Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cells ; 9(2)2020 02 12.
Article in English | MEDLINE | ID: mdl-32059483

ABSTRACT

Many recent studies have demonstrated the involvement of endoplasmic reticulum (ER) stress in the development of cardiac diseases and have suggested that modulation of ER stress response could be cardioprotective. Previously, we demonstrated that the deacetylase Sirtuin 1 (SIRT1) attenuates ER stress response and promotes cardiomyocyte survival. Here, we investigated whether and how autophagy plays a role in SIRT1-afforded cardioprotection against ER stress. The results revealed that protective autophagy was initiated before cell death in response to tunicamycin (TN)-induced ER stress in cardiac cells. SIRT1 inhibition decreased ER stress-induced autophagy, whereas its activation enhanced autophagy. In response to TN- or isoproterenol-induced ER stress, mice deficient for SIRT1 exhibited suppressed autophagy along with exacerbated cardiac dysfunction. At the molecular level, we found that in response to ER stress (i) the extinction of eEF2 or its kinase eEF2K not only reduced autophagy but further activated cell death, (ii) inhibition of SIRT1 inhibited the phosphorylation of eEF2, (iii) eIF2α co-immunoprecipitated with eEF2K, and (iv) knockdown of eIF2α reduced the phosphorylation of eEF2. Our results indicate that in response to ER stress, SIRT1 activation promotes cardiomyocyte survival by enhancing autophagy at least through activation of the eEF2K/eEF2 pathway.


Subject(s)
Autophagy , Elongation Factor 2 Kinase/metabolism , Endoplasmic Reticulum Stress , Sirtuin 1/metabolism , Animals , Autophagy/drug effects , Elongation Factor 2 Kinase/antagonists & inhibitors , Elongation Factor 2 Kinase/genetics , Endoplasmic Reticulum Stress/drug effects , Heat-Shock Proteins/metabolism , Isoproterenol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Sequestosome-1 Protein/metabolism , Signal Transduction/drug effects , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Tunicamycin/pharmacology
2.
Ann Biol Clin (Paris) ; 78(1): 27-34, 2020 02 01.
Article in French | MEDLINE | ID: mdl-32108577

ABSTRACT

Unfractionated heparin (UFH) is the main anticoagulante used in intensive care unit. The anticoagulant effect is monitored by activated partial thrombin time (aPTT) and anti-Xa activity (anti-Xa) measurement. However, delayed centrifugation induces platelet factor 4 (PF4) release and anti-Xa decrease. Several studies have concluded that aPTT and anti-Xa measurement should be performed within 2 hours in citrated anticoagulant but may be delayed longer in citrate theophylline adenosine and dypiridamol (CTAD) anticoagulant. The objective of this study was to compare the stability of both aPTT and anti-Xa in citrate and CTAD samples, and to determine the effect of delayed centrifugation on both aPTT, anti-Xa results, and PF4 release in citrate samples only. METHODS: aPTT and anti-Xa were measured in citrate and CTAD anticoagulant samples from 93 patients. Delayed centrifugation was performed in citrate samples from 31 additional patients, with hourly aPTT and anti-Xa measurement from 1 to 6 hours. In 14 of these last patients, PF4 release was also evaluated with Human CXCL4/PF4 Quantikine ELISA Kit. RESULTS: We observed a significant correlation between citrate and CTAD anticoagulant for aPTT (r2=0.94) and anti-Xa (r2=0.95). With Bland-Altman correlation, a minor bias was observed for anti-Xa (-0.025±0.041). Delayed centrifugation in citrated anticoagulant showed an excellent concordance from 1 to 4 hours for aPTT (-4.0±5.3 s) and anti-Xa (1.10-9±0.058 UI/mL) measurements. Moreover, PF4 release was not different between 1 hour (31.5±14.7 ng/mL) and 4 hours (33.8±11.8 ng/mL). CONCLUSION: We have demonstrated that anti-Xa measurement for unfractionated heparin should be done 4 hours in citrated plasma and that CTAD was not better than citrate. However, these initial findings require confirmation using other aPTT and calibrated anti-Xa assays.


Subject(s)
Anticoagulants/pharmacology , Blood Specimen Collection/methods , Drug Monitoring/methods , Heparin/therapeutic use , Adenosine/chemistry , Adenosine/pharmacology , Anticoagulants/chemistry , Anticoagulants/therapeutic use , Blood Coagulation Tests/methods , Blood Preservation/methods , Centrifugation/methods , Chemical Fractionation/methods , Citric Acid/chemistry , Citric Acid/pharmacology , Dipyridamole/chemistry , Dipyridamole/pharmacology , Factor Xa/metabolism , Factor Xa Inhibitors/analysis , Factor Xa Inhibitors/blood , Heparin/analysis , Humans , Partial Thromboplastin Time , Theophylline/chemistry , Theophylline/pharmacology , Thrombin Time , Time Factors
3.
J Thromb Thrombolysis ; 48(2): 277-283, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31098816

ABSTRACT

Unfractionated heparin (UFH) is the main anticoagulant used in intensive care unit. The anticoagulant effect is monitored by activated partial thrombin time (aPTT) and anti-Xa activity (anti-Xa) measurement. However, delayed centrifugation induces platelet factor 4 (PF4) release and anti-Xa decrease. Several studies have concluded that aPTT and anti-Xa measurement should be performed within 2 h in citrated anticoagulant but may be delayed longer in Citrate Theophylline Adenosine and Dypiridamol (CTAD) anticoagulant. The objective of this study was to compare the stability of both aPTT and anti-Xa in citrate and CTAD samples, and to determine the effect of delayed centrifugation on both aPTT, anti-Xa results, and PF4 release in citrate samples only. aPTT and anti-Xa were measured in citrate and CTAD anticoagulant samples from 93 patients. Delayed centrifugation was performed in citrate samples from 31 additional patients, with hourly aPTT and anti-Xa measurement from 1 to 6 h. In 14 of these last patients, PF4 release was also evaluated with Human CXCL4/PF4 Quantikine ELISA Kit. We observed a significant correlation between citrate and CTAD anticoagulant for aPTT (r2 = 0.94) and anti-Xa (r2 = 0.95). With Bland-Altman correlation, a minor bias was observed for anti-Xa (- 0.025 ± 0.041). Delayed centrifugation in citrated anticoagulant showed an excellent concordance from 1 to 4 h for aPTT (- 4.0 ± 5.3 s) and anti-Xa (1.10-9 ± 0.058 UI/ml) measurements. Moreover, PF4 release was not different between 1 h (31.5 ± 14.7 ng/ml) and 4 h (33.8 ± 11.8 ng/ml). We have demonstrated that anti-Xa measurement for unfractionated heparin should be done 4 h in citrated plasma and that CTAD was not better than citrate. However, these initial findings require confirmation using other aPTT and calibrated anti-Xa assays.


Subject(s)
Blood Preservation/methods , Citrates/pharmacology , Dipyridamole/pharmacology , Drug Monitoring/methods , Heparin/pharmacokinetics , Anticoagulants/pharmacology , Blood Coagulation Tests , Centrifugation , Heparin/therapeutic use , Humans , Phosphodiesterase Inhibitors/pharmacology , Theophylline , Time Factors
4.
Cardiovasc Res ; 115(2): 328-342, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30084984

ABSTRACT

Aims: Endoplasmic reticulum (ER) stress has recently emerged as an important mechanism involved in the pathogenesis of cardiovascular diseases. However, the molecular mechanisms by which ER stress leads to cardiac dysfunction remain poorly understood. Methods and results: In this study, we evaluated the early cardiac effects of ER stress induced by tunicamycin (TN) in mice. Echocardiographic analysis indicated that TN-induced ER stress led to a significant impairment of the cardiac function. Electron microscopic observations revealed that ultrastructural changes of cardiomyocytes in response to ER stress manifested extensively at the level of the reticular membrane system. Smooth tubules of sarcoplasmic reticulum in connection with short sections of rough ER were observed. The presence of rough instead of smooth reticulum was increased at the interfibrillar space, at the level of dyads, and in the vicinity of mitochondria. At the transcriptional level, ER stress resulted in a substantial decrease in the expression of the major regulator of mitochondrial biogenesis PGC-1α and of its targets NRF1, Tfam, CS, and COXIV. At the functional level, ER stress also induced an impairment of mitochondrial Ca2+ uptake, an alteration of mitochondrial oxidative phosphorylation, and a metabolic remodelling characterized by a shift from fatty acid to glycolytic substrate consumption. Conclusions: Our findings show that ER stress induces cytoarchitectural and metabolic alterations in cardiomyocytes and provide evidences that ER stress could represent a primary mechanism that contributes to the impairment of energy metabolism reported in most cardiac diseases.


Subject(s)
Endoplasmic Reticulum Stress , Heart Diseases/metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , ATP Citrate (pro-S)-Lyase/genetics , ATP Citrate (pro-S)-Lyase/metabolism , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Down-Regulation , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Fatty Acids/metabolism , Glycolysis , Heart Diseases/chemically induced , Heart Diseases/pathology , Heart Diseases/physiopathology , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Mice , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/ultrastructure , NF-E2-Related Factor 1/genetics , NF-E2-Related Factor 1/metabolism , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Signal Transduction , Tunicamycin
5.
Cell Death Differ ; 24(2): 343-356, 2017 02.
Article in English | MEDLINE | ID: mdl-27911441

ABSTRACT

Over the past decade, endoplasmic reticulum (ER) stress has emerged as an important mechanism involved in the pathogenesis of cardiovascular diseases including heart failure. Cardiac therapy based on ER stress modulation is viewed as a promising avenue toward effective therapies for the diseased heart. Here, we tested whether sirtuin-1 (SIRT1), a NAD+-dependent deacetylase, participates in modulating ER stress response in the heart. Using cardiomyocytes and adult-inducible SIRT1 knockout mice, we demonstrate that SIRT1 inhibition or deficiency increases ER stress-induced cardiac injury, whereas activation of SIRT1 by the SIRT1-activating compound STAC-3 is protective. Analysis of the expression of markers of the three main branches of the unfolded protein response (i.e., PERK/eIF2α, ATF6 and IRE1) showed that SIRT1 protects cardiomyocytes from ER stress-induced apoptosis by attenuating PERK/eIF2α pathway activation. We also present evidence that SIRT1 physically interacts with and deacetylates eIF2α. Mass spectrometry analysis identified lysines K141 and K143 as the acetylation sites on eIF2α targeted by SIRT1. Furthermore, mutation of K143 to arginine to mimic eIF2α deacetylation confers protection against ER stress-induced apoptosis. Collectively, our findings indicate that eIF2α deacetylation on lysine K143 by SIRT1 is a novel regulatory mechanism for protecting cardiac cells from ER stress and suggest that activation of SIRT1 has potential as a therapeutic approach to protect the heart against ER stress-induced injury.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/metabolism , Sirtuin 1/metabolism , Acetylation , Activating Transcription Factor 6/metabolism , Animals , Apoptosis/drug effects , Carbazoles/pharmacology , Cell Line , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/genetics , Heat-Shock Proteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Tunicamycin/pharmacology , Unfolded Protein Response/drug effects , Up-Regulation/drug effects
6.
Toxicol Appl Pharmacol ; 314: 82-90, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27889531

ABSTRACT

Zearalenone (ZEN) is a non-steroidal estrogenic mycotoxin produced by several species of Fusarium in cereals and agricultural products. The major ZEN metabolites are α-zearalenol (α-ZOL) and ß-zearalenol (ß-ZOL). In the present study, we investigated the underlying mechanism of the toxicity induced by ZEN, α-ZOL and ß-ZOL in cardiac cells (H9c2). We show that treatment with ZEN or its metabolites induces the activation of the mitochondrial pathway of apoptosis as characterized by an increase in ROS generation, a loss of mitochondrial transmembrane potential (ΔΨm) and an activation of caspases. Besides, we demonstrate that these mycotoxins promote the activation of autophagy before the onset of apoptosis. Indeed, we observed that a short-time (6h) treatment with ZEN, α-ZOL or ß-ZOL, increased the level of Beclin-1 and LC3-II and induced the accumulation of the CytoID® autophagy detection probe. Moreover, the inhibition of autophagy by Chloroquine significantly increased cell death induced by ZEN, α-ZOL or ß-ZOL, suggesting that the activation of autophagy serves as a cardioprotective mechanism against these mycotoxins. In addition, we found that the inhibition (EX527) or the knockdown of SIRT1 (siRNA) significantly increased apoptosis induced by ZEN or its derivatives, whereas SIRT1 activation with RSV greatly prevents the cytotoxic effects of these mycotoxins. By contrast, when autophagy was inhibited by CQ, the activation of SIRT1 by RSV had no protection against the cardiotoxicity of ZEN or its metabolites, suggesting that SIRT1 protects cardiac cells by an autophagy-dependent pathway.


Subject(s)
Apoptosis/physiology , Autophagy/drug effects , Sirtuin 1/physiology , Zearalenone/toxicity , Zeranol/analogs & derivatives , Animals , Apoptosis/drug effects , Cell Death , Cell Line , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Reactive Oxygen Species/metabolism , Zeranol/toxicity
7.
Environ Sci Pollut Res Int ; 23(15): 15504-14, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27121014

ABSTRACT

α-zearalenol (α-ZOL) and ß-zearalenol (ß-ZOL) are the major metabolites of Zearalenone (ZEN) and are known to induce many toxic effects. In the present study, we investigated the involvement of endoplasmic reticulum (ER) stress in α- and ß-ZOL-mediated toxicity in human kidney cells (HEK293) and evaluated the effect of a common dietary compound Crocin (CRO), from saffron. We show that α- and ß-ZOL treatment induces ER stress as evidenced by the upregulation of the 78 kDa glucose-regulated protein (GRP78) and the Growth arrest and DNA damage-inducible protein (GADD34). Activation of the ER stress response is associated with activation of the mitochondrial pathway of apoptosis. This apoptotic process is characterized by an increase in ROS generation and lipid peroxidation, a loss of mitochondrial transmembrane potential (ΔΨm) and activation of caspases. We also demonstrate that the antioxidant properties of CRO help to prevent ER stress and reduce α- and ß-ZOL-induced apoptosis in HEK293 cells. Our results suggest that saffron consumption might be helpful to prevent α- and ß-ZOL-induced ER stress and toxicity.


Subject(s)
Apoptosis/drug effects , Carotenoids/pharmacology , Endoplasmic Reticulum Stress/drug effects , Protective Agents/pharmacology , Zeranol/analogs & derivatives , Antioxidants/metabolism , Caspases/metabolism , Cell Line , DNA Damage/drug effects , Endoplasmic Reticulum Chaperone BiP , HEK293 Cells , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Zeranol/chemistry , Zeranol/toxicity
8.
Neurotoxicology ; 53: 334-342, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26584763

ABSTRACT

Zearalenone (ZEN) and its metabolites are found in many food products and are known to induce many toxic effects. The major ZEN metabolites are α-zearalenol (α-ZOL) and ß-zearalenol (ß-ZOL). The mechanisms by which they mediate their cytotoxic effects are not well known and seem to differ depending on the type of cells. We investigated the possible underlying mechanism in α-ZOL and ß-ZOL-induced toxicity in HCT116 cells. We showed that cell treatment with α-ZOL/ß-ZOL generated endoplasmic reticulum (ER) stress and activated the Unfolded Protein Response (UPR) as evidenced by XBP1 mRNA splicing and up-regulation of GADD34, GRP78, ATF4 and CHOP. Apoptosis was triggered by ZEN metabolites-induced ER stress, and executed through a mitochondria-dependent pathway, characterized by a loss of mitochondrial transmembrane potential (ΔΨm), a downstream generation of O2•(-) and caspase 3 activation. Cellular deficiency of the pro-apoptotic proteins Bax and Bak protected cells against α/ß-ZOL-induced toxicity. However, treatment with α-ZOL or ß-ZOL combined with Quercetin (QUER), a common dietary flavonoid with well-known antioxidant activity, significantly reduced damage induced by α and ß-ZOL in all tested markers. We concluded that QUER protects against the cellular toxicity of α and ß-ZOL.×.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Quercetin/pharmacology , Zearalenone/pharmacology , Zeranol/analogs & derivatives , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Analysis of Variance , Cell Line, Tumor , Dose-Response Relationship, Drug , Endoplasmic Reticulum Chaperone BiP , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Protein Disulfide-Isomerases/metabolism , Protein Phosphatase 1/metabolism , Reactive Oxygen Species/metabolism , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism , Zeranol/pharmacology
9.
Environ Toxicol ; 31(12): 1851-1858, 2016 Dec.
Article in English | MEDLINE | ID: mdl-26314699

ABSTRACT

Patulin (PAT) is a secondary metabolite produced by several species of the genera of Penicillium, Aspergillus, and Byssochlamys that can be found in rotting fruits, especially in apples and apple-based products. Exposure to this mycotoxin has been reported to induce intestinal and kidney injuries. The mechanism underlying such toxicity has been linked to the induction of apoptosis which occurred with reactive oxygen species production and endoplasmic reticulum (ER) stress induction. This study aimed to evaluate the effect of the two common dietary compounds Quercetin (QUER), a natural flavonoid, and Crocin (CRO), a natural carotenoid, on PAT-induced toxicity in human colon carcinoma (HCT116) and embryonic kidney cells (HEK293). We showed that antioxidant properties of QUER and CRO help to prevent ER stress activation and lipid peroxidation as evidenced by the reduction in GRP78 and GADD34 expressions and the decrease in malondialdehyde production. Furthermore, we demonstrated their ability to re-establish the loss of the mitochondrial membrane potential to inhibit caspase 3 activation and DNA fragmentation. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1851-1858, 2016.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Carotenoids/pharmacology , Endoplasmic Reticulum Stress/drug effects , Patulin/toxicity , Quercetin/pharmacology , Reactive Oxygen Species/metabolism , Caspase 3/metabolism , DNA Fragmentation/drug effects , Endoplasmic Reticulum Chaperone BiP , Enzyme Activation , HCT116 Cells , HEK293 Cells , Heat-Shock Proteins/metabolism , Humans , Lipid Peroxidation/drug effects , Malondialdehyde/metabolism , Membrane Potential, Mitochondrial/drug effects , Protein Phosphatase 1/metabolism
10.
Cell Calcium ; 58(5): 489-99, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26297240

ABSTRACT

BACKGROUND: Previous studies suggest that CaMKII activity is required for frequency-dependent acceleration of relaxation (FDAR) in ventricular myocytes. We propose that the underlying mechanism involves CaMKII-dependent regulation of myofilament Ca(2+) sensitivity. METHODS AND RESULTS: Cardiac function was measured in mice using murine echo machine. [Ca(2+)]i and sarcomere length were measured by IonOptix Ca(2+) image system. Increasing pacing rate from 0.5 to 4 Hz in left ventricular myocytes induced frequency-dependent myofilament Ca(2+) desensitization (FDMCD) and FDAR. Acute inhibition of PKA or PKC had no effect, whereas CaMKII inhibition abolished both FDMCD and FDAR. Co-immunoprecipitation of CaMKII and troponin I (TnI) has been detected and CaMKII inhibition significantly reduced serine residue phosphorylation of TnI. Finally, chronic inhibition of CaMKII in vivo reduced TnI phosphorylation and abolished both FDAR and FDMCD, leading to impaired diastolic function. CONCLUSIONS: Our results suggest that CaMKII-dependent TnI phosphorylation is involved in FDMCD and the consequent FDAR and that CaMKII inhibition removes this mechanism and thus induces diastolic dysfunction.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Myofibrils/metabolism , Acceleration , Animals , Male , Mice , Mice, Inbred C57BL
11.
Cell Stress Chaperones ; 20(6): 927-38, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26134454

ABSTRACT

Mycotoxins are considered to be significant contaminants of food and animal feed. Zearalenone (ZEN) is a non-steroidal estrogenic mycotoxin produced by several species of Fusarium in cereals and agricultural products. ZEN has been shown to be cytotoxic, genotoxic, and mutagenic in different cell types. In the present study, we investigated the involvement of endoplasmic reticulum (ER) stress in ZEN-mediated toxicity in human intestine (HCT116) and kidney (HEK293) cells and evaluated the effects of the two common dietary compounds Quercetin (QUER) and Crocin (CRO). We show that ZEN treatment induces ER stress and activates the unfolded protein response (UPR) as evidenced by XBP1 mRNA splicing and upregulation of GRP78, ATF4, GADD34, PDIA6, and CHOP. Activation of the ER stress response is associated with activation of the mitochondrial pathway of apoptosis. This apoptotic process is characterized by an increase in ROS generation and lipid peroxidation, a loss of mitochondrial transmembrane potential (ΔΨm), and an activation of caspases and DNA damages. We also demonstrate that the antioxidant properties of QUER and CRO help to prevent ER stress and reduce ZEN-induced apoptosis in HCT116 and HEK293 cells. Our results suggest that antioxidant molecule might be helpful to prevent ZEN-induced ER stress and toxicity.


Subject(s)
Apoptosis/drug effects , Carotenoids/pharmacology , Endoplasmic Reticulum Stress/drug effects , Quercetin/pharmacology , Zearalenone/pharmacology , Endoplasmic Reticulum Chaperone BiP , HCT116 Cells , HEK293 Cells , Humans
12.
Toxicol Sci ; 144(2): 328-37, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25577197

ABSTRACT

Patulin (PAT) is a toxic metabolite produced by several filamentous fungi of the genera of Penicillium, Aspergillus, and Byssochlamys. PAT is the most common mycotoxin found in apples and apple-based products including juice, compotes, cider, and baby food. Exposure to this mycotoxin has been reported to induce intestinal and kidney injuries. This study investigated the mechanism of PAT-induced toxicity in human colon carcinoma (HCT116) and embryonic kidney cells (HEK293). We demonstrated that PAT activated endoplasmic reticulum (ER) and unfolded protein response as evidenced by up-regulation of GRP78 and GADD34, splicing of XBP1 mRNA, and expression of the proapoptotic factor CHOP. This ER stress response was accompanied by the induction of the mitochondrial apoptotic pathway. Apoptosis occurred with ROS production, drop in mitochondrial membrane potential and caspase activation. Further, we showed that deficiency of the proapoptotic protein Bax or Bak protected cells against PAT-induced apoptosis. The treatment of cells with the ROS scavenger N-acetyl cysteine inhibits the ER stress response and prevents mitochondrial apoptosis. Collectively, our data provide new mechanistic insights in the signaling pathways of the cell death induced by PAT and demonstrate that PAT induces cytotoxicity through a ROS-dependent mechanism involving ER stress and activation of mitochondrial apoptotic pathway in human intestinal and kidney cells.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum/drug effects , Oxidative Stress/drug effects , Patulin/pharmacology , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Humans
13.
J Cardiovasc Electrophysiol ; 24(4): 457-63, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23279377

ABSTRACT

BACKGROUND: CaMKII activation is proarrhythmic in heart failure where myocardium is stretched. However, the arrhythmogenic role of CaMKII in stretched ventricle has not been well understood. OBJECTIVE: We tested abnormal impulse inducibility by stretch current in myocytes isolated from CaMKIIδ knockout (KO) mouse left ventricle (LV) where CaMKII activity is reduced by ≈ 62%. METHODS AND RESULTS: Action potentials were recorded by whole-cell patch clamp, and abnormal impulses were induced in LV myocytes by a simulation of stretch-activated channel (SAC) current. SAC activation failed to induce abnormal impulses in wild type (WT) myocytes but steadily produced early after-depolarizations and automaticity in KO myocytes in which an increase in L-type calcium channel (LTCC) current (I(Ca)) and a reduction of sarcoplasmic reticulum Ca(2+) leak and action potential duration (APD) were observed. The abnormal impulses were not suppressed by CaMKII inhibitor AIP whereas a low concentration of nifedipine eliminated abnormal impulses without shortening APD, implicating I(Ca) in promoting stretch-induced abnormal impulses. In addition, APD prolongation by LTCC opener S(-)Bay K 8644 or isoproterenol facilitated abnormal impulse induction in WT ventricular myocytes even in the presence of CaMKII inhibitor AIP, whereas APD prolongation by K(+) channel blocker 4-aminopyridine promoted abnormal impulses in KO myocytes but not in WT myocytes. CONCLUSION: I(Ca) activation plays a central role in stretch-induced abnormal impulses and APD prolongation is arrhythmogenic only when I(Ca) is highly activated. At increased I(Ca) activation, CaMKII inhibition cannot suppress abnormal impulse induction.


Subject(s)
Arrhythmias, Cardiac/enzymology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/deficiency , Heart Ventricles/enzymology , Mechanoreceptors/metabolism , Myocytes, Cardiac/enzymology , Action Potentials , Adrenergic beta-Agonists/pharmacology , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Calcium/metabolism , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Enzyme Activation , Evoked Potentials , Heart Ventricles/drug effects , Mice , Mice, Knockout , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Protein Kinase Inhibitors/pharmacology , Sarcoplasmic Reticulum/metabolism , Time Factors
14.
Am J Physiol Heart Circ Physiol ; 302(7): H1454-65, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22287581

ABSTRACT

Calmodulin-dependent protein kinase II (CaMKII) has been proposed to be a therapeutic target for heart failure (HF). However, the cardiac effect of chronic CaMKII inhibition in HF has not been well understood. We have tested alterations of Ca(2+) handling, excitation-contraction coupling, and in vivo ß-adrenergic regulation in pressure-overload HF mice with CaMKIIδ knockout (KO). HF was produced in wild-type (WT) and KO mice 1 wk after severe thoracic aortic banding (sTAB) with a continuous left ventricle (LV) dilation and reduction of ejection fraction for up to 3 wk postbanding. Cardiac hypertrophy was similar between WT HF and KO HF mice. However, KO HF mice manifested exacerbation of diastolic function and reduction in cardiac reserve to ß-adrenergic stimulation. Compared with WT HF, L-type calcium channel current (I(Ca)) density in KO HF LV was decreased without changes in I(Ca) activation and inactivation kinetics, whereas I(Ca) recovery from inactivation was accelerated and Ca(2+)-dependent I(Ca) facilitation, a positive staircase blunted in WT HF, was recovered. However, I(Ca) response to isoproterenol was reduced. KO HF myocytes manifested dramatic decrease in sarcoplasmic reticulum (SR) Ca(2+) leak and slowed cytostolic Ca(2+) concentration decline. Sarcomere shortening was increased, but relaxation was slowed. In addition, an increase in myofilament sensitivity to Ca(2+) and the slow skeletal muscle troponin I-to-cardiac troponin I ratio and interstitial fibrosis and a decrease in Na/Ca exchange function and myocyte apoptosis were observed in KO HF LV. CaMKIIδ KO cannot suppress severe pressure-overload-induced HF. Although cellular contractility is improved, it reduces in vivo cardiac reserve to ß-adrenergic regulation and deteriorates diastolic function. Our findings challenge the strategy of CaMKII inhibition in HF.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Heart Failure/drug therapy , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Adrenergic beta-Agonists/pharmacology , Animals , Aorta, Thoracic/physiology , Apoptosis/drug effects , Blotting, Western , Calcium Channels/drug effects , Calcium Channels/metabolism , Calcium Signaling/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cytosol/drug effects , Cytosol/metabolism , Fibrosis/pathology , Heart Failure/diagnostic imaging , Heart Ventricles/drug effects , Isoenzymes/metabolism , Male , Mice , Mice, Knockout , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Myofibrils/physiology , Protein Kinase Inhibitors/toxicity , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta/physiology , Sarcomeres/drug effects , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/physiology , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/metabolism , Ultrasonography
15.
Cell Physiol Biochem ; 28(5): 813-22, 2011.
Article in English | MEDLINE | ID: mdl-22178934

ABSTRACT

BACKGROUND: Transient Receptor Potential (TRP) channels are expressed in many solid tumors. However, their expression in breast cancer remains largely unknown. Here, we investigated the profile expression of 13 TRP channels in human breast ductal adenocarcinoma (hBDA) and performed a correlation between their overexpression and pathological parameters. METHODS: The TRP channels expression was determined by RT-PCR in hBDA tissue, in human breast cancer epithelial (hBCE) primary culture and in MCF-7 cell line. The TRP protein level was evaluated by immunohistochemistry in hBDA tissue samples of 59 patients. RESULTS: TRPC1, TRPC6, TRPM7, TRPM8, and TRPV6 channels were overexpressed in hBDA compared to the adjacent non-tumoral tissue. Most interestingly, TRPC1, TRPM7 and TRPM8 expression strongly correlated with proliferative parameters (SBR grade, Ki67 proliferation index, and tumor size), and TRPV6 was mainly overexpressed in the invasive breast cancer cells. Using laser capture microdissection, we found that TRPV6 expression was higher in invasive areas, compared to the corresponding non-invasive ones. Moreover, TRPV6 silencing inhibited MDA-MB-231 migration and invasion, and MCF-7 migration. CONCLUSION: TRP channels are aberrantly expressed in hBDA, hBCE primary cultures, and cell lines, and associated with pathological parameters. The high expression of TRP channels in tumors suggests the potential of these channels for diagnostic, prognosis and/or therapeutic approaches in human breast ductal adenocarcinoma.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Transient Receptor Potential Channels/metabolism , Breast Neoplasms/metabolism , Cells, Cultured , Female , Humans , Immunohistochemistry , Neoplasm Staging , Protein Serine-Threonine Kinases , RNA Interference , RNA, Small Interfering/metabolism , TRPC Cation Channels/metabolism , TRPC6 Cation Channel , TRPM Cation Channels/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/genetics
16.
BMC Cancer ; 10: 212, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20482834

ABSTRACT

BACKGROUND: The calcium-permeable cation channel TRPM8 (melastatin-related transient receptor potential member 8) is over-expressed in several cancers. The present study aimed at investigating the expression, function and potential regulation of TRPM8 channels by ER alpha (estrogen receptor alpha) in breast cancer. METHODS: RT-PCR, Western blot, immuno-histochemical, and siRNA techniques were used to investigate TRPM8 expression, its regulation by estrogen receptors, and its expression in breast tissue. To investigate the channel activity in MCF-7 cells, we used the whole cell patch clamp and the calcium imaging techniques. RESULTS: TRPM8 channels are expressed at both mRNA and protein levels in the breast cancer cell line MCF-7. Bath application of the potent TRPM8 agonist Icilin (20 microM) induced a strong outwardly rectifying current at depolarizing potentials, which is associated with an elevation of cytosolic calcium concentration, consistent with established TRPM8 channel properties. RT-PCR experiments revealed a decrease in TRPM8 mRNA expression following steroid deprivation for 48 and 72 hours. In steroid deprived medium, addition of 17-beta-estradiol (E2, 10 nM) increased both TRPM8 mRNA expression and the number of cells which respond to Icilin, but failed to affect the Ca2+ entry amplitude. Moreover, silencing ERalpha mRNA expression with small interfering RNA reduced the expression of TRPM8. Immuno-histochemical examination of the expression of TRPM8 channels in human breast tissues revealed an over-expression of TRPM8 in breast adenocarcinomas, which is correlated with estrogen receptor positive (ER+) status of the tumours. CONCLUSION: Taken together, these results show that TRPM8 channels are expressed and functional in breast cancer and that their expression is regulated by ER alpha.


Subject(s)
Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , TRPM Cation Channels/metabolism , Adenocarcinoma/genetics , Blotting, Western , Breast Neoplasms/genetics , Calcium/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Membrane Potentials , Patch-Clamp Techniques , Pyrimidinones/pharmacology , RNA Interference , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , TRPM Cation Channels/agonists , TRPM Cation Channels/genetics , Time Factors
17.
Am J Physiol Cell Physiol ; 297(3): C493-502, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19515901

ABSTRACT

Because transient receptor potential (TRP) channels have been implicated in tumor progression, we have investigated the potential role of TRPM7 channel in breast cancer cell proliferation. Under whole cell patch clamp, a Mg(2+)-inhibited cationic (MIC) current was observed in MCF-7 cells. This current was characterized by an inward current and a strong outward rectifying current that were both inhibited in a concentration-dependent manner by the presence of intracellular Mg(2+) or Mg(2+)-ATP. The inward current was reduced by La(3+), and the outward current was sensitive to 2-aminoethoxydiphenyl borate (2-APB), spermine, La(3+), and flufenamic acid. Importantly, a similar MIC current was also recorded in the primary culture of human breast cancerous epithelial cells (hBCE). Moreover, TRPM7 transcripts were found in both hBCE and MCF-7 cells. In MCF-7 cells, the MIC current was inhibited by TRPM7 small interfering RNA. Interestingly, we found that cell proliferation and intracellular Ca(2+) concentration were also reduced by TRPM7 silencing in MCF-7 cells. TRPM7 channels were also found in both human breast cancer and healthy tissues. Importantly, TRPM7 channel was overexpressed in grade III breast cancer samples associated with important Ki67 or tumor size. Our findings strongly suggest that TRPM7 is involved in the proliferative potentiality of breast cancer cells, probably by regulating Ca(2+) influx.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic/physiology , TRPM Cation Channels/metabolism , Calcium/metabolism , Cell Line, Tumor , Cell Proliferation , Female , Humans , Magnesium/metabolism , Manganese , Membrane Potentials/physiology , Protein Serine-Threonine Kinases , TRPM Cation Channels/genetics , Tissue Culture Techniques
18.
BMC Cancer ; 8: 125, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18452628

ABSTRACT

BACKGROUND: TRP channels have been shown to be involved in tumour generation and malignant growth. However, the expression of these channels in breast cancer remains unclear. Here we studied the expression and function of endogenous TRPC6 channels in a breast cancer cell line (MCF-7), a human breast cancer epithelial primary culture (hBCE) and in normal and tumour breast tissues. METHODS: Molecular (Western blot and RT-PCR), and immunohistochemical techniques were used to investigate TRPC6 expression. To investigate the channel activity in both MCF-7 cells and hBCE we used electrophysiological technique (whole cell patch clamp configuration). RESULTS: A non selective cationic current was activated by the oleoyl-2-acetyl-sn-glycerol (OAG) in both hBCE and MCF-7 cells. OAG-inward current was inhibited by 2-APB, SK&F 96365 and La3+. TRPC6, but not TRPM7, was expressed both in hBCE and in MCF-7 cells. TRPC3 was only expressed in hBCE. Clinically, TRPC6 mRNA and protein were elevated in breast carcinoma specimens in comparison to normal breast tissue. Furthermore, we found that the overexpression of TRPC6 protein levels were not correlated with tumour grades, estrogen receptor expression or lymph node positive tumours. CONCLUSION: Our results indicate that TRPC6 channels are strongly expressed and functional in breast cancer epithelial cells. Moreover, the overexpression of these channels appears without any correlation with tumour grade, ER expression and lymph node metastasis. Our findings support the idea that TRPC6 may have a role in breast carcinogenesis.


Subject(s)
Breast Neoplasms/metabolism , Mammary Glands, Human/metabolism , Neoplasms, Glandular and Epithelial/metabolism , TRPC Cation Channels/biosynthesis , Blotting, Western , Breast Neoplasms/pathology , Cell Culture Techniques , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Mammary Glands, Human/cytology , Neoplasm Staging , Neoplasms, Glandular and Epithelial/pathology , Patch-Clamp Techniques , Reverse Transcriptase Polymerase Chain Reaction , TRPC Cation Channels/physiology , TRPC6 Cation Channel
SELECTION OF CITATIONS
SEARCH DETAIL
...