Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Free Radic Biol Med ; 205: 244-261, 2023 08 20.
Article in English | MEDLINE | ID: mdl-37295539

ABSTRACT

Myocardial ischemia-reperfusion (IR) injury may result in cardiomyocyte dysfunction. Mitochondria play a critical role in cardiomyocyte recovery after IR injury. The mitochondrial uncoupling protein 3 (UCP3) has been proposed to reduce mitochondrial reactive oxygen species (ROS) production and to facilitate fatty acid oxidation. As both mechanisms might be protective following IR injury, we investigated functional, mitochondrial structural, and metabolic cardiac remodeling in wild-type mice and in mice lacking UCP3 (UCP3-KO) after IR. Results showed that infarct size in isolated perfused hearts subjected to IR ex vivo was larger in adult and old UCP3-KO mice than in equivalent wild-type mice, and was accompanied by higher levels of creatine kinase in the effluent and by more pronounced mitochondrial structural changes. The greater myocardial damage in UCP3-KO hearts was confirmed in vivo after coronary artery occlusion followed by reperfusion. S1QEL, a suppressor of superoxide generation from site IQ in complex I, limited infarct size in UCP3-KO hearts, pointing to exacerbated superoxide production as a possible cause of the damage. Metabolomics analysis of isolated perfused hearts confirmed the reported accumulation of succinate, xanthine and hypoxanthine during ischemia, and a shift to anaerobic glucose utilization, which all recovered upon reoxygenation. The metabolic response to ischemia and IR was similar in UCP3-KO and wild-type hearts, being lipid and energy metabolism the most affected pathways. Fatty acid oxidation and complex I (but not complex II) activity were equally impaired after IR. Overall, our results indicate that UCP3 deficiency promotes enhanced superoxide generation and mitochondrial structural changes that increase the vulnerability of the myocardium to IR injury.


Subject(s)
Coronary Artery Disease , Myocardial Ischemia , Myocardial Reperfusion Injury , Mice , Animals , Superoxides/metabolism , Myocardial Ischemia/metabolism , Myocytes, Cardiac/metabolism , Mitochondria/metabolism , Oxidative Stress , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Coronary Artery Disease/metabolism , Energy Metabolism , Ischemia/metabolism , Reperfusion , Fatty Acids/metabolism , Infarction/complications , Infarction/metabolism
2.
Antioxidants (Basel) ; 11(10)2022 Sep 25.
Article in English | MEDLINE | ID: mdl-36290620

ABSTRACT

The hexanucleotide expansion of the C9orf72 gene is found in 40% of familial amyotrophic lateral sclerosis (ALS) patients. This genetic alteration has been connected with impaired management of reactive oxygen species. In this study, we conducted targeted transcriptional profiling in leukocytes from C9orf72 patients and control subjects by examining the mRNA levels of 84 redox-related genes. The expression of ten redox genes was altered in samples from C9orf72 ALS patients compared to healthy controls. Considering that Nuclear factor erythroid 2-Related Factor 2 (NRF2) modulates the expression of a wide range of redox genes, we further investigated its status on an in vitro model of dipeptide repeat (DPR) toxicity. This model mimics the gain of function, toxic mechanisms attributed to C9orf72 pathology. We found that exposure to DPRs increased superoxide levels and reduced mitochondrial potential as well as cell survival. Importantly, cells overexpressing DPRs exhibited reduced protein levels of NRF2 and its target genes upon inhibition of the proteasome or its canonical repressor, the E3 ligase adapter KEAP1. However, NRF2 activation was sufficient to recover cell viability and redox homeostasis. This study identifies NRF2 as a putative target in precision medicine for the therapy of ALS patients harboring C9orf72 expansion repeats.

3.
Science ; 377(6606): 579-580, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35926037

ABSTRACT

Activation of a G protein-coupled receptor prevents cardiomyocyte death during ischemia.


Subject(s)
Mitochondria , Myocardial Ischemia , Receptors, G-Protein-Coupled , Humans , Mitochondria/metabolism , Myocardial Ischemia/metabolism , Myocytes, Cardiac/metabolism , Receptors, G-Protein-Coupled/metabolism
4.
Int J Mol Sci ; 22(21)2021 Nov 03.
Article in English | MEDLINE | ID: mdl-34769371

ABSTRACT

Nuclear factor erythroid-2 related factor 2 (Nrf2) is a transcription factor that controls cellular defense responses against toxic and oxidative stress by modulating the expression of genes involved in antioxidant response and drug detoxification. In addition to maintaining redox homeostasis, Nrf2 is also involved in various cellular processes including metabolism and inflammation. Nrf2 activity is tightly regulated at the transcriptional, post-transcriptional and post-translational levels, which allows cells to quickly respond to pathological stress. In the present review, we describe the molecular mechanisms underlying the transcriptional regulation of Nrf2. We also focus on the impact of Nrf2 in cardiac ischemia-reperfusion injury, a condition that stimulates the overproduction of reactive oxygen species. Finally, we analyze the protective effect of several natural and synthetic compounds that induce Nrf2 activation and protect against ischemia-reperfusion injury in the heart and other organs, and their potential clinical application.


Subject(s)
Antioxidants/pharmacology , Myocardial Reperfusion Injury/prevention & control , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/metabolism , Animals , Humans
5.
Arthritis Rheumatol ; 72(3): 477-487, 2020 03.
Article in English | MEDLINE | ID: mdl-31509349

ABSTRACT

OBJECTIVE: Pulmonary arterial hypertension (PAH), one of the major complications of systemic sclerosis (SSc), is a rare disease with unknown etiopathogenesis and noncurative treatments. As mice deficient in P-selectin glycoprotein ligand 1 (PSGL-1) develop a spontaneous SSc-like syndrome, we undertook this study to analyze whether they develop PAH and to examine the molecular mechanisms involved. METHODS: Doppler echocardiography was used to estimate pulmonary pressure, immunohistochemistry was used to assess vascular remodeling, and myography of dissected pulmonary artery rings was used to analyze vascular reactivity. Angiotensin II (Ang II) levels were quantified by enzyme-linked immunosorbent assay, and Western blotting was used to measure Ang II type 1 receptor (AT1 R), AT2 R, endothelial cell nitric oxide synthase (eNOS), and phosphorylated eNOS expression in lung lysates. Flow cytometry allowed us to determine cytokine production by immune cells and NO production by endothelial cells. In all cases, there were 4-8 mice per experimental group. RESULTS: PSGL-1-/- mice showed lung vessel wall remodeling and a reduced mean ± SD expression of pulmonary AT2 R (expression ratio [relative to ß-actin] in female mice age >18 months: wild-type mice 0.799 ± 0.508 versus knockout mice 0.346 ± 0.229). With aging, female PSGL-1-/- mice had impaired up-regulation of estrogen receptor α (ERα) and developed lung vascular endothelial dysfunction coinciding with an increase in mean ± SEM pulmonary Ang II levels (wild-type 48.70 ± 5.13 pg/gm lung tissue versus knockout 78.02 ± 28.09 pg/gm lung tissue) and a decrease in eNOS phosphorylation, leading to reduced endothelial NO production. These events led to a reduction in the pulmonary artery acceleration time:ejection time ratio in 33% of aged female PSGL-1-/- mice, indicating pulmonary hypertension. Importantly, we found expanded populations of interferon-γ-producing PSGL-1-/- T cells and B cells and a reduced presence of regulatory T cells. CONCLUSION: The absence of PSGL-1 induces a reduction in Treg cells, NO production, and ERα expression and causes an increase in Ang II in the lungs of female mice, favoring the development of PAH.


Subject(s)
Hypertension, Pulmonary/genetics , Membrane Glycoproteins/deficiency , Scleroderma, Systemic/genetics , Angiotensin II/metabolism , Animals , Disease Models, Animal , Endothelial Cells/metabolism , Female , Lung/metabolism , Male , Mice , Mice, Knockout , Nitric Oxide Synthase Type III/biosynthesis , Vascular Remodeling/genetics
6.
Front Pharmacol ; 10: 740, 2019.
Article in English | MEDLINE | ID: mdl-31333462

ABSTRACT

Massive intravascular hemolysis is associated with acute kidney injury (AKI). Nuclear factor erythroid-2-related factor 2 (Nrf2) plays a central role in the defense against oxidative stress by activating the expression of antioxidant proteins. We investigated the role of Nrf2 in intravascular hemolysis and whether Nrf2 activation protected against hemoglobin (Hb)/heme-mediated renal damage in vivo and in vitro. We observed renal Nrf2 activation in human hemolysis and in an experimental model of intravascular hemolysis promoted by phenylhydrazine intraperitoneal injection. In wild-type mice, Hb/heme released from intravascular hemolysis promoted AKI, resulting in decreased renal function, enhanced expression of tubular injury markers (KIM-1 and NGAL), oxidative and endoplasmic reticulum stress (ER), and cell death. These features were more severe in Nrf2-deficient mice, which showed decreased expression of Nrf2-related antioxidant enzymes, including heme oxygenase 1 (HO-1) and ferritin. Nrf2 activation with sulforaphane protected against Hb toxicity in mice and cultured tubular epithelial cells, ameliorating renal function and kidney injury and reducing cell stress and death. Nrf2 genotype or sulforaphane treatment did not influence the severity of hemolysis. In conclusion, our study identifies Nrf2 as a key molecule involved in protection against renal damage associated with hemolysis and opens novel therapeutic approaches to prevent renal damage in patients with severe hemolytic crisis. These findings provide new insights into novel aspects of Hb-mediated renal toxicity and may have important therapeutic implications for intravascular hemolysis-related diseases.

7.
Biochim Biophys Acta Bioenerg ; 1859(9): 940-950, 2018 09.
Article in English | MEDLINE | ID: mdl-29859845

ABSTRACT

Mitochondrial oxidative phosphorylation is incompletely coupled, since protons translocated to the intermembrane space by specific respiratory complexes of the electron transport chain can return to the mitochondrial matrix independently of the ATP synthase -a process known as proton leak- generating heat instead of ATP. Proton leak across the inner mitochondrial membrane increases the respiration rate and decreases the electrochemical proton gradient (Δp), and is an important mechanism for energy dissipation that accounts for up to 25% of the basal metabolic rate. It is well established that mitochondrial superoxide production is steeply dependent on Δp in isolated mitochondria and, correspondingly, mitochondrial uncoupling has been identified as a cytoprotective strategy under conditions of oxidative stress, including diabetes, drug-resistance in tumor cells, ischemia-reperfusion (IR) injury or aging. Mitochondrial uncoupling proteins (UCPs) are able to lower the efficiency of oxidative phosphorylation and are involved in the control of mitochondrial reactive oxygen species (ROS) production. There is strong evidence that UCP2 and UCP3, the UCP1 homologues expressed in the heart, protect against mitochondrial oxidative damage by reducing the production of ROS. This review first analyzes the relationship between mitochondrial proton leak and ROS generation, and then focuses on the cardioprotective role of chemical uncoupling and uncoupling mediated by UCPs. This includes their protective effects against cardiac IR, a condition known to increase ROS production, and their roles in modulating cardiovascular risk factors such as obesity, diabetes and atherosclerosis.


Subject(s)
Cardiovascular Diseases/prevention & control , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Uncoupling Agents/pharmacology , Animals , Humans , Mitochondria/drug effects
8.
Free Radic Biol Med ; 117: 76-89, 2018 03.
Article in English | MEDLINE | ID: mdl-29373843

ABSTRACT

Ischemia-reperfusion (IR) injury is central to the pathology of major cardiovascular diseases, such as stroke and myocardial infarction. IR injury is mediated by several factors including the elevated production of reactive oxygen species (ROS), which occurs particularly at reperfusion. The mitochondrial respiratory chain and NADPH oxidases of the NOX family are major sources of ROS in cardiomyocytes. The first part of this review discusses recent findings and controversies on the mechanisms of superoxide production by the mitochondrial electron transport chain during IR injury, as well as the contribution of the NOX isoforms expressed in cardiomyocytes, NOX1, NOX2 and NOX4, to this damage. It then focuses on the effects of ROS on the opening of the mitochondrial permeability transition pore (mPTP), an inner membrane non-selective pore that causes irreversible damage to the heart. The second part analyzes the redox mechanisms of cardiomyocyte mitochondrial protection; specifically, the activation of the hypoxia-inducible factor (HIF) pathway and the antioxidant transcription factor Nrf2, which are both regulated by the cellular redox state. Redox mechanisms involved in ischemic preconditioning, one of the most effective ways of protecting the heart against IR injury, are also reviewed. Interestingly, several of these protective pathways converge on the inhibition of mPTP opening during reperfusion. Finally, the clinical and translational implications of these cardioprotective mechanisms are discussed.


Subject(s)
Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Animals , Humans , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Oxidation-Reduction
9.
J Pathol ; 244(3): 296-310, 2018 03.
Article in English | MEDLINE | ID: mdl-29205354

ABSTRACT

Recurrent and massive intravascular haemolysis induces proteinuria, glomerulosclerosis, and progressive impairment of renal function, suggesting podocyte injury. However, the effects of haemoglobin (Hb) on podocytes remain unexplored. Our results show that cultured human podocytes or podocytes isolated from murine glomeruli bound and endocytosed Hb through the megalin-cubilin receptor system, thus resulting in increased intracellular Hb catabolism, oxidative stress, activation of the intrinsic apoptosis pathway, and altered podocyte morphology, with decreased expression of the slit diaphragm proteins nephrin and synaptopodin. Hb uptake activated nuclear factor erythroid-2-related factor 2 (Nrf2) and induced expression of the Nrf2-related antioxidant proteins haem oxygenase-1 (HO-1) and ferritin. Nrf2 activation and Hb staining was observed in podocytes of mice with intravascular haemolysis. These mice developed proteinuria and showed podocyte injury, characterized by foot process effacement, decreased synaptopodin and nephrin expression, and podocyte apoptosis. These pathological effects were enhanced in Nrf2-deficient mice, whereas Nrf2 activation with sulphoraphane protected podocytes against Hb toxicity both in vivo and in vitro. Supporting the translational significance of our findings, we observed podocyte damage and podocytes stained for Hb, HO-1, ferritin and phosphorylated Nrf2 in renal sections and urinary sediments of patients with massive intravascular haemolysis, such as atypical haemolytic uraemic syndrome and paroxysmal nocturnal haemoglobinuria. In conclusion, podocytes take up Hb both in vitro and during intravascular haemolysis, promoting oxidative stress, podocyte dysfunction, and apoptosis. Nrf2 may be a potential therapeutic target to prevent loss of renal function in patients with intravascular haemolysis. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Acute Kidney Injury/metabolism , Anemia, Hemolytic/metabolism , Apoptosis , Hemoglobins/metabolism , Podocytes/metabolism , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Adult , Anemia, Hemolytic/genetics , Anemia, Hemolytic/pathology , Animals , Cell Line , Disease Models, Animal , Endocytosis , Female , Ferritins/metabolism , Heme Oxygenase-1/metabolism , Hemolysis , Humans , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 1/genetics , NF-E2-Related Factor 1/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Phosphorylation , Podocytes/ultrastructure , Receptors, Cell Surface/metabolism , Young Adult
10.
Antioxid Redox Signal ; 27(2): 93-105, 2017 07 10.
Article in English | MEDLINE | ID: mdl-27554853

ABSTRACT

AIMS: A loss in brain acetylcholine and cholinergic markers, subchronic inflammation, and impaired mitochondrial function, which lead to low-energy production and high oxidative stress, are common pathological factors in several neurodegenerative diseases (NDDs). Glial cells are important for brain homeostasis, and microglia controls the central immune response, where α7 acetylcholine nicotinic receptors (nAChR) seem to play a pivotal role; however, little is known about the effects of this receptor in metabolism. Therefore, the aim of this study was to evaluate if glial mitochondrial energetics could be regulated through α7 nAChR. RESULTS: Primary glial cultures treated with the α7 nicotinic agonist PNU282987 increased their mitochondrial mass and their mitochondrial oxygen consumption without increasing oxidative stress; these changes were abolished when nuclear erythroid 2-related factor 2 (Nrf2) was absent, heme oxygenase-1 (HO-1) was inhibited, or peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α) was silenced. More specifically, microglia of animals treated intraperitoneally with the α7 nAChR agonist PNU282987 (10 mg/kg) showed a significant increase in mitochondrial mass. Interestingly, LysMcre-Hmox1Δ/Δ and PGC-1α-/- animals showed lower microglial mitochondrial levels and treatment with PNU282987 did not produce effects on mitochondrial levels. INNOVATION: Increases in microglial mitochondrial mass and metabolism can be achieved via α7 nAChR by a mechanism that implicates Nrf2, HO-1, and PGC-1α. This signaling pathway could open a new strategy for the treatment of NDDs, such as Alzheimer's, characterized by a reduction of cholinergic markers. CONCLUSION: α7 nAChR signaling increases glial mitochondrial mass, both in vitro and in vivo, via HO-1 and PCG-1α. These effects could be of potential benefit in the context of NDDs. Antioxid. Redox Signal. 27, 93-105.


Subject(s)
Benzamides/administration & dosage , Bridged Bicyclo Compounds/administration & dosage , Heme Oxygenase-1/metabolism , Mitochondria/drug effects , Neuroglia/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Animals , Benzamides/pharmacology , Bridged Bicyclo Compounds/pharmacology , Cells, Cultured , Injections, Intraperitoneal , Mice , Mitochondria/pathology , NF-E2-Related Factor 2/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Organelle Biogenesis , Rats , alpha7 Nicotinic Acetylcholine Receptor/metabolism
11.
Biochim Biophys Acta ; 1861(12 Pt A): 1929-1941, 2016 12.
Article in English | MEDLINE | ID: mdl-27686967

ABSTRACT

New onset diabetes after transplantation (NODAT) is a metabolic disorder that affects 40% of patients on immunosuppressive agent (IA) treatment, such as rapamycin (also known as sirolimus). IAs negatively modulate insulin action in peripheral tissues including skeletal muscle, liver and white fat. However, the effects of IAs on insulin sensitivity and thermogenesis in brown adipose tissue (BAT) have not been investigated. We have analyzed the impact of rapamycin on insulin signaling, thermogenic gene-expression and mitochondrial respiration in BAT. Treatment of brown adipocytes with rapamycin for 16h significantly decreased insulin receptor substrate 1 (IRS1) protein expression and insulin-mediated protein kinase B (Akt) phosphorylation. Consequently, both insulin-induced glucose transporter 4 (GLUT4) translocation to the plasma membrane and glucose uptake were decreased. Early activation of the N-terminal Janus activated kinase (JNK) was also observed, thereby increasing IRS1 Ser 307 phosphorylation. These effects of rapamycin on insulin signaling in brown adipocytes were partly prevented by a JNK inhibitor. In vivo treatment of rats with rapamycin for three weeks abolished insulin-mediated Akt phosphorylation in BAT. Rapamycin also inhibited norepinephrine (NE)-induced lipolysis, the expression of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) and uncoupling protein (UCP)-1 in brown adipocytes. Importantly, basal mitochondrial respiration, proton leak and maximal respiratory capacity were significantly decreased in brown adipocytes treated with rapamycin. In conclusion, we demonstrate, for the first time the important role of brown adipocytes as target cells of rapamycin, suggesting that insulin resistance in BAT might play a major role in NODAT development.


Subject(s)
Adipocytes, Brown/drug effects , Glucose/metabolism , Insulin/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , Uncoupling Protein 1/metabolism , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Cell Respiration/drug effects , Gene Expression/drug effects , Glucose Transporter Type 4/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance/physiology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar , Thermogenesis/drug effects
12.
Cell Rep ; 16(9): 2387-98, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27545886

ABSTRACT

Mitochondrial respiratory chain (MRC) complexes I, III, and IV associate into a variety of supramolecular structures known as supercomplexes and respirasomes. While COX7A2L was originally described as a supercomplex-specific factor responsible for the dynamic association of complex IV into these structures to adapt MRC function to metabolic variations, this role has been disputed. Here, we further examine the functional significance of COX7A2L in the structural organization of the mammalian respiratory chain. As in the mouse, human COX7A2L binds primarily to free mitochondrial complex III and, to a minor extent, to complex IV to specifically promote the stabilization of the III2+IV supercomplex without affecting respirasome formation. Furthermore, COX7A2L does not affect the biogenesis, stabilization, and function of the individual oxidative phosphorylation complexes. These data show that independent regulatory mechanisms for the biogenesis and turnover of different MRC supercomplex structures co-exist.


Subject(s)
Electron Transport Complex III/metabolism , Electron Transport Complex IV/metabolism , Electron Transport Complex I/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Membranes/metabolism , Oxidative Phosphorylation , Animals , Electron Transport , Electron Transport Complex I/genetics , Electron Transport Complex III/genetics , Electron Transport Complex IV/genetics , Gene Expression , HEK293 Cells , HeLa Cells , Humans , Mice , Mitochondria, Heart/chemistry , Myocardium/cytology , Myocardium/metabolism , Protein Binding , Protein Stability
13.
Redox Biol ; 6: 183-197, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26233704

ABSTRACT

Redox biological reactions are now accepted to bear the Janus faceted feature of promoting both physiological signaling responses and pathophysiological cues. Endogenous antioxidant molecules participate in both scenarios. This review focuses on the role of crucial cellular nucleophiles, such as glutathione, and their capacity to interact with oxidants and to establish networks with other critical enzymes such as peroxiredoxins. We discuss the importance of the Nrf2-Keap1 pathway as an example of a transcriptional antioxidant response and we summarize transcriptional routes related to redox activation. As examples of pathophysiological cellular and tissular settings where antioxidant responses are major players we highlight endoplasmic reticulum stress and ischemia reperfusion. Topologically confined redox-mediated post-translational modifications of thiols are considered important molecular mechanisms mediating many antioxidant responses, whereas redox-sensitive microRNAs have emerged as key players in the posttranscriptional regulation of redox-mediated gene expression. Understanding such mechanisms may provide the basis for antioxidant-based therapeutic interventions in redox-related diseases.


Subject(s)
Cardiovascular Diseases/metabolism , Endoplasmic Reticulum Stress/genetics , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Reperfusion Injury/metabolism , Adaptation, Physiological , Animals , Antioxidants/metabolism , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Gene Expression Regulation , Glutathione/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidative Stress , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
14.
Free Radic Biol Med ; 88(Pt B): 427-438, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25843654

ABSTRACT

4-Hydroxy-2-nonenal (HNE) is a highly cytotoxic product of lipid peroxidation. Nevertheless, at low concentrations, it is able to mediate cell signaling and to activate protective pathways, including that of the transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2). In addition, HNE activates uncoupling proteins (UCPs), mitochondrial inner membrane proteins that mediate uncoupling of oxidative phosphorylation and have been proposed to protect against oxidative stress. It is not known, however, whether HNE might induce UCP expression via Nrf2 to cause mitochondrial uncoupling. We investigated the effects of HNE on UCP3 expression in mouse cardiomyocytes and the involvement of Nrf2. HNE induced the nuclear accumulation of Nrf2 and enhanced UCP3 expression, effects prevented by the antioxidant N-acetylcysteine. ChIP assays indicated that Nrf2 bound to the Ucp3 promoter after HNE treatment, increasing its expression. Cardiomyocytes treated with Nrf2- or UCP3-specific siRNA were less tolerant to HNE as reflected by increased cell death, and Nrf2 siRNA prevented HNE-induced UCP3 upregulation. The treatment with HNE greatly altered cardiomyocyte bioenergetics, increasing the proton leak across the inner mitochondrial membrane and severely decreasing the maximal respiratory capacity and the respiratory reserve capacity. These findings confirm that low HNE doses activate Nrf2 in cardiomyocytes and provide the first evidence of Nrf2 binding to the Ucp3 promoter in response to HNE, leading to increased protein expression. These results suggest that the upregulation of UCP3 mediated by Nrf2 in response to HNE might be important in the protection of the heart under conditions of oxidative stress such as ischemia-reperfusion.


Subject(s)
Aldehydes/metabolism , Ion Channels/biosynthesis , Mitochondrial Proteins/biosynthesis , Myocytes, Cardiac/metabolism , NF-E2-Related Factor 2/metabolism , Aldehydes/pharmacology , Animals , Cells, Cultured , Chromatin Immunoprecipitation , Energy Metabolism/physiology , Flow Cytometry , Immunoblotting , Mice , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Transfection , Uncoupling Protein 3 , Up-Regulation
15.
Mol Cell Endocrinol ; 404: 151-8, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25662278

ABSTRACT

TSH receptor (TSHR) is present in the thyroid and other tissues, as adipose tissue. In brown adipose tissue (BAT) TSH increases UCP1 expression and lipolysis. We have studied the regulation of Tshr mRNA expression and the effect of TSH on Ucp1 and Dio2 mRNA, on D2 activity and O2 consumption in rat brown adipocytes and the TSH signaling pathways. Tshr increased during brown adipocyte differentiation, was up-regulated by insulin and low TSH concentrations and down-regulated by high TSH concentrations, T3 and/or NE. TSH increased basal Ucp1 mRNA in a dose-dependent way acting synergistically with T3, while had no effect when NE was present. High TSH concentrations increased basal Dio2 mRNA (12-fold) and were synergistic with T3 (100-fold), but decreased Dio2 mRNA in T3+NE-treated cells. TSH increased D2 activities in T3-treated cells and inhibition of ERK pathway decreased the TSH effect by 55%. In T3+NE treated-cells TSH decreased D2 activity by 50%, in a dose-dependent manner. TSH activated Akt and Erk phosphorylation, while inhibition of PKA promoted Akt phosphorylation. TSH inhibited leptin mRNA. TSH increased O2 consumption by 20% and T3 enhanced its effect. Tshr is expressed in brown adipocytes and is regulated by insulin, TSH, T3 and NE. TSH increases basal and T3-stimulated Ucp1 and Dio2 expression and D2 activity only when T3 is present, but decreases Dio2 mRNA and D2 activity stimulated by NE+T3. TSH increases O2 consumption, confirming the role of TSH in the maintenance of thermogenesis.


Subject(s)
Adipocytes, Brown/cytology , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Thermogenesis/drug effects , Thyrotropin/pharmacology , Adipocytes, Brown/drug effects , Animals , Cell Differentiation , Cells, Cultured , Dose-Response Relationship, Drug , Insulin/pharmacology , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Ion Channels/genetics , Leptin/genetics , MAP Kinase Signaling System/drug effects , Mitochondrial Proteins/genetics , Oxygen Consumption/drug effects , Rats , Rats, Sprague-Dawley , Uncoupling Protein 1 , Iodothyronine Deiodinase Type II
16.
J Proteomics ; 113: 38-56, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25239759

ABSTRACT

We have analyzed the cellular pathways and metabolic adaptations that take place in primary skin fibroblasts from patients with mutations in BCS1L, a major genetic cause of mitochondrial complex III enzyme deficiency. Mutant fibroblasts exhibited low oxygen consumption rates and intracellular ATP levels, indicating that the main altered molecular event probably is a limited respiration-coupled ATP production through the OXPHOS system. Two-dimensional DIGE and MALDI-TOF/TOF mass spectrometry analyses unambiguously identified 39 proteins whose expression was significantly altered in complex III-deficient fibroblasts. Extensive statistical and cluster analyses revealed a protein profile characteristic for the BCS1L mutant fibroblasts that included alterations in energy metabolism, cell signaling and gene expression regulation, cytoskeleton formation and maintenance, and intracellular stress responses. The physiological validation of the predicted functional adaptations of human cultured fibroblasts to complex III deficiency confirmed the up-regulation of glycolytic enzyme activities and the accumulation of branched-chain among other amino acids, suggesting the activation of anaerobic glycolysis and cellular catabolic states, in particular protein catabolism, together with autophagy as adaptive responses to mitochondrial respiratory chain dysfunction and ATP deficiency. Our data point to an overall metabolic and genetic reprogramming that could contribute to explain the clinical manifestations of complex III deficiency in patients. BIOLOGICAL SIGNIFICANCE: Despite considerable knowledge about their genetic origins, the pathophysiological mechanisms that contribute to the clinical manifestations of mitochondrial disorders remain poorly understood. We have investigated the molecular pathways and metabolic adaptations that take place in primary skin fibroblasts from patients with mutations in the BCS1L gene, a primary cause of mitochondrial complex III enzyme deficiency. Two-dimensional DIGE together with MALDI-TOF/TOF mass spectrometry and physiological validation analyses revealed a significant metabolic and genetic reprogramming as an adaptive response to mitochondrial respiratory chain dysfunction. Our data provide information about specific protein targets that regulate the transmitochondrial functional responses to complex III deficiency, thereby opening new doors for future research.


Subject(s)
Electron Transport Complex III/deficiency , Fibroblasts/metabolism , Genetic Diseases, Inborn/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/deficiency , Proteomics , ATPases Associated with Diverse Cellular Activities , Cells, Cultured , Cytoskeleton/genetics , Cytoskeleton/metabolism , Cytoskeleton/pathology , Energy Metabolism/genetics , Female , Fibroblasts/pathology , Gene Expression Regulation/genetics , Genetic Diseases, Inborn/genetics , Humans , Male , Mitochondria/genetics , Mitochondria/pathology , Signal Transduction/genetics
17.
Free Radic Biol Med ; 61: 395-407, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23597505

ABSTRACT

Uncoupling protein 3 (UCP3) is a member of the mitochondrial inner membrane carrier superfamily that modulates energy efficiency by catalyzing proton conductance and thus decreasing the production of superoxide anion. However, its role during oxidative stress and the underlying regulatory and molecular mechanisms remain poorly understood. We sought to investigate how UCP3 expression is regulated by oxidative stress and to evaluate the putative antioxidant role of this protein. H2O2 treatment increased UCP3 expression and the nuclear accumulation of the transcription factor Nrf2 in C2C12 and HL-1 cells. Nrf2 siRNA prevented H2O2-induced UCP3 expression, increasing oxidative stress and cell death. ChIP assays identified an antioxidant-response element (ARE) within the UCP3 promoter that bound Nrf2 after exposure to H2O2. Luciferase reporter experiments confirmed increased ARE activity in H2O2-treated HL-1 cells. Importantly, H2O2 increased the UCP3-mediated proton leak, suggesting a role for this protein in attenuating ROS-induced damage. Nrf2 nuclear accumulation and increased UCP3 protein were also detected in intact mouse heart subjected to a condition known to increase ROS generation. This is the first study to demonstrate that H2O2 augments UCP3 expression and it provides the first evidence of Nrf2 binding to the UCP3 promoter in response to oxidative challenge. These findings suggest that UCP3 functions as a member of the cellular antioxidant defense system that protects against oxidative stress in vivo. In conclusion, we have identified a novel regulatory process induced by an oxidative insult whereby the expression of the mitochondrial protein UCP3 is driven by the Nrf2 transcription factor, which decreases ROS production and prevents cell death.


Subject(s)
Ion Channels/physiology , Mitochondrial Proteins/physiology , NF-E2-Related Factor 2/physiology , Oxidative Stress , Animals , Antioxidant Response Elements/physiology , Base Sequence , Cells, Cultured , Hydrogen Peroxide/pharmacology , Ion Channels/genetics , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Molecular Sequence Data , Myocardial Reperfusion , Uncoupling Protein 3
18.
Biochem Pharmacol ; 85(2): 245-56, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23142710

ABSTRACT

UNLABELLED: Non-steroidal anti-inflammatory drugs (NSAIDs) induce the shedding of L-selectin in human neutrophils through a mechanism still not well understood. In this work we studied both the functional effect of NSAIDs on the neutrophils/endothelial cells dynamic interaction, and the potential involvement of reactive oxygen species (ROS) in the NSAIDs-mediated down-regulation of L-selectin. When human neutrophils were incubated with diclofenac, a significant reduction in the number of cells that rolled on activated endothelial cells was observed. Different NSAIDs (flufenamic acid, meclofenamic acid, diclofenac, indomethacin, nimesulide, flurbiprofen, meloxicam, phenylbutazone, piroxicam, ketoprofen and aspirin) caused variable increase in neutrophil intracellular ROS concentration, which was inversely proportional to the change produced in L-selectin surface expression. Pre-incubation of neutrophils with superoxide dismutase, but not with catalase, showed both a significant protective effect on the L-selectin down-regulation induced by several NSAIDs and a diminished effect of diclofenac on neutrophil rolling. Interestingly, diclofenac and flufenamic acid but not piroxicam significantly increased the extracellular superoxide anion production by neutrophils, and inhibition of nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase activity with diphenyleneiodonium prevented the down-regulation of L-selectin by diclofenac. In accordance with these results, neutrophils from patients with chronic granulomatous disease, a hereditary disease in which neutrophils show a reduced capacity to form superoxide radicals, exhibited a lower down-regulation of L-selectin (IC50: 15.3 µg/ml) compared to normal controls (IC50: 5.6 µg/ml) in response to diclofenac. CONCLUSION: A group of NSAIDs is capable of interfering with the ability of neutrophils to interact with endothelial cells by triggering L-selectin-shedding through the NADPH-oxidase-dependent generation of superoxide anion at the plasma membrane.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Diclofenac/pharmacology , Down-Regulation/drug effects , Flufenamic Acid/pharmacology , L-Selectin/metabolism , Neutrophils/drug effects , Superoxides/metabolism , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM17 Protein , Adolescent , Animals , Cell Communication/drug effects , Cell Line, Transformed , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Cells, Cultured , Child , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/immunology , Granulomatous Disease, Chronic/metabolism , Granulomatous Disease, Chronic/pathology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice , Mice, Mutant Strains , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology
19.
Mitochondrion ; 12(1): 126-31, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21664300

ABSTRACT

The possible existence of a mitochondrially localized nitric oxide (NO) synthase (mtNOS) is controversial. To clarify this, we studied the ability of intact mitochondria to generate NO and the effect of mitochondrial NO on respiration. Respiratory rates and oxygen kinetics (P(50) values) were determined by high-resolution respirometry in skeletal-muscle mitochondria from control mice and mice injected with Escherichia coli lipopolysaccharide (LPS). In the presence of the NOS substrate L-arginine, mitochondria from LPS-treated mice had lower respiration rates and higher P(50) values than control animals. These effects were prevented by the NOS inhibitor L-NMMA. Our results suggest that mitochondrially derived NO is generated by an LPS-inducible NOS protein other than iNOS and modulates oxygen consumption in mouse skeletal muscle.


Subject(s)
Lipopolysaccharides/metabolism , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Nitric Oxide/metabolism , Animals , Cell Respiration , Escherichia coli/chemistry , Escherichia coli/immunology , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Oxygen/metabolism
20.
Cell Metab ; 14(6): 768-79, 2011 Dec 07.
Article in English | MEDLINE | ID: mdl-22100406

ABSTRACT

The fine regulation of mitochondrial function has proved to be an essential metabolic adaptation to fluctuations in oxygen availability. During hypoxia, cells activate an anaerobic switch that favors glycolysis and attenuates the mitochondrial activity. This switch involves the hypoxia-inducible transcription factor-1 (HIF-1). We have identified a HIF-1 target gene, the mitochondrial NDUFA4L2 (NADH dehydrogenase [ubiquinone] 1 alpha subcomplex, 4-like 2). Our results, obtained employing NDUFA4L2-silenced cells and NDUFA4L2 knockout murine embryonic fibroblasts, indicate that hypoxia-induced NDUFA4L2 attenuates mitochondrial oxygen consumption involving inhibition of Complex I activity, which limits the intracellular ROS production under low-oxygen conditions. Thus, reducing mitochondrial Complex I activity via NDUFA4L2 appears to be an essential element in the mitochondrial reprogramming induced by HIF-1.


Subject(s)
Electron Transport Complex I/antagonists & inhibitors , Enzyme Induction/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/physiopathology , Mitochondria/physiology , Oxygen Consumption/physiology , Animals , Apoptosis/physiology , Cell Line , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Fibroblasts , HeLa Cells , Humans , Hypoxia/enzymology , Membrane Potential, Mitochondrial , Mice , Mice, Knockout , Microarray Analysis , Rats , Reactive Oxygen Species/metabolism , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL
...