Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 88
1.
Cell Metab ; 33(5): 1013-1026.e6, 2021 05 04.
Article En | MEDLINE | ID: mdl-33609439

Mitochondrial respiration is critical for cell proliferation. In addition to producing ATP, respiration generates biosynthetic precursors, such as aspartate, an essential substrate for nucleotide synthesis. Here, we show that in addition to depleting intracellular aspartate, electron transport chain (ETC) inhibition depletes aspartate-derived asparagine, increases ATF4 levels, and impairs mTOR complex I (mTORC1) activity. Exogenous asparagine restores proliferation, ATF4 and mTORC1 activities, and mTORC1-dependent nucleotide synthesis in the context of ETC inhibition, suggesting that asparagine communicates active respiration to ATF4 and mTORC1. Finally, we show that combination of the ETC inhibitor metformin, which limits tumor asparagine synthesis, and either asparaginase or dietary asparagine restriction, which limit tumor asparagine consumption, effectively impairs tumor growth in multiple mouse models of cancer. Because environmental asparagine is sufficient to restore tumor growth in the context of respiration impairment, our findings suggest that asparagine synthesis is a fundamental purpose of tumor mitochondrial respiration, which can be harnessed for therapeutic benefit to cancer patients.


Activating Transcription Factor 4/metabolism , Asparagine/metabolism , Mitochondria/metabolism , Animals , Asparagine/pharmacology , Aspartic Acid/deficiency , Aspartic Acid/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Diet/veterinary , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Electron Transport Chain Complex Proteins/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Mice , Mice, Inbred NOD , Mitochondria/drug effects , Neoplasms/drug therapy , Neoplasms/mortality , Neoplasms/pathology , Nucleotides/metabolism , Survival Rate
2.
Environ Toxicol Pharmacol ; 84: 103614, 2021 May.
Article En | MEDLINE | ID: mdl-33592315

Ethylene bisdithiocarbamate pesticides, including Mancozeb (MZ), are used as fungicides. Effects of MZ on apoptosis induction and mitochondrial activity of HT-29 colon cells were investigated. MZ exposed cells exhibited blebbing and cellular membrane disruption in scanning electron micrographs. Positive fluorescent staining with Annexin V at doses of 60-140 µM supports apoptosis as the mechanism of cell death. Activity of all electron transport chain complexes were evaluated. Mitochondrial Complex I activity was decreased in 100 µM treated cells. Mitochondrial Complex III activity was decreased in 60 and 100 µM MZ treated cells. Mitochondrial Complex II and Complex IV activities were decreased in cells treated with 60, 100, and 140 µM. Cells treated with 60 µM exhibited a decrease in Complex V enzyme activity. It is concluded that MZ exposure inhibits all mitochondrial complexes of HT-29 cells and that positive fluorescent microscopy and blebbing support previous studies of cell death via apoptosis.


Electron Transport Chain Complex Proteins/antagonists & inhibitors , Fungicides, Industrial/toxicity , Maneb/toxicity , Mitochondria/drug effects , Zineb/toxicity , Apoptosis/drug effects , Cell Survival/drug effects , Colon/cytology , Electron Transport Chain Complex Proteins/metabolism , HT29 Cells , Humans , Mitochondria/metabolism
3.
Arch Toxicol ; 95(2): 591-615, 2021 02.
Article En | MEDLINE | ID: mdl-33512557

Inhibition of complex I of the mitochondrial respiratory chain (cI) by rotenone and methyl-phenylpyridinium (MPP +) leads to the degeneration of dopaminergic neurons in man and rodents. To formally describe this mechanism of toxicity, an adverse outcome pathway (AOP:3) has been developed that implies that any inhibitor of cI, or possibly of other parts of the respiratory chain, would have the potential to trigger parkinsonian motor deficits. We used here 21 pesticides, all of which are described in the literature as mitochondrial inhibitors, to study the general applicability of AOP:3 or of in vitro assays that are assessing its activation. Five cI, three complex II (cII), and five complex III (cIII) inhibitors were characterized in detail in human dopaminergic neuronal cell cultures. The NeuriTox assay, examining neurite damage in LUHMES cells, was used as in vitro proxy of the adverse outcome (AO), i.e., of dopaminergic neurodegeneration. This test provided data on whether test compounds were unspecific cytotoxicants or specifically neurotoxic, and it yielded potency data with respect to neurite degeneration. The pesticide panel was also examined in assays for the sequential key events (KE) leading to the AO, i.e., mitochondrial respiratory chain inhibition, mitochondrial dysfunction, and disturbed proteostasis. Data from KE assays were compared to the NeuriTox data (AO). The cII-inhibitory pesticides tested here did not appear to trigger the AOP:3 at all. Some of the cI/cIII inhibitors showed a consistent AOP activation response in all assays, while others did not. In general, there was a clear hierarchy of assay sensitivity: changes of gene expression (biomarker of neuronal stress) correlated well with NeuriTox data; mitochondrial failure (measured both by a mitochondrial membrane potential-sensitive dye and a respirometric assay) was about 10-260 times more sensitive than neurite damage (AO); cI/cIII activity was sometimes affected at > 1000 times lower concentrations than the neurites. These data suggest that the use of AOP:3 for hazard assessment has a number of caveats: (i) specific parkinsonian neurodegeneration cannot be easily predicted from assays of mitochondrial dysfunction; (ii) deriving a point-of-departure for risk assessment from early KE assays may overestimate toxicant potency.


Electron Transport Chain Complex Proteins/antagonists & inhibitors , Electron Transport/drug effects , Enzyme Inhibitors/toxicity , Mitochondria/drug effects , Mitochondria/metabolism , Pesticides/toxicity , Biomarkers , Cell Line , Cell Line, Tumor , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Drug-Related Side Effects and Adverse Reactions , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex II/antagonists & inhibitors , Electron Transport Complex III/antagonists & inhibitors , Humans , Proteostasis/drug effects , Risk Assessment , Transcriptome
4.
Arch Toxicol ; 94(8): 2707-2729, 2020 08.
Article En | MEDLINE | ID: mdl-32607615

Evidence is mounting for the central role of mitochondrial dysfunction in several pathologies including metabolic diseases, accelerated ageing, neurodegenerative diseases and in certain xenobiotic-induced organ toxicity. Assessing mitochondrial perturbations is not trivial and the outcomes of such investigations are dependent on the cell types used and assays employed. Here we systematically investigated the effect of electron transport chain (ETC) inhibitors on multiple mitochondrial-related parameters in two human cell types, HepG2 and RPTEC/TERT1. Cells were exposed to a broad range of concentrations of 20 ETC-inhibiting agrochemicals and capsaicin, consisting of inhibitors of NADH dehydrogenase (Complex I, CI), succinate dehydrogenase (Complex II, CII) and cytochrome bc1 complex (Complex III, CIII). A battery of tests was utilised, including viability assays, lactate production, mitochondrial membrane potential (MMP) and the Seahorse bioanalyser, which simultaneously measures extracellular acidification rate [ECAR] and oxygen consumption rate [OCR]. CI inhibitors caused a potent decrease in OCR, decreased mitochondrial membrane potential, increased ECAR and increased lactate production in both cell types. Twenty-fourhour exposure to CI inhibitors decreased viability of RPTEC/TERT1 cells and 3D spheroid-cultured HepG2 cells in the presence of glucose. CI inhibitors decreased 2D HepG2 viability only in the absence of glucose. CII inhibitors had no notable effects in intact cells up to 10 µM. CIII inhibitors had similar effects to the CI inhibitors. Antimycin A was the most potent CIII inhibitor, with activity in the nanomolar range. The proposed CIII inhibitor cyazofamid demonstrated a mitochondrial uncoupling signal in both cell types. The study presents a comprehensive example of a mitochondrial assessment workflow and establishes measurable key events of ETC inhibition.


Agrochemicals/toxicity , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Energy Metabolism/drug effects , Hepatocytes/drug effects , Kidney Tubules, Proximal/drug effects , Mitochondria, Liver/drug effects , Uncoupling Agents/toxicity , Cell Survival/drug effects , Dose-Response Relationship, Drug , Electron Transport Chain Complex Proteins/metabolism , Hep G2 Cells , Hepatocytes/enzymology , Hepatocytes/pathology , Humans , Kidney Tubules, Proximal/enzymology , Kidney Tubules, Proximal/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/enzymology , Mitochondria, Liver/pathology , Oxygen Consumption/drug effects
5.
Int J Mol Sci ; 21(5)2020 Feb 25.
Article En | MEDLINE | ID: mdl-32106430

: Mitochondrial respiratory chain supercomplexes (RCS), particularly, the respirasome, which contains complexes I, III, and IV, have been suggested to participate in facilitating electron transport, reducing the production of reactive oxygen species (ROS), and maintaining the structural integrity of individual electron transport chain (ETC) complexes. Disassembly of the RCS has been observed in Barth syndrome, neurodegenerative and cardiovascular diseases, diabetes mellitus, and aging. However, the physiological role of RCS in high energy-demanding tissues such as the heart remains unknown. This study elucidates the relationship between RCS assembly and cardiac function. Adult male Sprague Dawley rats underwent Langendorff retrograde perfusion in the presence and absence of ethanol, isopropanol, or rotenone (an ETC complex I inhibitor). We found that ethanol had no effects on cardiac function, whereas rotenone reduced heart contractility, which was not recovered when rotenone was excluded from the perfusion medium. Blue native polyacrylamide gel electrophoresis revealed significant reductions of respirasome levels in ethanol- or rotenone-treated groups compared to the control group. In addition, rotenone significantly increased while ethanol had no effect on mitochondrial ROS production. In isolated intact mitochondria in vitro, ethanol did not affect respirasome assembly; however, acetaldehyde, a byproduct of ethanol metabolism, induced dissociation of respirasome. Isopropanol, a secondary alcohol which was used as an alternative compound, had effects similar to ethanol on heart function, respirasome levels, and ROS production. In conclusion, ethanol and isopropanol reduced respirasome levels without any noticeable effect on cardiac parameters, and cardiac function is not susceptible to moderate reductions of RCS.


Electron Transport Chain Complex Proteins/metabolism , Heart/physiology , Mitochondria, Heart/metabolism , 2-Propanol/pharmacology , Animals , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Ethanol/pharmacology , Male , Mitochondria, Heart/drug effects , Myocardial Contraction , Protein Multimerization , Rats , Rats, Sprague-Dawley , Rotenone/pharmacology
6.
Int J Mol Sci ; 21(3)2020 Jan 29.
Article En | MEDLINE | ID: mdl-32013250

Lung squamous cell carcinoma (LUSC) has a poor prognosis, in part due to poor therapeutic response and limited therapeutic alternatives. Lichens are symbiotic organisms, producing a variety of substances with multiple biological activities. (+)-Usnic acid, an important biologically active metabolite of lichens, has been shown to have high anti-cancer activity at low doses. However, there have been no reports regarding the effect of (+)-usnic acid on LUSC cells. This study found that (+)-usnic acid reduced viability and induced apoptosis in LUSC cells by reactive oxygen species (ROS) accumulation. (+)-Usnic acid induced mitochondria-derived ROS production via inhibition of complex I and complex III of the mitochondrial respiratory chain (MRC). Interestingly, the elimination of mitochondrial ROS by Mito-TEMPOL only partially reversed the effect of (+)-usnic acid on cellular ROS production. Further study showed that (+)-usnic acid also induced ROS production via reducing Nrf2 stability through disruption of the PI3K/Akt pathway. The in vitro and in vivo xenograft studies showed that combined treatment of (+)-usnic acid and paclitaxel synergistically suppressed LUSC cells. In conclusion, this study indicates that (+)-usnic acid induces apoptosis of LUSC cells through ROS accumulation, probably via disrupting the mitochondrial respiratory chain (MRC) and the PI3K/Akt/Nrf2 pathway. Therefore, although clinical use of (+)-usnic acid will be limited due to toxicity issues, derivatives thereof may turn out as promising anticancer candidates for adjuvant treatment of LUSC.


Apoptosis/drug effects , Benzofurans/pharmacology , Electron Transport Chain Complex Proteins/metabolism , Mitochondria/drug effects , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Benzofurans/chemistry , Benzofurans/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mitochondria/metabolism , NF-E2-Related Factor 2/antagonists & inhibitors , Paclitaxel/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Transplantation, Heterologous
7.
Cell Chem Biol ; 27(2): 158-171.e3, 2020 02 20.
Article En | MEDLINE | ID: mdl-31813848

We report detailed susceptibility profiling of asexual blood stages of the malaria parasite Plasmodium falciparum to clinical and experimental antimalarials, combined with metabolomic fingerprinting. Results revealed a variety of stage-specific and metabolic profiles that differentiated the modes of action of clinical antimalarials including chloroquine, piperaquine, lumefantrine, and mefloquine, and identified late trophozoite-specific peak activity and stage-specific biphasic dose-responses for the mitochondrial inhibitors DSM265 and atovaquone. We also identified experimental antimalarials hitting previously unexplored druggable pathways as reflected by their unique stage specificity and/or metabolic profiles. These included several ring-active compounds, ones affecting hemoglobin catabolism through distinct pathways, and mitochondrial inhibitors with lower propensities for resistance than either DSM265 or atovaquone. This approach, also applicable to other microbes that undergo multiple differentiation steps, provides an effective tool to prioritize compounds for further development within the context of combination therapies.


Antimalarials/pharmacology , Metabolome/drug effects , Metabolomics , Plasmodium falciparum/drug effects , Antimalarials/chemistry , Antimalarials/metabolism , Atovaquone/chemistry , Atovaquone/metabolism , Atovaquone/pharmacology , Drug Design , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Electron Transport Chain Complex Proteins/metabolism , Humans , Life Cycle Stages/drug effects , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Quinolines/chemistry , Quinolines/metabolism , Quinolines/pharmacology
8.
Sci Rep ; 9(1): 9413, 2019 06 28.
Article En | MEDLINE | ID: mdl-31253850

Coxsackievirus B3 (CVB3) is an important human pathogen associated with the development of acute pancreatitis, myocarditis, and type 1 diabetes. Currently, no vaccines or antiviral therapeutics are approved for the prevention and treatment of CVB3 infection. We found that Saururus chinensis Baill extract showed critical antiviral activity against CVB3 infection in vitro. Further, manassantin B inhibited replication of CVB3 and suppressed CVB3 VP1 protein expression in vitro. Additionally, oral administration of manassantin B in mice attenuated CVB3 infection-associated symptoms by reducing systemic production of inflammatory cytokines and chemokines including TNF-α, IL-6, IFN-γ, CCL2, and CXCL-1. We found that the antiviral activity of manassantin B is associated with increased levels of mitochondrial ROS (mROS). Inhibition of mROS generation attenuated the antiviral activity of manassantin B in vitro. Interestingly, we found that manassantin B also induced cytosolic release of mitochondrial DNA based on cytochrome C oxidase DNA levels. We further confirmed that STING and IRF-3 expression and STING and TBK-1 phosphorylation were increased by manassantin B treatment in CVB3-infected cells. Collectively, these results suggest that manassantin B exerts antiviral activity against CVB3 through activation of the STING/TKB-1/IRF3 antiviral pathway and increased production of mROS.


Antiviral Agents/pharmacology , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/virology , Enterovirus B, Human/drug effects , Furans/pharmacology , Interferon Regulatory Factor-3/metabolism , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Chlorocebus aethiops , Coxsackievirus Infections/drug therapy , Cytokines/metabolism , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Female , Humans , Inflammation Mediators/metabolism , Mice , Mitochondria/metabolism , Plant Extracts/pharmacology , Signal Transduction/drug effects , Vero Cells , Virus Replication/drug effects
9.
J Cell Biol ; 218(6): 1853-1870, 2019 06 03.
Article En | MEDLINE | ID: mdl-31085560

In childhood, skeletal growth is driven by transient expansion of cartilage in the growth plate. The common belief is that energy production in this hypoxic tissue mainly relies on anaerobic glycolysis and not on mitochondrial respiratory chain (RC) activity. However, children with mitochondrial diseases causing RC dysfunction often present with short stature, which indicates that RC activity may be essential for cartilage-mediated skeletal growth. To elucidate the role of the mitochondrial RC in cartilage growth and pathology, we generated mice with impaired RC function in cartilage. These mice develop normally until birth, but their later growth is retarded. A detailed molecular analysis revealed that metabolic signaling and extracellular matrix formation is disturbed and induces cell death at the cartilage-bone junction to cause a chondrodysplasia-like phenotype. Hence, the results demonstrate the overall importance of the metabolic switch from fetal glycolysis to postnatal RC activation in growth plate cartilage and explain why RC dysfunction can cause short stature in children with mitochondrial diseases.


Cartilage/pathology , Chondrocytes/pathology , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Growth Disorders/complications , Growth Plate/pathology , Mitochondrial Diseases/etiology , Animals , Cartilage/metabolism , Cell Differentiation , Chondrocytes/metabolism , Collagen Type II/physiology , DNA Helicases/physiology , Electron Transport , Energy Metabolism , Growth Disorders/metabolism , Growth Disorders/pathology , Growth Plate/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/physiology , Signal Transduction
10.
Biochimie ; 160: 141-147, 2019 May.
Article En | MEDLINE | ID: mdl-30790617

Microcin J25 (MccJ25), an antimicrobial peptide, targets the respiratory chain but the exact mechanism by which it does so remains unclear. Here, we reveal that MccJ25 is able to inhibit the enzymatic activity of the isolated cytochrome bd-I from E. coli and induces at the same time production of reactive oxygen species. MccJ25 behaves as a dose-dependent weak inhibitor. Intriguingly, MccJ25 is capable of producing a change in the oxidation state of cytochrome bd-I causing its partial reduction in the presence of cyanide. These effects are specific for cytochrome bd-I, since the peptide is not able to act on purified cytochrome bo3.


Anti-Bacterial Agents/pharmacology , Bacteriocins/pharmacology , Cytochromes/metabolism , Electron Transport Chain Complex Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/metabolism , Cyanides/pharmacology , Cytochrome b Group , Cytochromes/antagonists & inhibitors , Cytochromes/genetics , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Electron Transport Chain Complex Proteins/genetics , Escherichia coli/drug effects , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/genetics , Oxidation-Reduction , Oxidoreductases/genetics , Reactive Oxygen Species/metabolism
11.
Article En | MEDLINE | ID: mdl-30642937

Accumulating evidence suggests that the bactericidal activity of some antibiotics may not be directly initiated by target inhibition. The activity of isoniazid (INH), a key first-line bactericidal antituberculosis drug currently known to inhibit mycolic acid synthesis, becomes extremely poor under stress conditions, such as hypoxia and starvation. This suggests that the target inhibition may not fully explain the bactericidal activity of the drug. Here, we report that INH rapidly increased Mycobacterium bovis BCG cellular ATP levels and enhanced oxygen consumption. The INH-triggered ATP increase and bactericidal activity were strongly compromised by Q203 and bedaquiline, which inhibit mycobacterial cytochrome bc1 and FoF1 ATP synthase, respectively. Moreover, the antioxidant N-acetylcysteine (NAC) but not 4-hydroxy-2,2,6,6-tetramethylpiperidin-1-oxyl (TEMPOL) abrogated the INH-triggered ATP increase and killing. These results reveal a link between the energetic (ATP) perturbation and INH's killing. Furthermore, the INH-induced energetic perturbation and killing were also abrogated by chemical inhibition of NADH dehydrogenases (NDHs) and succinate dehydrogenases (SDHs), linking INH's bactericidal activity further to the electron transport chain (ETC) perturbation. This notion was also supported by the observation that INH dissipated mycobacterial membrane potential. Importantly, inhibition of cytochrome bd oxidase significantly reduced cell recovery during INH challenge in a culture settling model, suggesting that the respiratory reprogramming to the cytochrome bd oxidase contributes to the escape of INH killing. This study implicates mycobacterial ETC perturbation through NDHs, SDHs, cytochrome bc1, and FoF1 ATP synthase in INH's bactericidal activity and pinpoints the participation of the cytochrome bd oxidase in protection against this drug under stress conditions.


Antitubercular Agents/pharmacology , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Electron Transport/drug effects , Isoniazid/pharmacology , Mycobacterium bovis/drug effects , Mycobacterium tuberculosis/drug effects , Adenosine Triphosphate/metabolism , Cytochrome b Group/antagonists & inhibitors , Diarylquinolines/pharmacology , Electron Transport Complex IV/metabolism , Imidazoles/pharmacology , Membrane Potentials/drug effects , Mycobacterium bovis/metabolism , Mycobacterium tuberculosis/metabolism , Oxidation-Reduction/drug effects , Oxygen Consumption/drug effects , Piperidines/pharmacology , Pyridines/pharmacology
12.
J Biol Chem ; 294(6): 1936-1943, 2019 02 08.
Article En | MEDLINE | ID: mdl-30530783

Energy metabolism has recently gained interest as a target space for antibiotic drug development in mycobacteria. Of particular importance is bedaquiline (Sirturo), which kills mycobacteria by inhibiting the F1F0 ATP synthase. Other components of the electron transport chain such as the NADH dehydrogenases (NDH-2 and NdhA) and the terminal respiratory oxidase bc1:aa3 are also susceptible to chemical inhibition. Because antituberculosis drugs are prescribed as part of combination therapies, the interaction between novel drugs targeting energy metabolism and classical first and second line antibiotics must be considered to maximize treatment efficiency. Here, we show that subinhibitory concentration of drugs targeting the F1F0 ATP synthase and the cytochrome bc1:aa3, as well as energy uncouplers, interfere with the bactericidal potency of isoniazid and moxifloxacin. Isoniazid- and moxifloxacin-induced mycobacterial death correlated with a transient increase in intracellular ATP that was dissipated by co-incubation with energy metabolism inhibitors. Although oxidative phosphorylation is a promising target space for drug development, a better understanding of the link between energy metabolism and antibiotic-induced mycobacterial death is essential to develop potent drug combinations for the treatment of tuberculosis.


Anti-Bacterial Agents/pharmacology , Energy Metabolism/drug effects , Mycobacterium/drug effects , Adenosine Triphosphate/metabolism , Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Drug Design , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Isoniazid/pharmacology , Moxifloxacin/pharmacology , Mycobacterium/cytology , Oxidative Phosphorylation/drug effects , Proton-Translocating ATPases/antagonists & inhibitors
13.
Mol Cell ; 73(2): 354-363.e3, 2019 01 17.
Article En | MEDLINE | ID: mdl-30581146

Ferroptosis is a regulated necrosis process driven by iron-dependent lipid peroxidation. Although ferroptosis and cellular metabolism interplay with one another, whether mitochondria are involved in ferroptosis is under debate. Here, we demonstrate that mitochondria play a crucial role in cysteine-deprivation-induced ferroptosis but not in that induced by inhibiting glutathione peroxidase-4 (GPX4), the most downstream component of the ferroptosis pathway. Mechanistically, cysteine deprivation leads to mitochondrial membrane potential hyperpolarization and lipid peroxide accumulation. Inhibition of mitochondrial TCA cycle or electron transfer chain (ETC) mitigated mitochondrial membrane potential hyperpolarization, lipid peroxide accumulation, and ferroptosis. Blockage of glutaminolysis had the same inhibitory effect, which was counteracted by supplying downstream TCA cycle intermediates. Importantly, loss of function of fumarate hydratase, a tumor suppressor and TCA cycle component, confers resistance to cysteine-deprivation-induced ferroptosis. Collectively, this work demonstrates the crucial role of mitochondria in cysteine-deprivation-induced ferroptosis and implicates ferroptosis in tumor suppression.


Citric Acid Cycle , Electron Transport Chain Complex Proteins/metabolism , Fibroblasts/enzymology , Iron/metabolism , Lipid Peroxidation , Mitochondria/enzymology , Animals , Cell Line, Tumor , Citric Acid Cycle/drug effects , Electron Transport , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/pathology , Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Glutamine/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Humans , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/pathology , Mutation , Necrosis , Phospholipid Hydroperoxide Glutathione Peroxidase , Reactive Oxygen Species/metabolism , Signal Transduction
14.
Free Radic Res ; 52(9): 1052-1062, 2018 Sep.
Article En | MEDLINE | ID: mdl-30175632

The objectives of this study were to develop a robust protocol to measure the rate of hydrogen peroxide (H2O2) production in isolated perfused rat lungs, as an index of oxidative stress, and to determine the cellular sources of the measured H2O2 using the extracellular probe Amplex red (AR). AR was added to the recirculating perfusate in an isolated perfused rat lung. AR's highly fluorescent oxidation product resorufin was measured in the perfusate. Experiments were carried out without and with rotenone (complex I inhibitor), thenoyltrifluoroacetone (complex II inhibitor), antimycin A (complex III inhibitor), potassium cyanide (complex IV inhibitor), or diohenylene iodonium (inhibitor of flavin-containing enzymes, e.g. NAD(P)H oxidase or NOX) added to the perfusate. We also evaluated the effect of acute changes in oxygen (O2) concentration of ventilation gas on lung rate of H2O2 release into the perfusate. Baseline lung rate of H2O2 release was 8.45 ± 0.31 (SEM) nmol/min/g dry wt. Inhibiting mitochondrial complex II reduced this rate by 76%, and inhibiting flavin-containing enzymes reduced it by another 23%. Inhibiting complex I had a small (13%) effect on the rate, whereas inhibiting complex III had no effect. Inhibiting complex IV increased this rate by 310%. Increasing %O2 in the ventilation gas mixture from 15 to 95% had a small (27%) effect on this rate, and this O2-dependent increase was mostly nonmitochondrial. Results suggest complex II as a potentially important source and/or regulator of mitochondrial H2O2, and that most of acute hyperoxia-enhanced lung rate of H2O2 release is from nonmitochondrial rather than mitochondrial sources.


Electron Transport Chain Complex Proteins/antagonists & inhibitors , Hydrogen Peroxide/isolation & purification , Lung/chemistry , Oxidative Stress/drug effects , Animals , Antimycin A/pharmacology , Electron Transport Chain Complex Proteins/chemistry , Hydrogen Peroxide/chemistry , Lung/drug effects , Mitochondria/chemistry , Mitochondria/drug effects , Organ Culture Techniques , Oxazines/chemistry , Oxazines/pharmacology , Oxidation-Reduction/drug effects , Potassium Cyanide/pharmacology , Rats , Reactive Oxygen Species/chemistry , Rotenone/pharmacology , Thenoyltrifluoroacetone/pharmacology
15.
Int J Parasitol Drugs Drug Resist ; 8(2): 295-303, 2018 08.
Article En | MEDLINE | ID: mdl-29775797

Phenotypic screening has produced most of the new chemical entities currently in clinical development for malaria, plus many lead compounds active against Plasmodium falciparum asexual stages. However, lack of knowledge about the mode of action of these compounds delays and may even hamper their future development. Identifying the mode of action of the inhibitors greatly helps to prioritise compounds for further development as novel antimalarials. Here we describe a whole-cell method to detect inhibitors of the mitochondrial electron transport chain, using oxygen consumption as high throughput readout in 384-well plate format. The usefulness of the method has been confirmed with the Tres Cantos Antimalarial Compound Set (TCAMS). The assay identified 124 respiratory inhibitors in TCAMS, seven of which were novel anti-plasmodial chemical structures never before described as mitochondrial inhibitors.


Antimalarials/pharmacology , Drug Evaluation, Preclinical/methods , Mitochondria/drug effects , Plasmodium falciparum/drug effects , Drug Discovery/methods , Drug Evaluation, Preclinical/instrumentation , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Humans , Inhibitory Concentration 50 , Malaria/drug therapy , Malaria/parasitology , Malaria, Falciparum , Oxygen/metabolism , Plasmodium falciparum/cytology
16.
J Antibiot (Tokyo) ; 71(8): 731-740, 2018 08.
Article En | MEDLINE | ID: mdl-29691485

Biotransformation of wortmannilactone F (3) using the marine-derived fungus DL1103 generated wortmannilactone M (1), a novel analog of wortmannilactone, which was a reduction product of 3 at the C-3 carbonyl group. The in vitro inhibitory activities of 10 wortmannilactones, including 1, against electron transport enzymes indicated that all the wortmannilactones were selective inhibitors of NADH-fumarate reductase and NADH-rhodoquinone reductase. The structure-activity relationship analysis showed that the relative configuration of C1" and C5", the positions of double bonds, the oxygen atoms in the dihydropyran moiety, and the keto-carbonyl group in the oxabicyclo-[2.2.1]-heptane moiety were important to the inhibitory activity of wortmannilactones. In vivo studies indicated that 3 significantly decreased the number and size of adult worms in Trichinella spiralis-infected mice in a dose-dependent manner. Notable changes in the cuticle and microvilli of T. spiralis were also observed. Our data provided useful information in the research and development of polyketides with dihydropyran and oxabicyclo [2.2.1] heptane moieties as antihelminthics.


Anthelmintics/pharmacology , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Macrolides/pharmacology , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Quinone Reductases/antagonists & inhibitors , Trichinella spiralis/drug effects , Trichinellosis/drug therapy , Animals , Disease Models, Animal , Electron Transport/drug effects , Energy Metabolism/drug effects , Male , Mice , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/metabolism , Structure-Activity Relationship
17.
PLoS One ; 13(3): e0193386, 2018.
Article En | MEDLINE | ID: mdl-29538422

Betulin has a wide range of biological and pharmacological properties with its anticancer activity attracting most of the attention as it offers a possible alternative treatment to chemotherapy. However, betulin's in vivo biological effectiveness is limited by its poor solubility. As such, we synthesized polar glycosylated derivatives to increase its hydrosolubility and enhance its pharmacological properties. Among these synthesized compounds, 28-O-α-l-rhamnopyranosylbetulin 3ß-O-α-l-rhamnopyranoside (Bi-L-RhamBet) was assessed for its cytotoxic effects against a suite of lung cancer cell lines. We also investigated its mechanism of action using an A549 lung cancer cell line. Our results showed that Bi-L-RhamBet exhibited potent cytotoxic activity toward lung cancer cell lines including A549, NCI-H2087, NCI-H522, NCI-H1993 NCI-H1755, and LLC1 having IC50 values ranging from 2.9 to 5.9 µM. Moreover, Bi-L-RhamBet (50 mg/kg) significantly inhibited tumor growth with a treatment-to-control ratio (T/C) of 0.54 and a tumor growth inhibition rate of 46% at day 18 (p < 0.05). Microscopic observations of A549 cells, double stained with acridine orange and ethidium bromide, showed apoptotic features. Bi-L-RhamBet induced activation of pro-apoptotic caspases 8, 9, and 3/7 as well as causing DNA fragmentation. Moreover, a marked increase in mitochondrial ROS (mROS) was coupled with a reduction of mitochondrial potential. Interestingly, the presence of mitochondrial electron transport chain (ETC) inhibitors, including rotenone, malonate, and antimycin A, reduced mROS production, and the activation of caspases suggesting that Bi-L-RhamBet disturbs the ETC. Finally, dichloroacetate, a pyruvate dehydrogenase kinase inhibitor potentiated the cytotoxicity of Bi-L-RhamBet against A549 cells. Taken together, these data suggest that Bi-L-RhamBet can induce apoptotic cell death via disturbance of mitochondrial electron transfer chain, reduced ROS production, and decreased membrane potential.


Apoptosis/drug effects , Rhamnose/chemistry , Saponins/toxicity , Triterpenes/chemistry , Triterpenes/toxicity , A549 Cells , Animals , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Fragmentation/drug effects , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Electron Transport Chain Complex Proteins/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , M Phase Cell Cycle Checkpoints/drug effects , Male , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Rotenone/toxicity , Saponins/chemistry , Saponins/therapeutic use , Transplantation, Heterologous , Triterpenes/therapeutic use
18.
Neurochem Int ; 117: 23-34, 2018 07.
Article En | MEDLINE | ID: mdl-28732770

Mitochondrial respiratory chain (RC) disease is a heterogeneous and highly morbid group of energy deficiency disorders for which no proven effective therapies exist. Robust vertebrate animal models of primary RC dysfunction are needed to explore the effects of variation in RC disease subtypes, tissue-specific manifestations, and major pathogenic factors contributing to each disorder, as well as their pre-clinical response to therapeutic candidates. We have developed a series of zebrafish (Danio rerio) models that inhibit, to variable degrees, distinct aspects of RC function, and enable quantification of animal development, survival, behaviors, and organ-level treatment effects as well as effects on mitochondrial biochemistry and physiology. Here, we characterize four pharmacologic inhibitor models of mitochondrial RC dysfunction in early larval zebrafish, including rotenone (complex I inhibitor), azide (complex IV inhibitor), oligomycin (complex V inhibitor), and chloramphenicol (mitochondrial translation inhibitor that leads to multiple RC complex dysfunction). A range of concentrations and exposure times of each RC inhibitor were systematically evaluated on early larval development, animal survival, integrated behaviors (touch and startle responses), organ physiology (brain death, neurologic tone, heart rate), and fluorescence-based analyses of mitochondrial physiology in zebrafish skeletal muscle. Pharmacologic RC inhibitor effects were validated by spectrophotometric analysis of Complex I, II and IV enzyme activities, or relative quantitation of ATP levels in larvae. Outcomes were prioritized that utilize in vivo animal imaging and quantitative behavioral assessments, as may optimally inform the translational potential of pre-clinical drug screens for future clinical study in human mitochondrial disease subjects. The RC complex inhibitors each delayed early embryo development, with short-term exposures of these three agents or chloramphenicol from 5 to 7 days post fertilization also causing reduced larval survival and organ-specific defects ranging from brain death, behavioral and neurologic alterations, reduced mitochondrial membrane potential in skeletal muscle (rotenone), and/or cardiac edema with visible blood pooling (oligomycin). Remarkably, we demonstrate that treating animals with probucol, a nutrient-sensing signaling network modulating drug that has been shown to yield therapeutic effects in a range of other RC disease cellular and animal models, both prevented acute rotenone-induced brain death in zebrafish larvae, and significantly rescued early embryo developmental delay from either rotenone or oligomycin exposure. Overall, these zebrafish pharmacologic RC function inhibition models offer a unique opportunity to gain novel insights into diverse developmental, survival, organ-level, and behavioral defects of varying severity, as well as their individual response to candidate therapies, in a highly tractable and cost-effective vertebrate animal model system.


Electron Transport Chain Complex Proteins/antagonists & inhibitors , Electron Transport Chain Complex Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Rotenone/pharmacology , Sodium Azide/pharmacology , Animals , Enzyme Inhibitors/pharmacology , Insecticides/pharmacology , Zebrafish
19.
Sci Rep ; 7(1): 17922, 2017 12 20.
Article En | MEDLINE | ID: mdl-29263397

There exists an intricate relationship between hypercholesterolemia (elevated plasma cholesterol) and brain functions. The present study aims to understand the impact of hypercholesterolemia on pathological consequences in mouse brain. A chronic mouse model of hypercholesterolemia was induced by giving high-cholesterol diet for 12 weeks. The hypercholesterolemic mice developed cognitive impairment as evident from object recognition memory test. Cholesterol accumulation was observed in four discrete brain regions, such as cortex, striatum, hippocampus and substantia nigra along with significantly damaged blood-brain barrier by hypercholesterolemia. The crucial finding is the loss of acetylcholinesterase activity with mitochondrial dysfunction globally in the brain of hypercholesterolemic mice, which is related to the levels of cholesterol. Moreover, the levels of hydroxyl radical were elevated in the regions of brain where the activity of mitochondrial complexes was found to be reduced. Intriguingly, elevations of inflammatory stress markers in the cholesterol-rich brain regions were observed. As cognitive impairment, diminished brain acetylcholinesterase activity, mitochondrial dysfunctions, and inflammation are the prima facie pathologies of neurodegenerative diseases, the findings impose hypercholesterolemia as potential risk factor towards brain dysfunction.


Acetylcholinesterase/deficiency , Blood-Brain Barrier/pathology , Brain/pathology , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Hypercholesterolemia/complications , Inflammation/pathology , Mitochondrial Proteins/antagonists & inhibitors , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Cholesterol/metabolism , Disease Models, Animal , Hypercholesterolemia/physiopathology , Inflammation/etiology , Inflammation/metabolism , Male , Mice , Mitochondria/metabolism , Mitochondria/pathology
20.
Cell Rep ; 21(6): 1481-1494, 2017 Nov 07.
Article En | MEDLINE | ID: mdl-29117555

In Caenorhabditis elegans, the programmed repression of the heat shock response (HSR) accompanies the transition to reproductive maturity, leaving cells vulnerable to environmental stress and protein aggregation with age. To identify the factors driving this event, we performed an unbiased genetic screen for suppressors of stress resistance and identified the mitochondrial electron transport chain (ETC) as a central regulator of the age-related decline of the HSR and cytosolic proteostasis. Mild downregulation of ETC activity, either by genetic modulation or exposure to mitochondria-targeted xenobiotics, maintained the HSR in adulthood by increasing HSF-1 binding and RNA polymerase II recruitment at HSF-1 target genes. This resulted in a robust restoration of cytoplasmic proteostasis and increased vitality later in life, without detrimental effects on fecundity. We propose that low levels of mitochondrial stress regulate cytoplasmic proteostasis and healthspan during aging by coordinating the long-term activity of HSF-1 with conditions preclusive to optimal fitness.


Aging , Caenorhabditis elegans/metabolism , Heat-Shock Response/genetics , Mitochondria/metabolism , Animals , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cytoplasm/metabolism , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Electron Transport Chain Complex Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Longevity , Mitochondria/drug effects , Protein Binding , Proteostasis/physiology , RNA Interference , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Stress, Physiological , Temperature , Transcription Factors/genetics , Transcription Factors/metabolism , Xenobiotics/pharmacology
...