Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 488
Filter
1.
Nat Commun ; 15(1): 3367, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719808

ABSTRACT

Soil-transmitted helminths (STHs) are major pathogens infecting over a billion people. There are few classes of anthelmintics and there is an urgent need for new drugs. Many STHs use an unusual form of anaerobic metabolism to survive the hypoxic conditions of the host gut. This requires rhodoquinone (RQ), a quinone electron carrier. RQ is not made or used by vertebrate hosts making it an excellent therapeutic target. Here we screen 480 structural families of natural products to find compounds that kill Caenorhabditis elegans specifically when they require RQ-dependent metabolism. We identify several classes of compounds including a family of species-selective inhibitors of mitochondrial respiratory complex I. These identified complex I inhibitors have a benzimidazole core and we determine key structural requirements for activity by screening 1,280 related compounds. Finally, we show several of these compounds kill adult STHs. We suggest these species-selective complex I inhibitors are potential anthelmintics.


Subject(s)
Anthelmintics , Caenorhabditis elegans , Electron Transport Complex I , Ubiquinone/analogs & derivatives , Animals , Anthelmintics/pharmacology , Anthelmintics/chemistry , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Caenorhabditis elegans/metabolism , Benzimidazoles/pharmacology , Benzimidazoles/chemistry , Species Specificity , Quinones/chemistry , Quinones/pharmacology , Quinones/metabolism , Biological Products/pharmacology , Biological Products/chemistry
2.
Cell Death Dis ; 15(5): 311, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38697987

ABSTRACT

Cancer cells are highly dependent on bioenergetic processes to support their growth and survival. Disruption of metabolic pathways, particularly by targeting the mitochondrial electron transport chain complexes (ETC-I to V) has become an attractive therapeutic strategy. As a result, the search for clinically effective new respiratory chain inhibitors with minimized adverse effects is a major goal. Here, we characterize a new OXPHOS inhibitor compound called MS-L6, which behaves as an inhibitor of ETC-I, combining inhibition of NADH oxidation and uncoupling effect. MS-L6 is effective on both intact and sub-mitochondrial particles, indicating that its efficacy does not depend on its accumulation within the mitochondria. MS-L6 reduces ATP synthesis and induces a metabolic shift with increased glucose consumption and lactate production in cancer cell lines. MS-L6 either dose-dependently inhibits cell proliferation or induces cell death in a variety of cancer cell lines, including B-cell and T-cell lymphomas as well as pediatric sarcoma. Ectopic expression of Saccharomyces cerevisiae NADH dehydrogenase (NDI-1) partially restores the viability of B-lymphoma cells treated with MS-L6, demonstrating that the inhibition of NADH oxidation is functionally linked to its cytotoxic effect. Furthermore, MS-L6 administration induces robust inhibition of lymphoma tumor growth in two murine xenograft models without toxicity. Thus, our data present MS-L6 as an inhibitor of OXPHOS, with a dual mechanism of action on the respiratory chain and with potent antitumor properties in preclinical models, positioning it as the pioneering member of a promising drug class to be evaluated for cancer therapy. MS-L6 exerts dual mitochondrial effects: ETC-I inhibition and uncoupling of OXPHOS. In cancer cells, MS-L6 inhibited ETC-I at least 5 times more than in isolated rat hepatocytes. These mitochondrial effects lead to energy collapse in cancer cells, resulting in proliferation arrest and cell death. In contrast, hepatocytes which completely and rapidly inactivated this molecule, restored their energy status and survived exposure to MS-L6 without apparent toxicity.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Electron Transport Complex I , Mitochondria , Saccharomyces cerevisiae Proteins , Animals , Humans , Electron Transport Complex I/metabolism , Electron Transport Complex I/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Mice , Cell Line, Tumor , Mitochondria/metabolism , Mitochondria/drug effects , Cell Proliferation/drug effects , Uncoupling Agents/pharmacology , Oxidative Phosphorylation/drug effects , Xenograft Model Antitumor Assays , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/drug effects , Rats , NADH Dehydrogenase/metabolism , NADH Dehydrogenase/antagonists & inhibitors
3.
Nature ; 628(8006): 195-203, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38480879

ABSTRACT

Sustained smouldering, or low-grade activation, of myeloid cells is a common hallmark of several chronic neurological diseases, including multiple sclerosis1. Distinct metabolic and mitochondrial features guide the activation and the diverse functional states of myeloid cells2. However, how these metabolic features act to perpetuate inflammation of the central nervous system is unclear. Here, using a multiomics approach, we identify a molecular signature that sustains the activation of microglia through mitochondrial complex I activity driving reverse electron transport and the production of reactive oxygen species. Mechanistically, blocking complex I in pro-inflammatory microglia protects the central nervous system against neurotoxic damage and improves functional outcomes in an animal disease model in vivo. Complex I activity in microglia is a potential therapeutic target to foster neuroprotection in chronic inflammatory disorders of the central nervous system3.


Subject(s)
Electron Transport Complex I , Inflammation , Microglia , Neuroinflammatory Diseases , Animals , Female , Humans , Male , Mice , Central Nervous System/drug effects , Central Nervous System/metabolism , Central Nervous System/pathology , Disease Models, Animal , Electron Transport/drug effects , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Multiomics , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Reactive Oxygen Species/metabolism
4.
Pest Manag Sci ; 80(6): 2679-2688, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38284296

ABSTRACT

BACKGROUND: Bacterial leaf blight caused by Xanthomonas oryzae pv. oryzae (Xoo) is one of the most serious diseases of rice, and there is a lack of bactericides for controlling this disease. We previously found parthenolide (PTL) is a potential lead for developing bactericides against Xoo, and subunit F of respiratory chain complex I (NuoF) is an important target protein of PTL. However, the binding modes of PTL with NuoF need further elucidation. RESULTS: In this study, we obtained the crystal structure of Xoo NuoEF (complex of subunit E and F of respiratory chain complex I) with a resolution of 2.36 Å, which is the first report on the protein structure of NuoEF in plant-pathogenic bacteria. The possible binding sites of PTL with NuoF (Cys105 and Cys187) were predicted with molecular docking and mutated into alanine using a base mismatch method. The mutated proteins were expressed in Escherichia coli and purified with affinity chromatography. The binding abilities of PTL with mutated proteins were investigated via pull-down assay and BIAcore analysis, which revealed that double mutation of Cys105 and Cys187 in NuoF severely affected the binding ability of PTL with NuoF. In addition, the binding modes were further simulated with combined quantum mechanical/molecular mechanical calculations, and the results indicated that PTL may have a stronger binding with Cys105 than Cys187. CONCLUSION: NuoEF protein structure of Xoo was resolved, and Cys105 and Cys187 in NuoF are important binding sites of PTL. This study further clarified the action mechanism of PTL against Xoo, and will promote the innovation of bactericides targeting Xoo complex I. © 2024 Society of Chemical Industry.


Subject(s)
Bacterial Proteins , Molecular Docking Simulation , Sesquiterpenes , Xanthomonas , Xanthomonas/drug effects , Xanthomonas/genetics , Xanthomonas/enzymology , Xanthomonas/metabolism , Sesquiterpenes/pharmacology , Sesquiterpenes/metabolism , Sesquiterpenes/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex I/chemistry , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/genetics , Binding Sites
5.
Cell Rep ; 42(6): 112616, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37289585

ABSTRACT

Combined inhibition of oxidative phosphorylation (OXPHOS) and glycolysis has been shown to activate a PP2A-dependent signaling pathway, leading to tumor cell death. Here, we analyze highly selective mitochondrial complex I or III inhibitors in vitro and in vivo to elucidate the molecular mechanisms leading to cell death following OXPHOS inhibition. We show that IACS-010759 treatment (complex I inhibitor) induces a reactive oxygen species (ROS)-dependent dissociation of CIP2A from PP2A, leading to its destabilization and degradation through chaperone-mediated autophagy. Mitochondrial complex III inhibition has analogous effects. We establish that activation of the PP2A holoenzyme containing B56δ regulatory subunit selectively mediates tumor cell death, while the arrest in proliferation that is observed upon IACS-010759 treatment does not depend on the PP2A-B56δ complex. These studies provide a molecular characterization of the events subsequent to the alteration of critical bioenergetic pathways and help to refine clinical studies aimed to exploit metabolic vulnerabilities of tumor cells.


Subject(s)
Chaperone-Mediated Autophagy , Electron Transport Complex I , Neoplasms , Humans , Autoantigens/metabolism , Cell Line, Tumor , Energy Metabolism , Neoplasms/pathology , Oxidative Phosphorylation , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/metabolism , Signal Transduction , Electron Transport Complex I/antagonists & inhibitors
6.
Science ; 379(6630): 351-357, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36701435

ABSTRACT

The molecular mode of action of biguanides, including the drug metformin, which is widely used in the treatment of diabetes, is incompletely characterized. Here, we define the inhibitory drug-target interaction(s) of a model biguanide with mammalian respiratory complex I by combining cryo-electron microscopy and enzyme kinetics. We interpret these data to explain the selectivity of biguanide binding to different enzyme states. The primary inhibitory site is in an amphipathic region of the quinone-binding channel, and an additional binding site is in a pocket on the intermembrane-space side of the enzyme. An independent local chaotropic interaction, not previously described for any drug, displaces a portion of a key helix in the membrane domain. Our data provide a structural basis for biguanide action and enable the rational design of medicinal biguanides.


Subject(s)
Biguanides , Electron Transport Complex I , Animals , Cryoelectron Microscopy , Electron Transport Complex I/antagonists & inhibitors , Metformin/pharmacology , Mitochondria/metabolism , Biguanides/pharmacology
7.
Nature ; 607(7920): 756-761, 2022 07.
Article in English | MEDLINE | ID: mdl-35859172

ABSTRACT

Oocytes form before birth and remain viable for several decades before fertilization1. Although poor oocyte quality accounts for most female fertility problems, little is known about how oocytes maintain cellular fitness, or why their quality eventually declines with age2. Reactive oxygen species (ROS) produced as by-products of mitochondrial activity are associated with lower rates of fertilization and embryo survival3-5. Yet, how healthy oocytes balance essential mitochondrial activity with the production of ROS is unknown. Here we show that oocytes evade ROS by remodelling the mitochondrial electron transport chain through elimination of complex I. Combining live-cell imaging and proteomics in human and Xenopus oocytes, we find that early oocytes exhibit greatly reduced levels of complex I. This is accompanied by a highly active mitochondrial unfolded protein response, which is indicative of an imbalanced electron transport chain. Biochemical and functional assays confirm that complex I is neither assembled nor active in early oocytes. Thus, we report a physiological cell type without complex I in animals. Our findings also clarify why patients with complex-I-related hereditary mitochondrial diseases do not experience subfertility. Complex I suppression represents an evolutionarily conserved strategy that allows longevity while maintaining biological activity in long-lived oocytes.


Subject(s)
Electron Transport Complex I , Mitochondria , Oocytes , Reactive Oxygen Species , Animals , Electron Transport , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Female , Humans , Mitochondria/metabolism , Oocytes/cytology , Oocytes/enzymology , Oocytes/metabolism , Proteomics , Unfolded Protein Response , Xenopus laevis
8.
Science ; 376(6600): eabh2841, 2022 06 24.
Article in English | MEDLINE | ID: mdl-35737799

ABSTRACT

Tumor necrosis factor (TNF) is a critical host resistance factor against tuberculosis. However, excess TNF produces susceptibility by increasing mitochondrial reactive oxygen species (mROS), which initiate a signaling cascade to cause pathogenic necrosis of mycobacterium-infected macrophages. In zebrafish, we identified the mechanism of TNF-induced mROS in tuberculosis. Excess TNF in mycobacterium-infected macrophages elevates mROS production by reverse electron transport (RET) through complex I. TNF-activated cellular glutamine uptake leads to an increased concentration of succinate, a Krebs cycle intermediate. Oxidation of this elevated succinate by complex II drives RET, thereby generating the mROS superoxide at complex I. The complex I inhibitor metformin, a widely used antidiabetic drug, prevents TNF-induced mROS and necrosis of Mycobacterium tuberculosis-infected zebrafish and human macrophages; metformin may therefore be useful in tuberculosis therapy.


Subject(s)
Electron Transport Complex I , Macrophages , Metformin , Mycobacterium tuberculosis , Reactive Oxygen Species , Tuberculosis , Tumor Necrosis Factor-alpha , Animals , Citric Acid Cycle/drug effects , Electron Transport , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Humans , Macrophages/metabolism , Macrophages/microbiology , Macrophages/pathology , Metformin/pharmacology , Mycobacterium tuberculosis/metabolism , Necrosis , Reactive Oxygen Species/metabolism , Succinic Acid/metabolism , Tuberculosis/metabolism , Tuberculosis/microbiology , Tuberculosis/pathology , Tumor Necrosis Factor-alpha/metabolism , Zebrafish
9.
Sci Rep ; 12(1): 8020, 2022 05 16.
Article in English | MEDLINE | ID: mdl-35577908

ABSTRACT

Anticancer strategies aimed at inhibiting Complex I of the mitochondrial respiratory chain are increasingly being attempted in solid tumors, as functional oxidative phosphorylation is vital for cancer cells. Using ovarian cancer as a model, we show that a compensatory response to an energy crisis induced by Complex I genetic ablation or pharmacological inhibition is an increase in the mitochondrial biogenesis master regulator PGC1α, a pleiotropic coactivator of transcription regulating diverse biological processes within the cell. We associate this compensatory response to the increase in PGC1α target gene expression, setting the basis for the comprehension of the molecular pathways triggered by Complex I inhibition that may need attention as drawbacks before these approaches are implemented in ovarian cancer care.


Subject(s)
Electron Transport Complex I , Ovarian Neoplasms , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Female , Humans , Organelle Biogenesis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
10.
Chem Biol Interact ; 351: 109677, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34634269

ABSTRACT

Methylglyoxal (MG) is a reactive metabolite derived from different physiological pathways. Its production can be harmful to cells via glycation reactions of lipids, DNA, and proteins. But, the effects of MG on mitochondrial functioning and bioenergetic responses are still elusive. Then, the effects of MG on key parameters of mitochondrial functionality were examined here. Isolated rat liver mitochondria were exposed to 0.1-10 mM of MG to determine its toxicity in the mitochondrial viability, membrane potential (Δψm), swelling and the superoxide (O2•-) production. Besides, mitochondrial oxidative phosphorylation parameters were analyzed by high-resolution respiratory (HRR) assay. In this set of experiments, routine state, PM state (pyruvate/malate), oxidative phosphorylation (OXPHOS), LEAK respiration, electron transport system (ETS) and oxygen residual (ROX) states were evaluated. HRR showed that PM state, OXPHOS CI-Linked, LEAK respiration, ETS CI/CII-Linked and ETS CII-Linked/ROX were significantly inhibited by MG exposure. MG also inhibited the complex II activity, and decreased Δψm and the viability of mitochondria. Taken together, our data indicates that MG is an inductor of mitochondrial dysfunctions and impairs important steps of respiratory chain, effects that can alter bioenergetics responses.


Subject(s)
Enzyme Inhibitors/toxicity , Mitochondria/drug effects , Oxidative Phosphorylation/drug effects , Pyruvaldehyde/toxicity , Animals , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex II/antagonists & inhibitors , Male , Membrane Potential, Mitochondrial/drug effects , Rats, Wistar
11.
PLoS Genet ; 17(12): e1009971, 2021 12.
Article in English | MEDLINE | ID: mdl-34965247

ABSTRACT

Rothmund-Thomson syndrome (RTS) is an autosomal recessive genetic disorder characterized by poikiloderma, small stature, skeletal anomalies, sparse brows/lashes, cataracts, and predisposition to cancer. Type 2 RTS patients with biallelic RECQL4 pathogenic variants have multiple skeletal anomalies and a significantly increased incidence of osteosarcoma. Here, we generated RTS patient-derived induced pluripotent stem cells (iPSCs) to dissect the pathological signaling leading to RTS patient-associated osteosarcoma. RTS iPSC-derived osteoblasts showed defective osteogenic differentiation and gain of in vitro tumorigenic ability. Transcriptome analysis of RTS osteoblasts validated decreased bone morphogenesis while revealing aberrantly upregulated mitochondrial respiratory complex I gene expression. RTS osteoblast metabolic assays demonstrated elevated mitochondrial respiratory complex I function, increased oxidative phosphorylation (OXPHOS), and increased ATP production. Inhibition of mitochondrial respiratory complex I activity by IACS-010759 selectively suppressed cellular respiration and cell proliferation of RTS osteoblasts. Furthermore, systems analysis of IACS-010759-induced changes in RTS osteoblasts revealed that chemical inhibition of mitochondrial respiratory complex I impaired cell proliferation, induced senescence, and decreased MAPK signaling and cell cycle associated genes, but increased H19 and ribosomal protein genes. In summary, our study suggests that mitochondrial respiratory complex I is a potential therapeutic target for RTS-associated osteosarcoma and provides future insights for clinical treatment strategies.


Subject(s)
Electron Transport Complex I/genetics , Osteosarcoma/genetics , RNA, Long Noncoding/genetics , RecQ Helicases/genetics , Rothmund-Thomson Syndrome/genetics , Adenosine Triphosphate/biosynthesis , Cell Proliferation/drug effects , Cell Respiration/drug effects , Cellular Senescence/genetics , Electron Transport Complex I/antagonists & inhibitors , Gene Expression Regulation, Developmental/genetics , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Mitogen-Activated Protein Kinase Kinases/genetics , Mutation/genetics , Osteoblasts/drug effects , Osteogenesis/genetics , Osteosarcoma/complications , Osteosarcoma/pathology , Oxadiazoles/pharmacology , Oxidative Phosphorylation/drug effects , Piperidines/pharmacology , Rothmund-Thomson Syndrome/complications , Rothmund-Thomson Syndrome/pathology
12.
Sci Rep ; 11(1): 23549, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34876614

ABSTRACT

Mitochondrial oxidative phosphorylation (OXPHOS) has become an attractive target in anti-cancer studies in recent years. In this study, we found that a small molecule phenylbutenoid dimer NMac1 (Nm23-H1 activator 1), (±)-trans-3-(3,4-dimethoxyphenyl)-4-[(E)-3,4-dimethoxystyryl]cyclohex-1-ene, a previously identified anti-metastatic agent, has novel anti-proliferative effect only under glucose starvation in metastatic breast cancer cells. NMac1 causes significant activation of AMPK by decreasing ATP synthesis, lowers mitochondrial membrane potential (MMP, ΔΨm), and inhibits oxygen consumption rate (OCR) under glucose starvation. These effects of NMac1 are provoked by a consequence of OXPHOS complex I inhibition. Through the structure-activity relationship (SAR) study of NMac1 derivatives, NMac24 was identified as the most effective compound in anti-proliferation. NMac1 and NMac24 effectively suppress cancer cell proliferation in 3D-spheroid in vivo-like models only under glucose starvation. These results suggest that NMac1 and NMac24 have the potential as anti-cancer agents having cytotoxic effects selectively in glucose restricted cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cyclohexenes/pharmacology , NM23 Nucleoside Diphosphate Kinases/drug effects , Styrenes/pharmacology , Adenosine Triphosphate/biosynthesis , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclohexenes/chemistry , Electron Transport Complex I/antagonists & inhibitors , Enzyme Activators/chemistry , Enzyme Activators/pharmacology , Female , Gene Regulatory Networks/drug effects , Glucose/metabolism , Humans , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Metabolome/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Signal Transduction/drug effects , Structure-Activity Relationship , Styrenes/chemistry
13.
Angew Chem Int Ed Engl ; 60(52): 27277-27281, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34612584

ABSTRACT

NADH:ubiquinone oxidoreductase, respiratory complex I, plays a central role in cellular energy metabolism. As a major source of reactive oxygen species (ROS) it affects ageing and mitochondrial dysfunction. The novel inhibitor NADH-OH specifically blocks NADH oxidation and ROS production by complex I in nanomolar concentrations. Attempts to elucidate its structure by NMR spectroscopy have failed. Here, by using X-ray crystallographic analysis, we report the structure of NADH-OH bound in the active site of a soluble fragment of complex I at 2.0 Šresolution. We have identified key amino acid residues that are specific and essential for binding NADH-OH. Furthermore, the structure sheds light on the specificity of NADH-OH towards the unique Rossmann-fold of complex I and indicates a regulatory role in mitochondrial ROS generation. In addition, NADH-OH acts as a lead-structure for the synthesis of a novel class of ROS suppressors.


Subject(s)
Electron Transport Complex I/antagonists & inhibitors , Enzyme Inhibitors/chemistry , NAD/analogs & derivatives , Aquifex/enzymology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Bonding , Models, Molecular , NAD/chemistry , NAD/metabolism , NAD/pharmacology , Protein Binding
14.
Aging (Albany NY) ; 13(19): 22629-22648, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34607977

ABSTRACT

Green tea catechins are associated with a delay in aging. We have designed the current study to investigate the impact and to unveil the target of the most abundant green tea catechins, epigallocatechin gallate (EGCG) and epicatechin gallate (ECG). Experiments were performed in Caenorhabditis elegans to analyze cellular metabolism, ROS homeostasis, stress resistance, physical exercise capacity, health- and lifespan, and the underlying signaling pathways. Besides, we examined the impact of EGCG and ECG in isolated murine mitochondria. A concentration of 2.5 µM EGCG and ECG enhanced health- and lifespan as well as stress resistance in C. elegans. Catechins hampered mitochondrial respiration in C. elegans after 6-12 h and the activity of complex I in isolated rodent mitochondria. The impaired mitochondrial respiration was accompanied by a transient drop in ATP production and a temporary increase in ROS levels in C. elegans. After 24 h, mitochondrial respiration and ATP levels got restored, and ROS levels even dropped below control conditions. The lifespan increases induced by EGCG and ECG were dependent on AAK-2/AMPK and SIR-2.1/SIRT1, as well as on PMK-1/p38 MAPK, SKN-1/NRF2, and DAF-16/FOXO. Long-term effects included significantly diminished fat content and enhanced SOD and CAT activities, required for the positive impact of catechins on lifespan. In summary, complex I inhibition by EGCG and ECG induced a transient drop in cellular ATP levels and a temporary ROS burst, resulting in SKN-1 and DAF-16 activation. Through adaptative responses, catechins reduced fat content, enhanced ROS defense, and improved healthspan in the long term.


Subject(s)
Catechin/analogs & derivatives , Electron Transport Complex I/antagonists & inhibitors , Longevity/drug effects , Tea/chemistry , Animals , Caenorhabditis elegans , Catechin/chemistry , Catechin/pharmacology , Electron Transport Complex I/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Stress, Physiological/drug effects
15.
J Clin Invest ; 131(17)2021 09 01.
Article in English | MEDLINE | ID: mdl-34623325

ABSTRACT

Mitochondrial electron transport chain complex I (ETCC1) is the essential core of cancer metabolism, yet potent ETCC1 inhibitors capable of safely suppressing tumor growth and metastasis in vivo are limited. From a plant extract screening, we identified petasin (PT) as a highly potent ETCC1 inhibitor with a chemical structure distinct from conventional inhibitors. PT had at least 1700 times higher activity than that of metformin or phenformin and induced cytotoxicity against a broad spectrum of tumor types. PT administration also induced prominent growth inhibition in multiple syngeneic and xenograft mouse models in vivo. Despite its higher potency, it showed no apparent toxicity toward nontumor cells and normal organs. Also, treatment with PT attenuated cellular motility and focal adhesion in vitro as well as lung metastasis in vivo. Metabolome and proteome analyses revealed that PT severely depleted the level of aspartate, disrupted tumor-associated metabolism of nucleotide synthesis and glycosylation, and downregulated major oncoproteins associated with proliferation and metastasis. These findings indicate the promising potential of PT as a potent ETCC1 inhibitor to target the metabolic vulnerability of tumor cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Electron Transport Complex I/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Sesquiterpenes/pharmacology , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Electron Transport Complex I/metabolism , Endoplasmic Reticulum Stress/drug effects , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Metabolome/drug effects , Metformin/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasm Metastasis/prevention & control , Neoplasms, Experimental/pathology , Petasites/chemistry , Phenformin/pharmacology , Sesquiterpenes/chemistry , Unfolded Protein Response/drug effects , Xenograft Model Antitumor Assays
16.
Molecules ; 26(17)2021 Sep 02.
Article in English | MEDLINE | ID: mdl-34500765

ABSTRACT

Two series of novel amino acid Schiff base ligands containing heterocyclic moieties, such as quinazolinone 3-11 and indole 12-20 were successfully synthesized and confirmed by spectroscopic techniques and elemental analysis. Furthermore, all compounds were investigated in silico for their ability to inhibit mitochondrial NADH: ubiquinone oxidoreductase (complex I) by targeting the AMPK/mTOR signaling pathway and inhibiting hexokinase, a key glycolytic enzyme to prevent the Warburg effect in cancer cells. This inhibitory pathway may be an effective strategy to cause cancer cell death due to an insufficient amount of ATP. Our results revealed that, out of 18 compounds, two (11 and 20) were top-ranked as they exhibited the highest binding energies of -8.8, -13.0, -7.9, and -10.0 kcal/mol in the docking analysis, so they were then selected for in vitro assessment. Compound 11 promoted the best cytotoxic effect on MCF-7 with IC50 = 64.05 ± 0.14 µg/mL (0.135 mM) while compound 20 exhibited the best cytotoxic effect on MDA-231 with IC50 = 46.29 ± 0.09 µg/mL (0.166 mM) Compounds 11 and 20 showed significant activation of AMPK protein and oxidative stress, which led to elevated expression of p53 and Bax, reduced Bcl-2 expression, and caused cell cycle arrest at the sub-G0/G1 phase. Moreover, compounds 11 and 20 showed significant inhibition of the mTOR protein, which led to the activation of aerobic glycolysis for survival. This alternative pathway was also blocked as compounds 11 and 20 showed significant inhibitory effects on the hexokinase enzyme. These findings demonstrate that compounds 11 and 20 obeyed Lipinski's rule of five and could be used as privileged scaffolds for cancer therapy via their potential inhibition of mitochondrial complex I-associated hexokinase.


Subject(s)
Amino Acids/pharmacology , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Mitochondria/drug effects , Amino Acids/chemical synthesis , Amino Acids/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Drug Screening Assays, Antitumor , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Hexokinase/antagonists & inhibitors , Hexokinase/metabolism , Humans , Mitochondria/metabolism , Schiff Bases/chemical synthesis , Schiff Bases/chemistry , Schiff Bases/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured
17.
Pharmacol Res Perspect ; 9(5): e00854, 2021 10.
Article in English | MEDLINE | ID: mdl-34478236

ABSTRACT

Targeting the first protein complex of the mitochondrial electron transport chain (MC1) in cancer has become an attractive therapeutic approach in the recent years, given the metabolic vulnerabilities of cancer cells. The anticancer effect exerted by the pleiotropic drug metformin and the associated reduction in hypoxia-inducible factor 1α (HIF-1α) levels putatively mediated by MC1 inhibition led to the development of HIF-1α inhibitors, such as BAY87-2243, with a more specific MC1 targeting. However, the development of BAY87-2243 was stopped early in phase 1 due to dose-independent emesis and thus there is still no clinical proof of concept for the approach. Given the importance of mitochondrial metabolism during cancer progression, there is still a strong therapeutic need to develop specific and safe MC1 inhibitors. We recently reported the synthesis of compounds with a novel chemotype and potent action on HIF-1α degradation and MC1 inhibition. We describe here the selectivity, safety profile and anti-cancer activity in solid tumors of lead compound EVT-701. In addition, using murine models of lung cancer and of Non-Hodgkin's B cell lymphoma we demonstrated that EVT-701 reduced tumor growth and lymph node invasion when used as a single agent therapy. LKB1 deficiency in lung cancer was identified as a potential indicator of accrued sensitivity to EVT-701, allowing stratification and selection of patients in clinical trials. Altogether these results support further evaluation of EVT-701 alone or in combination in preclinical models and eventually in patients.


Subject(s)
Apoptosis/drug effects , Carcinoma, Lewis Lung/metabolism , Cell Proliferation/drug effects , Electron Transport Complex I/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Lung Neoplasms/metabolism , Lymph Nodes/drug effects , Lymphoma, B-Cell/metabolism , Mitochondria/drug effects , AMP-Activated Protein Kinases/genetics , Animals , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Cell Respiration , In Vitro Techniques , Lung Neoplasms/pathology , Lymph Nodes/pathology , Lymphoma, B-Cell/pathology , Mice , Mitochondria/metabolism , Neoplasm Invasiveness , Neoplasm Transplantation
18.
Nat Commun ; 12(1): 3299, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34083537

ABSTRACT

Bioenergetic perturbations driving neoplastic growth increase the production of reactive oxygen species (ROS), requiring a compensatory increase in ROS scavengers to limit oxidative stress. Intervention strategies that simultaneously induce energetic and oxidative stress therefore have therapeutic potential. Phenformin is a mitochondrial complex I inhibitor that induces bioenergetic stress. We now demonstrate that inflammatory mediators, including IFNγ and polyIC, potentiate the cytotoxicity of phenformin by inducing a parallel increase in oxidative stress through STAT1-dependent mechanisms. Indeed, STAT1 signaling downregulates NQO1, a key ROS scavenger, in many breast cancer models. Moreover, genetic ablation or pharmacological inhibition of NQO1 using ß-lapachone (an NQO1 bioactivatable drug) increases oxidative stress to selectively sensitize breast cancer models, including patient derived xenografts of HER2+ and triple negative disease, to the tumoricidal effects of phenformin. We provide evidence that therapies targeting ROS scavengers increase the anti-neoplastic efficacy of mitochondrial complex I inhibitors in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Phenformin/pharmacology , STAT1 Transcription Factor/metabolism , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Drug Synergism , Electron Transport Complex I/antagonists & inhibitors , Energy Metabolism/drug effects , Female , Glutathione/antagonists & inhibitors , Glutathione/biosynthesis , Humans , Interferon-gamma/administration & dosage , Interferon-gamma/deficiency , Interferon-gamma/metabolism , MCF-7 Cells , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/administration & dosage , Oxidative Stress/drug effects , Phenformin/administration & dosage , Poly I-C/administration & dosage , Reactive Oxygen Species/metabolism , STAT1 Transcription Factor/agonists , Xenograft Model Antitumor Assays
19.
Sci Rep ; 11(1): 9854, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33972583

ABSTRACT

We present the design and synthesis of a small library of substituted biguanidium salts and their capacity to inhibit the growth of pancreatic cancer cells. We first present their in vitro and membrane activity, before we address their mechanism of action in living cells and in vivo activity. We show that phenylethynyl biguanidium salts possess higher ability to cross hydrophobic barriers, improve mitochondrial accumulation and anticancer activity. Mechanistically, the most active compound, 1b, like metformin, activated AMPK, decreased the NAD+/NADH ratio and mitochondrial respiration, but at 800-fold lower concentration. In vivo studies show that compound 1b significantly inhibits the growth of pancreatic cancer xenografts in mice, while biguanides currently in clinical trials had little activity.


Subject(s)
Biguanides/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , AMP-Activated Protein Kinases/metabolism , Animals , Biguanides/chemistry , Biguanides/therapeutic use , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor/transplantation , Cell Proliferation/drug effects , Disease Models, Animal , Drug Screening Assays, Antitumor , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Fibroblasts , Humans , Inhibitory Concentration 50 , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Pancreatic Neoplasms/pathology
20.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33947138

ABSTRACT

Ovarian clear cell carcinoma (OCCC) is a rare but chemorefractory tumor. About 50% of all OCCC patients have inactivating mutations of ARID1A, a member of the SWI/SNF chromatin-remodeling complex. Members of the SWI/SNF remodeling have emerged as regulators of the energetic metabolism of mammalian cells; however, the role of ARID1A as a modulator of the mitochondrial metabolism in OCCCs is yet to be defined. Here, we show that ARID1A loss results in increased mitochondrial metabolism and renders ARID1A-mutated cells increasingly and selectively dependent on it. The increase in mitochondrial activity following ARID1A loss is associated with increase in c-Myc expression and increased mitochondrial number and reduction of their size consistent with a higher mitochondrial cristae/outer membrane ratio. Significantly, preclinical testing of the complex I mitochondrial inhibitor IACS-010759 showed it extends overall survival in a preclinical model of ARID1A-mutated OCCC. These findings provide for the targeting mitochondrial activity in ARID1A-mutated OCCCs.


Subject(s)
Adenocarcinoma, Clear Cell/drug therapy , Antineoplastic Agents/therapeutic use , DNA-Binding Proteins/antagonists & inhibitors , Mitochondria/drug effects , Neoplasm Proteins/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Oxadiazoles/therapeutic use , Piperidines/therapeutic use , Transcription Factors/antagonists & inhibitors , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/pathology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Electron Transport Complex I/antagonists & inhibitors , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Oxadiazoles/pharmacology , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Piperidines/pharmacology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Random Allocation , Spheroids, Cellular , Transcription Factors/deficiency , Transcription Factors/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...