1
|
Wu T, Zhou S, Qin M, Tang J, Yan X, Huang L, Huang M, Deng J, Xiao D, Hu X, Wu J, Yang X, Li G. Phenformin and ataxia-telangiectasia mutated inhibitors synergistically co-suppress liver cancer cell growth by damaging mitochondria. FEBS Open Bio 2021; 11:1440-1451. [PMID: 33742560 PMCID: PMC8091576 DOI: 10.1002/2211-5463.13152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/08/2021] [Accepted: 03/18/2021] [Indexed: 12/18/2022] Open
Abstract
Inhibitors of ataxia–telangiectasia mutated (ATM), such as KU‐55933 (Ku), represent a promising class of novel anticancer drugs. In addition, the biguanide derivative phenformin exhibits antitumor activity superior to that of the AMPK activator metformin. Herein, we assessed the potential combinatorial therapeutic efficacy of phenformin and Ku when used to inhibit the growth of liver cancer cells, and we assessed the mechanisms underlying such efficacy. The Hep‐G2 and SMMC‐7721 liver cancer cell lines were treated with phenformin and Ku either alone or in combination, after which the impact of these drugs on cellular proliferation was assessed via 3‐(4,5‐dimethylthiazol) 2, 5‐diphenyltetrazolium and colony formation assays, whereas Transwell assays were used to gauge cell migratory activity. The potential synergy between these two drugs was assessed using the compusyn software, while flow cytometry was employed to evaluate cellular apoptosis. In addition, western blotting was utilized to measure p‐ATM, p‐AMPK, p‐mTOR, and p‐p70s6k expression, while mitochondrial functionality was monitored via morphological analyses, JC‐1 staining, and measurements of ATP levels. Phenformin and Ku synergistically impacted the proliferation, migration, and apoptotic death of liver cancer cells. Together, these compounds were able to enhance AMPK phosphorylation while inhibiting the phosphorylation of mTOR and p70s6k. These data also revealed that phenformin and Ku induced mitochondrial dysfunction as evidenced by impaired ATP synthesis, mitochondrial membrane potential, and abnormal mitochondrial morphology. These findings suggest that combination treatment with phenformin and Ku may be an effective approach to treating liver cancer via damaging mitochondria within these tumor cells.
Collapse
Affiliation(s)
- Tianyu Wu
- Department of Oncology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Sichun Zhou
- Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy School of Medicine, Hunan Normal University, Changsha, China
| | - Mei Qin
- Department of Gynecologists, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Jing Tang
- Department of Oncology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Xinjian Yan
- Department of Oncology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Lingli Huang
- Department of Oncology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Meiyuan Huang
- Department of Pathology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Jun Deng
- Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy School of Medicine, Hunan Normal University, Changsha, China
| | - Di Xiao
- Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy School of Medicine, Hunan Normal University, Changsha, China
| | - Xin Hu
- Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy School of Medicine, Hunan Normal University, Changsha, China
| | - Jingtao Wu
- Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy School of Medicine, Hunan Normal University, Changsha, China
| | - Xiaoping Yang
- Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy School of Medicine, Hunan Normal University, Changsha, China
| | - Gaofeng Li
- Department of Oncology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| |
Collapse
|
2
|
Blignaut M, Harries S, Lochner A, Huisamen B. Ataxia Telangiectasia Mutated Protein Kinase: A Potential Master Puppeteer of Oxidative Stress-Induced Metabolic Recycling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8850708. [PMID: 33868575 PMCID: PMC8032526 DOI: 10.1155/2021/8850708] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/15/2021] [Accepted: 02/28/2021] [Indexed: 02/07/2023]
Abstract
Ataxia Telangiectasia Mutated protein kinase (ATM) has recently come to the fore as a regulatory protein fulfilling many roles in the fine balancing act of metabolic homeostasis. Best known for its role as a transducer of DNA damage repair, the activity of ATM in the cytosol is enjoying increasing attention, where it plays a central role in general cellular recycling (macroautophagy) as well as the targeted clearance (selective autophagy) of damaged mitochondria and peroxisomes in response to oxidative stress, independently of the DNA damage response. The importance of ATM activation by oxidative stress has also recently been highlighted in the clearance of protein aggregates, where the expression of a functional ATM construct that cannot be activated by oxidative stress resulted in widespread accumulation of protein aggregates. This review will discuss the role of ATM in general autophagy, mitophagy, and pexophagy as well as aggrephagy and crosstalk between oxidative stress as an activator of ATM and its potential role as a master regulator of these processes.
Collapse
Affiliation(s)
- Marguerite Blignaut
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | - Sarah Harries
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | - Amanda Lochner
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | - Barbara Huisamen
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| |
Collapse
|
3
|
Clark KL, Keating AF. Ataxia-telangiectasia mutated coordinates the ovarian DNA repair and atresia-initiating response to phosphoramide mustard. Biol Reprod 2020; 102:248-260. [PMID: 31435664 DOI: 10.1093/biolre/ioz160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/31/2019] [Accepted: 08/13/2019] [Indexed: 11/13/2022] Open
Abstract
Ataxia-telangiectasia-mutated (ATM) protein recognizes and repairs DNA double strand breaks through activation of cell cycle checkpoints and DNA repair proteins. Atm gene mutations increase female reproductive cancer risk. Phosphoramide mustard (PM) induces ovarian DNA damage and destroys primordial follicles, and pharmacological ATM inhibition prevents PM-induced follicular depletion. Wild-type (WT) C57BL/6 or Atm+/- mice were dosed once intraperitoneally with sesame oil (95%) or PM (25 mg/kg) in the proestrus phase of the estrous cycle and ovaries harvested 3 days thereafter. Atm+/- mice spent ~25% more time in diestrus phase than WT. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) on ovarian protein was performed and bioinformatically analyzed. Relative to WT, Atm+/- mice had 64 and 243 proteins increased or decreased in abundance, respectively. In WT mice, PM increased 162 and decreased 20 proteins. In Atm+/- mice, 173 and 37 proteins were increased and decreased, respectively, by PM. Exportin-2 (XPO2) was localized to granulosa cells of all follicle stages and was 7.2-fold greater in Atm+/- than WT mice. Cytoplasmic FMR1-interacting protein 1 was 6.8-fold lower in Atm+/- mice and was located in the surface epithelium with apparent translocation to the ovarian medulla post-PM exposure. PM induced γH2AX, but fewer γH2AX-positive foci were identified in Atm+/- ovaries. Similarly, cleaved caspase-3 was lower in the Atm+/- PM-treated, relative to WT mice. These findings support ATM involvement in ovarian DNA repair and suggest that ATM functions to regulate ovarian atresia.
Collapse
Affiliation(s)
- Kendra L Clark
- Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA
| | - Aileen F Keating
- Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA
| |
Collapse
|
4
|
Xie GH, Dai HJ, Liu F, Zhang YP, Zhu L, Nie JJ, Wu JH. A Dual Role of ATM in Ischemic Preconditioning and Ischemic Injury. Cell Mol Neurobiol 2019; 40:785-799. [PMID: 31845160 DOI: 10.1007/s10571-019-00773-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 12/05/2019] [Indexed: 11/25/2022]
Abstract
The ataxia-telangiectasia mutated (ATM) protein is regarded as the linchpin of cellular defenses to stress. Deletion of ATM results in strong oxidative stress and degenerative diseases in the nervous system. However, the role of ATM in neuronal ischemic preconditioning and lethal ischemic injury is still largely unknown. In this study, mice cortical neurons preconditioned with sublethal exposure to oxygen glucose deprivation (OGD) exhibited ATM/glucose-6-phosphate dehydrogenase pathway activation. Additionally, pharmacological inhibition of ATM prior to the preconditioning reversed neuroprotection provided by preconditioning in vitro and in vivo. Meanwhile, we found that ATM/P53 pro-apoptosis pathway was driven by lethal OGD injury, and pharmacological inhibition of ATM during fatal oxygen-glucose deprivation/reperfusion injury promoted neuronal survival. More importantly, inhibition of ATM activity after cerebral ischemia protected against cerebral ischemic-reperfusion damage in mice. In conclusion, our data show the dual role of ATM in neuronal ischemic preconditioning and lethal ischemic injury, involving in the protection of ischemic preconditioning, but promoting neuronal death in lethal ischemic injury. Thus, the present study provides new opportunity for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Guang-Hui Xie
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Han-Jun Dai
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Fang Liu
- General surgery department of Xinhua Hospital of Hubei Province, Wuhan, 430015, China
| | - Ying-Pei Zhang
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Dong-Hu Road #169, Wuhan, 430071, Hubei, China
| | - Li Zhu
- Department of Pharmacy, Tongren Hospital of Wuhan University, Wuhan, 430071, China
| | - Jun-Jie Nie
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Dong-Hu Road #169, Wuhan, 430071, Hubei, China
| | - Jian-Hua Wu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Dong-Hu Road #169, Wuhan, 430071, Hubei, China.
| |
Collapse
|
5
|
Synofzik M, Puccio H, Mochel F, Schöls L. Autosomal Recessive Cerebellar Ataxias: Paving the Way toward Targeted Molecular Therapies. Neuron 2019; 101:560-583. [PMID: 30790538 DOI: 10.1016/j.neuron.2019.01.049] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/20/2018] [Accepted: 01/23/2019] [Indexed: 12/22/2022]
Abstract
Autosomal-recessive cerebellar ataxias (ARCAs) comprise a heterogeneous group of rare degenerative and metabolic genetic diseases that share the hallmark of progressive damage of the cerebellum and its associated tracts. This Review focuses on recent translational research in ARCAs and illustrates the steps from genetic characterization to preclinical and clinical trials. The emerging common pathways underlying ARCAs include three main clusters: mitochondrial dysfunction, impaired DNA repair, and complex lipid homeostasis. Novel ARCA treatments might target common hubs in pathogenesis by modulation of gene expression, stem cell transplantation, viral gene transfer, or interventions in faulty pathways. All these translational steps are addressed in current ARCA research, leading to the expectation that novel treatments for ARCAs will be reached in the next decade.
Collapse
Affiliation(s)
- Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Hélène Puccio
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67404 Illkirch, France; INSERM, U1258, 67404 Illkirch, France; CNRS, UMR7104, 67404 Illkirch, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Fanny Mochel
- Sorbonne Université, UPMC-Paris 6, UMR S 1127 and Inserm U 1127, and CNRS UMR 7225, and Institut du Cerveau et de la Moelle épinière, 75013 Paris, France; Department of Genetics and Reference Centre for Adult Neurometabolic Diseases, AP-HP, La Pitié-Salpêtriere University Hospital, Paris, France
| | - Ludger Schöls
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
| |
Collapse
|
6
|
Blignaut M, Loos B, Botchway SW, Parker AW, Huisamen B. Ataxia-Telangiectasia Mutated is located in cardiac mitochondria and impacts oxidative phosphorylation. Sci Rep 2019; 9:4782. [PMID: 30886180 PMCID: PMC6423017 DOI: 10.1038/s41598-019-41108-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/26/2019] [Indexed: 01/16/2023] Open
Abstract
The absence of Ataxia-Telangiectasia mutated protein kinase (ATM) is associated with neurological, metabolic and cardiovascular defects. The protein has been associated with mitochondria and its absence results in mitochondrial dysfunction. Furthermore, it can be activated in the cytosol by mitochondrial oxidative stress and mediates a cellular anti-oxidant response through the pentose phosphate pathway (PPP). However, the precise location and function of ATM within mitochondria and its role in oxidative phosphorylation is still unknown. We show that ATM is found endogenously within cardiac myocyte mitochondria under normoxic conditions and is consistently associated with the inner mitochondrial membrane. Acute ex vivo inhibition of ATM protein kinase significantly decreased mitochondrial electron transfer chain complex I-mediated oxidative phosphorylation rate but did not decrease coupling efficiency or oxygen consumption rate during β-oxidation. Chemical inhibition of ATM in rat cardiomyoblast cells (H9c2) significantly decreased the excited-state autofluorescence lifetime of enzyme-bound reduced NADH and its phosphorylated form, NADPH (NAD(P)H; 2.77 ± 0.26 ns compared to 2.57 ± 0.14 ns in KU60019-treated cells). This suggests an interaction between ATM and the electron transfer chain in the mitochondria, and hence may have an important role in oxidative phosphorylation in terminally differentiated cells such as cardiomyocytes.
Collapse
Affiliation(s)
- Marguerite Blignaut
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, 7505, South Africa.
| | - Ben Loos
- Department of Physiological Sciences, Faculty of Sciences, Stellenbosch University, Stellenbosch, 7602, South Africa
| | - Stanley W Botchway
- Central Laser Facility, Research Complex at Harwell, STFC Rutherford Appleton Laboratory, Harwell Campus, Didcot, OX11 0QX, UK
- Oxford Brookes University, Department of Biological and Medical Sciences, Oxford, OX3 0BP, UK
| | - Anthony W Parker
- Central Laser Facility, Research Complex at Harwell, STFC Rutherford Appleton Laboratory, Harwell Campus, Didcot, OX11 0QX, UK
- Department of Physics, Faculty of Science, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - Barbara Huisamen
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, 7505, South Africa
- Biomedical, Research and Innovation Platform, South African Medical Research Council, Tygerberg, 7505, South Africa
| |
Collapse
|
7
|
Blatteau JE, Gaillard S, De Maistre S, Richard S, Louges P, Gempp E, Druelles A, Lehot H, Morin J, Castagna O, Abraini JH, Risso JJ, Vallée N. Reduction in the Level of Plasma Mitochondrial DNA in Human Diving, Followed by an Increase in the Event of an Accident. Front Physiol 2018; 9:1695. [PMID: 30555340 PMCID: PMC6282000 DOI: 10.3389/fphys.2018.01695] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/09/2018] [Indexed: 01/02/2023] Open
Abstract
Circulating mitochondrial DNA (mtDNA) is receiving increasing attention as a danger-associated molecular pattern in conditions such as autoimmunity or trauma. In the context of decompression sickness (DCS), the course of which is sometimes erratic, we hypothesize that mtDNA plays a not insignificant role particularly in neurological type accidents. This study is based on the comparison of circulating mtDNA levels in humans presenting with various types of diving accidents, and punctured upon their admission at the hyperbaric facility. One hundred and fourteen volunteers took part in the study. According to the clinical criteria there were 12 Cerebro DCS, 57 Medullary DCS, 15 Vestibular DCS, 8 Ctrl+ (accident-free divers), and 22 Ctrl- (non-divers). This work demonstrates that accident-free divers have less mtDNA than non-divers, which leads to the assumption that hyperbaric exposure degrades the mtDNA. mtDNA levels are on average greater in divers with DCS compared with accident-free divers. On another hand, the amount of double strand DNA (dsDNA) is neither significantly different between controls, nor between the different DCS types. Initially the increase in circulating oligonucleotides was attributed to the destruction of cells by bubble abrasion following necrotic phenomena. If there really is a significant difference between the Medullary DCS and the Ctrl-, this difference is not significant between these same DCS and the Ctrl+. This refutes the idea of massive degassing and suggests the need for new research in order to verify that oxidative stress could be a key element without necessarily being sufficient for the occurrence of a neurological type of accident.
Collapse
Affiliation(s)
- Jean-Eric Blatteau
- Institut de Recherche Biomédicale des Armées, Equipe Résidante de Recherche Subaquatique Opérationnelle, Département Environnement Opérationnel, Unité Environnements Extrêmes, Toulon, France
- Hôpital d’Instruction des Armées – Service de Médecine Hyperbare et Expertise Plongée, Toulon, France
| | | | - Sébastien De Maistre
- Hôpital d’Instruction des Armées – Service de Médecine Hyperbare et Expertise Plongée, Toulon, France
| | - Simone Richard
- Mediterranean Institute of Oceanography, Université de Toulon, Toulon, France
| | - Pierre Louges
- Hôpital d’Instruction des Armées – Service de Médecine Hyperbare et Expertise Plongée, Toulon, France
| | - Emmanuel Gempp
- Hôpital d’Instruction des Armées – Service de Médecine Hyperbare et Expertise Plongée, Toulon, France
| | - Arnaud Druelles
- Hôpital d’Instruction des Armées – Service de Médecine Hyperbare et Expertise Plongée, Toulon, France
| | - Henri Lehot
- Hôpital d’Instruction des Armées – Service de Médecine Hyperbare et Expertise Plongée, Toulon, France
| | - Jean Morin
- Hôpital d’Instruction des Armées – Service de Médecine Hyperbare et Expertise Plongée, Toulon, France
| | - Olivier Castagna
- Institut de Recherche Biomédicale des Armées, Equipe Résidante de Recherche Subaquatique Opérationnelle, Département Environnement Opérationnel, Unité Environnements Extrêmes, Toulon, France
| | - Jacques H. Abraini
- Institut de Recherche Biomédicale des Armées, Equipe Résidante de Recherche Subaquatique Opérationnelle, Département Environnement Opérationnel, Unité Environnements Extrêmes, Toulon, France
| | - Jean-Jacques Risso
- Institut de Recherche Biomédicale des Armées, Equipe Résidante de Recherche Subaquatique Opérationnelle, Département Environnement Opérationnel, Unité Environnements Extrêmes, Toulon, France
| | - Nicolas Vallée
- Institut de Recherche Biomédicale des Armées, Equipe Résidante de Recherche Subaquatique Opérationnelle, Département Environnement Opérationnel, Unité Environnements Extrêmes, Toulon, France
| |
Collapse
|
8
|
Trans-Plasma Membrane Electron Transport and Ascorbate Efflux by Skeletal Muscle. Antioxidants (Basel) 2017; 6:antiox6040089. [PMID: 29120354 PMCID: PMC5745499 DOI: 10.3390/antiox6040089] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 12/17/2022] Open
Abstract
Trans-plasma membrane electron transport (tPMET) and the antioxidant roles of ascorbate reportedly play a role in protection of cells from damage by reactive oxygen species, which have been implicated in causing metabolic dysfunction such as insulin resistance. Skeletal muscle comprises the largest whole-body organ fraction suggesting a potential role of tPMET and ascorbate export as a major source of extracellular antioxidant. We hypothesized that skeletal muscle is capable of tPMET and ascorbate efflux. To measure these processes, we assayed the ability of cultured muscle cells, satellite cells, and isolated extensor digitorum longus (EDL) and soleus (SOL) to reduce two extracellular electron acceptors, water soluble tetrazolium salt 1 (WST-1), and dichlorophenolindophenol (DPIP). Ascorbate oxidase (AO) was utilized to determine which portion of WST-1 reduction was dependent on ascorbate efflux. We found that muscle cells can reduce extracellular electron acceptors. In C2C12 myotubes and satellite cells, a substantial portion of this reduction was dependent on ascorbate. In myotubes, glucose transporter 1 (GLUT1) inhibitors along with a pan-GLUT inhibitor suppressed tPMET and ascorbate efflux, while a GLUT4 inhibitor had no effect. The adenosine 5′-monophosphate (AMP)-activated protein kinase activator 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR) suppressed both tPMET and ascorbate efflux by myotubes, while insulin had no effect. Taken together, our data suggest that muscle cells are capable of tPMET and ascorbate efflux supported by GLUT1, thus illustrating a model in which resting muscle exports electrons and antioxidant to the extracellular environment.
Collapse
|
9
|
Nayler SP, Powell JE, Vanichkina DP, Korn O, Wells CA, Kanjhan R, Sun J, Taft RJ, Lavin MF, Wolvetang EJ. Human iPSC-Derived Cerebellar Neurons from a Patient with Ataxia-Telangiectasia Reveal Disrupted Gene Regulatory Networks. Front Cell Neurosci 2017; 11:321. [PMID: 29081736 PMCID: PMC5645492 DOI: 10.3389/fncel.2017.00321] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 09/26/2017] [Indexed: 01/09/2023] Open
Abstract
Ataxia-telangiectasia (A-T) is a rare genetic disorder caused by loss of function of the ataxia-telangiectasia-mutated kinase and is characterized by a predisposition to cancer, pulmonary disease, immune deficiency and progressive degeneration of the cerebellum. As animal models do not faithfully recapitulate the neurological aspects, it remains unclear whether cerebellar degeneration is a neurodevelopmental or neurodegenerative phenotype. To address the necessity for a human model, we first assessed a previously published protocol for the ability to generate cerebellar neuronal cells, finding it gave rise to a population of precursors highly enriched for markers of the early hindbrain such as EN1 and GBX2, and later more mature cerebellar markers including PTF1α, MATH1, HOXB4, ZIC3, PAX6, and TUJ1. RNA sequencing was used to classify differentiated cerebellar neurons generated from integration-free A-T and control induced pluripotent stem cells. Comparison of RNA sequencing data with datasets from the Allen Brain Atlas reveals in vitro-derived cerebellar neurons are transcriptionally similar to discrete regions of the human cerebellum, and most closely resemble the cerebellum at 22 weeks post-conception. We show that patient-derived cerebellar neurons exhibit disrupted gene regulatory networks associated with synaptic vesicle dynamics and oxidative stress, offering the first molecular insights into early cerebellar pathogenesis of ataxia-telangiectasia.
Collapse
Affiliation(s)
- Sam P Nayler
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, QLD, Australia
| | - Joseph E Powell
- Queensland Brain Institute, University of Queensland, St. Lucia, QLD, Australia.,Institute for Molecular Bioscience, University of Queensland, St. Lucia, QLD, Australia
| | - Darya P Vanichkina
- Queensland Brain Institute, University of Queensland, St. Lucia, QLD, Australia
| | - Othmar Korn
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, QLD, Australia
| | - Christine A Wells
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, QLD, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, Australia
| | - Refik Kanjhan
- School of Biomedical Science, University of Queensland, St. Lucia, QLD, Australia
| | - Jian Sun
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, QLD, Australia
| | - Ryan J Taft
- Queensland Brain Institute, University of Queensland, St. Lucia, QLD, Australia.,Department of Integrated Systems Biology and Department of Pediatrics, School of Medicine and Health Services, George Washington University, Washington, DC, United States.,Illumina, Inc.,, San Diego, CA, United States
| | - Martin F Lavin
- UQ Centre for Clinical Research, University of Queensland, Brisbane, QLD, Australia
| | - Ernst J Wolvetang
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, QLD, Australia
| |
Collapse
|
10
|
Choy KR, Watters DJ. Neurodegeneration in ataxia-telangiectasia: Multiple roles of ATM kinase in cellular homeostasis. Dev Dyn 2017; 247:33-46. [PMID: 28543935 DOI: 10.1002/dvdy.24522] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/02/2017] [Accepted: 05/10/2017] [Indexed: 12/13/2022] Open
Abstract
Ataxia-telangiectasia (A-T) is characterized by neuronal degeneration, cancer, diabetes, immune deficiency, and increased sensitivity to ionizing radiation. A-T is attributed to the deficiency of the protein kinase coded by the ATM (ataxia-telangiectasia mutated) gene. ATM is a sensor of DNA double-strand breaks (DSBs) and signals to cell cycle checkpoints and the DNA repair machinery. ATM phosphorylates numerous substrates and activates many cell-signaling pathways. There has been considerable debate about whether a defective DNA damage response is causative of the neurological aspects of the disease. In proliferating cells, ATM is localized mainly in the nucleus; however, in postmitotic cells such as neurons, ATM is mostly cytoplasmic. Recent studies reveal an increasing number of roles for ATM in the cytoplasm, including activation by oxidative stress. ATM associates with organelles including mitochondria and peroxisomes, both sources of reactive oxygen species (ROS), which have been implicated in neurodegenerative diseases and aging. ATM is also associated with synaptic vesicles and has a role in regulating cellular homeostasis and autophagy. The cytoplasmic roles of ATM provide a new perspective on the neurodegenerative process in A-T. This review will examine the expanding roles of ATM in cellular homeostasis and relate these functions to the complex A-T phenotype. Developmental Dynamics 247:33-46, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kay Rui Choy
- School of Natural Sciences, Griffith University, Brisbane, Queensland, Australia
| | - Dianne J Watters
- School of Natural Sciences, Griffith University, Brisbane, Queensland, Australia
| |
Collapse
|
11
|
Resseguie EA, Staversky RJ, Brookes PS, O'Reilly MA. Hyperoxia activates ATM independent from mitochondrial ROS and dysfunction. Redox Biol 2015; 5:176-185. [PMID: 25967673 PMCID: PMC4430709 DOI: 10.1016/j.redox.2015.04.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 04/25/2015] [Indexed: 01/12/2023] Open
Abstract
High levels of oxygen (hyperoxia) are often used to treat individuals with respiratory distress, yet prolonged hyperoxia causes mitochondrial dysfunction and excessive reactive oxygen species (ROS) that can damage molecules such as DNA. Ataxia telangiectasia mutated (ATM) kinase is activated by nuclear DNA double strand breaks and delays hyperoxia-induced cell death through downstream targets p53 and p21. Evidence for its role in regulating mitochondrial function is emerging, yet it has not been determined if mitochondrial dysfunction or ROS activates ATM. Because ATM maintains mitochondrial homeostasis, we hypothesized that hyperoxia induces both mitochondrial dysfunction and ROS that activate ATM. In A549 lung epithelial cells, hyperoxia decreased mitochondrial respiratory reserve capacity at 12h and basal respiration by 48 h. ROS were significantly increased at 24h, yet mitochondrial DNA double strand breaks were not detected. ATM was not required for activating p53 when mitochondrial respiration was inhibited by chronic exposure to antimycin A. Also, ATM was not further activated by mitochondrial ROS, which were enhanced by depleting manganese superoxide dismutase (SOD2). In contrast, ATM dampened the accumulation of mitochondrial ROS during exposure to hyperoxia. Our findings suggest that hyperoxia-induced mitochondrial dysfunction and ROS do not activate ATM. ATM more likely carries out its canonical response to nuclear DNA damage and may function to attenuate mitochondrial ROS that contribute to oxygen toxicity.
Collapse
Affiliation(s)
- Emily A Resseguie
- Department of Environmental Medicine, University of Rochester, Rochester, NY 14642, USA
| | - Rhonda J Staversky
- Department of Pediatrics, University of Rochester, Rochester, NY 14642, USA
| | - Paul S Brookes
- Department of Anesthesiology, University of Rochester, Rochester, NY 14642, USA
| | - Michael A O'Reilly
- Department of Environmental Medicine, University of Rochester, Rochester, NY 14642, USA; Department of Pediatrics, University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
12
|
Abstract
Ataxia telangiectasia (A-T), a rare autosomal recessive disorder characterized by progressive cerebellar degeneration and a greatly increased incidence of cancer among other symptoms, is caused by a defective or missing ataxia telangiectasia mutated (ATM) gene. The ATM protein has roles in DNA repair and in the regulation of reactive oxygen species (ROS). Here, we provide, to our knowledge, the first evidence that NADPH oxidase 4 (NOX4) is involved in manifesting A-T disease. We showed that NOX4 expression levels are higher in A-T cells, and that ATM inhibition leads to increased NOX4 expression in normal cells. A-T cells exhibit elevated levels of oxidative DNA damage, DNA double-strand breaks and replicative senescence, all of which are partially abrogated by down-regulation of NOX4 with siRNA. Sections of degenerating cerebelli from A-T patients revealed elevated NOX4 levels. ATM-null mice exhibit A-T disease but they die from cancer before the neurological symptoms are manifested. Injecting Atm-null mice with fulvene-5, a specific inhibitor of NOX4 and NADPH oxidase 2 (NOX2), decreased their elevated cancer incidence to that of the controls. We conclude that, in A-T disease in humans and mice, NOX4 may be critical mediator and targeting it will open up new avenues for therapeutic intervention in neurodegeneration.
Collapse
|
13
|
Kalifa L, Gewandter JS, Staversky RJ, Sia EA, Brookes PS, O'Reilly MA. DNA double-strand breaks activate ATM independent of mitochondrial dysfunction in A549 cells. Free Radic Biol Med 2014; 75:30-9. [PMID: 25048973 PMCID: PMC4171189 DOI: 10.1016/j.freeradbiomed.2014.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 07/07/2014] [Accepted: 07/08/2014] [Indexed: 10/25/2022]
Abstract
Excessive nuclear or mitochondrial DNA damage can lead to mitochondrial dysfunction, decreased energy production, and increased generation of reactive oxygen species (ROS). Although numerous cell signaling pathways are activated when cells are injured, the ataxia telangiectasia mutant (ATM) protein has emerged as a major regulator of the response to both mitochondrial dysfunction and nuclear DNA double-strand breaks (DSBs). Because mitochondrial dysfunction is often a response to excessive DNA damage, it has been difficult to determine whether nuclear and/or mitochondrial DNA DSBs activate ATM independent of mitochondrial dysfunction. In this study, mitochondrial and nuclear DNA DSBs were generated in the A549 human lung adenocarcinoma cell line by infecting with retroviruses expressing the restriction endonuclease PstI fused to a mitochondrial targeting sequence (MTS) or nuclear localization sequence (NLS) and a hemagglutinin antigen epitope tag (HA). Expression of MTS-PstI-HA or NLS-PstI-HA activated the DNA damage response defined by phosphorylation of ATM, the tumor suppressor protein p53 (TP53), KRAB-associated protein (KAP)-1, and structural maintenance of chromosomes (SMC)-1. Phosphorylated ATM and SMC1 were detected in nuclear fractions, whereas phosphorylated TP53 and KAP1 were detected in both mitochondrial and nuclear fractions. PstI also enhanced expression of the cyclin-dependent kinase inhibitor p21 and inhibited cell growth. This response to DNA damage occurred in the absence of detectable mitochondrial dysfunction and excess production of ROS. These findings reveal that DNA DSBs are sufficient to activate ATM independent of mitochondrial dysfunction and suggest that the activated form of ATM and some of its substrates are restricted to the nuclear compartment, regardless of the site of DNA damage.
Collapse
Affiliation(s)
- Lidza Kalifa
- Department of Environmental Medicine, The University of Rochester, Rochester, NY 14642, USA
| | - Jennifer S Gewandter
- Department of Anesthesiology, The University of Rochester, Rochester, NY 14642, USA
| | - Rhonda J Staversky
- Department of Pediatrics, The University of Rochester, Rochester, NY 14642, USA
| | - Elaine A Sia
- Department of Biology, The University of Rochester, Rochester, NY 14642, USA
| | - Paul S Brookes
- Department of Anesthesiology, The University of Rochester, Rochester, NY 14642, USA
| | - Michael A O'Reilly
- Department of Pediatrics, The University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
14
|
Shadel GS. Live longer on MARS: a yeast paradigm of mitochondrial adaptive ROS signaling in aging. MICROBIAL CELL (GRAZ, AUSTRIA) 2014; 1:140-144. [PMID: 28357235 PMCID: PMC5354597 DOI: 10.15698/mic2014.05.143] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 04/14/2014] [Indexed: 01/05/2023]
Abstract
Adaptive responses to stress, including hormesis, have been implicated in longevity, but their mechanisms and outcomes are not fully understood. Here, I briefly summarize a longevity mechanism elucidated in the budding yeast chronological lifespan model by which Mitochondrial Adaptive ROS Signaling (MARS) promotes beneficial epigenetic and metabolic remodeling. The potential relevance of MARS to the human disease Ataxia-Telangiectasia and as a potential anti-aging target is discussed.
Collapse
Affiliation(s)
- Gerald S. Shadel
- Departments of Pathology and Genetics, Yale School of Medicine; New
Haven, CT 06437-8023, USA
| |
Collapse
|
15
|
Katou Y, Endo N, Suzuki T, Yu J, Kikuchi H, Oshima Y, Homma Y. Metarhizin A suppresses cell proliferation by inhibiting cytochrome c oxidase activity. Life Sci 2014; 103:1-7. [PMID: 24699005 DOI: 10.1016/j.lfs.2014.03.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 03/04/2014] [Accepted: 03/15/2014] [Indexed: 11/16/2022]
Abstract
AIMS Metarhizin A was originally isolated from Metarhizium flavoviride as a potent inhibitor of the growth of insect and mammalian cells. In this study, we aimed to understand the molecular targets of metarhizin A involved in its anti-proliferative activity against human cells. MAIN METHODS Cell cycle regulators and signaling molecules were examined by immunoblotting using specific antibodies. A mitochondria-enriched fraction was prepared from mouse liver, and mitochondrial activity was monitored using an oxygen electrode. Enzyme activity was measured using purified cytochrome c oxidase and permeabilized cells. KEY FINDINGS Metarhizin A inhibits the growth of MCF-7 cells with an IC50 value of ~0.2 μM and other cells in a similar manner; a cell cycle-dependent kinase inhibitor, p21, is selectively induced. Significant amounts of reactive oxygen species (ROS) are generated and ERK1/2 is activated in cells treated with metarhizin A. Metarhizin A completely suppresses oxygen consumption by mitochondria, and potently inhibits the activity of cytochrome c oxidase. It induces cell death when MCF-7 cells are cultured under limiting conditions. SIGNIFICANCE Metarhizin A is a potent inhibitor of cytochrome c oxidase and activates the MAPK pathway through the generation of ROS, which induces growth arrest of cells, and, under some conditions, enhances cell death. The cytochrome c oxidase system is a possible molecular target of metarhizin A.
Collapse
Affiliation(s)
- Yasuhiro Katou
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8678, Japan
| | - Naoya Endo
- Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Toshiyuki Suzuki
- Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Jiang Yu
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8678, Japan
| | - Haruhisa Kikuchi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8678, Japan
| | - Yoshiteru Oshima
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8678, Japan
| | - Yoshimi Homma
- Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan.
| |
Collapse
|
16
|
The Aspergillus nidulans ATM kinase regulates mitochondrial function, glucose uptake and the carbon starvation response. G3-GENES GENOMES GENETICS 2014; 4:49-62. [PMID: 24192833 PMCID: PMC3887539 DOI: 10.1534/g3.113.008607] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Mitochondria supply cellular energy and also perform a role in the adaptation to metabolic stress. In mammals, the ataxia-telangiectasia mutated (ATM) kinase acts as a redox sensor controlling mitochondrial function. Subsequently, transcriptomic and genetic studies were utilized to elucidate the role played by a fungal ATM homolog during carbon starvation. In Aspergillus nidulans, AtmA was shown to control mitochondrial function and glucose uptake. Carbon starvation responses that are regulated by target of rapamycin (TOR) were shown to be AtmA-dependent, including autophagy and hydrolytic enzyme secretion. AtmA also regulated a p53-like transcription factor, XprG, inhibiting starvation-induced XprG-dependent protease secretion and cell death. Thus, AtmA possibly represents a direct or indirect link between mitochondrial stress, metabolism, and growth through the influence of TOR and XprG function. The coordination of cell growth and division with nutrient availability is crucial for all microorganisms to successfully proliferate in a heterogeneous environment. Mitochondria supply cellular energy but also perform a role in the adaptation to metabolic stress and the cross-talk between prosurvival and prodeath pathways. The present study of Aspergillus nidulans demonstrated that AtmA also controlled mitochondrial mass, function, and oxidative phosphorylation, which directly or indirectly influenced glucose uptake. Carbon starvation responses, including autophagy, shifting metabolism to the glyoxylate cycle, and the secretion of carbon scavenging enzymes were AtmA-dependent. Transcriptomic profiling of the carbon starvation response demonstrated how TOR signaling and the retrograde response, which signals mitochondrial dysfunction, were directly or indirectly influenced by AtmA. The AtmA kinase was also shown to influence a p53-like transcription factor, inhibiting starvation-induced XprG-dependent protease secretion and cell death. Therefore, in response to metabolic stress, AtmA appears to perform a role in the regulation of TOR signaling, involving the retrograde and SnfA pathways. Thus, AtmA may represent a link between mitochondrial function and cell cycle or growth, possibly through the influence of the TOR and XprG function.
Collapse
|
17
|
Sharma NK, Lebedeva M, Thomas T, Kovalenko OA, Stumpf JD, Shadel GS, Santos JH. Intrinsic mitochondrial DNA repair defects in Ataxia Telangiectasia. DNA Repair (Amst) 2014; 13:22-31. [PMID: 24342190 PMCID: PMC6211587 DOI: 10.1016/j.dnarep.2013.11.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 11/12/2013] [Accepted: 11/26/2013] [Indexed: 12/28/2022]
Abstract
Ataxia Telangiectasia (A-T) is a progressive childhood disorder characterized most notably by cerebellar degeneration and predisposition to cancer. A-T is caused by mutations in the kinase ATM, a master regulator of the DNA double-strand break response. In addition to DNA-damage signaling defects, A-T cells display mitochondrial dysfunction that is thought to contribute to A-T pathogenesis. However, the molecular mechanism leading to mitochondrial dysfunction in A-T remains unclear. Here, we show that lack of ATM leads to reduced mitochondrial DNA (mtDNA) integrity and mitochondrial dysfunction, which are associated to defective mtDNA repair. While protein levels of mtDNA repair proteins are essentially normal, in the absence of ATM levels specifically of DNA ligase III (Lig3), the only DNA ligase working in mitochondria is reduced. The reduction of Lig3 is observed in different A-T patient cells, in brain and pre-B cells derived from ATM knockout mice as well as upon transient or stable knockdown of ATM. Furthermore, pharmacological inhibition of Lig3 in wild type cells phenocopies the mtDNA repair defects observed in A-T patient cells. As targeted deletion of LIG3 in the central nervous system causes debilitating ataxia in mice, reduced Lig3 protein levels and the consequent mtDNA repair defect may contribute to A-T neurodegeneration. A-T is thus the first disease characterized by diminished Lig3.
Collapse
Affiliation(s)
- Nilesh K Sharma
- Department of Pharmacology and Physiology, New Jersey Medical School of UMDNJ, 185 South Orange Avenue, Medical Sciences Building, Newark, NJ 07103, United States
| | - Maria Lebedeva
- Department of Genetics, Yale School of Medicine, 310 Cedar Street, BML 371, New Haven, CT 06520, United States
| | - Terace Thomas
- Department of Pharmacology and Physiology, New Jersey Medical School of UMDNJ, 185 South Orange Avenue, Medical Sciences Building, Newark, NJ 07103, United States
| | - Olga A Kovalenko
- Department of Pharmacology and Physiology, New Jersey Medical School of UMDNJ, 185 South Orange Avenue, Medical Sciences Building, Newark, NJ 07103, United States
| | - Jeffrey D Stumpf
- Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences (NIEHS), 111 TW Alexander Drive, Building 101, Durham, NC 27709, United States
| | - Gerald S Shadel
- Department of Genetics, Yale School of Medicine, 310 Cedar Street, BML 371, New Haven, CT 06520, United States; Department of Pathology, Yale School of Medicine, 310 Cedar Street, BML 371, New Haven, CT 06520, United States
| | - Janine H Santos
- Department of Pharmacology and Physiology, New Jersey Medical School of UMDNJ, 185 South Orange Avenue, Medical Sciences Building, Newark, NJ 07103, United States.
| |
Collapse
|
18
|
Abstract
In 1988, the gene responsible for the autosomal recessive disease ataxia- telangiectasia (A-T) was localized to 11q22.3-23.1. It was eventually cloned in 1995. Many independent laboratories have since demonstrated that in replicating cells, ataxia telangiectasia mutated (ATM) is predominantly a nuclear protein that is involved in the early recognition and response to double-stranded DNA breaks. ATM is a high-molecular-weight PI3K-family kinase. ATM also plays many important cytoplasmic roles where it phosphorylates hundreds of protein substrates that activate and coordinate cell-signaling pathways involved in cell-cycle checkpoints, nuclear localization, gene transcription and expression, the response to oxidative stress, apoptosis, nonsense-mediated decay, and others. Appreciating these roles helps to provide new insights into the diverse clinical phenotypes exhibited by A-T patients-children and adults alike-which include neurodegeneration, high cancer risk, adverse reactions to radiation and chemotherapy, pulmonary failure, immunodeficiency, glucose transporter aberrations, insulin-resistant diabetogenic responses, and distinct chromosomal and chromatin changes. An exciting recent development is the ATM-dependent pathology encountered in mitochondria, leading to inefficient respiration and energy metabolism and the excessive generation of free radicals that themselves create life-threatening DNA lesions that must be repaired within minutes to minimize individual cell losses.
Collapse
|
19
|
Suetomi K, Mereiter S, Mori C, Takanami T, Higashitani A. Caenorhabditis elegans ATR checkpoint kinase ATL-1 influences life span through mitochondrial maintenance. Mitochondrion 2013; 13:729-35. [PMID: 23434802 DOI: 10.1016/j.mito.2013.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 01/24/2013] [Accepted: 02/13/2013] [Indexed: 11/17/2022]
Abstract
ATR is highly conserved in all eukaryotes and functions as a cell-cycle nuclear checkpoint kinase. In mammals, ATR is essential whose complete absence results in early embryonic lethality and its hypomorphic mutation causes a complex disease known as Seckel syndrome. However, molecular mechanisms that cause a wide variety of symptoms including accelerated aging have remained unclear. Similarly, in the nematode Caenorhabditis elegans, a deletion mutant of ATR ortholog atl-1 appears to develop into normal adults, but their eggs do not hatch and die at early embryogenesis. Here we show that the parental worms of atl-1 defective mutant achieved longevity. Transcription levels of certain superoxide dismutase genes, sod-3 and -5 and enzymatic activity of superoxide dismutases significantly increased in the mutant. Furthermore, lipid peroxidation such as a formation of malondialdehyde was attenuated. Expressions of other genes regulated by DAF-16/FOXO transcription factor were also altered. In contrast, the mutant became hypersensitive to rotenone and ethidium bromide. Compared with the wild type the mitochondrial DNA copy number in the mutant was lesser and its proliferation is more severely inhibited in the presence of rotenone. These results suggest that C. elegans ATL-1 is involved not only in the nuclear checkpoint control but also in the mitochondrial maintenance, and its dysfunction activates mild oxidative stress response, resulting in an alteration of life span.
Collapse
Affiliation(s)
- Kazuhiro Suetomi
- Graduate School of Life Sciences, Tohoku University, Sendai, 980-8577, Japan
| | | | | | | | | |
Collapse
|
20
|
Ching JK, Spears LD, Armon JL, Renth AL, Andrisse S, Collins RL, Fisher JS. Impaired insulin-stimulated glucose transport in ATM-deficient mouse skeletal muscle. Appl Physiol Nutr Metab 2012; 38:589-96. [PMID: 23724874 DOI: 10.1139/apnm-2012-0175] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There are reports that ataxia telangiectasia mutated (ATM) plays a role in insulin-stimulated Akt phosphorylation, although this is not the case in some cell types. Because Akt plays a key role in insulin signaling, which leads to glucose transport in skeletal muscle, the predominant tissue in insulin-stimulated glucose disposal, we examined whether insulin-stimulated Akt phosphorylation and (or) glucose transport would be decreased in skeletal muscle of mice lacking functional ATM, compared with muscle from wild-type mice. We found that in vitro insulin-stimulated Akt phosphorylation was normal in soleus muscle from mice with 1 nonfunctional allele of ATM (ATM+/-) and from mice with 2 nonfunctional alleles (ATM-/-). However, insulin did not stimulate glucose transport or the phosphorylation of AS160 in ATM-/- soleus. ATM protein level was markedly higher in wild-type extensor digitorum longus (EDL) than in wild-type soleus. In EDL from ATM-/- mice, insulin did not stimulate glucose transport. However, in contrast to findings for soleus, insulin-stimulated Akt phosphorylation was blunted in ATM-/- EDL, concomitant with a tendency for insulin-stimulated phosphatidylinositol 3-kinase activity to be decreased. Together, the findings suggest that ATM plays a role in insulin-stimulated glucose transport at the level of AS160 in muscle comprised of slow and fast oxidative-glycolytic fibers (soleus) and at the level of Akt in muscle containing fast glycolytic fibers (EDL).
Collapse
Affiliation(s)
- James Kain Ching
- Department of Biology, Saint Louis University, 3507 Laclede Ave., St. Louis, MO 63103, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Zakikhani M, Bazile M, Hashemi S, Javeshghani S, Avizonis D, Pierre JS, Pollak MN. Alterations in cellular energy metabolism associated with the antiproliferative effects of the ATM inhibitor KU-55933 and with metformin. PLoS One 2012. [PMID: 23185347 PMCID: PMC3504012 DOI: 10.1371/journal.pone.0049513] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
KU-55933 is a specific inhibitor of the kinase activity of the protein encoded by Ataxia telangiectasia mutated (ATM), an important tumor suppressor gene with key roles in DNA repair. Unexpectedly for an inhibitor of a tumor suppressor gene, KU-55933 reduces proliferation. In view of prior preliminary evidence suggesting defective mitochondrial function in cells of patients with Ataxia Telangiectasia (AT), we examined energy metabolism of cells treated with KU-55933. The compound increased AMPK activation, glucose uptake and lactate production while reducing mitochondrial membrane potential and coupled respiration. The stimulation of glycolysis by KU-55933 did not fully compensate for the reduction in mitochondrial functions, leading to decreased cellular ATP levels and energy stress. These actions are similar to those previously described for the biguanide metformin, a partial inhibitor of respiratory complex I. Both compounds decreased mitochondrial coupled respiration and reduced cellular concentrations of fumarate, malate, citrate, and alpha-ketogluterate. Succinate levels were increased by KU-55933 levels and decreased by metformin, indicating that the effects of ATM inhibition and metformin are not identical. These observations suggest a role for ATM in mitochondrial function and show that both KU-55933 and metformin perturb the TCA cycle as well as oxidative phosphorylation.
Collapse
Affiliation(s)
- Mahvash Zakikhani
- Department of Oncology, McGill University, Montreal, Quebec, Canada
- Division of Cancer Prevention, McGill University, Montreal, Quebec, Canada
| | - Miguel Bazile
- Division of Cancer Prevention, McGill University, Montreal, Quebec, Canada
| | - Sina Hashemi
- Division of Cancer Prevention, McGill University, Montreal, Quebec, Canada
| | - Shiva Javeshghani
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Daina Avizonis
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Julie St Pierre
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Michael N. Pollak
- Department of Oncology, McGill University, Montreal, Quebec, Canada
- Division of Cancer Prevention, McGill University, Montreal, Quebec, Canada
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
22
|
Lin CS, Wang YC, Huang JL, Hung CC, Chen JYF. Autophagy and reactive oxygen species modulate cytotoxicity induced by suppression of ATM kinase activity in head and neck cancer cells. Oral Oncol 2012; 48:1152-8. [PMID: 22763242 DOI: 10.1016/j.oraloncology.2012.05.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 05/22/2012] [Accepted: 05/25/2012] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Because Ataxia Telangiectasia Mutated (ATM)-deficient cells are hypersensitive to ionizing irradiation and DNA-damaging agents, ATM kinase inhibition is thought to enhance radiochemotherapy efficacy. In this study, we investigated the roles of autophagy and reactive oxygen species (ROS) in modulating cytotoxicity induced by suppression of ATM kinase in head and neck cancer cells. MATERIALS AND METHODS We use KU55933 to inhibit ATM kinase activity. The cell viability was determined by MTT assays. Autophagy was examined by Western blot for LC3-II and microscopy for acidic vesicles and EGFP-LC3 punctate formation. DCF-DA staining and flow cytometry were used for analyzing ROS generation. RESULTS we found that KU55933 reduced cell viability in several head and neck cancer cell lines. KU55933-treated cells showed increased cytoplasmic vesicles, LC3-II accumulation, and EGFP-LC3 punctate formation, indicating that autophagy was induced. KU55933 also increased ROS generation, which was required for autophagy induction because the ROS scavenger N-acetyl-L-cysteine could reduce LC3-II accumulation. KU55933-induced autophagy played a cytoprotective role against ROS-mediated cytotoxicity because autophagy inhibition by chloroquine augmented KU55933's cytotoxicity. In addition, KU55933 reduced cisplatin-resistant head and neck cancer cell viabilities, and induced LC3-II accumulation in these cells. CONCLUSION Together, these results shed light on KU55933's therapeutic values as well as autophagy inhibitors in treating primary and cisplatin-resistant head and neck cancers.
Collapse
Affiliation(s)
- Chang-Shen Lin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | | | | | | | | |
Collapse
|
23
|
Nayler S, Gatei M, Kozlov S, Gatti R, Mar JC, Wells CA, Lavin M, Wolvetang E. Induced pluripotent stem cells from ataxia-telangiectasia recapitulate the cellular phenotype. Stem Cells Transl Med 2012. [PMID: 23197857 DOI: 10.5966/sctm.2012-0024] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Pluripotent stem cells can differentiate into every cell type of the human body. Reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) therefore provides an opportunity to gain insight into the molecular and cellular basis of disease. Because the cellular DNA damage response poses a barrier to reprogramming, generation of iPSCs from patients with chromosomal instability syndromes has thus far proven to be difficult. Here we demonstrate that fibroblasts from patients with ataxia-telangiectasia (A-T), a disorder characterized by chromosomal instability, progressive neurodegeneration, high risk of cancer, and immunodeficiency, can be reprogrammed to bona fide iPSCs, albeit at a reduced efficiency. A-T iPSCs display defective radiation-induced signaling, radiosensitivity, and cell cycle checkpoint defects. Bioinformatic analysis of gene expression in the A-T iPSCs identifies abnormalities in DNA damage signaling pathways, as well as changes in mitochondrial and pentose phosphate pathways. A-T iPSCs can be differentiated into functional neurons and thus represent a suitable model system to investigate A-T-associated neurodegeneration. Collectively, our data show that iPSCs can be generated from a chromosomal instability syndrome and that these cells can be used to discover early developmental consequences of ATM deficiency, such as altered mitochondrial function, that may be relevant to A-T pathogenesis and amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Sam Nayler
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Progressive DNA damage and mitochondrial decline are both considered to be prime instigators of natural ageing. Traditionally, these two pathways have been viewed largely in isolation. However, recent studies have revealed a molecular circuit that directly links DNA damage to compromised mitochondrial biogenesis and function via p53. This axis of ageing may account for both organ decline and disease development associated with advanced age and could illuminate a path for the development of relevant therapeutics.
Collapse
|
25
|
Oliveras-Ferraros C, Cufí S, Vazquez-Martin A, Menendez OJ, Bosch-Barrera J, Martin-Castillo B, Joven J, Menendez JA. Metformin rescues cell surface major histocompatibility complex class I (MHC-I) deficiency caused by oncogenic transformation. Cell Cycle 2012; 11:865-70. [PMID: 22333588 DOI: 10.4161/cc.11.5.19252] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Active avoidance by tumor cells from attack and elimination by immune cells is an emerging cancer hallmark that is achieved primarily through decreasing the levels of major histocompatibility complex class I (MHC-I) at the cancer cells' surface. Deficiencies in MHC-I antigen-restricted immunosurveillance may be intertwined with an altered, Warburg-like cancer cell-intrinsic metabolism, another emerging hallmark of cancer that involves a switch from mitochondrial respiration to glycolysis to efficiently support large-scale biosynthetic programs that are required for active cell proliferation. We recently envisioned that intervention strategies aimed at reversing the bioenergetic signature of cancer cells (e.g., the antidiabetic biguanide metformin) should correct oncogene (e.g., HER2)-driven MHC-I defects, thus preventing immune escape of oncogene transformants. First, we explored how metformin treatment impacted mitochondrial biogenesis in cultured breast cancer cells overexpressing the membrane tyrosine kinase receptor HER2, the best-characterized downregulator of MHC-I. Metformin exposure was found to dose-dependently increase the expression levels of cytochrome c oxidase I and mitochondrial succinate dehydrogenase, which are encoded by mitochondrial and nuclear DNA, respectively. Second, we explored whether metformin-enhanced mitochondrial biogenesis might significantly alter the MHC-I status in breast carcinoma cells. MHC-I expression, as assessed by flow cytometry using an anti-HLA-ABC monoclonal antibody, was fully restored (up to ~25-fold upregulation) in MHC-I-negative HER2 gene-amplified carcinoma cells. These findings may help delineate a previously unrecognized mechanism through which metformin (and metformin-like drugs) may enable a cancer patient's own immune system to mount an efficient anti-metastasis response that can prevent or delay disease recurrence. Restored antigenicity and immunogenicity of tumor cells may represent a previously unrecognized primary mode of action underlying the cancer-preventive effects of metformin.
Collapse
|
26
|
The ATM protein kinase and cellular redox signaling: beyond the DNA damage response. Trends Biochem Sci 2011; 37:15-22. [PMID: 22079189 DOI: 10.1016/j.tibs.2011.10.002] [Citation(s) in RCA: 247] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 10/06/2011] [Accepted: 10/07/2011] [Indexed: 01/29/2023]
Abstract
The ataxia-telangiectasia mutated (ATM) protein kinase is best known for its role in the DNA damage response, but recent findings suggest that it also functions as a redox sensor that controls the levels of reactive oxygen species in human cells. Here, we review evidence supporting the conclusion that ATM can be directly activated by oxidation, as well as various observations from ATM-deficient patients and mouse models that point to the importance of ATM in oxidative stress responses. We also discuss the roles of this kinase in regulating mitochondrial function and metabolic control through its action on tumor suppressor p53, AMP-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR) and hypoxia-inducible factor 1 (HIF1), and how the regulation of these enzymes may be affected in ATM-deficient patients and in cancer cells.
Collapse
|