochondrial chain transport, but also by NADPH oxidase (NOX) family members. Although NOX1 and NOX2 are expressed in the lungs, their precise function has not been determined until now. OBJECTIVES: To determine whether NOX1 and NOX2 contribute in vivo to hyperoxia-induced acute lung injury. METHODS: Wild-type and NOX1- and NOX2-deficient mice, as well as primary lung epithelial and endothelial cells, were exposed to room air or 100% O(2) for 72 hours. MEASUREMENTS AND MAIN RESULTS: Lung injury was significantly prevented in NOX1-deficient mice, but not in NOX2-deficient mice. Hyperoxia-dependent ROS production was strongly reduced in lung sections, in isolated epithelial type II cells, and lung endothelial cells from NOX1-deficient mice. Concomitantly, lung cell death in situ and in primary cells was markedly decreased in NOX1-deficient mice. In wild-type mice, hyperoxia led to phosphorylation of c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK), two mitogen-activated protein kinases involved in cell death signaling, and to caspase-3 activation. In NOX1-deficient mice, JNK phosphorylation was blunted, and ERK phosphorylation and caspase-3 activation were decreased. CONCLUSIONS: NOX1 is an important contributor to ROS production and cell death of the alveolocapillary barrier during hyperoxia and is an upstream actor in oxidative stress-induced acute lung injury involving JNK and ERK pathways in mice.


Subject(s)
Hypoxia/complications , Lung Injury/enzymology , NADPH Oxidases/physiology , Animals , Cell Death/physiology , Endothelium/cytology , Epithelial Cells/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lung/cytology , Lung Injury/etiology , Mice , Mice, Inbred C57BL , NADPH Oxidases/deficiency , Phosphorylation , Reactive Oxygen Species/metabolism
15.
Biochim Biophys Acta ; 1773(7): 1015-27, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17553579

ABSTRACT

Adipocyte differentiation is a complex process regulated among other factors by insulin and the production of reactive oxygen species (ROS). NOX4 is a ROS generating NADPH oxidase enzyme mediating insulin's action in 3T3L1 adipocytes. In the present paper we show that NOX4 is expressed at high levels both in white and brown preadipocytes and that differentiation into adipocytes results in a decrease in their NOX4 mRNA content. These in vitro results were confirmed in vivo by demonstrating that in intact adipose tissue the majority of NOX4 expressing cells are localized within the preadipocyte containing stromal/vascular fraction, rather than in the portion consisting of mature adipocytes. In line with these observations, quantification of NOX4 mRNA in fat derived from different rodent models of insulin resistance indicated that alteration in NOX4 expression reflects changes in the ratio of adipocyte/interstitial fractions. In conclusion, we reveal that decreased NOX4 mRNA content is a hallmark of adipocyte differentiation and that NOX4 expression measured in whole adipose tissue is not an unequivocal indicator of intact or impaired insulin action.


Subject(s)
Adipocytes/enzymology , Adipocytes/physiology , Cell Differentiation , Gene Expression Regulation, Enzymologic , NADPH Oxidases/metabolism , 3T3 Cells , Adipocytes/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/enzymology , Animals , Catalase/metabolism , Cells, Cultured , Dietary Fats , Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factor 2/metabolism , Humans , Insulin/metabolism , Insulin Resistance/physiology , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Knockout , Mice, Obese , NADP/metabolism , NADPH Oxidase 4 , NADPH Oxidases/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction/physiology , Superoxide Dismutase/metabolism
16.
Free Radic Biol Med ; 42(7): 1062-74, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17349933

ABSTRACT

Bcl-2 is an antiapoptotic molecule that prevents oxidative stress damage and cell death. We investigated the possible protective mechanisms mediated by Bcl-2 during hyperoxia-induced cell death in L929 cells. In these cells, hyperoxia promoted apoptosis without DNA fragmentation. Overexpression of Bcl-2 significantly protected cells from oxygen-induced apoptosis, as shown by measurement of lactate dehydrogenase release, quantification of apoptotic nuclei, and detection of Annexin-V-positive cells. Bcl-2 partially prevented mitochondrial damage and interfered with the mitochondrial proapoptotic signaling pathway: it reduced Bax translocation to mitochondria, decreased the release of cytochrome c, and inhibited caspase 3 activation. However, treatment with the caspase inhibitor Z-VAD.fmk failed to rescue the cells from death, indicating that protection provided by Bcl-2 was due not only to caspase inhibition. Bcl-2 also prevented the release of mitochondrial apoptotic inducing factor, a mediator of caspase-independent apoptosis, correlating with the absence of oligonucleosomal DNA fragmentation. In addition, Bcl-2-overexpressing cells showed significantly higher intracellular amounts of glutathione after 72 h of oxygen exposure. In conclusion, our results demonstrate that the overexpression of Bcl-2 is able to prevent hyperoxia-induced cell death, by affecting mitochondria-dependent apoptotic pathways and increasing intracellular antioxidant compounds.


Subject(s)
Apoptosis/physiology , Hyperoxia/physiopathology , Mitochondria/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , Caspase Inhibitors , Caspases/metabolism , Cell Line , Cytochromes c/metabolism , Glutathione/metabolism , Immunohistochemistry , Mice , Microscopy, Electron , Reactive Oxygen Species/metabolism
17.
Curr Pharm Des ; 21(41): 5977-87, 2015.
Article in English | MEDLINE | ID: mdl-26510434

ABSTRACT

Inner ear pathologies are associated with major morbidity and loss of life quality in affected patients. In many of these conditions, production of reactive oxygen-species (ROS) is thought to be a key pathological mechanism. While the sources of ROS are complex (including for example mitochondria), there is increasing evidence that activation of NOX enzymes, in particular NOX3, plays a key role. NOX3 is a multi-subunit NADPH oxidase, functionally and structurally closely related to NOX1 and NOX2. In both the vestibular and the cochlear compartments of the inner ear, high levels of NOX3 mRNA are expressed. In NOX3 mutant mice, the vestibular function is perturbed due to a lack of otoconia, while only minor alterations of hearing have been documented. However, there is increasing evidence that activation of NOX3 through drugs, noise and probably also aging, leads to hearing loss. Thus, NOX3 is an interesting target to treat and prevent inner ear pathologies and a few first animal models based on drug - or molecular therapy have been reported. So far however, there are no specific NOX3 inhibitors with a documented penetration into the inner ear. Nevertheless, certain antioxidants and non-specific NOX inhibitors diminish hearing loss in animal models. Development of small molecules inhibitors or molecular strategies against NOX3 could improve specificity and efficiency of redox-targeted treatments. In this review, we will discuss arguments for the involvement of NOX3 in inner ear pathologies and therapeutic approaches to target NOX3 activity.


Subject(s)
Labyrinth Diseases/drug therapy , Membrane Proteins/antagonists & inhibitors , NADPH Oxidases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Humans , Labyrinth Diseases/metabolism , Labyrinth Diseases/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry
18.
Cancer Lett ; 215(1): 53-9, 2004 Nov 08.
Article in English | MEDLINE | ID: mdl-15374632

ABSTRACT

We investigated on colon cancer cells the effect of geraniol on thymidylate synthase and thymidine kinase expression, two enzymes related to 5-fluorouracil cytotoxicity. The anti-tumoral efficacy of geraniol and 5-fluorouracil were also evaluated on TC-118 human tumors transplanted in Swiss nu/nu mice. Geraniol (150 microM) but not 5-fluorouracil caused a 2-fold reduction of thymidylate synthase and thymidine kinase expression in cancer cells. In nude mice, the combined administration of 5-fluorouracil (20 mg/kg) and geraniol (150 mg/kg) caused a 53% reduction of the tumor volume, whereas a 26% reduction was obtained with geraniol alone, 5-fluorouracil alone showed no effect.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Colonic Neoplasms/drug therapy , DNA/metabolism , Fluorouracil/therapeutic use , Oils, Volatile , Terpenes/administration & dosage , Acyclic Monoterpenes , Animals , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Drug Therapy, Combination , Female , Humans , Mice , Mice, Nude , Plant Oils , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
19.
Cancer Lett ; 175(2): 147-55, 2002 Jan 25.
Article in English | MEDLINE | ID: mdl-11741742

ABSTRACT

The effects of cocoa powder and extracts with different amounts of flavanols and related procyanidin oligomers were investigated on the growth of Caco-2 cells. Treatment of the cells with 50 microg/ml of procyanidin-enriched (PE) extracts caused a 70% growth inhibition with a blockade of the cell cycle at the G2/M phase. PE extracts caused a significant decrease of ornithine decarboxylase and S-adenosylmethionine decarboxylase activities, two key enzymes of polyamine biosynthesis. This led to a decrease in the intracellular pool of the polyamines. These observations indicate that polyamine metabolism might be an important target in the anti-proliferative effects of cocoa polyphenols.


Subject(s)
Antioxidants/pharmacology , Biflavonoids , Cacao/chemistry , Catechin/pharmacology , Cell Division/drug effects , Flavonoids/pharmacology , Plant Extracts/pharmacology , Polyamines/metabolism , Proanthocyanidins , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Survival/drug effects , Colonic Neoplasms , Humans , Kinetics , Phytotherapy , Tumor Cells, Cultured
20.
Free Radic Biol Med ; 76: 173-84, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25151121

ABSTRACT

Nicotine contributes to the onset and progression of several pulmonary diseases. Among the various pathophysiological mechanisms triggered by nicotine, oxidative stress and cell death are reported in several cell types. We found that chronic exposure to nicotine (48h) induced NOX1-dependent oxidative stress and apoptosis in primary pulmonary cells. In murine (MLE-12) and human (BEAS-2B) lung epithelial cell lines, nicotine acted as a sensitizer to cell death and synergistically enhanced apoptosis when cells were concomitantly exposed to hyperoxia. The precise signaling pathway was investigated in MLE-12 cells in which NOX1 was abrogated by a specific inhibitor or stably silenced by shRNA. In the early phase of exposure (1h), nicotine mediated intracellular Ca(2+) fluxes and activation of protein kinase C, which in its turn activated NOX1, leading to cellular and mitochondrial oxidative stress. The latter triggered the intrinsic apoptotic machinery by modulating the expression of Bcl-2 and Bax. Overexpression of Bcl-2 completely prevented nicotine's detrimental effects, suggesting Bcl-2as a downstream key regulator in nicotine/NOX1-induced cell damage. These results suggest that NOX1 is a major contributor to the generation of intracellular oxidative stress induced by nicotine and might be an important molecule to target in nicotine-related lung pathologies.


Subject(s)
Apoptosis/drug effects , Epithelial Cells/pathology , Lung/pathology , NADH, NADPH Oxidoreductases/physiology , Nicotine/pharmacology , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Blotting, Western , Calcium/metabolism , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADPH Oxidase 1 , Nicotinic Agonists/pharmacology , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL