Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Biomed Pharmacother ; 174: 116552, 2024 May.
Article En | MEDLINE | ID: mdl-38599061

AIMS: Pulmonary hypertension (PH) is characterised by an increase in pulmonary arterial pressure, ultimately leading to right ventricular failure and death. We have previously shown that nerve growth factor (NGF) plays a critical role in PH. Our objectives here were to determine whether NGF controls Connexin-43 (Cx43) expression and function in the pulmonary arterial smooth muscle, and whether this mechanism contributes to NGF-induced pulmonary artery hyperreactivity. METHODS AND RESULTS: NGF activates its TrkA receptor to increase Cx43 expression, phosphorylation, and localization at the plasma membrane in human pulmonary arterial smooth muscle cells, thus leading to enhanced activity of Cx43-dependent GAP junctions as shown by Lucifer Yellow dye assay transfer and fluorescence recovery after photobleaching -FRAP- experiments. Using both in vitro pharmacological and in vivo SiRNA approaches, we demonstrate that NGF-dependent increase in Cx43 expression and activity in the rat pulmonary circulation causes pulmonary artery hyperreactivity. We also show that, in a rat model of PH induced by chronic hypoxia, in vivo blockade of NGF or of its TrkA receptor significantly reduces Cx43 increased pulmonary arterial expression induced by chronic hypoxia and displays preventive effects on pulmonary arterial pressure increase and right heart hypertrophy. CONCLUSIONS: Modulation of Cx43 by NGF in pulmonary arterial smooth muscle cells contributes to NGF-induced alterations of pulmonary artery reactivity. Since NGF and its TrkA receptor play a role in vivo in Cx43 increased expression in PH induced by chronic hypoxia, these NGF/Cx43-dependent mechanisms may therefore play a significant role in human PH pathophysiology.


Connexin 43 , Myocytes, Smooth Muscle , Nerve Growth Factor , Pulmonary Artery , Animals , Humans , Male , Rats , Cells, Cultured , Connexin 43/metabolism , Gap Junctions/metabolism , Gap Junctions/drug effects , Hypertension, Pulmonary/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Nerve Growth Factor/metabolism , Phosphorylation , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Rats, Sprague-Dawley , Rats, Wistar , Receptor, trkA/metabolism
2.
Br J Pharmacol ; 180(21): 2802-2821, 2023 11.
Article En | MEDLINE | ID: mdl-37351910

BACKGROUND AND PURPOSE: Pulmonary hypertension (PH) is a cardiovascular disease characterised by an increase in pulmonary arterial (PA) resistance leading to right ventricular (RV) failure. Reactive oxygen species (ROS) play a major role in PH. OP2113 is a drug with beneficial effects on cardiac injuries that targets mitochondrial ROS. The aim of the study was to address the in vivo therapeutic effect of OP2113 in PH. EXPERIMENTAL APPROACH: PH was induced by 3 weeks of chronic hypoxia (CH-PH) in rats treated with OP2113 or its vehicle via subcutaneous osmotic mini-pumps. Haemodynamic parameters and both PA and heart remodelling were assessed. Reactivity was quantified in PA rings and in RV or left ventricular (LV) cardiomyocytes. Oxidative stress was detected by electron paramagnetic resonance and western blotting. Mitochondrial mass and respiration were measured by western blotting and oxygraphy, respectively. KEY RESULTS: In CH-PH rats, OP2113 reduced the mean PA pressure, PA remodelling, PA hyperreactivity in response to 5-HT, the contraction slowdown in RV and LV and increased the mitochondrial mass in RV. Interestingly, OP2113 had no effect on haemodynamic parameters, both PA and RV wall thickness and PA reactivity, in control rats. Whereas oxidative stress was evidenced by an increase in protein carbonylation in CH-PH, this was not affected by OP2113. CONCLUSION AND IMPLICATIONS: Our study provides evidence for a selective protective effect of OP2113 in vivo on alterations in both PA and RV from CH-PH rats without side effects in control rats.


Heart Failure , Hypertension, Pulmonary , Ventricular Dysfunction, Right , Rats , Animals , Hypertension, Pulmonary/metabolism , Reactive Oxygen Species/metabolism , Heart Ventricles/metabolism , Pulmonary Artery , Heart Failure/metabolism , Hypoxia/complications , Hypoxia/drug therapy , Hypoxia/metabolism , Ventricular Dysfunction, Right/metabolism , Ventricular Function, Right , Disease Models, Animal
3.
J Toxicol ; 2022: 1786810, 2022.
Article En | MEDLINE | ID: mdl-36310640

Particulate matter (PM) is one of the main air pollutants with 257,000 deaths per year in Africa. Studying their toxic mechanisms of action could provide a better understanding of their effects on the population health. The objective of this study was to describe the PM10 toxic mechanism of action collected in 3 districts of Ouagadougou. Once per month and per site between November 2015 and February 2016, PM10 was sampled for 24 hours using the MiniVol TAS (AirMetrics, Eugene, USA). The collected filters were then stored in Petri dishes at room temperature for in vitro toxicological studies using human pulmonary artery endothelial cells (HPAEC) at the Bordeaux INSERM-U1045 Cardio-thoracic Research Center. The three study districts were classified based on PM10 level (high, intermediate, and low, respectively, for districts 2, 3, and 4). PM10 induced a concentration-dependent decrease in cell viability. A significant decrease in cell viability was observed at 1 µg/cm2, 10 µg/cm2, and 25 µg/cm2 for, respectively, districts 2, 3, and 4. A significant increase in the production of reactive oxygen species (ROS) was observed at 10 µg/cm2 for district 2 versus 5 µg/cm2 and 1 µg/cm2 for districts 3 and 4, respectively. Finally, a significant production of IL-6 was recorded from 5 µg/cm2 for district 4 versus 10 µg/cm2 for districts 2 and 3. Consequently, Ouagadougou is subjected to PM10 pollution, which can induce a significant production of ROS and IL-6 to cause adverse effects on the health of the population.

4.
Cells ; 11(18)2022 09 07.
Article En | MEDLINE | ID: mdl-36139373

Expression of the nerve growth factor NGF is increased in pulmonary hypertension (PH). We have here studied whether oxidative stress and inflammation, two pathological conditions associated with transforming growth factor-ß1 (TGF-ß1) in PH, may trigger NGF secretion by pulmonary arterial (PA) cells. Effects of hydrogen peroxide (H2O2) and interleukin-1ß (IL-1ß) were investigated ex vivo on rat pulmonary arteries, as well as in vitro on human PA smooth muscle (hPASMC) or endothelial cells (hPAEC). TßRI expression was assessed by Western blotting. NGF PA secretion was assessed by ELISA after TGF-ß1 blockade (anti-TGF-ß1 siRNA, TGF-ß1 blocking antibodies, TßRI kinase, p38 or Smad3 inhibitors). TßRI PA expression was evidenced by Western blotting both ex vivo and in vitro. H2O2 or IL-1ß significantly increased NGF secretion by hPASMC and hPAEC, and this effect was significantly reduced when blocking TGF-ß1 expression, binding to TßRI, TßRI activity, or signaling pathways. In conclusion, oxidative stress and inflammation may trigger TGF-ß1 secretion by hPASMC and hPAEC. TGF-ß1 may then act as an autocrine factor on these cells, increasing NGF secretion via TßRI activation. Since NGF and TGF-ß1 are relevant growth factors involved in PA remodeling, such mechanisms may therefore be relevant to PH pathophysiology.


Hypertension, Pulmonary , Transforming Growth Factor beta1 , Animals , Antibodies, Blocking , Endothelial Cells/metabolism , Humans , Hydrogen Peroxide/metabolism , Hypertension, Pulmonary/metabolism , Inflammation/pathology , Interleukin-1beta/metabolism , Nerve Growth Factor/metabolism , Oxidative Stress , Pulmonary Artery/metabolism , RNA, Small Interfering/metabolism , Rats , Transforming Growth Factor beta1/metabolism
5.
Cells ; 11(15)2022 07 30.
Article En | MEDLINE | ID: mdl-35954193

In intrapulmonary arteries (IPAs), mechanical forces due to blood flow control vessel tone, and these forces change during pulmonary hypertension (PH). Piezo1, a stretch-activated calcium channel, is a sensor of mechanical stress present in both endothelial cells (ECs) and smooth muscle cells (SMCs). The present study investigated the role of Piezo1 on IPA in the chronic hypoxia model of PH. Rats were raised in chronically hypoxic conditions for 1 (1W-CH, early stage) or 3 weeks (3W-CH, late-stage) of PH or in normoxic conditions (Nx). Immunofluorescence labeling and patch-clamping revealed the presence of Piezo1 in both ECs and SMCs. The Piezo1 agonist, Yoda1, induced an IPA contraction in Nx and 3W-CH. Conversely, Yoda1 induced an endothelial nitric oxide (eNOS) dependent relaxation in 1W-CH. In ECs, the Yoda1-mediated intracellular calcium concentration ([Ca2+]i) increase was greater in 1W-CH as compared to Nx. Yoda1 induced an EC hyperpolarization in 1W-CH. The eNOS levels were increased in 1W-CH IPA compared to Nx or 3W-CH PH and Yoda1 activated phosphorylation of Akt (Ser473) and eNOS (Ser1177). Thus, we demonstrated that endothelial Piezo1 contributes to intrapulmonary vascular relaxation by controlling endothelial [Ca2+]i, endothelial-dependent hyperpolarization, and Akt-eNOS pathway activation in the early stage of PH.


Hypertension, Pulmonary , Animals , Endothelial Cells/metabolism , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Nitric Oxide/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/metabolism , Rats , Vasoconstriction/physiology
6.
Biomolecules ; 12(7)2022 07 07.
Article En | MEDLINE | ID: mdl-35883510

Transient receptor potential vanilloid 4 (TRPV4) is a polymodal Ca2+-permeable channel involved in various hypoxia-sensitive pathophysiological phenomena. Different tools are available to study channel activity, requiring cells to be cultured at specific optimal densities. In the present study, we examined if cell density may influence the effect of hypoxia on TRPV4 activity. Transiently TRPV4-transfected HEK293T cells were seeded at low or high densities corresponding to non-confluent or confluent cells, respectively, on the day of experiments, and cultured under in vitro normoxia or hypoxia. TRPV4-mediated cytosolic Ca2+ responses, single-channel currents, and Ca2+ influx through the channel were measured using Ca2+ imaging/microspectrofluorimetric assay, patch-clamp, and Bioluminescence Resonance Energy Transfer (BRET), respectively. TRPV4 plasma membrane translocation was studied using confocal microscopy, biotinylation of cell surface proteins, and BRET. Our results show that hypoxia exposure has a differential effect on TRPV4 activation depending on cell confluence. At low confluence levels, TRPV4 response is increased in hypoxia, whereas at high confluence levels, TRPV4 response is strongly inhibited, due to channel internalization. Thus, cell density appears to be a crucial parameter for TRPV4 channel activity.


TRPV Cation Channels , Transient Receptor Potential Channels , Calcium/metabolism , HEK293 Cells , Humans , Hypoxia/metabolism , Patch-Clamp Techniques , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
7.
J Hazard Mater ; 436: 129285, 2022 08 15.
Article En | MEDLINE | ID: mdl-35739794

New Caledonia is particularly affected by nickel open pit mining activities because of the presence of ultramafic soils rich in metals. The particles dispersed by atmospheric transport and soil erosion during the excavation of nickel end up by deposition or leaching in rivers where they may be bioaccumulated by organisms living downstream the mines. Despite alarming freshwater metals concentrations, no study investigated the level of their bioaccumulation in eels, and if high bioaccumulation levels occur, the potential consequences on their health. The aim of this study was to determine how eels Anguilla marmorata are impacted in situ by metals issued from mining activity by measuring: morphometric parameters; metal concentrations in tissues and organs and transcription levels of target genes encoding proteins involved in several metabolic key functions. Among organs, liver was found to be the most affected by mining with average nickel concentrations of 5.14 mg/kg versus 1.63 mg/kg for eels away from mines leading to dysregulation of numerous genes involved in oxidative stress, DNA repair, apoptosis, reproduction and both lipid and mitochondrial metabolisms. This study should allow us to define in an integrated way if metals released by mining activities influence metals bioaccumulation in eels and induce biological effects.


Anguilla , Anguilla/physiology , Animals , Metals/toxicity , Mining , New Caledonia , Nickel/toxicity , Rivers
8.
Antioxidants (Basel) ; 11(5)2022 Apr 26.
Article En | MEDLINE | ID: mdl-35624710

The development and use of nanomaterials, especially of nickel oxide nanoparticles (NiONPs), is expected to provide many benefits but also has raised concerns about the potential human health risks. Inhaled NPs are known to exert deleterious cardiovascular side effects, including pulmonary hypertension. Consequently, patients with pulmonary hypertension (PH) could be at increased risk for morbidity. The objective of this study was to compare the toxic effects of NiONPs on human pulmonary artery endothelial cells (HPAEC) under physiological and pathological conditions. The study was conducted with an in vitro model mimicking the endothelial dysfunction observed in PH. HPAEC were cultured under physiological (static and normoxic) or pathological (20% cycle stretch and hypoxia) conditions and exposed to NiONPs (0.5-5 µg/cm2) for 4 or 24 h. The following endpoints were studied: (i) ROS production using CM-H2DCF-DA and MitoSOX probes, (ii) nitrite production by the Griess reaction, (iii) IL-6 secretion by ELISA, (iv) calcium signaling with a Fluo-4 AM probe, and (v) mitochondrial dysfunction with TMRM and MitoTracker probes. Our results evidenced that under pathological conditions, ROS and nitrite production, IL-6 secretions, calcium signaling, and mitochondria alterations increased compared to physiological conditions. Human exposure to NiONPs may be associated with adverse effects in vulnerable populations with cardiovascular risks.

9.
Chemosphere ; 303(Pt 2): 135158, 2022 Sep.
Article En | MEDLINE | ID: mdl-35640691

Anthropic activities such as open pit mining, amplify the natural erosion of metals contained in the soils, particularly in New Caledonia, leading to atmospheric emission of nickel oxide nanoparticles (NiONPs). These particles are produced during extraction end up in aquatic ecosystems through deposition or leaching in the rivers. Despite alarming freshwater Ni concentrations, only few studies have focused on the cellular and molecular mechanisms of NiONPs toxicity on aquatic organisms and particularly on eels. Those fish are known to be sensitive to metal contamination, especially their liver, which is a key organ for lipid metabolism, detoxification and reproduction. The objective of this study was to assess in vitro the cytotoxic effects of NiONPs on Anguilla japonica hepatocytes, HEPA-E1. HEPA-E1 were exposed to NiONPs (0.5-5 µg/cm2) for 4 or 24 h. Several endpoints were studied: (i) viability, (ii) ROS production, SOD activity and selected anti-oxidant genes expression, (iii) inflammation, (iv) calcium signalling, (v) mitochondrial function and (vi) apoptosis. The results evidenced that NiONPs induce a decrease of cell viability and an increase in oxidative stress with a significant superoxide anion production. An increase of mitochondrial calcium concentration and a decrease of mitochondrial membrane potential were observed, leading to apoptosis. These results underline the potential toxic impact of NiONPs on eels living in mining areas. Therefore, eel exposure to NiONPs can affect their migration and reproduction in New Caledonia.


Anguilla , Ecosystem , Anguilla/metabolism , Animals , Calcium/metabolism , Hepatocytes , New Caledonia
10.
Nanotoxicology ; 16(1): 29-51, 2022 02.
Article En | MEDLINE | ID: mdl-35090355

In New Caledonia, anthropic activities, such as mining, increase the natural erosion of soils in nickel mines, which in turn, releases nickel oxide nanoparticles (NiONPs) into the atmosphere. Pulmonary vascular endothelial cells represent one of the primary targets for inhaled nanoparticles. The objective of this in vitro study was to assess the cytotoxic effects of NiONPs on human pulmonary artery endothelial cells (HPAEC). Special attention will be given to the level of oxidative stress and calcium signaling, which are involved in the physiopathology of cardiovascular diseases. HPAEC were exposed to NiONPs (0.5-150 µg/cm2) for 4 or 24 h. The following different endpoints were studied: (i) ROS production using CM-H2DCF-DA probe, electron spin resonance, and MitoSOX probe; the SOD activity was also measured (ii) calcium signaling with Fluo4-AM, Rhod-2, and Fluo4-FF probes; (iii) inflammation by IL-6 production and secretion and, (iv) mitochondrial dysfunction and apoptosis with TMRM and MitoTracker probes, and AnnexinV/PI. Our results have evidenced that NiONPs induced oxidative stress in HPAEC. This was demonstrated by an increase in ROS production and a decrease in SOD activity, the two mechanisms seem to trigger a pro-inflammatory response with IL-6 secretion. In addition, NiONPs exposure altered calcium homeostasis inducing an increased cytosolic calcium concentration ([Ca2+]i) that was significantly reduced by the extracellular calcium chelator EGTA and the TRPV4 inhibitor HC-067047. Interestingly, exposure to NiONPs also altered TRPV4 activity. Finally, HPAEC exposure to NiONPs increased intracellular levels of both ROS and calcium ([Ca2+]m) in mitochondria, leading to mitochondrial dysfunction and HPAEC apoptosis.


Calcium Signaling , Endothelial Cells , Metal Nanoparticles , Mitochondria , Oxidative Stress , TRPV Cation Channels , Calcium/metabolism , Cells, Cultured , Endothelial Cells/drug effects , Humans , Interleukin-6/metabolism , Metal Nanoparticles/adverse effects , Mitochondria/pathology , Nickel/adverse effects , Pulmonary Artery/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , TRPV Cation Channels/metabolism
11.
Biomolecules ; 11(9)2021 09 21.
Article En | MEDLINE | ID: mdl-34572602

A variety of cell types in pulmonary arteries (endothelial cells, fibroblasts, and smooth muscle cells) are continuously exposed to mechanical stimulations such as shear stress and pulsatile blood pressure, which are altered under conditions of pulmonary hypertension (PH). Most functions of such vascular cells (e.g., contraction, migration, proliferation, production of extracellular matrix proteins, etc.) depend on a key event, i.e., the increase in intracellular calcium concentration ([Ca2+]i) which results from an influx of extracellular Ca2+ and/or a release of intracellular stored Ca2+. Calcium entry from the extracellular space is a major step in the elevation of [Ca2+]i, involving a variety of plasmalemmal Ca2+ channels including the superfamily of stretch-activated channels (SAC). A common characteristic of SAC is that their gating depends on membrane stretch. In general, SAC are non-selective Ca2+-permeable cation channels, including proteins of the TRP (Transient Receptor Potential) and Piezo channel superfamily. As membrane mechano-transducers, SAC convert physical forces into biological signals and hence into a cell response. Consequently, SAC play a major role in pulmonary arterial calcium homeostasis and, thus, appear as potential novel drug targets for a better management of PH.


Calcium Channels/metabolism , Hypertension, Pulmonary/physiopathology , Pulmonary Circulation/physiology , Animals , Biomechanical Phenomena , Biophysical Phenomena , Humans , Models, Biological
12.
Environ Sci Technol ; 54(4): 2360-2369, 2020 02 18.
Article En | MEDLINE | ID: mdl-31961142

Regulations on ambient particulate matter (PM) are becoming more stringent because of adverse health effects arising from PM exposure. PM-induced oxidant production is a key mechanism behind the observed health effects and is heavily dependent on PM composition. Measurement of the intrinsic oxidative potential (OP) of PM could provide an integrated indicator of PM bioreactivity and could serve as a better metric of PM hazard exposure than PM mass concentration. The OP of two chemically contrasted PM2.5 samples was compared through four acellular assays, and OP predictive capability was evaluated in different cellular assays on two in vitro lung cell models. PM2.5 collected in Paris at a site close to the traffic exhibited a systematically higher OP in all assays compared to PM2.5 enriched in particles from domestic wood burning. Similar results were obtained for oxidative stress, expression of antioxidant enzymes, and pro-inflammatory chemokine in human bronchial epithelial and endothelial cells. The strongest correlations between OP assays and cellular responses were observed with the antioxidant (ascorbic acid and glutathione) depletion (OPAO) assay. Multivariate regression analysis from OP daily measurements suggested that OPAO was strongly correlated with polycyclic aromatic hydrocarbons at the traffic site while it was correlated with potassium for the domestic wood burning sample.


Air Pollutants , Antioxidants , Endothelial Cells , Humans , Oxidation-Reduction , Oxidative Stress , Particle Size , Particulate Matter
13.
Am J Respir Cell Mol Biol ; 60(6): 650-658, 2019 06.
Article En | MEDLINE | ID: mdl-30562052

In intrapulmonary arteries (IPA), endothelial cells (EC) respond to mechanical stimuli by releasing vasoactive factors to set the vascular tone. Piezo1, a stretch-activated, calcium-permeable channel, is a sensor of mechanical stress in EC. The present study was undertaken to investigate the implication of Piezo1 in the endothelium-dependent regulation of IPA tone and potential involvement of Piezo1 in pulmonary hypertension, the main disease of this circulation. IPA tone was quantified by means of a myograph in control Piezo1+/+ mice and in mice lacking endothelial Piezo1 (EC-Piezo1-/-). Endothelial intracellular calcium concentration ([Ca2+]i) and nitric oxide (NO) production were measured, in mouse or human EC, with Fluo-4 or DAF-FM probe, respectively. Immunofluorescent labeling and patch-clamp experiments revealed the presence of Piezo1 channels in EC. Yoda1, a Piezo1 agonist, induced an endothelium-dependent relaxation that was significantly reduced in pulmonary arteries in EC-Piezo1-/- compared with Piezo1+/+ mice. Yoda1 as well as mechanical stimulation (by osmotic stress) increased [Ca2+]i in mouse or human EC. Consequently, both stimuli increased the production of NO. NO and [Ca2+]i increases were reduced in EC from Piezo1-/- mice or in the presence of Piezo1 inhibitors. Furthermore, deletion of Piezo1 increased α-adrenergic agonist-mediated contraction. Finally, in chronically hypoxic mice, a model of pulmonary hypertension, Piezo1 still mediated arterial relaxation, and deletion of this channel did not impair the development of the disease. The present study thus demonstrates that endothelial Piezo1 contributes to intrapulmonary vascular relaxation by controlling endothelial [Ca2+]i and NO production and that this effect is still present in pulmonary hypertension.


Endothelial Cells/metabolism , Ion Channels/metabolism , Pulmonary Artery/metabolism , Animals , Calcium/metabolism , Chronic Disease , Humans , Hypoxia/metabolism , Hypoxia/pathology , Ion Channels/agonists , Mice, Inbred C57BL , Nitric Oxide/biosynthesis , Pulmonary Artery/pathology , Vasoconstriction , Vasodilation
14.
Toxicology ; 375: 37-47, 2017 Jan 15.
Article En | MEDLINE | ID: mdl-27939335

The development and use of nanomaterials, especially engineered nanoparticles (NP), is expected to provide many benefits. But at the same time the development of such materials is also feared because of their potential human health risks. Indeed, NP display some characteristics similar to ultrafine environmental particles which are known to exert deleterious cardiovascular effects including pro-hypertensive ones. In this context, the effect of NP on calcium signalling, whose deregulation is often involved in hypertensive diseases, remain poorly described. We thus assessed the effect of SiO2 NP on calcium signalling by fluorescence imaging and on the proliferation response in rat pulmonary artery smooth muscle cells (PASMC). In PASMC, acute exposure to SiO2 NP, from 1 to 500µg/mL, produced an increase of the [Ca2+]i. In addition, when PASMC were exposed to NP at 200µg/mL, a proliferative response was observed. This calcium increase was even greater in PASMC isolated from rats suffering from pulmonary hypertension. The absence of extracellular calcium, addition of diltiazem or nicardipine (L-type voltage-operated calcium channel inhibitors both used at 10µM), and addition of capsazepine or HC067047 (TRPV1 and TRPV4 inhibitors used at 10µM and 5µM, respectively) significantly reduced this response. Moreover, this response was also inhibited by thapsigargin (SERCA inhibitor, 1µM), ryanodine (100µM) and dantrolene (ryanodine receptor antagonists, 10µM) but not by xestospongin C (IP3 receptor antagonist, 10µM). Thus, NP induce an intracellular calcium rise in rat PASMC originating from both extracellular and intracellular calcium sources. This study also provides evidence for the implication of TRPV channels in NP induced calcium rise that may highlight the role of these channels in the deleterious cardiovascular effects of NP.


Calcium Signaling/drug effects , Myocytes, Smooth Muscle/drug effects , Nanoparticles/toxicity , Pulmonary Artery/drug effects , Silicon Dioxide/toxicity , Animals , Calcium Signaling/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Male , Myocytes, Smooth Muscle/physiology , Pulmonary Artery/physiology , Rats , Rats, Wistar
15.
Toxicol In Vitro ; 32: 205-11, 2016 Apr.
Article En | MEDLINE | ID: mdl-26780163

Particulate air pollution exerts deleterious effects on cardiovascular system. We previously described that exposure to urban particulate matter (SRM1648) impairs nitric oxide (NO, a major vasculoprotective factor) responsiveness in intrapulmonary arteries. As Heme Oxygenase-1 (HO-1) is induced by urban particles in some cell types and is known to alter NO-dependent signaling pathway, the objective was to characterize HO-1 involvement in SRM1648-induced impairment of NO-dependent relaxation in intrapulmonary arteries. Rat intrapulmonary artery rings were exposed or not to Co (III) Protoporphyrin IX Chloride (HO-1 inducer) or SRM1648 in the absence or presence of Cr (III) Mesoporphyrin IX Chloride (HO-1 activity inhibitor). NO-dependent relaxation was assessed with DEA-NOnoate (DEA-NO) on pre-contracted arteries. HO-1 and soluble guanylyl-cyclase (sGC) mRNA and protein expressions were assessed by qRT-PCR and Western blotting, respectively. SRM1648 or Co (III) Protoporphyrin IX Chloride exposure (24) impaired DEA-NO-dependent relaxation. The SRM-induced alteration of DEA-NO responsiveness was partially prevented by Cr (III) Mesoporphyrin IX Chloride. Co (III) Protoporphyrin IX Chloride induced HO-1 mRNA and protein expressions, whereas SRM1648 only induced HO-1 protein expression without affecting its mRNA level. Exposure to either SRM1648 or to Co (III) Protoporphyrin IX Chloride did not affect the expression levels of sGC. In conclusion, this study provides some evidence that impairment of NO signaling pathway in intrapulmonary arteries involves HO-1. Therefore it highlights the role of HO-1 in particulate matter-induced detrimental effects in pulmonary circulation.


Air Pollutants/toxicity , Heme Oxygenase (Decyclizing)/physiology , Nitric Oxide/physiology , Particulate Matter/toxicity , Pulmonary Artery/drug effects , Animals , Heme Oxygenase (Decyclizing)/metabolism , In Vitro Techniques , Male , Protoporphyrins/pharmacology , Pulmonary Artery/physiology , Rats, Wistar , Vasodilation
16.
J Appl Toxicol ; 34(6): 667-74, 2014 Jun.
Article En | MEDLINE | ID: mdl-23881823

We have previously shown that exposure to urban particulate matter (UPM) impairs endothelial nitric oxide (NO) bioactivity in intrapulmonary arteries. As UPM is composed of heterogeneous constituents, the aim of this study was to clarify the class of pollutants responsible for such effect. Extracts (aqueous, acidic or organic) were prepared from SRM1648, an UPM sample collected in St. Louis (MO, USA). The metal composition of extracts as well as endotoxin content was determined. The effects of each extract, metal mixture and endotoxin were evaluated on endothelium-dependent relaxation to acetylcholine (reflecting endothelial NO production) in rat isolated intrapulmonary arteries. Aqueous or organic SRM1648 pretreatment altered acetylcholine-induced relaxation, similar to that induced by native SRM1648. Organic extract induced similar attenuation of acetylcholine relaxation than organic-treated SRM1648, whereas aqueous extract had no effect. Acidic pretreatment, which impoverished metal and endotoxin content of SRM1648, prevented the impairment of acetylcholine-induced relaxation. However, neither the acidic extract enriched in metals, nor a metal mixture representative of SRM1648 content, modified acetylcholine relaxation, while endotoxin impaired it. Polymyxin B, which chelates endotoxin, prevented SRM1648-induced decrease in relaxation to acetylcholine. It is concluded that SRM1648-induced impairment of endothelial NO-dependent relaxation in intrapulmonary arteries unlikely involved a soluble factor released by vascular cells during UPM exposure, but rather an organic extractible and acidic-sensitive constituents of UPM. Endotoxin, but not metals, may be responsible for UPM-induced impairment of endothelial NO-dependent relaxation.


Endotoxins/toxicity , Metals/toxicity , Nitric Oxide/metabolism , Particulate Matter/toxicity , Pulmonary Artery/drug effects , Vasodilation/drug effects , Animals , Dose-Response Relationship, Drug , Endotoxins/analysis , Male , Metals/analysis , Particulate Matter/analysis , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Rats, Wistar , Risk Assessment , Tissue Culture Techniques , Vasodilator Agents/pharmacology
17.
Article En | MEDLINE | ID: mdl-18955370

Abnormal Savda Munziq (ASMq) is a traditional Uighur medicinal herbal preparation, commonly used for the treatment and prevention of cancer. We tested the effects of ethanol extract of ASMq on cultured human hepatoma cells (HepG2) to explore the mechanism of its putative anticancer properties, using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) bromide, neutral red and lactate dehydrogenase (LDH) leakage assays, testing the incorporation of (3)[H]-leucine and (3)[H]-nucleosides into protein, DNA and RNA, and quantifying the formation of malondialdehyde-thiobarbituric acid (MDA) adducts. ASMq ethanol extract significantly inhibited the growth of HepG2 and cell viability, increased the leakage of LDH after 48 hours or 72 hours treatment, in a concentration- and time-dependent manner (P < .05). Cellular protein, DNA and RNA synthesis were inhibited in a concentration- and time-dependent manner (P < .05). No significant MDA release in culture medium and no lipid peroxidation in cells were observed. The results suggest that the cytotoxic effects of ASMq ethanol extract might be related to inhibition of cancer cell growth, alteration of cell membrane integrity and inhibition of cellular protein, DNA and RNA synthesis.

18.
Toxicol Appl Pharmacol ; 245(2): 203-10, 2010 Jun 01.
Article En | MEDLINE | ID: mdl-20214918

Pulmonary circulation could be one of the primary vascular targets of finest particles that can deeply penetrate into the lungs after inhalation. We investigated the effects of engineered nanoparticles on vasomotor responses of small intrapulmonary arteries using isometric tension measurements. Acute in vitro exposure to carbon nanoparticles (CNP) decreased, and in some case abolished, the vasomotor responses induced by several vasoactive agents, whereas acute exposure to titanium dioxide nanoparticles (TiO(2)NP) did not. This could be attributed to a decrease in the activity of those vasoactive agents (including PGF(2)(alpha), serotonin, endothelin-1 and acetylcholine), as suggested when they were exposed to CNP before being applied to arteries. Also, CNP decreased the contraction induced by 30 mM KCl, without decreasing its activity. After endoplasmic reticulum calcium stores depletion (by caffeine and thapsigargin), CaCl(2) addition induced a contraction, dependent on Store-Operated Calcium Channels that was not modified by acute CNP exposure. Further addition of 30 mM KCl elicited a contraction, originating from activation of Voltage-Operated Calcium Channels that was diminished by CNP. Contractile responses to PGF(2)(alpha) or KCl, and relaxation to acetylcholine were modified neither in pulmonary arteries exposed in vitro for prolonged time to CNP or TiO(2)NP, nor in those removed from rats intratracheally instilled with CNP or TiO(2)NP. In conclusion, prolonged in vitro or in vivo exposure to CNP or TiO(2)NP does not affect vasomotor responses of pulmonary arteries. However, acute exposure to CNP decreases contraction mediated by activation of Voltage-Operated, but not Store-Operated, Calcium Channels. Moreover, interaction of some vasoactive agents with CNP decreases their biological activity that might lead to misinterpretation of experimental data.


Carbon/pharmacology , Isometric Contraction/drug effects , Nanoparticles , Pulmonary Artery/drug effects , Titanium/pharmacology , Animals , Calcium Channels/physiology , Dose-Response Relationship, Drug , Endoplasmic Reticulum/metabolism , Inhalation Exposure/adverse effects , Male , Pulmonary Artery/physiology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology
19.
Environ Health Perspect ; 116(10): 1294-9, 2008 Oct.
Article En | MEDLINE | ID: mdl-18941568

BACKGROUND AND OBJECTIVES: Because pulmonary circulation is the primary vascular target of inhaled particulate matter (PM), and nitric oxide is a major vasculoprotective agent, in this study we investigated the effect of various particles on the NO-cyclic guanosine monophosphate (cGMP) pathway in pulmonary arteries. METHODS: We used intrapulmonary arteries and/or endothelial cells, either exposed in vitro to particles or removed from PM-instilled animals for assessment of vasomotricity, cGMP and reactive oxygen species (ROS) levels, and cytokine/chemokine release. RESULTS: Endothelial NO-dependent relaxation and cGMP accumulation induced by acetylcholine (ACh) were both decreased after 24 hr exposure of rat intrapulmonary arteries to standard reference material 1648 (SRM1648; urban PM). Relaxation due to NO donors was also decreased by SRM1648, whereas responsiveness to cGMP analogue remained unaffected. Unlike SRM1648, ultrafine carbon black and ultrafine and fine titanium dioxide (TiO2) manufactured particles did not impair NO-mediated relaxation. SRM1648-induced decrease in relaxation response to ACh was prevented by dexamethasone (an anti-inflammatory agent) but not by antioxidants. Accordingly, SRM1648 increased the release of proinflammatory mediators (tumor necrosis factor-alpha, interleukin-8) from intrapulmonary arteries or pulmonary artery endothelial cells, but did not elevate ROS levels within intrapulmonary arteries. Decreased relaxation in response to ACh was also evidenced in intrapulmonary arteries removed from rats intratracheally instilled with SRM1648, but not with fine TiO2. CONCLUSION: In contrast to manufactured particles (including nanoparticles), urban PM impairs NO but not cGMP responsiveness in intrapulmonary arteries. We attribute this effect to oxidative-stress-independent inflammatory response, resulting in decreased guanylyl cyclase activation by NO. Such impairment of the NO pathway may contribute to urban-PM-induced cardiovascular dysfunction.


Air Pollutants/toxicity , Nanoparticles , Nitric Oxide/physiology , Pulmonary Artery/drug effects , Animals , Cattle , In Vitro Techniques , Male , Muscle Relaxation/drug effects , Pulmonary Artery/physiology , Rats , Rats, Wistar
20.
Zhongguo Zhong Yao Za Zhi ; 32(11): 1068-71, 2007 Jun.
Article Zh | MEDLINE | ID: mdl-17672346

OBJECTIVE: To investigate the effect of Abnormal Savda Munziq (ASMq) flavonoids on proliferation, apoptosis and apoptosis-related gene expression in human hepatoma (HepG2) cells in vitro and to probe the mechanism. METHOD: The effects of ASMq flavonoids on proliferation, apoptosis and apoptosis-related gene expression of HepG2 cells were investigated respectively by MTT assay, gel electrophoresis, flow cytometry and RT-PCR. RESULT: ASMq flavonoids significantly inhibited growth of HepG2 cells in vitro, arrested HepG2 in the sub-G, phase, induced cell apoptosis and significantly down-regulated expression level of Bcl-2 mRNA, and up-regulated expression of p53, p21, Bax gene mRNA expressions. CONCLUSION: ASMq flavonoids has significantly regulative action on growth, apoptosis and apoptosis-related gene expression of cancer cells in vitro, which possibly are the important way to excert anticancer effect, and flavonoids are possibly a main active component of ASMq for exerting the anticancer effect.


Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Flavonoids/pharmacology , Plants, Medicinal/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Combinations , Flavonoids/isolation & purification , Flow Cytometry , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins p21(ras)/biosynthesis , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/genetics
...