Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Basic Res Cardiol ; 118(1): 4, 2023 01 20.
Article in English | MEDLINE | ID: mdl-36670288

ABSTRACT

During embryonic development, cardiomyocytes undergo differentiation and maturation, processes that are tightly regulated by tissue-specific signaling cascades. Although redox signaling pathways involved in cardiomyogenesis are established, the exact sources responsible for reactive oxygen species (ROS) formation remain elusive. The present study investigates whether ROS produced by the mitochondrial flavoenzyme monoamine oxidase A (MAO-A) play a role in cardiomyocyte differentiation from human induced pluripotent stem cells (hiPSCs). Wild type (WT) and MAO-A knock out (KO) hiPSCs were generated by CRISPR/Cas9 genome editing and subjected to cardiomyocyte differentiation. Mitochondrial ROS levels were lower in MAO-A KO compared to the WT cells throughout the differentiation process. MAO-A KO hiPSC-derived cardiomyocytes (hiPSC-CMs) displayed sarcomere disarray, reduced α- to ß-myosin heavy chain ratio, GATA4 upregulation and lower macroautophagy levels. Functionally, genetic ablation of MAO-A negatively affected intracellular Ca2+ homeostasis in hiPSC-CMs. Mechanistically, MAO-A generated ROS contributed to the activation of AKT signaling that was considerably attenuated in KO cells. In addition, MAO-A ablation caused a reduction in WNT pathway gene expression consistent with its reported stimulation by ROS. As a result of WNT downregulation, expression of MESP1 and NKX2.5 was significantly decreased in MAO-A KO cells. Finally, MAO-A re-expression during differentiation rescued expression levels of cardiac transcription factors, contractile structure, and intracellular Ca2+ homeostasis. Taken together, these results suggest that MAO-A mediated ROS generation is necessary for the activation of AKT and WNT signaling pathways during cardiac lineage commitment and for the differentiation of fully functional human cardiomyocytes.


Subject(s)
Induced Pluripotent Stem Cells , Myocytes, Cardiac , Humans , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Monoamine Oxidase/genetics , Monoamine Oxidase/metabolism , Cell Differentiation/physiology , Wnt Signaling Pathway
2.
Antioxidants (Basel) ; 11(5)2022 May 05.
Article in English | MEDLINE | ID: mdl-35624771

ABSTRACT

The control of radical damage and oxidative stress, phenomena involved in a large number of human pathologies, is a major pharmaceutical and medical goal. We here show that two biocompatible formulations of Pluronic-stabilized, poly (lipoic acid)-based nanoparticles (NP) effectively antagonized the formation of radicals and reactive oxygen species (ROS). These NPs, not only intrinsically scavenged radicals in a-cellular DPPH/ABTS assays, but also inhibited the overproduction of ROS induced by tert-Butyl hydroperoxide (t-BHP) in tumor cells (HeLa), human macrophages and neonatal rat ventricular myocytes (NRVMs). NPs were captured by macrophages and cardiomyocytes much more effectively as compared to HeLa cells and non-phagocytic leukocytes, eventually undergoing intracellular disassembly. Notably, NPs decreased the mitochondrial ROS generation induced by simulated Ischemia/Reperfusion Injury (IRI) in isolated cardiomyocytes. NPs also prevented IRI-triggered cardiomyocyte necrosis, mitochondrial dysfunction, and alterations of contraction-related intracellular Ca2+ waves. Hence, NPs appear to be an effective and cardiomyocyte-selective drug to protect against damages induced by post-ischemic reperfusion.

3.
Cell Calcium ; 94: 102344, 2021 03.
Article in English | MEDLINE | ID: mdl-33556741

ABSTRACT

Mitochondrial reactive oxygen species (mROS) are routinely produced at several sites within the organelle. The balance in their formation and elimination is maintained by a complex and robust antioxidant system. mROS may act as second messengers and regulate a number of physiological processes, such as insulin signaling, cell differentiation and proliferation, wound healing, etc. Nevertheless, when a sudden or sustained increase in ROS formation is not efficiently neutralized by the endogenous antioxidant defense system, the detrimental impact of high mROS levels on cell function and viability eventually results in disease development. In this review, we will focus on the dual role of mROS in pathophysiology, emphasizing the physiological role exerted by a regulated mROS production/elimination, and discussing the detrimental effects evoked by an imbalance in mitochondrial redox state. Furthermore, we will touch upon the interplay between mROS and Ca2+ homeostasis.


Subject(s)
Disease , Mitochondria/metabolism , Physiological Phenomena , Reactive Oxygen Species/metabolism , Animals , Calcium/metabolism , Humans , Oxidation-Reduction , Oxidative Stress
4.
Antioxid Redox Signal ; 34(7): 531-550, 2021 03 01.
Article in English | MEDLINE | ID: mdl-32524823

ABSTRACT

Aims: Doxorubicin cardiomyopathy is a lethal pathology characterized by oxidative stress, mitochondrial dysfunction, and contractile impairment, leading to cell death. Although extensive research has been done to understand the pathophysiology of doxorubicin cardiomyopathy, no effective treatments are available. We investigated whether monoamine oxidases (MAOs) could be involved in doxorubicin-derived oxidative stress, and in the consequent mitochondrial, cardiomyocyte, and cardiac dysfunction. Results: We used neonatal rat ventricular myocytes (NRVMs) and adult mouse ventricular myocytes (AMVMs). Doxorubicin alone (i.e., 0.5 µM doxorubicin) or in combination with H2O2 induced an increase in mitochondrial formation of reactive oxygen species (ROS), which was prevented by the pharmacological inhibition of MAOs in both NRVMs and AMVMs. The pharmacological approach was supported by the genetic ablation of MAO-A in NRVMs. In addition, doxorubicin-derived ROS caused lipid peroxidation and alterations in mitochondrial function (i.e., mitochondrial membrane potential, permeability transition, redox potential), mitochondrial morphology (i.e., mitochondrial distribution and perimeter), sarcomere organization, intracellular [Ca2+] homeostasis, and eventually cell death. All these dysfunctions were abolished by MAO inhibition. Of note, in vivo MAO inhibition prevented chamber dilation and cardiac dysfunction in doxorubicin-treated mice. Innovation and Conclusion: This study demonstrates that the severe oxidative stress induced by doxorubicin requires the involvement of MAOs, which modulate mitochondrial ROS generation. MAO inhibition provides evidence that mitochondrial ROS formation is causally linked to all disorders caused by doxorubicin in vitro and in vivo. Based upon these results, MAO inhibition represents a novel therapeutic approach for doxorubicin cardiomyopathy.


Subject(s)
Doxorubicin/pharmacology , Heart Ventricles/drug effects , Monoamine Oxidase/metabolism , Myocytes, Cardiac/drug effects , Reactive Oxygen Species/metabolism , Animals , Heart Ventricles/metabolism , Mice , Mitochondria , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/analysis
6.
Cell Rep ; 32(9): 108095, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32877677

ABSTRACT

The mitochondrial permeability transition pore (PTP) is a Ca2+-activated channel that plays a key role in cell death. Thiol oxidation facilitates PTP opening, yet the targets and molecular mechanisms still await a definition. Here, we investigate the role of C141 of F-ATP synthase oligomycin sensitivity conferral protein (OSCP) subunit in PTP modulation by oxidation. We find that the OSCP C141S mutation confers resistance to PTP opening and cell death by diamide and MitoParaquat only when cyclophilin D (CyPD) has been ablated, a protective role that can be explained by CyPD shielding C141 from oxidants. The mutation decreases apoptosis in zebrafish embryos, indicating that this OSCP residue is involved in development. Site-directed mutagenesis in yeast suggests that other conserved cysteines in the α, γ, and c subunits of F-ATP synthase are not involved in PTP modulation. Thus, OSCP provides a strategic site that regulates PTP opening by the interplay between CyPD (un)binding and thiol oxidation-reduction.


Subject(s)
Cysteine/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Permeability , Humans
7.
Pharmacol Res ; 151: 104548, 2020 01.
Article in English | MEDLINE | ID: mdl-31759087

ABSTRACT

Ischemia/reperfusion (I/R) injury is mediated in large part by opening of the mitochondrial permeability transition pore (PTP). Consequently, inhibitors of the PTP hold great promise for the treatment of a variety of cardiovascular disorders. At present, PTP inhibition is obtained only through the use of drugs (e.g. cyclosporine A, CsA) targeting cyclophilin D (CyPD) which is a key modulator, but not a structural component of the PTP. This limitation might explain controversial findings in clinical studies. Therefore, we investigated the protective effects against I/R injury of small-molecule inhibitors of the PTP (63 and TR002) that do not target CyPD. Both compounds exhibited a dose-dependent inhibition of PTP opening in isolated mitochondria and were more potent than CsA. Notably, PTP inhibition was observed also in mitochondria devoid of CyPD. Compounds 63 and TR002 prevented PTP opening and mitochondrial depolarization induced by Ca2+ overload and by reactive oxygen species in neonatal rat ventricular myocytes (NRVMs). Remarkably, both compounds prevented cell death, contractile dysfunction and sarcomeric derangement induced by anoxia/reoxygenation injury in NRVMs at sub-micromolar concentrations, and were more potent than CsA. Cardioprotection was observed also in adult mouse ventricular myocytes and human iPSc-derived cardiomyocytes, as well as ex vivo in perfused hearts. Thus, this study demonstrates that 63 and TR002 represent novel cardioprotective agents that inhibit PTP opening independent of CyPD targeting.


Subject(s)
Cardiotonic Agents/therapeutic use , Mitochondrial Permeability Transition Pore/antagonists & inhibitors , Myocardial Reperfusion Injury/drug therapy , Small Molecule Libraries/therapeutic use , Animals , Cardiotonic Agents/pharmacology , Cell Line , Cells, Cultured , Humans , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondrial Permeability Transition Pore/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Rats, Sprague-Dawley , Rats, Wistar , Small Molecule Libraries/pharmacology
8.
Free Radic Biol Med ; 134: 678-687, 2019 04.
Article in English | MEDLINE | ID: mdl-30731114

ABSTRACT

Reactive oxygen species (ROS) have an equivocal role in myocardial ischaemia reperfusion injury. Within the cardiomyocyte, mitochondria are both a major source and target of ROS. We evaluate the effects of a selective, dose-dependent increase in mitochondrial ROS levels on cardiac physiology using the mitochondria-targeted redox cycler MitoParaquat (MitoPQ). Low levels of ROS decrease the susceptibility of neonatal rat ventricular myocytes (NRVMs) to anoxia/reoxygenation injury and also cause profound protection in an in vivo mouse model of ischaemia/reperfusion. However higher doses of MitoPQ resulted in a progressive alteration of intracellular [Ca2+] homeostasis and mitochondrial function in vitro, leading to dysfunction and death at high doses. Our data show that a primary increase in mitochondrial ROS can alter cellular function, and support a hormetic model in which low levels of ROS are cardioprotective while higher levels of ROS are cardiotoxic.


Subject(s)
Disease Models, Animal , Hormesis , Mitochondria, Heart/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/cytology , Paraquat/pharmacology , Superoxides/metabolism , Animals , Animals, Newborn , Apoptosis , Herbicides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Mitochondria, Heart/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Rats, Wistar
9.
Basic Res Cardiol ; 113(5): 39, 2018 08 17.
Article in English | MEDLINE | ID: mdl-30120595
10.
Methods Mol Biol ; 1782: 403-418, 2018.
Article in English | MEDLINE | ID: mdl-29851014

ABSTRACT

Reactive oxygen species (ROS) are involved in both physiological and pathological processes. This widely accepted concept is based more on the effects of antioxidant interventions than on reliable assessments of rates and sites of intracellular ROS formation. This argument applies also to mitochondria that are generally considered the major site for ROS formation, especially in skeletal and cardiac myocytes.Detection of oxidative modifications of intracellular or circulating molecules is frequently used as a marker of ROS formation. However, this approach provides limited information on spatiotemporal aspects of ROS formation that have to be defined in order to elucidate the role of ROS in a given pathophysiological condition. This information can be obtained by means of fluorescent probes that allow monitoring ROS formation in cell-free extracts and isolated cells. Thus, this approach can be used to characterize ROS formation in both isolated mitochondria and mitochondria within intact cells. This chapter describes three major examples of the use of fluorescent probes for monitoring mitochondrial ROS formation. Detailed methods description is accompanied by a critical analysis of the limitations of each technique, highlighting the possible sources of errors in performing the assay and results interpretation.


Subject(s)
Fluorescent Dyes/chemistry , Fluorometry/methods , Mitochondria, Heart/metabolism , Primary Cell Culture/methods , Reactive Oxygen Species/analysis , Animals , Animals, Newborn , Calibration , Cells, Cultured , Fluorometry/instrumentation , Image Processing, Computer-Assisted/instrumentation , Image Processing, Computer-Assisted/methods , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Myocytes, Cardiac , Oxidation-Reduction , Primary Cell Culture/instrumentation , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Software
11.
Front Physiol ; 9: 38, 2018.
Article in English | MEDLINE | ID: mdl-29467663

ABSTRACT

The ErbB2 blocker trastuzumab improves survival in oncologic patients, but can cause cardiotoxicity. The late Na+ current inhibitor ranolazine has been shown to counter experimental HF, including doxorubicin cardiotoxicity (a condition characterized by derangements in redox balance), by lowering the levels of reactive oxygen species (ROS). Since ErbB2 can modulate ROS signaling, we tested whether trastuzumab cardiotoxicity could be blunted by ranolazine via redox-mediated mechanisms. Trastuzumab decreased fractional shortening and ejection fraction in mice, but ranolazine prevented heart dysfunction when co-administered with trastuzumab. Trastuzumab cardiotoxicity was accompanied by elevations in natriuretic peptides and matrix metalloproteinase 2 (MMP2) mRNAs, which were not elevated with co-treatment with ranolazine. Trastuzumab also increased cleavage of caspase-3, indicating activation of the proapoptotic machinery. Again, ranolazine prevented this activation. Interestingly, Neonatal Rat Ventricular Myocytes (NRVMs), labeled with MitoTracker Red and treated with trastuzumab, showed only a small increase in ROS compared to baseline conditions. We then stressed trastuzumab-treated cells with the beta-agonist isoproterenol to increase workload, and we observed a significant increase of probe fluorescence, compared with cells treated with isoproterenol alone, reflecting induction of oxidative stress. These effects were blunted by ranolazine, supporting a role for INa inhibition in the regulation of redox balance also in trastuzumab cardiotoxicity.

12.
Cell Death Differ ; 25(9): 1671-1685, 2018 09.
Article in English | MEDLINE | ID: mdl-29459772

ABSTRACT

Monoamine oxidase (MAO) inhibitors ameliorate contractile function in diabetic animals, but the mechanisms remain unknown. Equally elusive is the interplay between the cardiomyocyte alterations induced by hyperglycemia and the accompanying inflammation. Here we show that exposure of primary cardiomyocytes to high glucose and pro-inflammatory stimuli leads to MAO-dependent increase in reactive oxygen species that causes permeability transition pore opening and mitochondrial dysfunction. These events occur upstream of endoplasmic reticulum (ER) stress and are abolished by the MAO inhibitor pargyline, highlighting the role of these flavoenzymes in the ER/mitochondria cross-talk. In vivo, streptozotocin administration to mice induced oxidative changes and ER stress in the heart, events that were abolished by pargyline. Moreover, MAO inhibition prevented both mast cell degranulation and altered collagen deposition, thereby normalizing diastolic function. Taken together, these results elucidate the mechanisms underlying MAO-induced damage in diabetic cardiomyopathy and provide novel evidence for the role of MAOs in inflammation and inter-organelle communication. MAO inhibitors may be considered as a therapeutic option for diabetic complications as well as for other disorders in which mast cell degranulation is a dominant phenomenon.


Subject(s)
Cell Degranulation/drug effects , Endoplasmic Reticulum Stress , Mitochondria/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Ventricular Remodeling/drug effects , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Endoplasmic Reticulum Stress/drug effects , Glucose/pharmacology , Interleukin-1beta/pharmacology , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Monoamine Oxidase/chemistry , Monoamine Oxidase/genetics , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle Cells/physiology , Myocardium/metabolism , Myocardium/pathology , RNA Interference , RNA, Small Interfering/metabolism , Rats , Reactive Oxygen Species/metabolism
13.
EMBO Rep ; 19(2): 257-268, 2018 02.
Article in English | MEDLINE | ID: mdl-29217657

ABSTRACT

The permeability transition pore (PTP) is a Ca2+-dependent mitochondrial channel whose opening causes a permeability increase in the inner membrane to ions and solutes. The most potent inhibitors are matrix protons, with channel block at pH 6.5. Inhibition is reversible, mediated by histidyl residue(s), and prevented by their carbethoxylation by diethylpyrocarbonate (DPC), but their assignment is unsolved. We show that PTP inhibition by H+ is mediated by the highly conserved histidyl residue (H112 in the human mature protein) of oligomycin sensitivity conferral protein (OSCP) subunit of mitochondrial F1FO (F)-ATP synthase, which we also show to undergo carbethoxylation after reaction of mitochondria with DPC. Mitochondrial PTP-dependent swelling cannot be inhibited by acidic pH in H112Q and H112Y OSCP mutants, and the corresponding megachannels (the electrophysiological counterpart of the PTP) are insensitive to inhibition by acidic pH in patch-clamp recordings of mitoplasts. Cells harboring the H112Q and H112Y mutations are sensitized to anoxic cell death at acidic pH. These results demonstrate that PTP channel formation and its inhibition by H+ are mediated by the F-ATP synthase.


Subject(s)
Histidine/metabolism , Hydrogen-Ion Concentration , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Animals , Calcium/metabolism , Calcium Signaling , Cattle , Cell Line , Cell Membrane Permeability , Histidine/chemistry , Humans , Hydrolysis , Hypoxia/metabolism , Mice , Mitochondria, Liver/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Proton-Translocating ATPases/chemistry , Models, Molecular , Molecular Dynamics Simulation , Oxygen Consumption , Protein Conformation , Protein Subunits
14.
Islets ; 7(4): e1107255, 2015.
Article in English | MEDLINE | ID: mdl-26732126

ABSTRACT

Disturbances in pulsatile insulin secretion and Ca(2+) oscillations in pancreatic ß-cells are early markers of diabetes, but the underlying mechanisms are still incompletely understood. Reactive oxygen/nitrogen species (ROS/RNS) are implicated in reduced ß-cell function, and ROS/RNS target several Ca(2+) pumps and channels. Thus, we hypothesized that ROS/RNS could disturb Ca(2+) oscillations and downstream insulin pulsatility. We show that ROS/RNS production by photoactivation of aluminum phthalocyanine chloride (AlClPc) abolish or accelerate Ca(2+) oscillations in the MIN6 ß-cell line, depending on the amount of ROS/RNS. Application of the sarcoplasmic/endoplasmic reticulum Ca(2+) ATPase (SERCA) inhibitor thapsigargin modifies the Ca(2+) response to high concentrations of ROS/RNS. Further, thapsigargin produces effects that resemble those elicited by moderate ROS/RNS production. These results indicate that ROS/RNS interfere with endoplasmic reticulum Ca(2+) handling. This idea is supported by theoretical studies using a mathematical model of Ca(2+) handling adapted to MIN6 cells. Our results suggest a putative link between ROS/RNS and disturbed pulsatile insulin secretion.


Subject(s)
Calcium Signaling/drug effects , Insulin-Secreting Cells/drug effects , Reactive Nitrogen Species/pharmacology , Reactive Oxygen Species/pharmacology , Animals , Cells, Cultured , Glucose/pharmacology , Indoles/pharmacology , Insulin-Secreting Cells/metabolism , Mice , Organometallic Compounds/pharmacology , Potassium Channels/drug effects , Potassium Channels/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL