Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Chem Res Toxicol ; 33(9): 2310-2323, 2020 09 21.
Article in English | MEDLINE | ID: mdl-32786544

ABSTRACT

We have previously shown that phenyl saligenin phosphate (PSP), an organophosphorus compound which is classed as a weak inhibitor of acetylcholinesterase, triggered cytotoxicity in mitotic and differentiated H9c2 cardiomyoblasts. The aim of this study was to assess whether sublethal concentrations of PSP could disrupt the morphology of differentiating rat H9c2 cardiomyoblasts and human-induced pluripotent stem-cell-derived cardiomyocyte progenitor cells (hiPSC-CMs) and to assess the underlying cytoskeletal changes. PSP-induced changes in protein expression were monitored via Western blotting, immunocytochemistry, and proteomic analysis. PSP-mediated cytotoxicity was determined by measuring MTT reduction, LDH release, and caspase-3 activity. Sublethal exposure to PSP (3 µM) induced morphological changes in differentiating H9c2 cells (7, 9, and 13 days), reflected by reduced numbers of spindle-shaped cells. Moreover, this treatment (7 days) attenuated the expression of the cytoskeletal proteins cardiac troponin I, tropomyosin-1, and α-actin. Further proteomic analysis identified nine proteins (e.g., heat shock protein 90-ß and calumenin) which were down-regulated by PSP exposure in H9c2 cells. To assess the cytotoxic effects of organophosphorus compounds in a human cell model, we determined their effects on human-induced pluripotent stem-cell-derived cardiomyocyte progenitor cells. Chlorpyrifos and diazinon-induced cytotoxicity (48 h) was evident only at concentrations >100 µM. By contrast, PSP exhibited cytotoxicity in hiPSC-CMs at a concentration of 25 µM following 48 h exposure. Finally, sublethal exposure to PSP (3 µM; 7 days) induced morphological changes and decreased the expression of cardiac troponin I, tropomyosin-1, and α-actin in hiPSC-CMs. In summary, our data suggest cardiomyocyte morphology is disrupted in both cell models by sublethal concentrations of PSP via modulation of cytoskeletal protein expression.


Subject(s)
Actin Cytoskeleton/drug effects , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Organophosphorus Compounds/pharmacology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Rats
2.
Eur J Pharmacol ; 819: 144-160, 2018 Jan 15.
Article in English | MEDLINE | ID: mdl-29208472

ABSTRACT

Pharmacologically-induced pre- and post-conditioning represent attractive therapeutic strategies to reduce ischaemia/reperfusion injury during cardiac surgery and following myocardial infarction. We have previously reported that transglutaminase 2 (TG2) activity is modulated by the A1 adenosine receptor and ß2-adrenoceptor in H9c2 cardiomyoblasts. The primary aim of this study was to determine the role of TG2 in A1 adenosine receptor and ß2-adrenoceptor-induced pharmacological pre- and post-conditioning in the H9c2 cells. H9c2 cells were exposed to 8h hypoxia (1% O2) followed by 18h reoxygenation, after which cell viability was assessed by monitoring mitochondrial reduction of MTT, lactate dehydrogenase release and caspase-3 activation. N6-cyclopentyladenosine (CPA; A1 adenosine receptor agonist), formoterol (ß2-adrenoceptor agonist) or isoprenaline (non-selective ß-adrenoceptor agonist) were added before hypoxia/reoxygenation (pre-conditioning) or at the start of reoxygenation following hypoxia (post-conditioning). Pharmacological pre- and post-conditioning with CPA and isoprenaline significantly reduced hypoxia/reoxygenation-induced cell death. In contrast, formoterol did not elicit protection. Pre-treatment with pertussis toxin (Gi/o-protein inhibitor), DPCPX (A1 adenosine receptor antagonist) or TG2 inhibitors (Z-DON and R283) attenuated the A1 adenosine receptor-induced pharmacological pre- and post-conditioning. Similarly, pertussis toxin, ICI 118,551 (ß2-adrenoceptor antagonist) or TG2 inhibition attenuated the isoprenaline-induced cell survival. Knockdown of TG2 using small interfering RNA (siRNA) attenuated CPA and isoprenaline-induced pharmacological pre- and post-conditioning. Finally, proteomic analysis following isoprenaline treatment identified known (e.g. protein S100-A6) and novel (e.g. adenine phosphoribosyltransferase) protein substrates for TG2. These results have shown that A1 adenosine receptor and ß2-adrenoceptor-induced protection against simulated hypoxia/reoxygenation occurs in a TG2 and Gi/o-protein dependent manner in H9c2 cardiomyoblasts.


Subject(s)
Cell Death/drug effects , GTP-Binding Proteins/metabolism , Ischemic Postconditioning , Ischemic Preconditioning , Oxygen/metabolism , Receptor, Adenosine A1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Transglutaminases/metabolism , Animals , Caspase 3/metabolism , Cell Hypoxia/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , GTP-Binding Proteins/deficiency , GTP-Binding Proteins/genetics , Gene Knockdown Techniques , Protein Glutamine gamma Glutamyltransferase 2 , Rats , Time Factors , Transglutaminases/deficiency , Transglutaminases/genetics
3.
Eur J Pharmacol ; 813: 105-121, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28754379

ABSTRACT

Tissue transglutaminase 2 (TG2) is modulated by protein kinase A (PKA) mediated phosphorylation: however, the precise mechanism(s) of its modulation by G-protein coupled receptors coupled to PKA activation are not fully understood. In the current study we investigated the potential regulation of TG2 activity by the ß2-adrenoceptor in rat H9c2 cardiomyoblasts. Transglutaminase transamidation activity was assessed using amine-incorporating and protein cross-linking assays. TG2 phosphorylation was determined via immunoprecipitation and Western blotting. The long acting ß2-adrenoceptor agonist formoterol induced time- and concentration-dependent increases in TG2 transamidation. Increases in TG2 activity were reduced by the TG2 inhibitors Z-DON (Benzyloxycarbonyl-(6-Diazo-5-oxonorleucinyl)-L-valinyl-L-prolinyl-L-leucinmethylester) and R283 ((1,3,dimethyl-2[2-oxo-propyl]thio)imidazole chloride). Responses to formoterol were blocked by pharmacological inhibition of PKA, extracellular signal-regulated kinase 1 and 2 (ERK1/2), or phosphatidylinositol 3-kinase (PI-3K) signalling. Furthermore, the removal of extracellular Ca2+ also attenuated formoterol-induced TG2 activation. Fluorescence microscopy demonstrated TG2-induced biotin-X-cadaverine incorporation into proteins. Formoterol increased the levels of TG2-associated phosphoserine and phosphothreonine, which were blocked by inhibition of PKA, ERK1/2 or PI-3K signalling. Subsequent proteomic analysis identified known (e.g. lactate dehydrogenase A chain) and novel (e.g. Protein S100-A6) protein substrates for TG2. Taken together, the data obtained suggest that ß2-adrenoceptor-induced modulation of TG2 represents a novel paradigm in ß2-adrenoceptor cell signalling, expanding the repertoire of cellular functions responsive to catecholamine stimulation.


Subject(s)
GTP-Binding Proteins/metabolism , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, beta-2/metabolism , Transglutaminases/metabolism , Animals , Calcium/metabolism , Catecholamines/pharmacology , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Phosphorylation/drug effects , Protein Glutamine gamma Glutamyltransferase 2 , Rats , Signal Transduction/drug effects , Transglutaminases/antagonists & inhibitors
4.
Biochem Pharmacol ; 107: 41-58, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27005940

ABSTRACT

The regulation of tissue transglutaminase (TG2) activity by the GPCR family is poorly understood. In this study, we investigated the modulation of TG2 activity by the A1 adenosine receptor in cardiomyocyte-like H9c2 cells. H9c2 cells were lysed following stimulation with the A1 adenosine receptor agonist N(6)-cyclopentyladenosine (CPA). Transglutaminase activity was determined using an amine incorporating and a protein cross linking assay. TG2 phosphorylation was assessed via immunoprecipitation and Western blotting. The role of TG2 in A1 adenosine receptor-induced cytoprotection was investigated by monitoring hypoxia-induced cell death. CPA induced time and concentration-dependent increases in amine incorporating and protein crosslinking activity of TG2. CPA-induced increases in TG2 activity were attenuated by the TG2 inhibitors Z-DON and R283. Responses to CPA were blocked by PKC (Ro 31-8220), MEK1/2 (PD 98059), p38 MAPK (SB 203580) and JNK1/2 (SP 600125) inhibitors and by removal of extracellular Ca(2+). CPA triggered robust increases in the levels of TG2-associated phosphoserine and phosphothreonine, which were attenuated by PKC, MEK1/2 and JNK1/2 inhibitors. Fluorescence microscopy revealed TG2-mediated biotin-X-cadaverine incorporation into proteins and proteomic analysis identified known (Histone H4) and novel (Hexokinase 1) protein substrates for TG2. CPA pre-treatment reversed hypoxia-induced LDH release and decreases in MTT reduction. TG2 inhibitors R283 and Z-DON attenuated A1 adenosine receptor-induced cytoprotection. TG2 activity was stimulated by the A1 adenosine receptor in H9c2 cells via a multi protein kinase dependent pathway. These results suggest a role for TG2 in A1 adenosine receptor-induced cytoprotection.


Subject(s)
Adenosine A1 Receptor Agonists/pharmacology , GTP-Binding Proteins/agonists , MAP Kinase Signaling System/drug effects , Myoblasts, Cardiac/drug effects , Protein Processing, Post-Translational/drug effects , Receptor, Adenosine A1/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Calcium Signaling/drug effects , Cell Hypoxia/drug effects , Cell Line , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/metabolism , Hexokinase/metabolism , Histones/metabolism , Kinetics , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/metabolism , Phosphorylation/drug effects , Protein Glutamine gamma Glutamyltransferase 2 , Protein Kinase Inhibitors/pharmacology , Rats , Receptor, Adenosine A1/blood , Substrate Specificity , Transglutaminases/antagonists & inhibitors , Transglutaminases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...