1
|
Ali R, Algethami M, Sheha A, Alqahtani S, Altayyar A, Lashen A, Rakha E, Sulaiman AA, Madhusudan S, Ramotar D. PRDX1 protects ATM from arsenite-induced proteotoxicity and maintains its stability during DNA damage signaling. Oncotarget 2025; 16:362-378. [PMID: 40387816 PMCID: PMC12088036 DOI: 10.18632/oncotarget.28720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/17/2025] [Indexed: 05/20/2025] Open
Abstract
Redox regulation and DNA repair coordination are essential for genomic stability. Peroxiredoxin 1 (PRDX1) is a thiol-dependent peroxidase and a chaperone that protects proteins from excessive oxidation. ATM kinase (Ataxia-Telangiectasia Mutated) and the MRN (MRE11-RAD50-NBS1) complex are DNA damage signaling and repair proteins. We previously showed that cells lacking PRDX1 are sensitive to arsenite, a toxic metal that induces DNA single- and double-strand breaks (DSBs). Herein, we showed that PRDX1 interacts with ATM. PRDX1-deleted cells have reduced ATM, MRE11, and RAD50 protein levels, but not NBS1. In control cells treated with arsenite, we observed γH2AX foci formation due to arsenite-induced DSBs, and not from PRDX1-deleted cells. Arsenite caused profound depletion of ATM in PRDX1-deleted cells, suggesting that PRDX1 protects and stabilizes ATM required to form γH2AX foci. Importantly, arsenite pretreatment of PRDX1-deleted cells caused hypersensitivity to chemotherapeutic agents that generate DSBs. Analysis of a clinical cohort of ovarian cancers treated with platinum chemotherapy revealed that tumours with high PRDX1/high ATM or high PRDX1/high MRE11 expression manifested aggressive phenotypes and poor patient survival. The data suggest that PRDX1 can predict responses to chemotherapy, and targeting PRDX1 could be a viable strategy to improve the efficacy of platinum chemotherapy.
Collapse
Affiliation(s)
- Reem Ali
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Mashael Algethami
- Naaz Coker Ovarian Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK
| | - Amera Sheha
- Naaz Coker Ovarian Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK
| | - Shatha Alqahtani
- Naaz Coker Ovarian Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK
| | - Ahmad Altayyar
- Naaz Coker Ovarian Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK
| | - Ayat Lashen
- Naaz Coker Ovarian Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK
| | - Emad Rakha
- Naaz Coker Ovarian Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK
- Department of Pathology, Nottingham University Hospitals, City Hospital Campus, Nottingham NG5 1PB, UK
| | | | - Srinivasan Madhusudan
- Naaz Coker Ovarian Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK
- Department of Oncology, Nottingham University Hospitals, City Hospital Campus, Nottingham NG5 1PB, UK
| | - Dindial Ramotar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
2
|
Dematteis G, Lecchi G, Boni G, Pendin D, Distasi C, Grilli M, Lim D, Fresu LG, Talmon M. ATM knock out alters calcium signalling and augments contraction in skeletal muscle cells differentiated from human urine-derived stem cells. Cell Death Discov 2025; 11:177. [PMID: 40234386 PMCID: PMC12000312 DOI: 10.1038/s41420-025-02485-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 04/17/2025] Open
Abstract
Ataxia-telangiectasia (A-T) is a rare neurodegenerative disorder caused by the deficiency of the serine/threonine kinase ataxia telangiectasia mutated (ATM) protein, whose loss of function leads to altered cell cycle, apoptosis, oxidative stress balance and DNA repair after damage. The clinical manifestations are multisystemic, among them cerebellar degeneration and muscular ataxia. The molecular mechanism by which ATM loss leads to A-T is still uncertain and, currently only symptomatic treatments are available. In this study, we generated a functional skeletal muscle cell model that recapitulates A-T and highlights the role of ATM in calcium signalling and muscle contraction. To this aim, by using CRISPR/Cas9 technology, we knocked out the ATM protein in urine-derived stem cells (USCs) from healthy donors. The resulting USCs-ATM-KO maintained stemness but showed G2/S cell cycle progression and an inability to repair DNA after UV damage. Moreover, they showed increased cytosolic calcium release after ATP stimulation to the detriment of the mitochondria. The alterations of calcium homoeostasis were maintained after differentiation of USCs-ATM-KO into skeletal muscle cells (USC-SkMCs) and correlated with impaired cell contraction. Indeed, USC-SkMCs-ATM-KO contraction kinetics were dramatically accelerated compared to control cells. These results highlight the relevant function of ATM in skeletal muscle, which is not only dependent on a non-functional neuronal communication, paving the way for future studies on a muscular interpretation of A-T ataxia.
Collapse
Grants
- C13C22000590006 - ID T4-AN-04 Ministry of Health, Italy | Agenzia Italiana del Farmaco, Ministero della Salute (Italian Medicines Agency)
- C13C22000590006 - ID T4-AN-04 Ministry of Health, Italy | Agenzia Italiana del Farmaco, Ministero della Salute (Italian Medicines Agency)
- C13C22000590006 - ID T4-AN-04 Ministry of Health, Italy | Agenzia Italiana del Farmaco, Ministero della Salute (Italian Medicines Agency)
- C13C22000590006 - ID T4-AN-04 Ministry of Health, Italy | Agenzia Italiana del Farmaco, Ministero della Salute (Italian Medicines Agency)
- C13C22000590006 - ID T4-AN-04 Ministry of Health, Italy | Agenzia Italiana del Farmaco, Ministero della Salute (Italian Medicines Agency)
- C13C22000590006 - ID T4-AN-04 Ministry of Health, Italy | Agenzia Italiana del Farmaco, Ministero della Salute (Italian Medicines Agency)
- C13C22000590006 - ID T4-AN-04 Ministry of Health, Italy | Agenzia Italiana del Farmaco, Ministero della Salute (Italian Medicines Agency)
- C13C22000590006 - ID T4-AN-04 Ministry of Health, Italy | Agenzia Italiana del Farmaco, Ministero della Salute (Italian Medicines Agency)
Collapse
Affiliation(s)
- Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Giulia Lecchi
- Department of Health Sciences, School of Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Giulia Boni
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Diana Pendin
- Neuroscience Institute, Padua Section, National Research Council, Padua, Italy
| | - Carla Distasi
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Mariagrazia Grilli
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy.
| | - Luigia Grazia Fresu
- Department of Health Sciences, School of Medicine, Università del Piemonte Orientale, Novara, Italy.
| | - Maria Talmon
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
3
|
Abrahams RR, Majumder K. Small Genomes, Big Disruptions: Parvoviruses and the DNA Damage Response. Viruses 2025; 17:494. [PMID: 40284937 PMCID: PMC12031541 DOI: 10.3390/v17040494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Parvoviruses are small, single-stranded DNA viruses that have evolved sophisticated mechanisms to hijack host cell machinery for replication and persistence. One critical aspect of this interaction involves the manipulation of the host's DNA Damage Response (DDR) pathways. While the viral genome is comparatively simple, parvoviruses have developed strategies that cause significant DNA damage, activate DDR pathways, and disrupt the host cell cycle. This review will explore the impact of parvovirus infections on host genome stability, focusing on key viral species such as Adeno-Associated Virus (AAV), Minute Virus of Mice (MVM), and Human Bocavirus (HBoV), and their interactions with DDR proteins. Since parvoviruses are used as oncolytic agents and gene therapy vectors, a better understanding of cellular DDR pathways will aid in engineering potent anti-cancer agents and gene therapies for chronic diseases.
Collapse
Affiliation(s)
| | - Kinjal Majumder
- Institute for Molecular Virology and McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI 53707, USA;
| |
Collapse
|
4
|
Li H, Furusawa T, Cavero R, Xiao Y, Chari R, Wu X, Sun D, Hartmann O, Dhall A, Holewinski R, Andresson T, Karim B, Villamor-Payà M, Gallardo D, Day CP, Pal LR, Nair NU, Ruppin E, Aladjem MI, Pommier Y, Diefenbacher ME, Lim JM, Levine RL, Stracker TH, Weyemi U. Metabolic dependency mapping identifies Peroxiredoxin 1 as a driver of resistance to ATM inhibition. Redox Biol 2025; 80:103503. [PMID: 39854937 PMCID: PMC11795153 DOI: 10.1016/j.redox.2025.103503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/12/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Metabolic pathways fuel tumor progression and resistance to stress conditions including chemotherapeutic drugs, such as DNA damage response (DDR) inhibitors. Yet, significant gaps persist in how metabolic pathways confer resistance to DDR inhibition in cancer cells. Here, we employed a metabolism-focused CRISPR knockout screen and identified genetic vulnerabilities to DDR inhibitors. We unveiled Peroxiredoxin 1 (PRDX1) as a synthetic lethality partner with Ataxia Telangiectasia Mutated (ATM) kinase. Tumor cells depleted of PRDX1 displayed heightened sensitivity to ATM inhibition in vitro and in mice in a manner dependent on p53 status. Mechanistically, we discovered that the ribosomal protein RPL32 undergoes redox modification on active cysteine residues 91 and 96 upon ATM inhibition, promoting p53 stability and altered cell fitness. Our findings reveal a new pathway whereby RPL32 senses stress and induces p53 activation impairing tumor cell survival.
Collapse
Affiliation(s)
- Haojian Li
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute/National Institutes of Health, 37 Convent Drive, Bethesda, MD, 20892, USA; Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Takashi Furusawa
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute/National Institutes of Health, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Renzo Cavero
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute/National Institutes of Health, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Yunjie Xiao
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute/National Institutes of Health, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Raj Chari
- Genome Modification Core, Laboratory Animal Sciences Program, Frederick, MD, USA
| | - Xiaolin Wu
- NCI Genomics Technology Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research/ Frederick, Maryland, USA
| | - David Sun
- NCI Genomics Technology Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research/ Frederick, Maryland, USA
| | - Oliver Hartmann
- Comprehensive Pneumology Center (CPC)/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL/CPC-M), Munich, Germany
| | - Anjali Dhall
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute/National Institutes of Health, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Ronald Holewinski
- Protein Characterization Laboratory/Cancer Research Technology Program/Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Thorkell Andresson
- Protein Characterization Laboratory/Cancer Research Technology Program/Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Baktiar Karim
- Molecular Histopathology Laboratory/ Cancer Research Technology Program/Frederick National Laboratory for Cancer Research/ Frederick, Maryland, USA
| | | | - Devorah Gallardo
- Laboratory Animal Sciences Program, Leidos Biomedical Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chi-Ping Day
- Cancer Data Science Lab/ Center for Cancer Research/National Cancer Institute/National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lipika R Pal
- Cancer Data Science Lab/ Center for Cancer Research/National Cancer Institute/National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nishanth Ulhas Nair
- Cancer Data Science Lab/ Center for Cancer Research/National Cancer Institute/National Institutes of Health, Bethesda, MD, 20892, USA
| | - Eytan Ruppin
- Cancer Data Science Lab/ Center for Cancer Research/National Cancer Institute/National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mirit I Aladjem
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute/National Institutes of Health, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Yves Pommier
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute/National Institutes of Health, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Markus E Diefenbacher
- Comprehensive Pneumology Center (CPC)/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL/CPC-M), Munich, Germany
| | - Jung Mi Lim
- Laboratory of Biochemistry, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Rodney L Levine
- Laboratory of Biochemistry, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | | | - Urbain Weyemi
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute/National Institutes of Health, 37 Convent Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
5
|
Lee JH. ATM in immunobiology: From lymphocyte development to cancer immunotherapy. Transl Oncol 2025; 52:102268. [PMID: 39752906 PMCID: PMC11754496 DOI: 10.1016/j.tranon.2024.102268] [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: 08/13/2024] [Revised: 11/14/2024] [Accepted: 12/30/2024] [Indexed: 01/25/2025] Open
Abstract
Ataxia Telangiectasia Mutated (ATM) is a protein kinase traditionally known for its role in DNA damage response and cell cycle regulation. However, emerging research has revealed its multifaceted and crucial functions in the immune system. This comprehensive review explores the diverse roles of ATM in immune regulation, from lymphocyte development to its involvement in cancer immunotherapy. The review describes ATM's critical functions in V(D)J recombination and class switch recombination, highlighting its importance in adaptive immunity. It examines ATM's role in innate immunity, particularly in NF-κB signaling and cytokine production. Furthermore, the review analyzes the impact of ATM deficiency on oxidative stress and mitochondrial function in immune cells, providing insights into the immunological defects observed in Ataxia Telangiectasia (A-T). The article explores ATM's significance in maintaining hematopoietic stem cell function and its implications for bone marrow transplantation and gene therapy. Additionally, it addresses ATM's involvement in inflammation and immune senescence, linking DNA damage response to age-related immune decline. Finally, this review highlights the emerging role of ATM in cancer immunotherapy, where its inhibition shows promise in enhancing immune checkpoint blockade therapy. This review synthesizes current knowledge on ATM's functions in the immune system, offering insights into the pathophysiology of ATM-related disorders and potential therapeutic strategies for immune-related conditions and cancer immunotherapy.
Collapse
Affiliation(s)
- Ji-Hoon Lee
- Department of Biological Sciences, Research Center of Ecomimetics, Chonnam National University, Gwangju 61186, South Korea.
| |
Collapse
|
6
|
Cendali FI, Lisk C, Dzieciatkowska M, LaCroix IS, Reisz JA, Harral J, Stephenson D, Hay AM, Wartchow EP, Darehshouri A, Dziewulska-Cronk KH, Buehler PW, Norris PJ, Deng X, Busch MP, Earley EJ, Page GP, Hansen KC, Zimring JC, Irwin DC, Nemkov T, D’Alessandro A. Increased exercise tolerance in humanized G6PD-deficient mice. Blood Adv 2025; 9:321-334. [PMID: 39514761 PMCID: PMC11786683 DOI: 10.1182/bloodadvances.2024013968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/30/2024] [Accepted: 09/19/2024] [Indexed: 11/16/2024] Open
Abstract
ABSTRACT Glucose-6-phosphate dehydrogenase (G6PD) deficiency affects 500 million people globally, affecting red blood cell (RBC) antioxidant pathways and increasing susceptibility to hemolysis under oxidative stress. Despite the systemic generation of reactive oxygen species during exercise, the effects of exercise on individuals with G6PD deficiency remain poorly understood This study used humanized mouse models expressing the G6PD Mediterranean variant (S188F, with 10% enzymatic activity) to investigate exercise performance and molecular outcomes. Surprisingly, despite decreased enzyme activity, G6PD-deficient mice have faster critical speed than mice expressing human canonical G6PD. After exercise, deficient mice did not exhibit differences in RBC morphology or hemolysis, but had improved cardiac function, including cardiac output, stroke volume, sarcomere length, and mitochondrial content. Proteomics analyses of cardiac and skeletal muscles (gastrocnemius, soleus) from G6PD-deficient compared with sufficient mice revealed improvements in mitochondrial function and increased protein turnover via ubiquitination, especially for mitochondrial and structural myofibrillar proteins. Mass spectrometry-based metabolomics revealed alterations in energy metabolism and fatty acid oxidation. These findings challenge the traditional assumptions regarding hemolytic risk during exercise in G6PD deficiency, suggesting a potential metabolic advantage in exercise performance for individuals carrying noncanonical G6PD variants.
Collapse
Affiliation(s)
- Francesca I. Cendali
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Christina Lisk
- Department of Medicine, Cardiovascular and Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Ian S. LaCroix
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Julie Harral
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Ariel M. Hay
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA
| | - Eric P. Wartchow
- Department of Pathology, Children's Hospital Colorado, Aurora, CO
| | - Anza Darehshouri
- Electron Microscopy Core Facility, University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | - Paul W. Buehler
- Department of Pediatrics, Center for Blood Oxygen Transport, University of Maryland School of Medicine, Baltimore, MD
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD
| | | | - Xutao Deng
- Vitalant Research Institute, San Francisco, CA
| | | | | | - Grier P. Page
- Research Triangle Institute International, Atlanta, GA
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - James C. Zimring
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA
| | - David C. Irwin
- Department of Medicine, Cardiovascular and Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
7
|
Gong GQ, Anandapadamanaban M, Islam MS, Hay IM, Bourguet M, Špokaitė S, Dessus AN, Ohashi Y, Perisic O, Williams RL. Making PI3K superfamily enzymes run faster. Adv Biol Regul 2025; 95:101060. [PMID: 39592347 DOI: 10.1016/j.jbior.2024.101060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024]
Abstract
The phosphoinositide 3-kinase (PI3K) superfamily includes lipid kinases (PI3Ks and type III PI4Ks) and a group of PI3K-like Ser/Thr protein kinases (PIKKs: mTOR, ATM, ATR, DNA-PKcs, SMG1 and TRRAP) that have a conserved C-terminal kinase domain. A common feature of the superfamily is that they have very low basal activity that can be greatly increased by a range of regulatory factors. Activators reconfigure the active site, causing a subtle realignment of the N-lobe of the kinase domain relative to the C-lobe. This realignment brings the ATP-binding loop in the N-lobe closer to the catalytic residues in the C-lobe. In addition, a conserved C-lobe feature known as the PIKK regulatory domain (PRD) also can change conformation, and PI3K activators can alter an analogous PRD-like region. Recent structures have shown that diverse activating influences can trigger these conformational changes, and a helical region clamping onto the kinase domain transmits regulatory interactions to bring about the active site realignment for more efficient catalysis. A recent report of a small-molecule activator of PI3Kα for application in nerve regeneration suggests that flexibility of these regulatory elements might be exploited to develop specific activators of all PI3K superfamily members. These activators could have roles in wound healing, anti-stroke therapy and treating neurodegeneration. We review common structural features of the PI3K superfamily that may make them amenable to activation.
Collapse
Affiliation(s)
- Grace Q Gong
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK; University College London Cancer Institute, University College London, London, UK
| | | | - Md Saiful Islam
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Iain M Hay
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Maxime Bourguet
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Saulė Špokaitė
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Antoine N Dessus
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Yohei Ohashi
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Olga Perisic
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Roger L Williams
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK.
| |
Collapse
|
8
|
Nimalasena S, Anbalagan S, Box C, Yu S, Boult JKR, Bush N, Howell L, Sinnett V, Murphy W, Yarnold J, Robinson SP, Somaiah N. Tumour reoxygenation after intratumoural hydrogen peroxide (KORTUC) injection: a novel approach to enhance radiosensitivity. BJC REPORTS 2024; 2:78. [PMID: 39391329 PMCID: PMC11461272 DOI: 10.1038/s44276-024-00098-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/08/2024] [Accepted: 09/02/2024] [Indexed: 10/12/2024]
Abstract
Background KORTUC (0.5% hydrogen peroxide (H2O2) in 1% sodium-hyaluronate) releases cytotoxic levels of H2O2 in tissues after intratumoural injection. High levels of tumour control after radiotherapy plus KORTUC are reported in breast cancer patients. Here, we use human xenograft models to test the hypothesis that oxygen microbubbles released post-KORTUC are effective in modifying the hypoxic tumour microenvironment. Methods and materials Pimonidazole and Image-iT™ Red (live hypoxia marker) were utilised to assess dose-dependent changes in hypoxia post-H2O2 in HCT116 and LICR-LON-HN5 spheroids. Using a dual 2-nitroimidazole-marker technique and phospho-ATM we evaluated changes in hypoxia and reactive oxygen species (ROS) respectively, in HCT116 and LICR-LON-HN5 xenografts following intratumoural KORTUC. Results A significant reduction in Image-iT™ Red fluorescence was observed in spheroids 1 h post-H2O2 at ≥1.2 mM, maintained at 24 h. Ultrasound demonstrated sustained release of oxygen microbubbles within tumours, 1 h post-KORTUC. Hypoxia markers demonstrated significant tissue reoxygenation in both models post-KORTUC and significantly increased phospho-ATM foci reflecting increased ROS production. Conclusion Intratumoural KORTUC represents a novel oxygen delivery method, which can be exploited to enhance radiation response. If efficacy is confirmed in the ongoing phase 2 breast trial it could improve treatment of several tumour types where hypoxia is known to affect radiotherapy outcomes.
Collapse
Affiliation(s)
- Samantha Nimalasena
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, London, UK
| | - Selvakumar Anbalagan
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, UK
| | - Carol Box
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, UK
| | - Sheng Yu
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, UK
| | - Jessica K. R. Boult
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, UK
| | - Nigel Bush
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, UK
| | - Louise Howell
- Core Research Facilities, The Institute of Cancer Research, London, UK
| | | | - William Murphy
- Biological Service Unit, The Institute of Cancer Research, London, UK
| | - John Yarnold
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, UK
| | - Simon P. Robinson
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, UK
| | - Navita Somaiah
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|
9
|
Qian C, Li X, Zhang J, Wang Y. Small Molecular Inhibitors That Target ATM for Drug Discovery: Current Research and Potential Prospective. J Med Chem 2024; 67:14742-14767. [PMID: 39149790 DOI: 10.1021/acs.jmedchem.4c01064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The protein kinase ataxia telangiectasia mutated (ATM) is a constituent of the phosphatidylinositol 3-kinase-related kinase (PIKK) family, exerting a pivotal influence on diverse cellular processes, notably the signaling of double-strand DNA breaks (DSB) and stress response. The dysregulation of ATM is implicated in the pathogenesis of cancer and other diseases such as neurodegeneration. Hence, ATM is deemed a promising candidate for potential therapeutic interventions across a spectrum of diseases. Presently, while ATM small molecule inhibitors are not commercially available, various selective inhibitors have progressed to the clinical research phase. Specifically, AZD1390, WSD0628, SYH2051, and ZN-B-2262 are under investigation in clinical studies pertaining to glioblastoma multiforme and advanced solid tumors, respectively. In this Perspective, we encapsulate the structure, biological functions, and disease relevance of ATM. Subsequently, we concentrate on the design concepts and structure-activity relationships (SAR) of ATM inhibitors, delineating potential avenues for the development of more efficacious ATM-targeted inhibitors.
Collapse
Affiliation(s)
- Chunlin Qian
- Department of Respiratory and Critical Care Medicine and Targeted Tracer Research and Development Laboratory and Institute of Respiratory Health and State Key Laboratory of Respiratory Health and Multimorbidity and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, Sichuan China
| | - Xiaoxue Li
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan China
| | - Jifa Zhang
- Department of Respiratory and Critical Care Medicine and Targeted Tracer Research and Development Laboratory and Institute of Respiratory Health and State Key Laboratory of Respiratory Health and Multimorbidity and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, Sichuan China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan China
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine and Targeted Tracer Research and Development Laboratory and Institute of Respiratory Health and State Key Laboratory of Respiratory Health and Multimorbidity and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, Sichuan China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan China
| |
Collapse
|
10
|
Lee JH. Oxidative stress and the multifaceted roles of ATM in maintaining cellular redox homeostasis. Redox Biol 2024; 75:103269. [PMID: 39018798 PMCID: PMC11301354 DOI: 10.1016/j.redox.2024.103269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024] Open
Abstract
The ataxia telangiectasia mutated (ATM) protein kinase is best known as a master regulator of the DNA damage response. However, accumulating evidence has unveiled an equally vital function for ATM in sensing oxidative stress and orchestrating cellular antioxidant defenses to maintain redox homeostasis. ATM can be activated through a non-canonical pathway involving intermolecular disulfide crosslinking of the kinase dimers, distinct from its canonical activation by DNA double-strand breaks. Structural studies have elucidated the conformational changes that allow ATM to switch into an active redox-sensing state upon oxidation. Notably, loss of ATM function results in elevated reactive oxygen species (ROS) levels, altered antioxidant profiles, and mitochondrial dysfunction across multiple cell types and tissues. This oxidative stress arising from ATM deficiency has been implicated as a central driver of the neurodegenerative phenotypes in ataxia-telangiectasia (A-T) patients, potentially through mechanisms involving oxidative DNA damage, PARP hyperactivation, and widespread protein aggregation. Moreover, defective ATM oxidation sensing disrupts transcriptional programs and RNA metabolism, with detrimental impacts on neuronal homeostasis. Significantly, antioxidant therapy can ameliorate cellular and organismal abnormalities in various ATM-deficient models. This review synthesizes recent advances illuminating the multifaceted roles of ATM in preserving redox balance and mitigating oxidative insults, providing a unifying paradigm for understanding the complex pathogenesis of A-T disease.
Collapse
Affiliation(s)
- Ji-Hoon Lee
- Department of Biological Sciences, Research Center of Ecomimetics, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
11
|
Yu P, Gu T, Rao Y, Liang W, Zhang X, Jiang H, Lu J, She J, Guo J, Yang W, Liu Y, Tu Y, Tang L, Zhou X. A novel marine-derived anti-acute kidney injury agent targeting peroxiredoxin 1 and its nanodelivery strategy based on ADME optimization. Acta Pharm Sin B 2024; 14:3232-3250. [PMID: 39027260 PMCID: PMC11252462 DOI: 10.1016/j.apsb.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/28/2024] [Accepted: 02/07/2024] [Indexed: 07/20/2024] Open
Abstract
Insufficient therapeutic strategies for acute kidney injury (AKI) necessitate precision therapy targeting its pathogenesis. This study reveals the new mechanism of the marine-derived anti-AKI agent, piericidin glycoside S14, targeting peroxiredoxin 1 (PRDX1). By binding to Cys83 of PRDX1 and augmenting its peroxidase activity, S14 alleviates kidney injury efficiently in Prdx1-overexpression (Prdx1-OE) mice. Besides, S14 also increases PRDX1 nuclear translocation and directly activates the Nrf2/HO-1/NQO1 pathway to inhibit ROS production. Due to the limited druggability of S14 with low bioavailability (2.6%) and poor renal distribution, a pH-sensitive kidney-targeting dodecanamine-chitosan nanoparticle system is constructed to load S14 for precise treatment of AKI. l-Serine conjugation to chitosan imparts specificity to kidney injury molecule-1 (Kim-1)-overexpressed cells. The developed S14-nanodrug exhibits higher therapeutic efficiency by improving the in vivo behavior of S14 significantly. By encapsulation with micelles, the AUC0‒t , half-life time, and renal distribution of S14 increase 2.5-, 1.8-, and 3.1-fold, respectively. The main factors contributing to the improved druggability of S14 nanodrugs include the lower metabolic elimination rate and UDP-glycosyltransferase (UGT)-mediated biotransformation. In summary, this study identifies a new therapeutic target for the marine-derived anti-AKI agent while enhancing its ADME properties and druggability through nanotechnology, thereby driving advancements in marine drug development for AKI.
Collapse
Affiliation(s)
- Ping Yu
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tanwei Gu
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yueyang Rao
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Weimin Liang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xi Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Huanguo Jiang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jindi Lu
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jianglian She
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Jianmin Guo
- Guangdong Lewwin Pharmaceutical Research Institute Co., Ltd., Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, Guangdong Engineering Research Center for Innovative Drug Evaluation and Research, Guangzhou 510990, China
| | - Wei Yang
- Guangdong Lewwin Pharmaceutical Research Institute Co., Ltd., Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, Guangdong Engineering Research Center for Innovative Drug Evaluation and Research, Guangzhou 510990, China
| | - Yonghong Liu
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Yingfeng Tu
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lan Tang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xuefeng Zhou
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
12
|
Lai J, Demirbas D, Kim J, Jeffries AM, Tolles A, Park J, Chittenden TW, Buckley PG, Yu TW, Lodato MA, Lee EA. ATM-deficiency-induced microglial activation promotes neurodegeneration in ataxia-telangiectasia. Cell Rep 2024; 43:113622. [PMID: 38159274 PMCID: PMC10908398 DOI: 10.1016/j.celrep.2023.113622] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/26/2023] [Accepted: 12/08/2023] [Indexed: 01/03/2024] Open
Abstract
While ATM loss of function has long been identified as the genetic cause of ataxia-telangiectasia (A-T), how it leads to selective and progressive degeneration of cerebellar Purkinje and granule neurons remains unclear. ATM expression is enriched in microglia throughout cerebellar development and adulthood. Here, we find evidence of microglial inflammation in the cerebellum of patients with A-T using single-nucleus RNA sequencing. Pseudotime analysis revealed that activation of A-T microglia preceded upregulation of apoptosis-related genes in granule and Purkinje neurons and that microglia exhibited increased neurotoxic cytokine signaling to granule and Purkinje neurons in A-T. To confirm these findings experimentally, we performed transcriptomic profiling of A-T induced pluripotent stem cell (iPSC)-derived microglia, which revealed cell-intrinsic microglial activation of cytokine production and innate immune response pathways compared to controls. Furthermore, A-T microglia co-culture with either control or A-T iPSC-derived neurons was sufficient to induce cytotoxicity. Taken together, these studies reveal that cell-intrinsic microglial activation may promote neurodegeneration in A-T.
Collapse
Affiliation(s)
- Jenny Lai
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Program in Neuroscience, Harvard University, Boston, MA 02115, USA
| | - Didem Demirbas
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Junho Kim
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ailsa M Jeffries
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Allie Tolles
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Junseok Park
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Thomas W Chittenden
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA; Computational Statistics and Bioinformatics Group, Genuity AI Research Institute, Genuity Science, Boston, MA 02114, USA
| | | | - Timothy W Yu
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michael A Lodato
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Eunjung Alice Lee
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
13
|
Bastianello G, Porcella G, Beznoussenko GV, Kidiyoor G, Ascione F, Li Q, Cattaneo A, Matafora V, Disanza A, Quarto M, Mironov AA, Oldani A, Barozzi S, Bachi A, Costanzo V, Scita G, Foiani M. Cell stretching activates an ATM mechano-transduction pathway that remodels cytoskeleton and chromatin. Cell Rep 2023; 42:113555. [PMID: 38088930 DOI: 10.1016/j.celrep.2023.113555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/01/2023] [Accepted: 11/20/2023] [Indexed: 12/30/2023] Open
Abstract
Ataxia telangiectasia mutated (ATM) and ataxia telangiectasia and Rad3-related (ATR) DNA damage response (DDR) kinases contain elastic domains. ATM also responds to reactive oxygen species (ROS) and ATR to nuclear mechanical stress. Mre11 mediates ATM activation following DNA damage; ATM mutations cause ataxia telangiectasia (A-T). Here, using in vivo imaging, electron microscopy, proteomic, and mechano-biology approaches, we study how ATM responds to mechanical stress. We report that cytoskeleton and ROS, but not Mre11, mediate ATM activation following cell deformation. ATM deficiency causes hyper-stiffness, stress fiber accumulation, Yes-associated protein (YAP) nuclear enrichment, plasma and nuclear membrane alterations during interstitial migration, and H3 hyper-methylation. ATM locates to the actin cytoskeleton and, following cytoskeleton stress, promotes phosphorylation of key cytoskeleton and chromatin regulators. Our data contribute to explain some clinical features of patients with A-T and pinpoint the existence of an integrated mechano-response in which ATM and ATR have distinct roles unrelated to their canonical DDR functions.
Collapse
Affiliation(s)
- Giulia Bastianello
- IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy; Oncology and Haemato-Oncology Department, University of Milan, 20122 Milan, Italy.
| | | | | | - Gururaj Kidiyoor
- IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Flora Ascione
- IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Qingsen Li
- IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | | | | | - Andrea Disanza
- IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Micaela Quarto
- IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | | | - Amanda Oldani
- IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Sara Barozzi
- IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Angela Bachi
- IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Vincenzo Costanzo
- IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy; Oncology and Haemato-Oncology Department, University of Milan, 20122 Milan, Italy
| | - Giorgio Scita
- IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy; Oncology and Haemato-Oncology Department, University of Milan, 20122 Milan, Italy
| | - Marco Foiani
- IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy; Oncology and Haemato-Oncology Department, University of Milan, 20122 Milan, Italy.
| |
Collapse
|
14
|
Ali R, Alhaj Sulaiman A, Memon B, Pradhan S, Algethami M, Aouida M, McKay G, Madhusudan S, Abdelalim EM, Ramotar D. Altered Regulation of the Glucose Transporter GLUT3 in PRDX1 Null Cells Caused Hypersensitivity to Arsenite. Cells 2023; 12:2682. [PMID: 38067110 PMCID: PMC10705171 DOI: 10.3390/cells12232682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/13/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
Targeting tumour metabolism through glucose transporters is an attractive approach. However, the role these transporters play through interaction with other signalling proteins is not yet defined. The glucose transporter SLC2A3 (GLUT3) is a member of the solute carrier transporter proteins. GLUT3 has a high affinity for D-glucose and regulates glucose uptake in the neurons, as well as other tissues. Herein, we show that GLUT3 is involved in the uptake of arsenite, and its level is regulated by peroxiredoxin 1 (PRDX1). In the absence of PRDX1, GLUT3 mRNA and protein expression levels are low, but they are increased upon arsenite treatment, correlating with an increased uptake of glucose. The downregulation of GLUT3 by siRNA or deletion of the gene by CRISPR cas-9 confers resistance to arsenite. Additionally, the overexpression of GLUT3 sensitises the cells to arsenite. We further show that GLUT3 interacts with PRDX1, and it forms nuclear foci, which are redistributed upon arsenite exposure, as revealed by immunofluorescence analysis. We propose that GLUT3 plays a role in mediating the uptake of arsenite into cells, and its homeostatic and redox states are tightly regulated by PRDX1. As such, GLUT3 and PRDX1 are likely to be novel targets for arsenite-based cancer therapy.
Collapse
Affiliation(s)
- Reem Ali
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha 34110, Qatar; (R.A.); (A.A.S.); (B.M.); (M.A.); (E.M.A.)
| | - Abdallah Alhaj Sulaiman
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha 34110, Qatar; (R.A.); (A.A.S.); (B.M.); (M.A.); (E.M.A.)
| | - Bushra Memon
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha 34110, Qatar; (R.A.); (A.A.S.); (B.M.); (M.A.); (E.M.A.)
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha 34110, Qatar
| | - Singdhendubala Pradhan
- Division of Sustainable Development, College of Science and Engineering, Hamad Bin Khalifa University, Doha 34110, Qatar; (S.P.); (G.M.)
| | - Mashael Algethami
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (M.A.); (S.M.)
| | - Mustapha Aouida
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha 34110, Qatar; (R.A.); (A.A.S.); (B.M.); (M.A.); (E.M.A.)
| | - Gordon McKay
- Division of Sustainable Development, College of Science and Engineering, Hamad Bin Khalifa University, Doha 34110, Qatar; (S.P.); (G.M.)
| | - Srinivasan Madhusudan
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (M.A.); (S.M.)
- Department of Oncology, Nottingham University Hospitals, Nottingham NG5 1PB, UK
| | - Essam M. Abdelalim
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha 34110, Qatar; (R.A.); (A.A.S.); (B.M.); (M.A.); (E.M.A.)
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha 34110, Qatar
| | - Dindial Ramotar
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha 34110, Qatar; (R.A.); (A.A.S.); (B.M.); (M.A.); (E.M.A.)
| |
Collapse
|
15
|
Jin Y, Li Y, He S, Ge Y, Zhao Y, Zhu K, He A, Li S, Yan S, Cao C. ATM participates in fine particulate matter-induced airway inflammation through regulating DNA damage and DNA damage response. ENVIRONMENTAL TOXICOLOGY 2023; 38:2668-2678. [PMID: 37483094 DOI: 10.1002/tox.23901] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/20/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023]
Abstract
The relationship between fine particulate matter (PM2.5) and chronic airway inflammatory diseases, such as chronic obstructive pulmonary disease and asthma, have garnered public attention, while the detailed mechanisms of PM2.5-induced airway inflammation remain unclear. This study reveals that PM2.5 induces airway inflammation both in vivo and in vitro, and, moreover, identifies DNA damage and DNA damage repair (DDR) as results of this exposure. Ataxia telangiectasia-mutated heterozygous (ATM+/- ) and wild-type C57BL/6 (WT) mice were exposed to PM2.5. The results show that, following exposure to PM2.5, the number of neutrophils in broncho alveolar lavage fluid and the mRNA expression of CXCL-1 in lung tissues of the ATM+/- mice were lower than those of the WT mice. The mRNA expression of FANCD2 and FANCI were also down-regulated. Human bronchial epithelial (HBE) cells were transfected with ATM-siRNA to induce down-regulation of ATM gene expression and were subsequently stimulated with PM2.5. The results show that the mRNA expression of TNF-α decreased in the ATM-siRNA-transfected cells. The mRNA expression of CXCL-1 and CXCL-2 in peritoneal macrophages, derived from ATM-null mice in which experiments showed that the protein expression of FANCD2 and FANCI decreased, were also decreased after PM2.5 exposure in ATM-siRNA-transfected HBE cells. In conclusion, PM2.5-induced airway inflammation is alleviated in ATM+/- mice compared with WT mice. ATM promotes PM2.5-induced airway inflammation, which may be attributed to the regulation of DNA damage and DDR.
Collapse
Affiliation(s)
- Yan Jin
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Respiratory and Critical Care Medicine, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Yiting Li
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Shiyi He
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yijun Ge
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yun Zhao
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Ke Zhu
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Andong He
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Siyu Li
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Siyu Yan
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Chao Cao
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
16
|
Xun J, Ohtsuka H, Hirose K, Douchi D, Nakayama S, Ishida M, Miura T, Ariake K, Mizuma M, Nakagawa K, Morikawa T, Furukawa T, Unno M. Reduced expression of phosphorylated ataxia-telangiectasia mutated gene is related to poor prognosis and gemcitabine chemoresistance in pancreatic cancer. BMC Cancer 2023; 23:835. [PMID: 37674118 PMCID: PMC10481509 DOI: 10.1186/s12885-023-11294-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 08/12/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND Loss of expression of the gene ataxia-telangiectasia mutated (ATM), occurring in patients with multiple primary malignancies, including pancreatic cancer, is associated with poor prognosis. In this study, we investigated the detailed molecular mechanism through which ATM expression affects the prognosis of patients with pancreatic cancer. METHODS The levels of expression of ATM and phosphorylated ATM in patients with pancreatic cancer who had undergone surgical resection were analyzed using immunohistochemistry staining. RNA sequencing was performed on ATM-knockdown pancreatic-cancer cells to elucidate the mechanism underlying the invlovement of ATM in pancreatic cancer. RESULTS Immunohistochemical analysis showed that 15.3% and 27.8% of clinical samples had low levels of ATM and phosphorylated ATM, respectively. Low expression of phosphorylated ATM substantially reduced overall and disease-free survival in patients with pancreatic cancer. In the pancreatic cancer cell lines with ATM low expression, resistance to gemcitabine was demonstrated. The RNA sequence demonstrated that ATM knockdown induced the expression of MET and NTN1. In ATM knockdown cells, it was also revealed that the protein expression levels of HIF-1α and antiapoptotic BCL-2/BAD were upregulated. CONCLUSIONS These findings demonstrate that loss of ATM expression increases tumor development, suppresses apoptosis, and reduces gemcitabine sensitivity. Additionally, loss of phosphorylated ATM is associated with a poor prognosis in patients with pancreatic cancer. Thus, phosphorylated ATM could be a possible target for pancreatic cancer treatment as well as a molecular marker to track patient prognosis.
Collapse
Affiliation(s)
- Jingyu Xun
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Miyagi, Japan
| | - Hideo Ohtsuka
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Miyagi, Japan.
| | - Katsuya Hirose
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Japan
| | - Daisuke Douchi
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Miyagi, Japan
| | - Shun Nakayama
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Miyagi, Japan
| | - Masaharu Ishida
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Miyagi, Japan
| | - Takayuki Miura
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Miyagi, Japan
| | - Kyohei Ariake
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Miyagi, Japan
| | - Masamichi Mizuma
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Miyagi, Japan
| | - Kei Nakagawa
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Miyagi, Japan
| | - Takanori Morikawa
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Miyagi, Japan
| | - Toru Furukawa
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Miyagi, Japan
| |
Collapse
|
17
|
TeSlaa T, Ralser M, Fan J, Rabinowitz JD. The pentose phosphate pathway in health and disease. Nat Metab 2023; 5:1275-1289. [PMID: 37612403 PMCID: PMC11251397 DOI: 10.1038/s42255-023-00863-2] [Citation(s) in RCA: 161] [Impact Index Per Article: 80.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 07/12/2023] [Indexed: 08/25/2023]
Abstract
The pentose phosphate pathway (PPP) is a glucose-oxidizing pathway that runs in parallel to upper glycolysis to produce ribose 5-phosphate and nicotinamide adenine dinucleotide phosphate (NADPH). Ribose 5-phosphate is used for nucleotide synthesis, while NADPH is involved in redox homoeostasis as well as in promoting biosynthetic processes, such as the synthesis of tetrahydrofolate, deoxyribonucleotides, proline, fatty acids and cholesterol. Through NADPH, the PPP plays a critical role in suppressing oxidative stress, including in certain cancers, in which PPP inhibition may be therapeutically useful. Conversely, PPP-derived NADPH also supports purposeful cellular generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) for signalling and pathogen killing. Genetic deficiencies in the PPP occur relatively commonly in the committed pathway enzyme glucose-6-phosphate dehydrogenase (G6PD). G6PD deficiency typically manifests as haemolytic anaemia due to red cell oxidative damage but, in severe cases, also results in infections due to lack of leucocyte oxidative burst, highlighting the dual redox roles of the pathway in free radical production and detoxification. This Review discusses the PPP in mammals, covering its roles in biochemistry, physiology and disease.
Collapse
Affiliation(s)
- Tara TeSlaa
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Markus Ralser
- Department of Biochemistry, Charité Universitätsmedizin, Berlin, Germany
- The Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Jing Fan
- Morgride Institute for Research, Madison, WI, USA
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua D Rabinowitz
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
- Department of Chemistry, Princeton University, Princeton, NJ, USA.
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA.
| |
Collapse
|
18
|
Averbeck D. Low-Dose Non-Targeted Effects and Mitochondrial Control. Int J Mol Sci 2023; 24:11460. [PMID: 37511215 PMCID: PMC10380638 DOI: 10.3390/ijms241411460] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Non-targeted effects (NTE) have been generally regarded as a low-dose ionizing radiation (IR) phenomenon. Recently, regarding long distant abscopal effects have also been observed at high doses of IR) relevant to antitumor radiation therapy. IR is inducing NTE involving intracellular and extracellular signaling, which may lead to short-ranging bystander effects and distant long-ranging extracellular signaling abscopal effects. Internal and "spontaneous" cellular stress is mostly due to metabolic oxidative stress involving mitochondrial energy production (ATP) through oxidative phosphorylation and/or anaerobic pathways accompanied by the leakage of O2- and other radicals from mitochondria during normal or increased cellular energy requirements or to mitochondrial dysfunction. Among external stressors, ionizing radiation (IR) has been shown to very rapidly perturb mitochondrial functions, leading to increased energy supply demands and to ROS/NOS production. Depending on the dose, this affects all types of cell constituents, including DNA, RNA, amino acids, proteins, and membranes, perturbing normal inner cell organization and function, and forcing cells to reorganize the intracellular metabolism and the network of organelles. The reorganization implies intracellular cytoplasmic-nuclear shuttling of important proteins, activation of autophagy, and mitophagy, as well as induction of cell cycle arrest, DNA repair, apoptosis, and senescence. It also includes reprogramming of mitochondrial metabolism as well as genetic and epigenetic control of the expression of genes and proteins in order to ensure cell and tissue survival. At low doses of IR, directly irradiated cells may already exert non-targeted effects (NTE) involving the release of molecular mediators, such as radicals, cytokines, DNA fragments, small RNAs, and proteins (sometimes in the form of extracellular vehicles or exosomes), which can induce damage of unirradiated neighboring bystander or distant (abscopal) cells as well as immune responses. Such non-targeted effects (NTE) are contributing to low-dose phenomena, such as hormesis, adaptive responses, low-dose hypersensitivity, and genomic instability, and they are also promoting suppression and/or activation of immune cells. All of these are parts of the main defense systems of cells and tissues, including IR-induced innate and adaptive immune responses. The present review is focused on the prominent role of mitochondria in these processes, which are determinants of cell survival and anti-tumor RT.
Collapse
Affiliation(s)
- Dietrich Averbeck
- Laboratory of Cellular and Molecular Radiobiology, PRISME, UMR CNRS 5822/IN2P3, IP2I, Lyon-Sud Medical School, University Lyon 1, 69921 Oullins, France
| |
Collapse
|
19
|
Vardar Acar N, Özgül RK. The bridge between cell survival and cell death: reactive oxygen species-mediated cellular stress. EXCLI JOURNAL 2023; 22:520-555. [PMID: 37534225 PMCID: PMC10390897 DOI: 10.17179/excli2023-6221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 06/15/2023] [Indexed: 08/04/2023]
Abstract
As a requirement of aerobic metabolism, regulation of redox homeostasis is indispensable for the continuity of living homeostasis and life. Since the stability of the redox state is necessary for the maintenance of the biological functions of the cells, the balance between the pro-oxidants, especially ROS and the antioxidant capacity is kept in balance in the cells through antioxidant defense systems. The pleiotropic transcription factor, Nrf2, is the master regulator of the antioxidant defense system. Disruption of redox homeostasis leads to oxidative and reductive stress, bringing about multiple pathophysiological conditions. Oxidative stress characterized by high ROS levels causes oxidative damage to biomolecules and cell death, while reductive stress characterized by low ROS levels disrupt physiological cell functions. The fact that ROS, which were initially attributed as harmful products of aerobic metabolism, at the same time function as signal molecules at non-toxic levels and play a role in the adaptive response called mithormesis points out that ROS have a dose-dependent effect on cell fate determination. See also Figure 1(Fig. 1).
Collapse
Affiliation(s)
- Nese Vardar Acar
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Riza Köksal Özgül
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
20
|
Ajouaou Y, Magnani E, Madakashira B, Jenkins E, Sadler KC. atm Mutation and Oxidative Stress Enhance the Pre-Cancerous Effects of UHRF1 Overexpression in Zebrafish Livers. Cancers (Basel) 2023; 15:cancers15082302. [PMID: 37190230 DOI: 10.3390/cancers15082302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 03/29/2023] [Indexed: 05/17/2023] Open
Abstract
The ataxia-telangiectasia mutated (atm) gene is activated in response to genotoxic stress and leads to activation of the tp53 tumor suppressor gene which induces either senescence or apoptosis as tumor suppressive mechanisms. Atm also serves non-canonical functions in the response to oxidative stress and chromatin reorganization. We previously reported that overexpression of the epigenetic regulator and oncogene Ubiquitin Like with PHD and Ring Finger Domains 1 (UHRF1) in zebrafish hepatocytes resulted in tp53-dependent hepatocyte senescence, a small liver and larval lethality. We investigated the role of atm on UHRF1-mediated phenotypes by generating zebrafish atm mutants. atm-/- adults were viable but had reduction in fertility. Embryos developed normally but were protected from lethality caused by etoposide or H2O2 exposure and failed to fully upregulate Tp53 targets or oxidative stress response genes in response to these treatments. In contrast to the finding that Tp53 prevents the small liver phenotype caused by UHRF1 overexpression, atm mutation and exposure to H2O2 further reduced the liver size in UHRF1 overexpressing larvae whereas treatment with the antioxidant N-acetyl cysteine suppressed this phenotype. We conclude that UHRF1 overexpression in hepatocytes causes oxidative stress, and that loss of atm further enhances this, triggering elimination of these precancerous cells, leading to a small liver.
Collapse
Affiliation(s)
- Yousra Ajouaou
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
- Center for Genomics and Systems Biology (CGSB), New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Elena Magnani
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Bhavani Madakashira
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Eleanor Jenkins
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Kirsten C Sadler
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
- Center for Genomics and Systems Biology (CGSB), New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| |
Collapse
|
21
|
Chen W, Chiang J, Shang Z, Palchik G, Newman C, Zhang Y, Davis AJ, Lee H, Chen BPC. DNA-PKcs and ATM modulate mitochondrial ADP-ATP exchange as an oxidative stress checkpoint mechanism. EMBO J 2023; 42:e112094. [PMID: 36727301 PMCID: PMC10015379 DOI: 10.15252/embj.2022112094] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 01/13/2023] [Accepted: 01/21/2023] [Indexed: 02/03/2023] Open
Abstract
DNA-PKcs is a key regulator of DNA double-strand break repair. Apart from its canonical role in the DNA damage response, DNA-PKcs is involved in the cellular response to oxidative stress (OS), but its exact role remains unclear. Here, we report that DNA-PKcs-deficient human cells display depolarized mitochondria membrane potential (MMP) and reoriented metabolism, supporting a role for DNA-PKcs in oxidative phosphorylation (OXPHOS). DNA-PKcs directly interacts with mitochondria proteins ANT2 and VDAC2, and formation of the DNA-PKcs/ANT2/VDAC2 (DAV) complex supports optimal exchange of ADP and ATP across mitochondrial membranes to energize the cell via OXPHOS and to maintain MMP. Moreover, we demonstrate that the DAV complex temporarily dissociates in response to oxidative stress to attenuate ADP-ATP exchange, a rate-limiting step for OXPHOS. Finally, we found that dissociation of the DAV complex is mediated by phosphorylation of DNA-PKcs at its Thr2609 cluster by ATM kinase. Based on these findings, we propose that the coordination between the DAV complex and ATM serves as a novel oxidative stress checkpoint to decrease ROS production from mitochondrial OXPHOS and to hasten cellular recovery from OS.
Collapse
Affiliation(s)
- Wei‐Min Chen
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
- Department of Life ScienceNational Taiwan UniversityTaipeiTaiwan
| | - Jui‐Chung Chiang
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
- Department of Life ScienceNational Taiwan UniversityTaipeiTaiwan
| | - Zengfu Shang
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
| | - Guillermo Palchik
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
| | - Ciara Newman
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
| | - Yuanyuan Zhang
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
| | - Anthony J Davis
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
| | - Hsinyu Lee
- Department of Life ScienceNational Taiwan UniversityTaipeiTaiwan
| | - Benjamin PC Chen
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
| |
Collapse
|
22
|
Adipose-Derived Mesenchymal Stem Cells Inhibit JNK-Mediated Mitochondrial Retrograde Pathway to Alleviate Acetaminophen-Induced Liver Injury. Antioxidants (Basel) 2023; 12:antiox12010158. [PMID: 36671020 PMCID: PMC9854665 DOI: 10.3390/antiox12010158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/31/2022] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
Acetaminophen (APAP) is the major cause of drug-induced liver injury, with limited treatment options. APAP overdose invokes excessive oxidative stress that triggers mitochondria-to-nucleus retrograde pathways, contributing to APAP-induced liver injury (AILI). Mesenchymal stem cell therapy is a promising tool for acute liver failure. Therefore, the purpose of this study was to investigate the beneficial effects of adipose-derived mesenchymal stem cell (AMSC) therapy on AILI and reveal the potential therapeutic mechanisms. C57BL/6 mice were used as the animal model and AML12 normal murine hepatocytes as the cellular model of APAP overdose. Immunohistochemical staining, Western blotting, immunofluorescence staining, and RNA sequencing assays were used for assessing the efficacy and validating mechanisms of AMSC therapy. We found AMSC therapy effectively ameliorated AILI, while delayed AMSC injection lost its efficacy related to the c-Jun N-terminal kinase (JNK)-mediated mitochondrial retrograde pathways. We further found that AMSC therapy inhibited JNK activation and mitochondrial translocation, reducing APAP-induced mitochondrial damage. The downregulation of activated ataxia telangiectasia-mutated (ATM) and DNA damage response proteins in AMSC-treated mouse liver indicated AMSCs blocked the JNK-ATM pathway. Overall, AMSCs may be an effective treatment for AILI by inhibiting the JNK-ATM mitochondrial retrograde pathway, which improves APAP-induced mitochondrial dysfunction and liver injury.
Collapse
|
23
|
Barzilai A, Mitiagin Y. Ataxia-telangiectasia mutated plays an important role in cerebellar integrity and functionality. Neural Regen Res 2023; 18:497-502. [DOI: 10.4103/1673-5374.350194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
24
|
Malin SK, Stewart NR, Ude AA, Alderman BL. Brain insulin resistance and cognitive function: influence of exercise. J Appl Physiol (1985) 2022; 133:1368-1380. [PMID: 36269295 PMCID: PMC9744647 DOI: 10.1152/japplphysiol.00375.2022] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 12/15/2022] Open
Abstract
Exercise has systemic health benefits in people, in part, through improving whole body insulin sensitivity. The brain is an insulin-sensitive organ that is often underdiscussed relative to skeletal muscle, liver, and adipose tissue. Although brain insulin action may have only subtle impacts on peripheral regulation of systemic glucose homeostasis, it is important for weight regulation as well as mental health. In fact, brain insulin signaling is also involved in processes that support healthy cognition. Furthermore, brain insulin resistance has been associated with age-related declines in memory and executive function as well as Alzheimer's disease pathology. Herein, we provide an overview of brain insulin sensitivity in relation to cognitive function from animal and human studies, with particular emphasis placed on the impact exercise may have on brain insulin sensitivity. Mechanisms discussed include mitochondrial function, brain growth factors, and neurogenesis, which collectively help combat obesity-related metabolic disease and Alzheimer's dementia.
Collapse
Affiliation(s)
- Steven K Malin
- Department of Kinesiology & Health, Rutgers University, New Brunswick, New Jersey
- Division of Endocrinology, Metabolism & Nutrition, Rutgers University, New Brunswick, New Jersey
- New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, New Jersey
- Institute of Translational Medicine and Science, Rutgers University, New Brunswick, New Jersey
| | - Nathan R Stewart
- Department of Kinesiology & Health, Rutgers University, New Brunswick, New Jersey
| | - Andrew A Ude
- Department of Kinesiology & Health, Rutgers University, New Brunswick, New Jersey
| | - Brandon L Alderman
- Department of Kinesiology & Health, Rutgers University, New Brunswick, New Jersey
- Center of Alcohol and Substance Use Studies, Rutgers University, New Brunswick, New Jersey
| |
Collapse
|
25
|
Abstract
ATM, a member of the PIKK-like protein family, plays a central role in responding to DNA double-strand breaks and other lesions to protect the genome against DNA damage. Loss of ATM's kinase function has been shown to increase the sensitivity of most cells to ionizing radiation. Therefore, ATM is thought to be a promising target for chemotherapy as a radiotherapy sensitizer. The mechanism of ATM in cancer treatment and the development of its inhibitors have become research hotspots. Here we present an overview of research concerning ATM protein domains, functions and inhibitors, as well as perspectives and insights for future development of ATM-targeting agents.
Collapse
|
26
|
Bar-Peled L, Kory N. Principles and functions of metabolic compartmentalization. Nat Metab 2022; 4:1232-1244. [PMID: 36266543 PMCID: PMC10155461 DOI: 10.1038/s42255-022-00645-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/24/2022] [Indexed: 01/20/2023]
Abstract
Metabolism has historically been studied at the levels of whole cells, whole tissues and whole organisms. As a result, our understanding of how compartmentalization-the spatial and temporal separation of pathways and components-shapes organismal metabolism remains limited. At its essence, metabolic compartmentalization fulfils three important functions or 'pillars': establishing unique chemical environments, providing protection from reactive metabolites and enabling the regulation of metabolic pathways. However, how these pillars are established, regulated and maintained at both the cellular and systemic levels remains unclear. Here we discuss how the three pillars are established, maintained and regulated within the cell and discuss the consequences of dysregulation of metabolic compartmentalization in human disease. Organelles are increasingly emerging as 'command-and-control centres' and the increased understanding of metabolic compartmentalization is revealing new aspects of metabolic homeostasis, with this knowledge being translated into therapies for the treatment of cancer and certain neurodegenerative diseases.
Collapse
Affiliation(s)
- Liron Bar-Peled
- Center for Cancer Research, Massachusetts General Hospital and Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Nora Kory
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
27
|
Thirunavukkarasu C, Sharma Y, Tchaikovskaya T, Maslov AY, Gupta S. Transcriptional profiling reveals ataxia telangiectasia mutated pathways regulate joint copper and arsenic toxicity for hepatic metalloplasia and anti-cancer therapies. Life Sci 2022; 305:120787. [PMID: 35809665 DOI: 10.1016/j.lfs.2022.120787] [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: 04/28/2022] [Revised: 06/27/2022] [Accepted: 07/03/2022] [Indexed: 10/17/2022]
Abstract
AIMS Exposures to toxic metals, including arsenic (As), pose health risks but joint effects of physiologically needed metals, e.g., copper (Cu), are ill-defined for regulated metal-dependent cell proliferation (or metalloplasia). This study elucidated hepatic toxicities of As and Cu. MAIN METHODS Human HuH-7 cells were exposed to As and Cu and mRNA profiling obtained for molecular networks, regulators and signaling pathways. This followed biological testing of ATM signaling-related DNA damage response, mitochondrial dysfunction and lysosome activity using HuH-7 cells and primary hepatocytes. Free Cu ions were bound to 3-indole propionic acid for finding their contribution in toxicity. KEY FINDINGS The As or As plus Cu toxicities in HuH-7 cells produced dimorphic down- or up-regulation patterns in mRNA profiles. Significant differences extended for ontologies in protein synthesis, intermediary metabolism, mitochondrial function, autophagy, or cell survival and growth. Bioassays revealed ATM signaling regulated As and Cu toxicity for oxidative phosphorylation, mitochondrial membrane potential, lysosomal activity, DNA damage response, and cell growth-arrest. Removal of reactive Cu ions decreased As and Cu toxicity. Primary hepatocytes withstood Cu and As toxicity better. SIGNIFICANCE This joint As and Cu toxicity offers further mechanisms for metalloplasia, carcinogenesis and tissue damage in other settings, e.g., during excess Cu accumulation in Wilson disease. Moreover, joint As and Cu toxicities are relevant for anti-cancer therapies, potentially including manipulations to increase intracellular Cu through altered uptake or efflux processes and incorporating ATM-related checkpoint inhibitors. Superior tolerance of healthy hepatocytes to Cu and As toxicity should improve safety margins for anti-cancer therapies.
Collapse
Affiliation(s)
- Chinnasamy Thirunavukkarasu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry 605 014, India
| | - Yogeshwar Sharma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Tatyana Tchaikovskaya
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Alexander Y Maslov
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Laboratory of Applied Genomic Technologies, Voronezh State University of Engineering Technology, Voronezh, Russia
| | - Sanjeev Gupta
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry 605 014, India; Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Diabetes Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
28
|
Oanh N, Lee HS, Kim YH, Min S, Park YJ, Heo J, Park YY, Lim WC, Cho H. Regulation of nuclear DNA damage response by mitochondrial morphofunctional pathway. Nucleic Acids Res 2022; 50:9247-9259. [PMID: 35979947 PMCID: PMC9458461 DOI: 10.1093/nar/gkac690] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/22/2022] [Accepted: 08/03/2022] [Indexed: 12/24/2022] Open
Abstract
Cells are constantly challenged by genotoxic stresses that can lead to genome instability. The integrity of the nuclear genome is preserved by the DNA damage response (DDR) and repair. Additionally, these stresses can induce mitochondria to transiently hyperfuse; however, it remains unclear whether canonical DDR is linked to these mitochondrial morphological changes. Here, we report that the abolition of mitochondrial fusion causes a substantial defect in the ATM-mediated DDR signaling. This deficiency is overcome by the restoration of mitochondria fusion. In cells with fragmented mitochondria, genotoxic stress-induced activation of JNK and its translocation to DNA lesion are lost. Importantly, the mitochondrial fusion machinery of MFN1/MFN2 associates with Sab (SH3BP5) and JNK, and these interactions are indispensable for the Sab-mediated activation of JNK and the ATM-mediated DDR signaling. Accordingly, the formation of BRCA1 and 53BP1 foci, as well as homology and end-joining repair are impaired in cells with fragmented mitochondria. Together, these data show that mitochondrial fusion-dependent JNK signaling is essential for the DDR, providing vital insight into the integration of nuclear and cytoplasmic stress signals.
Collapse
Affiliation(s)
| | | | - Yong-Hyun Kim
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea,Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
| | - Sunwoo Min
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Yeon-Ji Park
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - June Heo
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea,Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
| | - Yong-Yea Park
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Won-Chung Lim
- Correspondence may also be addressed to Won-Chung Lim.
| | - Hyeseong Cho
- To whom correspondence should be addressed. Tel: +82 312195052; Fax: +82 312195059;
| |
Collapse
|
29
|
Patel SB, Nemkov T, D'Alessandro A, Welner RS. Deciphering Metabolic Adaptability of Leukemic Stem Cells. Front Oncol 2022; 12:846149. [PMID: 35756656 PMCID: PMC9213881 DOI: 10.3389/fonc.2022.846149] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Therapeutic targeting of leukemic stem cells is widely studied to control leukemia. An emerging approach gaining popularity is altering metabolism as a potential therapeutic opportunity. Studies have been carried out on hematopoietic and leukemic stem cells to identify vulnerable pathways without impacting the non-transformed, healthy counterparts. While many metabolic studies have been conducted using stem cells, most have been carried out in vitro or on a larger population of progenitor cells due to challenges imposed by the low frequency of stem cells found in vivo. This creates artifacts in the studies carried out, making it difficult to interpret and correlate the findings to stem cells directly. This review discusses the metabolic difference seen between hematopoietic stem cells and leukemic stem cells across different leukemic models. Moreover, we also shed light on the advancements of metabolic techniques and current limitations and areas for additional research of the field to study stem cell metabolism.
Collapse
Affiliation(s)
- Sweta B Patel
- Department of Medicine, Division of Hematology/Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at, Birmingham, AL, United States.,Divison of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Robert S Welner
- Department of Medicine, Division of Hematology/Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at, Birmingham, AL, United States
| |
Collapse
|
30
|
Aguado J, Gómez-Inclán C, Leeson HC, Lavin MF, Shiloh Y, Wolvetang EJ. The hallmarks of aging in Ataxia-Telangiectasia. Ageing Res Rev 2022; 79:101653. [PMID: 35644374 DOI: 10.1016/j.arr.2022.101653] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/28/2022] [Accepted: 05/24/2022] [Indexed: 01/10/2023]
Abstract
Ataxia-telangiectasia (A-T) is caused by absence of the catalytic activity of ATM, a protein kinase that plays a central role in the DNA damage response, many branches of cellular metabolism, redox and mitochondrial homeostasis, and cell cycle regulation. A-T is a complex disorder characterized mainly by progressive cerebellar degeneration, immunodeficiency, radiation sensitivity, genome instability, and predisposition to cancer. It is increasingly recognized that the premature aging component of A-T is an important driver of this disease, and A-T is therefore an attractive model to study the aging process. This review outlines the current state of knowledge pertaining to the molecular and cellular signatures of aging in A-T and proposes how these new insights can guide novel therapeutic approaches for A-T.
Collapse
Affiliation(s)
- Julio Aguado
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Queensland 4072, Australia.
| | - Cecilia Gómez-Inclán
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Queensland 4072, Australia
| | - Hannah C Leeson
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Queensland 4072, Australia
| | - Martin F Lavin
- University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Brisbane, Australia
| | - Yosef Shiloh
- The David and Inez Myers Laboratory of Cancer Genetics, Department of Human Molecular Genetics and Biochemistry, Tel Aviv University School of Medicine, Tel Aviv, Israel
| | - Ernst J Wolvetang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Queensland 4072, Australia.
| |
Collapse
|
31
|
Wang H, Huang J, Yi W, Li J, He N, Kang L, He Z, Chen C. Identification of Immune-Related Key Genes as Potential Diagnostic Biomarkers of Sepsis in Children. J Inflamm Res 2022; 15:2441-2459. [PMID: 35444449 PMCID: PMC9015049 DOI: 10.2147/jir.s359908] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/05/2022] [Indexed: 12/13/2022] Open
Abstract
Objective The pathogenesis of sepsis is still unclear due to its complexity, especially in children. This study aimed to analyse the immune microenvironment and regulatory networks related to sepsis in children at the molecular level and to identify key immune-related genes to provide a new basis for the early diagnosis of sepsis. Methods The GSE145227 and GSE26440 datasets were downloaded from the Gene Expression Omnibus. The analyses included differentially expressed genes (DEGs), functional enrichment, immune cell infiltration, the competing endogenous RNA (ceRNA) interaction network, weighted gene coexpression network analysis (WGCNA), protein–protein interaction (PPI) network, key gene screening, correlation of sepsis molecular subtypes/immune infiltration with key gene expression, the diagnostic capabilities of key genes, and networks describing the interaction of key genes with transcription factors and small-molecule compounds. Finally, real-time quantitative PCR (RT–qPCR) was performed to verify the expression of key genes. Results A total of 236 immune-related DEGs, most of which were enriched in immune-related biological functions, were found. Further analysis of immune cell infiltration showed that M0 macrophages and neutrophils infiltrated more in the sepsis group, while fewer activated memory CD4+ T cells, resting memory CD4+ T cells, and CD8+ T cells did. The interaction network of ceRNA was successfully constructed. Six key genes (FYN, FBL, ATM, WDR75, FOXO1 and ITK) were identified by WGCNA and PPI analysis. We found strong associations between key genes and constructed septic molecular subtypes or immune cell infiltration. Receiver operating characteristic analysis showed that the area under the curve values of the key genes for diagnosis were all greater than 0.84. Subsequently, we successfully constructed an interaction network of key genes and transcription factors/small-molecule compounds. Finally, the key genes in the samples were verified by RT–qPCR. Conclusion Our results offer new insights into the pathogenesis of sepsis in children and provide new potential diagnostic biomarkers for the disease.
Collapse
Affiliation(s)
- Huabin Wang
- Division of Hematology/Oncology, Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People’s Republic of China
- Department of Pediatric Intensive Care Unit, the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People’s Republic of China
| | - Junbin Huang
- Division of Hematology/Oncology, Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People’s Republic of China
- Department of Pediatric Intensive Care Unit, the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People’s Republic of China
| | - Wenfang Yi
- Division of Hematology/Oncology, Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People’s Republic of China
- Department of Pediatric Intensive Care Unit, the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People’s Republic of China
| | - Jiahong Li
- Department of Neonatal Intensive Care Unit, the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People’s Republic of China
| | - Nannan He
- Department of Pediatric Intensive Care Unit, Shenzhen Children’s Hospital, Shenzhen, 518000, People’s Republic of China
| | - Liangliang Kang
- Department of Pediatric Intensive Care Unit, Shenzhen Children’s Hospital, Shenzhen, 518000, People’s Republic of China
| | - Zhijie He
- Department of Intensive Care Unit, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, 510000, People’s Republic of China
- Correspondence: Zhijie He; Chun Chen, Email ;
| | - Chun Chen
- Division of Hematology/Oncology, Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People’s Republic of China
- Department of Pediatric Intensive Care Unit, the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People’s Republic of China
| |
Collapse
|
32
|
Subramanian GN, Yeo AJ, Gatei MH, Coman DJ, Lavin MF. Metabolic Stress and Mitochondrial Dysfunction in Ataxia-Telangiectasia. Antioxidants (Basel) 2022; 11:653. [PMID: 35453338 PMCID: PMC9032508 DOI: 10.3390/antiox11040653] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 02/04/2023] Open
Abstract
The ataxia-telangiectasia mutated (ATM) protein kinase is, as the name implies, mutated in the human genetic disorder ataxia-telangiectasia (A-T). This protein has its "finger in many pies", being responsible for the phosphorylation of many thousands of proteins in different signaling pathways in its role in protecting the cell against a variety of different forms of stress that threaten to perturb cellular homeostasis. The classical role of ATM is the protection against DNA damage, but it is evident that it also plays a key role in maintaining cell homeostasis in the face of oxidative and other forms of non-DNA damaging stress. The presence of ATM is not only in the nucleus to cope with damage to DNA, but also in association with other organelles in the cytoplasm, which suggests a greater protective role. This review attempts to address this greater role of ATM in protecting the cell against both external and endogenous damage.
Collapse
Affiliation(s)
| | - Abrey Jie Yeo
- University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, QLD 4029, Australia
| | - Magtouf Hnaidi Gatei
- University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, QLD 4029, Australia
| | - David John Coman
- Queensland Children's Hospital, Brisbane, QLD 4101, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD 4006, Australia
| | - Martin Francis Lavin
- University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, QLD 4029, Australia
| |
Collapse
|
33
|
Circulating primitive murine erythroblasts undergo complex proteomic and metabolomic changes during terminal maturation. Blood Adv 2022; 6:3072-3089. [PMID: 35139174 PMCID: PMC9131905 DOI: 10.1182/bloodadvances.2021005975] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 01/31/2022] [Indexed: 11/20/2022] Open
Abstract
Terminal maturation of primary murine primitive erythroid precursors is characterized by loss of organelles and anabolic components. Metabolic reprogramming includes depression of mitochondrial metabolism and upregulation of the pentose phosphate pathway and redox metabolism.
Primitive erythropoiesis is a critical component of the fetal cardiovascular network and is essential for the growth and survival of the mammalian embryo. The need to rapidly establish a functional cardiovascular system is met, in part, by the intravascular circulation of primitive erythroid precursors that mature as a single semisynchronous cohort. To better understand the processes that regulate erythroid precursor maturation, we analyzed the proteome, metabolome, and lipidome of primitive erythroblasts isolated from embryonic day (E) 10.5 and E12.5 of mouse gestation, representing their transition from basophilic erythroblast to orthochromatic erythroblast (OrthoE) stages of maturation. Previous transcriptional and biomechanical characterizations of these precursors have highlighted a transition toward the expression of protein elements characteristic of mature red blood cell structure and function. Our analysis confirmed a loss of organelle-specific protein components involved in messenger RNA processing, proteostasis, and metabolism. In parallel, we observed metabolic rewiring toward the pentose phosphate pathway, glycolysis, and the Rapoport-Luebering shunt. Activation of the pentose phosphate pathway in particular may have stemmed from increased expression of hemoglobin chains and band 3, which together control oxygen-dependent metabolic modulation. Increased expression of several antioxidant enzymes also indicated modification to redox homeostasis. In addition, accumulation of oxylipins and cholesteryl esters in primitive OrthoE cells was paralleled by increased transcript levels of the p53-regulated cholesterol transporter (ABCA1) and decreased transcript levels of cholesterol synthetic enzymes. The present study characterizes the extensive metabolic rewiring that occurs in primary embryonic erythroid precursors as they prepare to enucleate and continue circulating without internal organelles.
Collapse
|
34
|
Ueno S, Sudo T, Hirasawa A. ATM: Functions of ATM Kinase and Its Relevance to Hereditary Tumors. Int J Mol Sci 2022; 23:523. [PMID: 35008949 PMCID: PMC8745051 DOI: 10.3390/ijms23010523] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 02/04/2023] Open
Abstract
Ataxia-telangiectasia mutated (ATM) functions as a key initiator and coordinator of DNA damage and cellular stress responses. ATM signaling pathways contain many downstream targets that regulate multiple important cellular processes, including DNA damage repair, apoptosis, cell cycle arrest, oxidative sensing, and proliferation. Over the past few decades, associations between germline ATM pathogenic variants and cancer risk have been reported, particularly for breast and pancreatic cancers. In addition, given that ATM plays a critical role in repairing double-strand breaks, inhibiting other DNA repair pathways could be a synthetic lethal approach. Based on this rationale, several DNA damage response inhibitors are currently being tested in ATM-deficient cancers. In this review, we discuss the current knowledge related to the structure of the ATM gene, function of ATM kinase, clinical significance of ATM germline pathogenic variants in patients with hereditary cancers, and ongoing efforts to target ATM for the benefit of cancer patients.
Collapse
Affiliation(s)
- Sayaka Ueno
- Section of Translational Research, Hyogo Cancer Center, 13-70 Kita-Oji-cho, Akashi-shi 673-8558, Japan;
- Department of Clinical Genomic Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan;
| | - Tamotsu Sudo
- Section of Translational Research, Hyogo Cancer Center, 13-70 Kita-Oji-cho, Akashi-shi 673-8558, Japan;
| | - Akira Hirasawa
- Department of Clinical Genomic Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan;
| |
Collapse
|
35
|
Edara VV, Manning KE, Ellis M, Lai L, Moore KM, Foster SL, Floyd K, Davis-Gardner ME, Mantus G, Nyhoff LE, Bechnak S, Alaaeddine G, Naji A, Samaha H, Lee M, Bristow L, Hussaini L, Ciric CR, Nguyen PV, Gagne M, Roberts-Torres J, Henry AR, Godbole S, Grakoui A, Sexton M, Piantadosi A, Waggoner JJ, Douek DC, Anderson EJ, Rouphael N, Wrammert J, Suthar MS. mRNA-1273 and BNT162b2 mRNA vaccines have reduced neutralizing activity against the SARS-CoV-2 Omicron variant. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34981056 DOI: 10.1101/2021.09.09.459619] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The BNT162b2 (Pfizer-BioNTech) and mRNA-1273 (Moderna) vaccines generate potent neutralizing antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, the global emergence of SARS-CoV-2 variants with mutations in the spike protein, the principal antigenic target of these vaccines, has raised concerns over the neutralizing activity of vaccine-induced antibody responses. The Omicron variant, which emerged in November 2021, consists of over 30 mutations within the spike protein. Here, we used an authentic live virus neutralization assay to examine the neutralizing activity of the SARS-CoV-2 Omicron variant against mRNA vaccine-induced antibody responses. Following the 2nd dose, we observed a 30-fold reduction in neutralizing activity against the omicron variant. Through six months after the 2nd dose, none of the sera from naïve vaccinated subjects showed neutralizing activity against the Omicron variant. In contrast, recovered vaccinated individuals showed a 22-fold reduction with more than half of the subjects retaining neutralizing antibody responses. Following a booster shot (3rd dose), we observed a 14-fold reduction in neutralizing activity against the omicron variant and over 90% of boosted subjects showed neutralizing activity against the omicron variant. These findings show that a 3rd dose is required to provide robust neutralizing antibody responses against the Omicron variant.
Collapse
|
36
|
Yeo AJ, Subramanian GN, Chong KL, Gatei M, Parton RG, Coman D, Lavin MF. An anaplerotic approach to correct the mitochondrial dysfunction in ataxia-telangiectasia (A-T). Mol Metab 2021; 54:101354. [PMID: 34637921 PMCID: PMC8599162 DOI: 10.1016/j.molmet.2021.101354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/22/2021] [Accepted: 10/06/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND ATM, the protein defective in the human genetic disorder, ataxia-telangiectasia (A-T) plays a central role in response to DNA double-strand breaks (DSBs) and in protecting the cell against oxidative stress. We showed that A-T cells are hypersensitive to metabolic stress which can be accounted for by a failure to exhibit efficient endoplasmic reticulum (ER)-mitochondrial signalling and Ca2+ transfer in response to nutrient deprivation resulting in mitochondrial dysfunction. The objective of the current study is to use an anaplerotic approach using the fatty acid, heptanoate (C7), a metabolic product of the triglyceride, triheptanoin to correct the defect in ER-mitochondrial signalling and enhance cell survival of A-T cells in response to metabolic stress. METHODS We treated control cells and A-T cells with the anaplerotic agent, heptanoate to determine their sensitivity to metabolic stress induced by inhibition of glycolysis with 2- deoxyglucose (2DG) using live-cell imaging to monitor cell survival for 72 h using the Incucyte system. We examined ER-mitochondrial signalling in A-T cells exposed to metabolic stress using a suite of techniques including immunofluorescence staining of Grp75, ER-mitochondrial Ca2+ channel, the VAPB-PTPIP51 ER-mitochondrial tether complexes as well as proximity ligation assays between Grp75-IP3R1 and VAPB1-PTPIP51 to establish a functional interaction between ER and mitochondria. Finally, we also performed metabolomic analysis using LC-MS/MS assay to determine altered levels of TCA intermediates A-T cells compared to healthy control cells. RESULTS We demonstrate that heptanoate corrects all aspects of the defective ER-mitochondrial signalling observed in A-T cells. Heptanoate enhances ER-mitochondrial contacts; increases the flow of calcium from the ER to the mitochondrion; restores normal mitochondrial function and mitophagy and increases the resistance of ATM-deficient cells and cells from A-T patients to metabolic stress-induced killing. The defect in mitochondrial function in ATM-deficient cells was accompanied by more reliance on aerobic glycolysis as shown by increased lactate dehydrogenase A (LDHA), accumulation of lactate, and reduced levels of both acetyl CoA and ATP which are all restored by heptanoate. CONCLUSIONS We conclude that heptanoate corrects metabolic stress in A-T cells by restoring ER-mitochondria signalling and mitochondrial function and suggest that the parent compound, triheptanoin, has immense potential as a novel therapeutic agent for patients with A-T.
Collapse
Affiliation(s)
- A J Yeo
- University of Queensland Centre for Clinical Research, University of Queensland, Herston, Brisbane, Australia.
| | - G N Subramanian
- University of Queensland Centre for Clinical Research, University of Queensland, Herston, Brisbane, Australia
| | - K L Chong
- University of Queensland Centre for Clinical Research, University of Queensland, Herston, Brisbane, Australia
| | - M Gatei
- University of Queensland Centre for Clinical Research, University of Queensland, Herston, Brisbane, Australia
| | - R G Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, University of Queensland, St Lucia, Brisbane, Australia
| | - D Coman
- Queensland Children's Hospital, Brisbane, Australia; Faculty of Medicine, University of Queensland, Herston, Brisbane, Australia
| | - M F Lavin
- University of Queensland Centre for Clinical Research, University of Queensland, Herston, Brisbane, Australia.
| |
Collapse
|
37
|
Wu Z, Sainz AG, Shadel GS. Mitochondrial DNA: cellular genotoxic stress sentinel. Trends Biochem Sci 2021; 46:812-821. [PMID: 34088564 PMCID: PMC9809014 DOI: 10.1016/j.tibs.2021.05.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/24/2021] [Accepted: 05/08/2021] [Indexed: 02/08/2023]
Abstract
High copy number, damage prone, and lean on repair mechanisms are unique features of mitochondrial DNA (mtDNA) that are hard to reconcile with its essentiality for oxidative phosphorylation, the primary function ascribed to this maternally inherited component of our genome. We propose that mtDNA is also a genotoxic stress sentinel, as well as a direct second messenger of this type of cellular stress. Here, we discuss existing evidence for this sentinel/effector role through the ability of mtDNA to escape the confines of the mitochondrial matrix and activate nuclear DNA damage/repair responses via interferon-stimulated gene products and other downstream effectors. However, this arrangement may come at a cost, leading to cancer chemoresistance and contributing to inflammation, disease pathology, and aging.
Collapse
Affiliation(s)
- Zheng Wu
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA,Graduate Program in Genetics, Yale School of Medicine, New Haven, CT 06437, USA,These authors contributed equally to this work
| | - Alva G. Sainz
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA,Graduate Program in Experimental Pathology, Yale School of Medicine, New Haven, CT 06437, USA,These authors contributed equally to this work
| | - Gerald S. Shadel
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA,Correspondence: (G.S. Shadel)
| |
Collapse
|
38
|
Kwok M, Agathanggelou A, Davies N, Stankovic T. Targeting the p53 Pathway in CLL: State of the Art and Future Perspectives. Cancers (Basel) 2021; 13:4681. [PMID: 34572908 PMCID: PMC8468925 DOI: 10.3390/cancers13184681] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 12/20/2022] Open
Abstract
The p53 pathway is a desirable therapeutic target, owing to its critical role in the maintenance of genome integrity. This is exemplified in chronic lymphocytic leukemia (CLL), one of the most common adult hematologic malignancies, in which functional loss of p53 arising from genomic aberrations are frequently associated with clonal evolution, disease progression, and therapeutic resistance, even in the contemporary era of CLL targeted therapy and immunotherapy. Targeting the 'undruggable' p53 pathway therefore arguably represents the holy grail of cancer research. In recent years, several strategies have been proposed to exploit p53 pathway defects for cancer treatment. Such strategies include upregulating wild-type p53, restoring tumor suppressive function in mutant p53, inducing synthetic lethality by targeting collateral genome maintenance pathways, and harnessing the immunogenicity of p53 pathway aberrations. In this review, we will examine the biological and clinical implications of p53 pathway defects, as well as our progress towards development of therapeutic approaches targeting the p53 pathway, specifically within the context of CLL. We will appraise the opportunities and pitfalls associated with these therapeutic strategies, and evaluate their place amongst the array of new biological therapies for CLL.
Collapse
Affiliation(s)
- Marwan Kwok
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SY, UK; (A.A.); (N.D.)
- Centre for Clinical Haematology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2SY, UK
| | - Angelo Agathanggelou
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SY, UK; (A.A.); (N.D.)
| | - Nicholas Davies
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SY, UK; (A.A.); (N.D.)
| | - Tatjana Stankovic
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SY, UK; (A.A.); (N.D.)
| |
Collapse
|
39
|
Cellular functions of the protein kinase ATM and their relevance to human disease. Nat Rev Mol Cell Biol 2021; 22:796-814. [PMID: 34429537 DOI: 10.1038/s41580-021-00394-2] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
The protein kinase ataxia telangiectasia mutated (ATM) is a master regulator of double-strand DNA break (DSB) signalling and stress responses. For three decades, ATM has been investigated extensively to elucidate its roles in the DNA damage response (DDR) and in the pathogenesis of ataxia telangiectasia (A-T), a human neurodegenerative disease caused by loss of ATM. Although hundreds of proteins have been identified as ATM phosphorylation targets and many important roles for this kinase have been identified, it is still unclear how ATM deficiency leads to the early-onset cerebellar degeneration that is common in all individuals with A-T. Recent studies suggest the existence of links between ATM deficiency and other cerebellum-specific neurological disorders, as well as the existence of broader similarities with more common neurodegenerative disorders. In this Review, we discuss recent structural insights into ATM regulation, and possible aetiologies of A-T phenotypes, including reactive oxygen species, mitochondrial dysfunction, alterations in transcription, R-loop metabolism and alternative splicing, defects in cellular proteostasis and metabolism, and potential pathogenic roles for hyper-poly(ADP-ribosyl)ation.
Collapse
|
40
|
Somyajit K, Spies J, Coscia F, Kirik U, Rask MB, Lee JH, Neelsen KJ, Mund A, Jensen LJ, Paull TT, Mann M, Lukas J. Homology-directed repair protects the replicating genome from metabolic assaults. Dev Cell 2021; 56:461-477.e7. [PMID: 33621493 DOI: 10.1016/j.devcel.2021.01.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/14/2020] [Accepted: 01/20/2021] [Indexed: 12/18/2022]
Abstract
Homology-directed repair (HDR) safeguards DNA integrity under various forms of stress, but how HDR protects replicating genomes under extensive metabolic alterations remains unclear. Here, we report that besides stalling replication forks, inhibition of ribonucleotide reductase (RNR) triggers metabolic imbalance manifested by the accumulation of increased reactive oxygen species (ROS) in cell nuclei. This leads to a redox-sensitive activation of the ATM kinase followed by phosphorylation of the MRE11 nuclease, which in HDR-deficient settings degrades stalled replication forks. Intriguingly, nascent DNA degradation by the ROS-ATM-MRE11 cascade is also triggered by hypoxia, which elevates signaling-competent ROS and attenuates functional HDR without arresting replication forks. Under these conditions, MRE11 degrades daughter-strand DNA gaps, which accumulate behind active replisomes and attract error-prone DNA polymerases to escalate mutation rates. Thus, HDR safeguards replicating genomes against metabolic assaults by restraining mutagenic repair at aberrantly processed nascent DNA. These findings have implications for cancer evolution and tumor therapy.
Collapse
Affiliation(s)
- Kumar Somyajit
- Protein Signaling Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark.
| | - Julian Spies
- Protein Signaling Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark
| | - Fabian Coscia
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark
| | - Ufuk Kirik
- Disease Systems Biology Program, Novo Nordisk Foundation Center for Protein, Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark
| | - Maj-Britt Rask
- Protein Signaling Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark
| | - Ji-Hoon Lee
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Kai John Neelsen
- Protein Signaling Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark
| | - Andreas Mund
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark
| | - Lars Juhl Jensen
- Disease Systems Biology Program, Novo Nordisk Foundation Center for Protein, Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark
| | - Tanya T Paull
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Matthias Mann
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark
| | - Jiri Lukas
- Protein Signaling Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark.
| |
Collapse
|
41
|
Mechanisms of Ataxia Telangiectasia Mutated (ATM) Control in the DNA Damage Response to Oxidative Stress, Epigenetic Regulation, and Persistent Innate Immune Suppression Following Sepsis. Antioxidants (Basel) 2021; 10:antiox10071146. [PMID: 34356379 PMCID: PMC8301080 DOI: 10.3390/antiox10071146] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
Cells have evolved extensive signaling mechanisms to maintain redox homeostasis. While basal levels of oxidants are critical for normal signaling, a tipping point is reached when the level of oxidant species exceed cellular antioxidant capabilities. Myriad pathological conditions are characterized by elevated oxidative stress, which can cause alterations in cellular operations and damage to cellular components including nucleic acids. Maintenance of nuclear chromatin are critically important for host survival and eukaryotic organisms possess an elaborately orchestrated response to initiate repair of such DNA damage. Recent evidence indicates links between the cellular antioxidant response, the DNA damage response (DDR), and the epigenetic status of the cell under conditions of elevated oxidative stress. In this emerging model, the cellular response to excessive oxidants may include redox sensors that regulate both the DDR and an orchestrated change to the epigenome in a tightly controlled program that both protects and regulates the nuclear genome. Herein we use sepsis as a model of an inflammatory pathophysiological condition that results in elevated oxidative stress, upregulation of the DDR, and epigenetic reprogramming of hematopoietic stem cells (HSCs) to discuss new evidence for interplay between the antioxidant response, the DNA damage response, and epigenetic status.
Collapse
|
42
|
Ovchinnikov DA, Withey SL, Leeson HC, Lei UW, Sundarrajan A, Junday K, Pewarchuk M, Yeo AJ, Kijas AW, Lavin MF, Wolvetang EJ. Correction of ATM mutations in iPS cells from two ataxia-telangiectasia patients restores DNA damage and oxidative stress responses. Hum Mol Genet 2021; 29:990-1001. [PMID: 32037450 DOI: 10.1093/hmg/ddaa023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 01/06/2020] [Accepted: 02/04/2020] [Indexed: 01/23/2023] Open
Abstract
Patients with ataxia-telangiectasia (A-T) lack a functional ATM kinase protein and exhibit defective repair of DNA double-stranded breaks and response to oxidative stress. We show that CRISPR/Cas9-assisted gene correction combined with piggyBac (PB) transposon-mediated excision of the selection cassette enables seamless restoration of functional ATM alleles in induced pluripotent stem cells from an A-T patient carrying compound heterozygous exonic missense/frameshift mutations, and from a patient with a homozygous splicing acceptor mutation of an internal coding exon. We show that the correction of one allele restores expression of ~ 50% of full-length ATM protein and ameliorates DNA damage-induced activation (auto-phosphorylation) of ATM and phosphorylation of its downstream targets, KAP-1 and H2AX. Restoration of ATM function also normalizes radiosensitivity, mitochondrial ROS production and oxidative-stress-induced apoptosis levels in A-T iPSC lines, demonstrating that restoration of a single ATM allele is sufficient to rescue key ATM functions. Our data further show that despite the absence of a functional ATM kinase, homology-directed repair and seamless correction of a pathogenic ATM mutation is possible. The isogenic pairs of A-T and gene-corrected iPSCs described here constitute valuable tools for elucidating the role of ATM in ageing and A-T pathogenesis.
Collapse
Affiliation(s)
- Dmitry A Ovchinnikov
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia.,StemCore, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Sarah L Withey
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Hannah C Leeson
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - U Wang Lei
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Ashmitha Sundarrajan
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Keerat Junday
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Michelle Pewarchuk
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Abrey J Yeo
- UQ Centre for Clinical Research (UQCCR), The University of Queensland, Herston, Brisbane, QLD 4006, Australia
| | - Amanda W Kijas
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Martin F Lavin
- UQ Centre for Clinical Research (UQCCR), The University of Queensland, Herston, Brisbane, QLD 4006, Australia
| | - Ernst J Wolvetang
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| |
Collapse
|
43
|
Li H, Zimmerman SE, Weyemi U. Genomic instability and metabolism in cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 364:241-265. [PMID: 34507785 DOI: 10.1016/bs.ircmb.2021.05.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Genomic instability and metabolic reprogramming are among the key hallmarks discriminating cancer cells from normal cells. The two phenomena contribute to the robust and evasive nature of cancer, particularly when cancer cells are exposed to chemotherapeutic agents. Genomic instability is defined as the increased frequency of mutations within the genome, while metabolic reprogramming is the alteration of metabolic pathways that cancer cells undergo to adapt to increased bioenergetic demand. An underlying source of these mutations is the aggregate product of damage to the DNA, and a defective repair pathway, both resulting in the expansion of genomic lesions prior to uncontrolled proliferation and survival of cancer cells. Exploitation of DNA damage and the subsequent DNA damage response (DDR) have aided in defining therapeutic approaches in cancer. Studies have demonstrated that targeting metabolic reprograming yields increased sensitivity to chemo- and radiotherapies. In the past decade, it has been shown that these two key features are interrelated. Metabolism impacts DNA damage and DDR via regulation of metabolite pools. Conversely, DDR affects the response of metabolic pathways to therapeutic agents. Because of the interplay between genomic instability and metabolic reprogramming, we have compiled findings which more selectively highlight the dialog between metabolism and DDR, with a particular focus on glucose metabolism and double-strand break (DSB) repair pathways. Decoding this dialog will provide significant clues for developing combination cancer therapies.
Collapse
Affiliation(s)
- Haojian Li
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States; Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, United States
| | - Susan E Zimmerman
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States; Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, United States
| | - Urbain Weyemi
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States; Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, United States.
| |
Collapse
|
44
|
Shimura T. ATM-Mediated Mitochondrial Radiation Responses of Human Fibroblasts. Genes (Basel) 2021; 12:genes12071015. [PMID: 34208940 PMCID: PMC8305810 DOI: 10.3390/genes12071015] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 01/15/2023] Open
Abstract
Ataxia telangiectasia (AT) is characterized by extreme sensitivity to ionizing radiation. The gene mutated in AT, Ataxia Telangiectasia Mutated (ATM), has serine/threonine protein kinase activity and mediates the activation of multiple signal transduction pathways involved in the processing of DNA double-strand breaks. Reactive oxygen species (ROS) created as a byproduct of the mitochondria's oxidative phosphorylation (OXPHOS) has been proposed to be the source of intracellular ROS. Mitochondria are uniquely vulnerable to ROS because they are the sites of ROS generation. ROS-induced mitochondrial mutations lead to impaired mitochondrial respiration and further increase the likelihood of ROS generation, establishing a vicious cycle of further ROS production and mitochondrial damage. AT patients and ATM-deficient mice display intrinsic mitochondrial dysfunction and exhibit constitutive elevations in ROS levels. ATM plays a critical role in maintaining cellular redox homeostasis. However, the precise mechanism of ATM-mediated mitochondrial antioxidants remains unclear. The aim of this review paper is to introduce our current research surrounding the role of ATM on maintaining cellular redox control in human fibroblasts. ATM-mediated signal transduction is important in the mitochondrial radiation response. Perturbation of mitochondrial redox control elevates ROS which are key mediators in the development of cancer by many mechanisms, including ROS-mediated genomic instability, tumor microenvironment formation, and chronic inflammation.
Collapse
Affiliation(s)
- Tsutomu Shimura
- Department of Environmental Health, National Institute of Public Health 2-3-6 Minami, Wako 351-0197, Saitama, Japan
| |
Collapse
|
45
|
Oka SI, Byun J, Huang CY, Imai N, Ralda G, Zhai P, Xu X, Kashyap S, Warren JS, Alan Maschek J, Tippetts TS, Tong M, Venkatesh S, Ikeda Y, Mizushima W, Kashihara T, Sadoshima J. Nampt Potentiates Antioxidant Defense in Diabetic Cardiomyopathy. Circ Res 2021; 129:114-130. [PMID: 33928788 PMCID: PMC8513534 DOI: 10.1161/circresaha.120.317943] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
MESH Headings
- Animals
- Antioxidants/metabolism
- Apoptosis
- Autophagy
- Cells, Cultured
- Cytokines/genetics
- Cytokines/metabolism
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/genetics
- Diabetic Cardiomyopathies/pathology
- Diet, High-Fat
- Disease Models, Animal
- Fibrosis
- Glutathione/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/genetics
- Mitochondria, Heart/pathology
- Mitophagy
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- NAD/metabolism
- NADP/metabolism
- Nicotinamide Phosphoribosyltransferase/genetics
- Nicotinamide Phosphoribosyltransferase/metabolism
- Oxidative Stress
- Rats, Wistar
- Sirtuins/genetics
- Sirtuins/metabolism
- Thioredoxins/metabolism
- Mice
- Rats
Collapse
Affiliation(s)
- Shin-Ichi Oka
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Jaemin Byun
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Chun-Yang Huang
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taiwan (C.-y.H.)
- Institute of Clinical Medicine, School of Medicine National Yang-Ming University, Taipei, Taiwan (C.-y.H.)
| | - Nobushige Imai
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Guersom Ralda
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Xiaoyong Xu
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Zhejiang, China (X.X.)
| | - Sanchita Kashyap
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Junco S Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute (J.S.W.), University of Utah, Salt Lake City
| | - John Alan Maschek
- Metabolomics, Proteomics, and Mass Spectrometry Cores (J.A.M.), University of Utah, Salt Lake City
- Department of Nutrition and Integrative Physiology (J.A.M.), University of Utah, Salt Lake City
| | - Trevor S Tippetts
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center (T.S.T., S.V.), University of Utah, Salt Lake City
| | - Mingming Tong
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Sundararajan Venkatesh
- Department of Microbiology, Biochemistry, and Molecular Genetics (S.V.), Rutgers New Jersey Medical School, Newark
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center (T.S.T., S.V.), University of Utah, Salt Lake City
| | - Yoshiyuki Ikeda
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Wataru Mizushima
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Toshihide Kashihara
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| |
Collapse
|
46
|
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
|
47
|
Yeo AJ, Chong KL, Gatei M, Zou D, Stewart R, Withey S, Wolvetang E, Parton RG, Brown AD, Kastan MB, Coman D, Lavin MF. Impaired endoplasmic reticulum-mitochondrial signaling in ataxia-telangiectasia. iScience 2021; 24:101972. [PMID: 33437944 PMCID: PMC7788243 DOI: 10.1016/j.isci.2020.101972] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/18/2020] [Accepted: 12/16/2020] [Indexed: 12/21/2022] Open
Abstract
There is evidence that ATM mutated in ataxia-telangiectasia (A-T) plays a key role in protecting against mitochondrial dysfunction, the mechanism for which remains unresolved. We demonstrate here that ATM-deficient cells are exquisitely sensitive to nutrient deprivation, which can be explained by defective cross talk between the endoplasmic reticulum (ER) and the mitochondrion. Tethering between these two organelles in response to stress was reduced in cells lacking ATM, and consistent with this, Ca2+ release and transfer between ER and mitochondria was reduced dramatically when compared with control cells. The impact of this on mitochondrial function was evident from an increase in oxygen consumption rates and a defect in mitophagy in ATM-deficient cells. Our findings reveal that ER-mitochondrial connectivity through IP3R1-GRP75-VDAC1, to maintain Ca2+ homeostasis, as well as an abnormality in mitochondrial fusion defective in response to nutrient stress, can account for at least part of the mitochondrial dysfunction observed in A-T cells.
Collapse
Affiliation(s)
- Abrey J. Yeo
- University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Brisbane, Australia
| | - Kok L. Chong
- University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Brisbane, Australia
| | - Magtouf Gatei
- University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Brisbane, Australia
| | - Dongxiu Zou
- University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Brisbane, Australia
| | | | - Sarah Withey
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, Australia
| | - Ernst Wolvetang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, Australia
| | - Robert G. Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, The University of Queensland, St Lucia, Brisbane, Australia
| | | | | | - David Coman
- Queensland Children's Hospital, Brisbane, Australia
| | - Martin F. Lavin
- University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Brisbane, Australia
| |
Collapse
|
48
|
Cirotti C, Rizza S, Giglio P, Poerio N, Allega MF, Claps G, Pecorari C, Lee J, Benassi B, Barilà D, Robert C, Stamler JS, Cecconi F, Fraziano M, Paull TT, Filomeni G. Redox activation of ATM enhances GSNOR translation to sustain mitophagy and tolerance to oxidative stress. EMBO Rep 2021; 22:e50500. [PMID: 33245190 PMCID: PMC7788447 DOI: 10.15252/embr.202050500] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 10/01/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
The denitrosylase S-nitrosoglutathione reductase (GSNOR) has been suggested to sustain mitochondrial removal by autophagy (mitophagy), functionally linking S-nitrosylation to cell senescence and aging. In this study, we provide evidence that GSNOR is induced at the translational level in response to hydrogen peroxide and mitochondrial ROS. The use of selective pharmacological inhibitors and siRNA demonstrates that GSNOR induction is an event downstream of the redox-mediated activation of ATM, which in turn phosphorylates and activates CHK2 and p53 as intermediate players of this signaling cascade. The modulation of ATM/GSNOR axis, or the expression of a redox-insensitive ATM mutant influences cell sensitivity to nitrosative and oxidative stress, impairs mitophagy and affects cell survival. Remarkably, this interplay modulates T-cell activation, supporting the conclusion that GSNOR is a key molecular effector of the antioxidant function of ATM and providing new clues to comprehend the pleiotropic effects of ATM in the context of immune function.
Collapse
Affiliation(s)
- Claudia Cirotti
- Department of BiologyTor Vergata UniversityRomeItaly
- Laboratory of Cell SignalingIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa LuciaRomeItaly
| | - Salvatore Rizza
- Redox Signaling and Oxidative Stress GroupDanish Cancer Society Research CenterCopenhagenDenmark
| | - Paola Giglio
- Department of BiologyTor Vergata UniversityRomeItaly
| | - Noemi Poerio
- Department of BiologyTor Vergata UniversityRomeItaly
| | - Maria Francesca Allega
- Redox Signaling and Oxidative Stress GroupDanish Cancer Society Research CenterCopenhagenDenmark
- Present address:
Cancer Research UK Beatson InstituteGarscube EstateGlasgowUK
| | | | - Chiara Pecorari
- Redox Signaling and Oxidative Stress GroupDanish Cancer Society Research CenterCopenhagenDenmark
| | - Ji‐Hoon Lee
- Department of Molecular BiosciencesThe University of Texas at AustinAustinTXUSA
| | - Barbara Benassi
- Division of Health Protection TechnologiesENEA‐CasacciaRomeItaly
| | - Daniela Barilà
- Department of BiologyTor Vergata UniversityRomeItaly
- Laboratory of Cell SignalingIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa LuciaRomeItaly
| | - Caroline Robert
- INSERM, U981VillejuifFrance
- Université Paris SudUniversité Paris‐SaclayKremlin‐BicêtreFrance
- Oncology DepartmentGustave RoussyUniversité Paris‐SaclayVillejuifFrance
| | - Jonathan S Stamler
- Institute for Transformative Molecular MedicineCase Western Reserve University and Harrington Discovery InstituteUniversity Hospitals Case Medical CenterClevelandOHUSA
| | - Francesco Cecconi
- Department of BiologyTor Vergata UniversityRomeItaly
- Cell Stress and Survival UnitDanish Cancer Society Research CenterCopenhagenDenmark
- Department of Pediatric Hematology and OncologyIRCCS Bambino Gesù Children's HospitalRomeItaly
| | | | - Tanya T Paull
- Department of Molecular BiosciencesThe University of Texas at AustinAustinTXUSA
| | - Giuseppe Filomeni
- Department of BiologyTor Vergata UniversityRomeItaly
- Redox Signaling and Oxidative Stress GroupDanish Cancer Society Research CenterCopenhagenDenmark
- Center for Healthy AgingCopenhagen UniversityCopenhagenDenmark
| |
Collapse
|
49
|
ATM inhibition enhances Auranofin-induced oxidative stress and cell death in lung cell lines. PLoS One 2020; 15:e0244060. [PMID: 33338048 PMCID: PMC7748142 DOI: 10.1371/journal.pone.0244060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/02/2020] [Indexed: 12/04/2022] Open
Abstract
Ataxia-Telangiectasia (A-T), a pleiotropic chromosomal breakage syndrome, is caused by the loss of the kinase Ataxia-telangiectasia mutated (ATM). ATM is not only involved in the response to DNA damage, but also in sensing and counteracting oxidative stress. Since a disturbed redox balance has been implicated in the pathophysiology of A-T lung disease, we aimed to further explore the interplay between ATM and oxidative stress in lung cells. Using a kinetic trapping approach, we could demonstrate an interaction between the trapping mutant TRX1-CS and ATM upon oxidative stress. We could further show that combined inhibition of thioredoxin reductase (TrxR) and ATM kinase activity, using Auranofin and KU55933 respectively, induced an increase in cellular reactive oxygen species (ROS) levels and protein oxidation in lung cells. Furthermore, ATM inhibition sensitized lung cells to Auranofin-induced cell death that could be rescued by ROS scavengers. As a consequence, targeted reduction of ATM by TRX1 could serve as a regulator of oxidative ATM activation and contribute to the maintenance of the cellular redox homeostasis. These results highlight the importance of the redox-active function of ATM in preventing ROS accumulation and cell death in lung cells.
Collapse
|
50
|
Bhalla K, Jaber S, Reagan K, Hamburg A, Underwood KF, Jhajharia A, Singh M, Bhandary B, Bhat S, Nanaji NM, Hisa R, McCracken C, Creasy HH, Lapidus RG, Kingsbury T, Mayer D, Polster B, Gartenhaus RB. SIRT3, a metabolic target linked to ataxia-telangiectasia mutated (ATM) gene deficiency in diffuse large B-cell lymphoma. Sci Rep 2020; 10:21159. [PMID: 33273545 PMCID: PMC7712916 DOI: 10.1038/s41598-020-78193-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 11/10/2020] [Indexed: 12/16/2022] Open
Abstract
Inactivation of Ataxia-telangiectasia mutated (ATM) gene results in an increased risk to develop cancer. We show that ATM deficiency in diffuse large B-cell lymphoma (DLBCL) significantly induce mitochondrial deacetylase sirtuin-3 (SIRT3) activity, disrupted mitochondrial structure, decreased mitochondrial respiration, and compromised TCA flux compared with DLBCL cells expressing wild type (WT)-ATM. This corresponded to enrichment of glutamate receptor and glutamine pathways in ATM deficient background compared to WT-ATM DLBCL cells. ATM-/- DLBCL cells have decreased apoptosis in contrast to radiosensitive non-cancerous A-T cells. In vivo studies using gain and loss of SIRT3 expression showed that SIRT3 promotes growth of ATM CRISPR knockout DLBCL xenografts compared to wild-type ATM control xenografts. Importantly, screening of DLBCL patient samples identified SIRT3 as a putative therapeutic target, and validated an inverse relationship between ATM and SIRT3 expression. Our data predicts SIRT3 as an important therapeutic target for DLBCL patients with ATM null phenotype.
Collapse
Affiliation(s)
- Kavita Bhalla
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Sausan Jaber
- Department of Anesthesiology, University of Maryland, Baltimore, MD, 21201, USA
| | - Kayla Reagan
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Arielle Hamburg
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Karen F Underwood
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Aditya Jhajharia
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, USA
| | - Maninder Singh
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, USA
| | - Binny Bhandary
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Shambhu Bhat
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Nahid M Nanaji
- Veterans Administration Medical Center, Baltimore, MD, 21201, USA
| | - Ruching Hisa
- Electron Microscopy Core Imaging Facility, Department of Medicine, University of Maryland, Baltimore, USA
| | - Carrie McCracken
- Institute of Genome Sciences, University of Maryland, Baltimore, MD, 21201, USA
| | - Heather Huot Creasy
- Institute of Genome Sciences, University of Maryland, Baltimore, MD, 21201, USA
| | - Rena G Lapidus
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Tami Kingsbury
- Department of Physiology, The Center for Stem Cell Biology and Regenerative Medicine, Baltimore, MD, 21201, USA
| | - Dirk Mayer
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, USA
| | - Brian Polster
- Department of Anesthesiology, University of Maryland, Baltimore, MD, 21201, USA
| | - Ronald B Gartenhaus
- Hunter Holmes McGuire Veterans Administration Medical Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| |
Collapse
|