Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Physiol Rev ; 104(3): 1387-1408, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38451234

ABSTRACT

Effective data management is crucial for scientific integrity and reproducibility, a cornerstone of scientific progress. Well-organized and well-documented data enable validation and building on results. Data management encompasses activities including organization, documentation, storage, sharing, and preservation. Robust data management establishes credibility, fostering trust within the scientific community and benefiting researchers' careers. In experimental biomedicine, comprehensive data management is vital due to the typically intricate protocols, extensive metadata, and large datasets. Low-throughput experiments, in particular, require careful management to address variations and errors in protocols and raw data quality. Transparent and accountable research practices rely on accurate documentation of procedures, data collection, and analysis methods. Proper data management ensures long-term preservation and accessibility of valuable datasets. Well-managed data can be revisited, contributing to cumulative knowledge and potential new discoveries. Publicly funded research has an added responsibility for transparency, resource allocation, and avoiding redundancy. Meeting funding agency expectations increasingly requires rigorous methodologies, adherence to standards, comprehensive documentation, and widespread sharing of data, code, and other auxiliary resources. This review provides critical insights into raw and processed data, metadata, high-throughput versus low-throughput datasets, a common language for documentation, experimental and reporting guidelines, efficient data management systems, sharing practices, and relevant repositories. We systematically present available resources and optimal practices for wide use by experimental biomedical researchers.


Subject(s)
Biomedical Research , Data Management , Information Dissemination , Biomedical Research/standards , Biomedical Research/methods , Information Dissemination/methods , Humans , Animals , Data Management/methods
2.
Clin Sci (Lond) ; 137(17): 1347-1372, 2023 09 13.
Article in English | MEDLINE | ID: mdl-37565250

ABSTRACT

Maternal obesity (MO) is rising worldwide, affecting half of all gestations, constituting a possible risk-factor for some pregnancy-associated liver diseases (PALD) and hepatic diseases. PALD occur in approximately 3% of pregnancies and are characterized by maternal hepatic oxidative stress (OS) and mitochondrial dysfunction. Maternal hepatic disease increases maternal and fetal morbidity and mortality. Understanding the role of MO on liver function and pathophysiology could be crucial for better understanding the altered pathways leading to PALD and liver disease, possibly paving the way to prevention and adequate management of disease. We investigated specific hepatic metabolic alterations in mitochondria and oxidative stress during MO at late-gestation. Maternal hepatic tissue was collected at 90% gestation in Control and MO ewes (fed 150% of recommended nutrition starting 60 days before conception). Maternal hepatic redox state, mitochondrial respiratory chain (MRC), and OS markers were investigated. MO decreased MRC complex-II activity and its subunits SDHA and SDHB protein expression, increased complex-I and complex-IV activities despite reduced complex-IV subunit mtCO1 protein expression, and increased ATP synthase ATP5A subunit. Hepatic MO-metabolic remodeling was characterized by decreased adenine nucleotide translocator 1 and 2 (ANT-1/2) and voltage-dependent anion channel (VDAC) protein expression and protein kinase A (PKA) activity (P<0.01), and augmented NAD+/NADH ratio due to reduced NADH levels (P<0.01). MO showed an altered redox state with increased OS, increased lipid peroxidation (P<0.01), decreased GSH/GSSG ratio (P=0.005), increased superoxide dismutase (P=0.03) and decreased catalase (P=0.03) antioxidant enzymatic activities, lower catalase, glutathione peroxidase (GPX)-4 and glutathione reductase protein expression (P<0.05), and increased GPX-1 abundance (P=0.03). MO-related hepatic changes were more evident in the right lobe, corroborated by the integrative data analysis. Hepatic tissue from obese pregnant ewes showed alterations in the redox state, consistent with OS and MRC and metabolism remodeling. These are hallmarks of PALD and hepatic disease, supporting MO as a risk-factor and highlighting OS and mitochondrial dysfunction as mechanisms responsible for liver disease predisposition.


Subject(s)
Liver Diseases , NAD , Humans , Female , Pregnancy , Animals , Sheep , Catalase/metabolism , NAD/metabolism , Liver/metabolism , Oxidative Stress , Obesity/metabolism , Antioxidants/metabolism , Liver Diseases/metabolism , Superoxide Dismutase/metabolism , Glutathione/metabolism
3.
Int J Mol Sci ; 24(20)2023 Oct 14.
Article in English | MEDLINE | ID: mdl-37894873

ABSTRACT

Intra-uterine growth restriction (IUGR) is a common cause of fetal/neonatal morbidity and mortality and is associated with increased offspring predisposition for cardiovascular disease (CVD) development. Mitochondria are essential organelles in maintaining cardiac function, and thus, fetal cardiac mitochondria could be responsive to the IUGR environment. In this study, we investigated whether in utero fetal cardiac mitochondrial programming can be detectable in an early stage of IUGR pregnancy. Using a well-established nonhuman IUGR primate model, we induced IUGR by reducing by 30% the maternal diet (MNR), both in males (MNR-M) and in female (MNR-F) fetuses. Fetal cardiac left ventricle (LV) tissue and blood were collected at 90 days of gestation (0.5 gestation, 0.5 G). Blood biochemical parameters were determined and heart LV mitochondrial biology assessed. MNR fetus biochemical blood parameters confirm an early fetal response to MNR. In addition, we show that in utero cardiac mitochondrial MNR adaptations are already detectable at this early stage, in a sex-divergent way. MNR induced alterations in the cardiac gene expression of oxidative phosphorylation (OXPHOS) subunits (mostly for complex-I, III, and ATP synthase), along with increased protein content for complex-I, -III, and -IV subunits only for MNR-M in comparison with male controls, highlight the fetal cardiac sex-divergent response to MNR. At this fetal stage, no major alterations were detected in mitochondrial DNA copy number nor markers for oxidative stress. This study shows that in 90-day nonhuman primate fetuses, a 30% decrease in maternal nutrition generated early in utero adaptations in fetal blood biochemical parameters and sex-specific alterations in cardiac left ventricle gene and protein expression profiles, affecting predominantly OXPHOS subunits. Since the OXPHOS system is determinant for energy production in mitochondria, our findings suggest that these early IUGR-induced mitochondrial adaptations play a role in offspring's mitochondrial dysfunction and can increase predisposition to CVD in a sex-specific way.


Subject(s)
Cardiovascular Diseases , Fetal Development , Pregnancy , Humans , Animals , Male , Female , Fetus/metabolism , Fetal Growth Retardation/metabolism , Primates , Nutrients , Cardiovascular Diseases/metabolism
4.
Eur J Clin Invest ; 52(9): e13798, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35467758

ABSTRACT

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly progressing neurodegenerative disease that affects motor neurons. This disease is associated with oxidative stress especially in mutant superoxide dismutase 1 (mutSOD1) patients. However, less is known for the most prevalent sporadic ALS form, due to a lack of disease models. Here, we studied oxidative stress profiles in lymphoblasts from ALS patients with mutSOD1 or unknown (undSOD1) mutations. METHODS: mutSOD1 and undSOD1 lymphoblasts, as well as sex/age-matched controls (3/group) were obtained from Coriell and divided into 46 years-old-men (C1), 46 years-old-women (C2) or 26/27 years-old-men (C3) cohorts. Growth curves were performed, and several parameters associated with redox homeostasis were evaluated, including SOD activity and expression, general oxidative stress levels, lipid peroxidation, response to oxidative stimulus, glutathione redox cycle, catalase expression, and activity, and Nrf2 transcripts. Pooled (all cohorts) and paired (intra-cohort) statistical analyses were performed, followed by clustering and principal component analyses (PCA). RESULTS: Although a high heterogeneity among lymphoblast redox profiles was found between cohorts, clustering analysis based on 7 parameters with high chi-square ranking (total SOD activity, oxidative stress levels, catalase transcripts, SOD1 protein levels, metabolic response to mM concentrations of tert-butyl hydroperoxide, glutathione reductase activity, and Nrf2 transcript levels) provided a perfect cluster segregation between samples from healthy controls and ALS (undSOD1 and mutSOD1), also visualized in the PCA. CONCLUSIONS: Our results show distinct redox signatures in lymphoblasts from mutSOD1, undSOD1 and healthy controls that can be used as therapeutic targets for ALS drug development.


Subject(s)
Amyotrophic Lateral Sclerosis , Superoxide Dismutase-1 , Adult , Amyotrophic Lateral Sclerosis/genetics , Catalase/metabolism , Female , Humans , Male , Middle Aged , Mutation , NF-E2-Related Factor 2/genetics , Oxidation-Reduction , Superoxide Dismutase-1/genetics
5.
FASEB J ; 35(12): e22024, 2021 12.
Article in English | MEDLINE | ID: mdl-34751984

ABSTRACT

Alterations in mitochondrial dynamics, including their intracellular trafficking, are common early manifestations of neuronal degeneration. However, current methodologies used to study mitochondrial trafficking events rely on parameters that are primarily altered in later stages of neurodegeneration. Our objective was to establish a reliable applied statistical analysis to detect early alterations in neuronal mitochondrial trafficking. We propose a novel quantitative analysis of mitochondria trajectories based on innovative movement descriptors, including straightness, efficiency, anisotropy, and kurtosis. We evaluated time- and dose-dependent alterations in trajectory descriptors using biological data from differentiated SH-SY5Y cells treated with the mitochondrial toxicants 6-hydroxydopamine and rotenone. MitoTracker Red CMXRos-labelled mitochondria movement was analyzed by total internal reflection fluorescence microscopy followed by computational modelling to describe the process. Based on the aforementioned trajectory descriptors, this innovative analysis of mitochondria trajectories provides insights into mitochondrial movement characteristics and can be a consistent and sensitive method to detect alterations in mitochondrial trafficking occurring in the earliest time points of neurodegeneration.


Subject(s)
Mitochondria/pathology , Mitochondrial Dynamics , Neuroblastoma/pathology , Neurons/pathology , Oxidopamine/adverse effects , Rotenone/adverse effects , Adrenergic Agents/adverse effects , Cell Differentiation , Humans , Mitochondria/drug effects , Neuroblastoma/chemically induced , Neurons/drug effects , Uncoupling Agents/adverse effects
6.
Int J Mol Sci ; 23(6)2022 Mar 10.
Article in English | MEDLINE | ID: mdl-35328430

ABSTRACT

With the increase in life expectancy and consequent aging of the world's population, the prevalence of many neurodegenerative diseases is increasing, without concomitant improvement in diagnostics and therapeutics. These diseases share neuropathological hallmarks, including mitochondrial dysfunction. In fact, as mitochondrial alterations appear prior to neuronal cell death at an early phase of a disease's onset, the study and modulation of mitochondrial alterations have emerged as promising strategies to predict and prevent neurotoxicity and neuronal cell death before the onset of cell viability alterations. In this work, differentiated SH-SY5Y cells were treated with the mitochondrial-targeted neurotoxicants 6-hydroxydopamine and rotenone. These compounds were used at different concentrations and for different time points to understand the similarities and differences in their mechanisms of action. To accomplish this, data on mitochondrial parameters were acquired and analyzed using unsupervised (hierarchical clustering) and supervised (decision tree) machine learning methods. Both biochemical and computational analyses resulted in an evident distinction between the neurotoxic effects of 6-hydroxydopamine and rotenone, specifically for the highest concentrations of both compounds.


Subject(s)
Neuroprotective Agents , Neurotoxicity Syndromes , Apoptosis , Cell Death , Cell Line, Tumor , Cell Survival , Humans , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/etiology , Oxidopamine/toxicity , Rotenone/toxicity
7.
Eur J Clin Invest ; 51(2): e13375, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32780417

ABSTRACT

BACKGROUND: Changes in the nutritional environment in utero induced by maternal obesity (MO) lead to foetal metabolic dysfunction predisposing offspring to later-life metabolic diseases. Since mitochondria play a crucial role in hepatic metabolism and function, we hypothesized that MO prior to conception and throughout pregnancy programmes foetal sheep liver mitochondrial phenotype. MATERIAL AND METHODS: Ewes ate an obesogenic diet (150% requirements; MO), or 100% requirements (CTR), from 60 days prior to conception. Foetal livers were removed at 0.9 gestation. We measured foetal liver mitochondrial DNA copy number, activity of superoxide dismutase, cathepsins B and D and selected protein content, total phospholipids and cardiolipin and activity of mitochondrial respiratory chain complexes. RESULTS: A significant decrease in activities of mitochondrial complexes I, II-III and IV, but not aconitase, was observed in MO. In the antioxidant machinery, there was a significant increase in activity of total superoxide dismutase (SOD) and SOD2 in MO. However, no differences were found regarding autophagy-related protein content (p62, beclin-I, LC3-I, LC3-II and Lamp2A) and cathepsin B and D activities. A 21.5% decrease in total mitochondrial phospholipid was observed in MO. CONCLUSIONS: The data indicate that MO impairs foetal hepatic mitochondrial oxidative capacity and affects total mitochondrial phospholipid content. In addition, MO affects the regulation of foetal liver redox pathways, indicating metabolic adaptations to the higher foetal lipid environment. Consequences of in utero programming of foetal hepatic metabolism may persist and compromise mitochondrial bioenergetics in later life, and increase susceptibility to metabolic diseases.


Subject(s)
Autophagy/physiology , Electron Transport/physiology , Fetus/metabolism , Liver/metabolism , Mitochondria, Liver/metabolism , Obesity, Maternal/metabolism , Animals , Beclin-1/metabolism , Cardiolipins/metabolism , Cathepsin B/metabolism , Cathepsin D/metabolism , Female , Microtubule-Associated Proteins/metabolism , Phospholipids/metabolism , Pregnancy , Sheep , Superoxide Dismutase/metabolism
8.
Clin Sci (Lond) ; 135(9): 1103-1126, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33899910

ABSTRACT

Poor maternal nutrition in pregnancy affects fetal development, predisposing offspring to cardiometabolic diseases. The role of mitochondria during fetal development on later-life cardiac dysfunction caused by maternal nutrient reduction (MNR) remains unexplored. We hypothesized that MNR during gestation causes fetal cardiac bioenergetic deficits, compromising cardiac mitochondrial metabolism and reserve capacity. To enable human translation, we developed a primate baboon model (Papio spp.) of moderate MNR in which mothers receive 70% of control nutrition during pregnancy, resulting in intrauterine growth restriction (IUGR) offspring and later exhibiting myocardial remodeling and heart failure at human equivalent ∼25 years. Term control and MNR baboon offspring were necropsied following cesarean-section, and left ventricle (LV) samples were collected. MNR adversely impacted fetal cardiac LV mitochondria in a sex-dependent fashion. Increased maternal plasma aspartate aminotransferase, creatine phosphokinase (CPK), and elevated cortisol levels in MNR concomitant with decreased blood insulin in male fetal MNR were measured. MNR resulted in a two-fold increase in fetal LV mitochondrial DNA (mtDNA). MNR resulted in increased transcripts for several respiratory chain (NDUFB8, UQCRC1, and cytochrome c) and adenosine triphosphate (ATP) synthase proteins. However, MNR fetal LV mitochondrial complex I and complex II/III activities were significantly decreased, possibly contributing to the 73% decreased ATP content and increased lipid peroxidation. MNR fetal LV showed mitochondria with sparse and disarranged cristae dysmorphology. Conclusion: MNR disruption of fetal cardiac mitochondrial fitness likely contributes to the documented developmental programming of adult cardiac dysfunction, indicating a programmed mitochondrial inability to deliver sufficient energy to cardiac tissues as a chronic mechanism for later-life heart failure.


Subject(s)
Fetal Nutrition Disorders/metabolism , Maternal Nutritional Physiological Phenomena , Mitochondria, Heart/metabolism , Adenine Nucleotides/metabolism , Animals , Female , Fetal Nutrition Disorders/pathology , Mitochondria, Heart/ultrastructure , Oxidative Stress , Papio , Pregnancy
9.
Arch Toxicol ; 94(1): 257-271, 2020 01.
Article in English | MEDLINE | ID: mdl-31768571

ABSTRACT

Circadian rhythms disruption can be the cause of chronic diseases. External cues, including therapeutic drugs, have been shown to modulate peripheral-circadian clocks. Since anthracycline cardiotoxicity is associated with loss of mitochondrial function and metabolic remodeling, we investigated whether the energetic failure induced by sub-chronic doxorubicin (DOX) treatment in juvenile mice was associated with persistent disruption of circadian regulators. Juvenile C57BL/6J male mice were subjected to a sub-chronic DOX treatment (4 weekly injections of 5 mg/kg DOX) and several cardiac parameters, as well as circadian-gene expression and acetylation patterns, were analyzed after 6 weeks of recovery time. Complementary experiments were performed with Mouse Embryonic Fibroblasts (MEFs) and Human Embryonic Kidney 293 cells. DOX-treated juvenile mice showed cardiotoxicity markers and persistent alterations of transcriptional- and signaling cardiac circadian homeostasis. The results showed a delayed influence of DOX on gene expression, accompanied by changes in SIRT1-mediated cyclic deacetylation. The mechanism behind DOX interference with the circadian clock was further studied in vitro, in which were observed alterations of circadian-gene expression and increased BMAL1 SIRT1-mediated deacetylation. In conclusion, DOX treatment in juvenile mice resulted in disruption of oscillatory molecular mechanisms including gene expression and acetylation profiles.


Subject(s)
Circadian Rhythm/drug effects , Doxorubicin/adverse effects , Heart Diseases/chemically induced , Acetylation/drug effects , Animals , Antibiotics, Antineoplastic/adverse effects , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Fibroblasts/drug effects , Fibroblasts/physiology , Gene Expression Regulation/genetics , HEK293 Cells , Heart Diseases/physiopathology , Homeostasis/drug effects , Humans , Male , Mice, Inbred C57BL , Sirtuin 1/metabolism , Sirtuins/genetics , Toxicity Tests, Subchronic
10.
Int J Mol Sci ; 20(19)2019 Oct 03.
Article in English | MEDLINE | ID: mdl-31623305

ABSTRACT

Worldwide, several million workers are employed in the various chromium (Cr) industries. These workers may suffer from a variety of adverse health effects produced by dusts, mists and fumes containing Cr in the hexavalent oxidation state, Cr(VI). Of major importance, occupational exposure to Cr(VI) compounds has been firmly associated with the development of lung cancer. Counterintuitively, Cr(VI) is mostly unreactive towards most biomolecules, including nucleic acids. However, its intracellular reduction produces several species that react extensively with biomolecules. The diversity and chemical versatility of these species add great complexity to the study of the molecular mechanisms underlying Cr(VI) toxicity and carcinogenicity. As a consequence, these mechanisms are still poorly understood, in spite of intensive research efforts. Here, we discuss the impact of Cr(VI) on the stress response-an intricate cellular system against proteotoxic stress which is increasingly viewed as playing a critical role in carcinogenesis. This discussion is preceded by information regarding applications, chemical properties and adverse health effects of Cr(VI). A summary of our current understanding of cancer initiation, promotion and progression is also provided, followed by a brief description of the stress response and its links to cancer and by an overview of potential molecular mechanisms of Cr(VI) carcinogenicity.


Subject(s)
Carcinogens, Environmental/pharmacology , Chromium/pharmacology , Stress, Physiological/drug effects , Carcinogens, Environmental/toxicity , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chromium/toxicity , DNA Damage , Humans , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Oxidative Stress/drug effects , Oxidative Stress/genetics
11.
J Neurosci ; 37(10): 2776-2794, 2017 03 08.
Article in English | MEDLINE | ID: mdl-28123081

ABSTRACT

Transcriptional deregulation and changes in mitochondrial bioenergetics, including pyruvate dehydrogenase (PDH) dysfunction, have been described in Huntington's disease (HD). We showed previously that the histone deacetylase inhibitors (HDACIs) trichostatin A and sodium butyrate (SB) ameliorate mitochondrial function in cells expressing mutant huntingtin. In this work, we investigated the effect of HDACIs on the regulation of PDH activity in striatal cells derived from HD knock-in mice and YAC128 mice. Mutant cells exhibited decreased PDH activity and increased PDH E1alpha phosphorylation/inactivation, accompanied by enhanced protein levels of PDH kinases 1 and 3 (PDK1 and PDK3). Exposure to dichloroacetate, an inhibitor of PDKs, increased mitochondrial respiration and decreased production of reactive oxygen species in mutant cells, emphasizing PDH as an interesting therapeutic target in HD. Treatment with SB and sodium phenylbutyrate, another HDACI, recovered cell viability and overall mitochondrial metabolism in mutant cells. Exposure to SB also suppressed hypoxia-inducible factor-1 (HIF-1α) stabilization and decreased the transcription of the two most abundant PDK isoforms, PDK2 and PDK3, culminating in increased PDH activation in mutant cells. Concordantly, PDK3 knockdown improved mitochondrial function, emphasizing the role of PDK3 inactivation on the positive effects achieved by SB treatment. YAC128 mouse brain presented higher mRNA levels of PDK1-3 and PDH phosphorylation and decreased energy levels that were significantly ameliorated after SB treatment. Furthermore, enhanced motor learning and coordination were observed in SB-treated YAC128 mice. These results suggest that HDACIs, particularly SB, promote the activity of PDH in the HD brain, helping to counteract HD-related deficits in mitochondrial bioenergetics and motor function.SIGNIFICANCE STATEMENT The present work provides a better understanding of mitochondrial dysfunction in Huntington's disease (HD) by showing that the pyruvate dehydrogenase (PDH) complex is a promising therapeutic target. In particular, the histone deacetylase inhibitor sodium butyrate (SB) may indirectly (through reduced hypoxia-inducible factor 1 alpha stabilization) decrease the expression of the most abundant PDH kinase isoforms (e.g., PDK3), ameliorating PDH activity and mitochondrial metabolism and further affecting motor behavior in HD mice, thus constituting a promising agent for HD neuroprotective treatment.


Subject(s)
Histone Deacetylase Inhibitors/administration & dosage , Huntington Disease/drug therapy , Huntington Disease/metabolism , Neurons/enzymology , Neuroprotective Agents/administration & dosage , Pyruvate Dehydrogenase Complex/metabolism , Animals , Cells, Cultured , Enzyme Activation/drug effects , Male , Mice , Mitochondria/drug effects , Mitochondria/enzymology , Neurons/drug effects , Treatment Outcome
12.
Toxicol Appl Pharmacol ; 348: 1-13, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29653124

ABSTRACT

Doxorubicin (DOX) is a widely used anticancer drug that could be even more effective if its clinical dosage was not limited because of delayed cardiotoxicity. Beating stem cell-derived cardiomyocytes are a preferred in vitro model to further uncover the mechanisms of DOX-induced cardiotoxicity. Our objective was to use cultured induced-pluripotent stem cell(iPSC)-derived mouse cardiomyocytes (Cor.At) to investigate the effects of DOX on cell and mitochondrial metabolism, as well as on stress responses. Non-proliferating and beating Cor.At cells were treated with 0.5 or 1 µM DOX for 24 h, and morphological, functional and biochemical changes associated with mitochondrial bioenergetics, DNA-damage response and apoptosis were measured. Both DOX concentrations decreased ATP levels and SOD2 protein levels and induced p53-dependent caspase activation. However, differential effects were observed for the two DOX concentrations. The highest concentration induced a high degree of apoptosis, with increased nuclear apoptotic morphology, PARP-1 cleavage and decrease of some OXPHOS protein subunits. At the lowest concentration, DOX increased the expression of p53 target transcripts associated with mitochondria-dependent apoptosis and decreased transcripts related with DNA-damage response and glycolysis. Interestingly, cells treated with 0.5 µM DOX presented an increase in PDK4 transcript levels, accompanied by an increase in phospho-PDH and decreased PDH activity. This was accompanied by an apparent decrease in basal and maximal oxygen consumption rates (OCR) and in basal extracellular acidification rate (ECAR). Cells pre-treated with the PDK inhibitor dichloroacetate (DCA), with the aim of restoring PDH activity, partially recovered OCR and ECAR. The results suggest that the higher DOX concentration mainly induces p53-dependent apoptosis, whereas for the lower DOX concentration the cardiotoxic effects involve bioenergetic failure, unveiling PDH as a possible therapeutic target to decrease DOX cardiotoxicity.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , Energy Metabolism/drug effects , Heart Diseases/chemically induced , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Tumor Suppressor Protein p53/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Cardiotoxicity , Cell Differentiation , Cell Line , DNA Damage , Dose-Response Relationship, Drug , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/pathology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mice , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Poly (ADP-Ribose) Polymerase-1/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Pyruvate Dehydrogenase Complex/metabolism , Superoxide Dismutase/metabolism , Tumor Suppressor Protein p53/genetics
13.
Biometals ; 31(4): 477-487, 2018 08.
Article in English | MEDLINE | ID: mdl-29549560

ABSTRACT

Exposure to hexavalent chromium [Cr(VI)], a lung carcinogen, triggers several types of cellular stresses, namely oxidative, genotoxic and proteotoxic stresses. Given the evolutionary character of carcinogenesis, it is tempting to speculate that cells that survive the stresses produced by this carcinogen become more resistant to subsequent stresses, namely those encountered during neoplastic transformation. To test this hypothesis, we determined whether pre-incubation with Cr(VI) increased the resistance of human bronchial epithelial cells (BEAS-2B cells) to the antiproliferative action of acute thermal shock, used here as a model for stress. In line with the proposed hypothesis, it was observed that, at mildly cytotoxic concentrations, Cr(VI) attenuated the antiproliferative effects of both cold and heat shock. Mechanistically, Cr(VI) interfered with the expression of two components of the stress response pathway: heat shock proteins Hsp72 and Hsp90α. Specifically, Cr(VI) significantly depleted the mRNA levels of the former and the protein levels of the latter. Significantly, these two proteins are members of heat shock protein (Hsp) families (Hsp70 and Hsp90, respectively) that have been implicated in carcinogenesis. Thus, our results confirm and extend previous studies showing the capacity of Cr(VI) to interfere with the expression of stress response components.


Subject(s)
Carcinogens/toxicity , Cell Proliferation/drug effects , Chromium/toxicity , Lung Neoplasms/genetics , Bronchi/drug effects , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogens/pharmacology , Cell Line , Chromium/pharmacology , Epithelial Cells/drug effects , Heat-Shock Proteins/genetics , Heat-Shock Response/drug effects , Humans , Lung/drug effects , Lung/pathology , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Reactive Oxygen Species/chemistry
14.
Biochim Biophys Acta Mol Basis Dis ; 1863(11): 2904-2923, 2017 11.
Article in English | MEDLINE | ID: mdl-28760703

ABSTRACT

Doxorubicin (DOX) is one of the most widely used anti-neoplastic agents. However, treatment with DOX is associated with cumulative cardiotoxicity inducing progressive cardiomyocyte death. Sirtuin 3 (Sirt3), a mitochondrial deacetylase, regulates the activity of proteins involved in apoptosis, autophagy and metabolism. Our hypothesis is that pharmacological modulation by berberine (BER) pre-conditioning of Sirt3 protein levels decreases DOX-induced cardiotoxicity. Our results showed that DOX induces cell death in all experimental groups. Increase in Sirt3 content by transfection-mediated overexpression decreased DOX cytotoxicity, mostly by maintaining mitochondrial network integrity and reducing oxidative stress. p53 was upregulated by DOX, and appeared to be a direct target of Sirt3, suggesting that Sirt3-mediated protection against cell death could be related to this protein. BER pre-treatment increased Sirt3 and Sirt1 protein levels in the presence of DOX and inhibited DOX-induced caspase 9 and 3-like activation. Moreover, BER modulated autophagy in DOX-treated H9c2 cardiomyoblasts. Interestingly, mitochondrial biogenesis markers were upregulated in in BER/DOX-treated cells. Sirt3 over-expression contributes to decrease DOX cytotoxicity on H9c2 cardiomyoblasts, while BER can be used as a modulator of Sirtuin function and cell quality control pathways to decrease DOX toxicity.


Subject(s)
Berberine/pharmacology , Cardiotonic Agents/pharmacology , Doxorubicin/adverse effects , Myoblasts, Cardiac/enzymology , Oxidative Stress/drug effects , Sirtuin 3/metabolism , Cell Line , Doxorubicin/pharmacology , Humans , Muscle Proteins/metabolism , Myoblasts, Cardiac/pathology
15.
Eur J Clin Invest ; 46(3): 285-98, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26782788

ABSTRACT

BACKGROUND: The employment of dietary strategies such as ketogenic diets, which force cells to alter their energy source, has shown efficacy in the treatment of several diseases. Ketogenic diets are composed of high fat, moderate protein and low carbohydrates, which favour mitochondrial respiration rather than glycolysis for energy metabolism. DESIGN: This review focuses on how oncological, neurological and mitochondrial disorders have been targeted by ketogenic diets, their metabolic effects, and the possible mechanisms of action on mitochondrial energy homeostasis. The beneficial and adverse effects of the ketogenic diets are also highlighted. RESULTS AND CONCLUSIONS: Although the full mechanism by which ketogenic diets improve oncological and neurological conditions still remains to be elucidated, their clinical efficacy has attracted many new followers, and ketogenic diets can be a good option as a co-adjuvant therapy, depending on the situation and the extent of the disease.


Subject(s)
Diet, Ketogenic/methods , Epilepsy/diet therapy , Mitochondrial Diseases/diet therapy , Neoplasms/diet therapy , Energy Metabolism , Glycolysis , Humans , Mitochondria/metabolism , Nervous System Diseases/diet therapy
16.
Biochim Biophys Acta ; 1842(12 Pt A): 2468-78, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25283819

ABSTRACT

The cardiotoxicity induced by the anti-cancer doxorubicin involves increased oxidative stress, disruption of calcium homeostasis and activation of cardiomyocyte death. Nevertheless, antioxidants and caspase inhibitors often show little efficacy in preventing cell death. We hypothesize that a caspase-independent cell death mechanism with the release of the apoptosis-inducing factor from mitochondria is involved in doxorubicin toxicity. To test the hypothesis, H9c2 cardiomyoblasts were used as model for cardiac cells. Our results demonstrate that z-VAD-fmk, a pan-caspase inhibitor, does not prevent doxorubicin toxicity in this cell line. Doxorubicin treatment results in AIF translocation to the nuclei, as confirmed by Western Blotting of cell fractions and confocal microscopy. Also, doxorubicin treatment of H9c2 cardiomyoblasts resulted in the appearance of 50kbp DNA fragments, a hallmark of apoptosis-inducing factor nuclear effects. Apoptosis-inducing factor knockdown using a small-interfering RNA approach in H9c2 cells resulted in a reduction of doxorubicin toxicity, including decreased p53 activation and poly-ADP-ribose-polymerase cleavage. Among the proteases that could be responsible for apoptosis-inducing factor cleavage, doxorubicin decreased calpain activity but increased cathepsin B activation, with inhibition of the latter partly decreasing doxorubicin toxicity. Altogether, the results support that apoptosis-inducing factor release is involved in doxorubicin-induced H9c2 cell death, which explains the limited ability of caspase inhibitors to prevent toxicity.


Subject(s)
Apoptosis Inducing Factor/metabolism , Apoptosis/drug effects , Doxorubicin/pharmacology , Myocytes, Cardiac/drug effects , Active Transport, Cell Nucleus/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/genetics , Apoptosis Inducing Factor/genetics , Blotting, Western , Caspase Inhibitors/pharmacology , Caspases/metabolism , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cell Survival/genetics , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Microscopy, Confocal , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , RNA Interference , Rats , Time Factors
17.
Eur J Clin Invest ; 45 Suppl 1: 44-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25524586

ABSTRACT

BACKGROUND: Abnormal mitochondrial function has long been associated with the development and the progression of cancer. Multiple defects in the mitochondrial genome have been reported for various cancers, however the often disregarded mitochondrial epigenetic landscape provides an additional source of deregulation that may contribute to carcinogenesis. DESIGN: This article reviews the current understanding of mitochondrial epigenetics and how it may relate to cancer progression and development. Relevant studies were found through electronic databases (Web of Science and PubMed). RESULTS AND CONCLUSIONS: The remarkably unexplored field of mitochondrial epigenetics has the potential to shed light on several cancer-related mitochondrial abnormalities. More studies using innovative, genome-wide sequencing technologies are highly warranted to assess whether and how altered mtDNA methylation patterns affect cancer initiation and progression.


Subject(s)
DNA, Mitochondrial/genetics , Epigenesis, Genetic , Mitochondria/genetics , Neoplasms/genetics , DNA Methylation , DNA, Mitochondrial/metabolism , Humans , Mitochondria/metabolism , Neoplasms/metabolism
18.
Biochim Biophys Acta Mol Basis Dis ; 1871(1): 167495, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39241844

ABSTRACT

Aging and lifestyle-related diseases, such as cardiovascular diseases, diabetes, cancer, and neurodegenerative disorders, are major global health challenges. These conditions are often linked to redox imbalances, where cells fail to regulate reactive redox species (RRS), leading to oxidative stress and cellular damage. Although antioxidants are known to neutralize harmful RRS, their clinical efficacy remains inconsistent. One reason for this inconsistency is the inadequacy of current in vitro models to accurately mimic in vivo redox conditions. This study addresses the gap in understanding the heterogeneity of redox responses in cells by using metabolically primed human dermal fibroblasts (NHDF), a model relevant for precision mitochondrial medicine. We investigated how metabolic priming, which enhances mitochondrial bioenergetics, influences redox responses to oxidative stress induced by hydrogen peroxide (H2O2) and tert-butyl hydroperoxide (tBHP). Specifically, we explored the impact of cell population density and cell cycle distribution on redox dynamics. Our findings indicate that NHDF cells cultured in oxidative phosphorylation-promoting medium (OXm) exhibit significantly larger variability in oxidative stress responses. This variability suggests that enhanced mitochondrial bioenergetics necessitates a constant regulation of the cellular redox machinery, potentially leading to heterogeneous responses. Additionally, cells grown in OXm showed increased mitochondrial polarization and a lower percentage of cells in the G2/M phase, contributing to the observed heterogeneity. Key factors influencing this variability included cell population density at the time of oxidant exposure and fluctuations in cell cycle distribution. Our results highlight the necessity of employing multiple oxidants in metabolic priming models to achieve a comprehensive understanding of oxidative stress responses and redox regulation mechanisms. Furthermore, the study emphasizes the need to refine in vitro models to better reflect in vivo conditions, which is crucial for the development of effective redox-based therapeutic strategies.

19.
Cells ; 13(3)2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38334639

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by the progressive loss of motor neurons, for which current treatment options are limited. Recent studies have shed light on the role of mitochondria in ALS pathogenesis, making them an attractive therapeutic intervention target. This review contains a very comprehensive critical description of the involvement of mitochondria and mitochondria-mediated mechanisms in ALS. The review covers several key areas related to mitochondria in ALS, including impaired mitochondrial function, mitochondrial bioenergetics, reactive oxygen species, metabolic processes and energy metabolism, mitochondrial dynamics, turnover, autophagy and mitophagy, impaired mitochondrial transport, and apoptosis. This review also highlights preclinical and clinical studies that have investigated various mitochondria-targeted therapies for ALS treatment. These include strategies to improve mitochondrial function, such as the use of dichloroacetate, ketogenic and high-fat diets, acetyl-carnitine, and mitochondria-targeted antioxidants. Additionally, antiapoptotic agents, like the mPTP-targeting agents minocycline and rasagiline, are discussed. The paper aims to contribute to the identification of effective mitochondria-targeted therapies for ALS treatment by synthesizing the current understanding of the role of mitochondria in ALS pathogenesis and reviewing potential convergent therapeutic interventions. The complex interplay between mitochondria and the pathogenic mechanisms of ALS holds promise for the development of novel treatment strategies to combat this devastating disease.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Humans , Amyotrophic Lateral Sclerosis/metabolism , Neurodegenerative Diseases/metabolism , Mitochondria/metabolism , Motor Neurons/pathology , Apoptosis
20.
Bioorg Med Chem ; 21(23): 7239-49, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24156937

ABSTRACT

Triterpenoids are a large class of naturally occurring compounds, and some potentially interesting as anticancer agents have been found to target mitochondria. The objective of the present work was to investigate the mechanisms of mitochondrial toxicity induced by novel dimethylaminopyridine (DMAP) derivatives of pentacyclic triterpenes, which were previously shown to inhibit the growth of melanoma cells in vitro. MCF-7, Hs 578T and BJ cell lines, as well as isolated hepatic mitochondria, were used to investigate direct mitochondrial effects. On isolated mitochondrial hepatic fractions, respiratory parameters, mitochondrial transmembrane electric potential, induction of the mitochondrial permeability transition (MPT) pore and ion transport-dependent osmotic swelling were measured. Our results indicate that the DMAP triterpenoid derivatives lead to fragmentation and depolarization of the mitochondrial network in situ, and to inhibition of uncoupled respiration, induction of the permeability transition pore and depolarization of isolated hepatic mitochondria. The results show that mitochondrial toxicity is an important component of the biological interaction of DMAP derivatives, which can explain the effects observed in cancer cells.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Mitochondria, Liver/drug effects , Permeability/drug effects , Triterpenes/chemistry , Triterpenes/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Cell Respiration/drug effects , Female , Humans , Male , Melanoma/drug therapy , Melanoma/metabolism , Membrane Potentials/drug effects , Mitochondria, Liver/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Pyridines/chemistry , Pyridines/pharmacology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL