1
|
Zhu Q, McElroy R, Machhar JS, Cassel J, Zheng Z, Mansoori B, Guo H, Guo S, Pangilinan C, Liang J, Shen D, Zhang L, Liu Q, Kossenkov AV, Altieri DC, Lieberman PM, Gao SJ, Feng P, Murphy ME, Song J, Salvino JM, Liang Q, Jung JU, Liang C. Kaposi's sarcoma-associated herpesvirus induces mitochondrial fission to evade host immune responses and promote viral production. Nat Microbiol 2025:10.1038/s41564-025-02018-3. [PMID: 40404827 DOI: 10.1038/s41564-025-02018-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 04/16/2025] [Indexed: 05/24/2025]
Abstract
Mitochondrial dynamics are pivotal for host immune responses upon infection, yet how viruses manipulate these processes to impair host defence and enhance viral fitness remains unclear. Here we show that Kaposi's sarcoma-associated herpesvirus (KSHV), an oncogenic virus also known as human herpesvirus 8, encodes Bcl-2 (vBcl-2), which reprogrammes mitochondrial architecture. It binds with NM23-H2, a host nucleoside diphosphate (NDP) kinase, to stimulate GTP loading of the dynamin-related protein (DRP1) GTPase, which triggers mitochondrial fission, inhibits mitochondrial antiviral signalling protein (MAVS) aggregation and impairs interferon responses in cell lines. An NM23-H2-binding-defective vBcl-2 mutant fails to evoke fission, leading to defective virion assembly due to activated MAVS-IFN signalling. Notably, we identify two key interferon-stimulated genes restricting vBcl-2-dependent virion morphogenesis. Using a high-throughput drug screening, we discover an inhibitor targeting vBcl-2-NM23-H2 interaction that blocks virion production in vitro. Our study identifies a mechanism in which KSHV manipulates mitochondrial dynamics to allow for virus assembly and shows that targeting the virus-mitochondria interface represents a potential therapeutic strategy.
Collapse
Affiliation(s)
- Qing Zhu
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robert McElroy
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Janvhi Suresh Machhar
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Joel Cassel
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Zihan Zheng
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Behzad Mansoori
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Hongrui Guo
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sen Guo
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Christian Pangilinan
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Jinghui Liang
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Dongliang Shen
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Lu Zhang
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Qin Liu
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Andrew V Kossenkov
- Program in Genome Regulation and Cell Signaling, The Wistar Institute, Philadelphia, PA, USA
| | - Dario C Altieri
- Program in Genome Regulation and Cell Signaling, The Wistar Institute, Philadelphia, PA, USA
| | - Paul M Lieberman
- Program in Genome Regulation and Cell Signaling, The Wistar Institute, Philadelphia, PA, USA
| | - Shou-Jiang Gao
- Cancer Virology Program, University of Pittsburgh Medical Center Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Maureen E Murphy
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Jikui Song
- Department of Biochemistry, University of California, Riverside, CA, USA
| | - Joseph M Salvino
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Qiming Liang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jae U Jung
- Department of Cancer Biology, Department of Infection Biology, and Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chengyu Liang
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA.
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Lechner S, Sha S, Sethiya JP, Szczupak P, Dolot R, Lomada S, Sakhteman A, Tushaus J, Prokofeva P, Krauss M, Breu F, Vögerl K, Morgenstern M, Hrabě de Angelis M, Haucke V, Wieland T, Wagner C, Médard G, Bracher F, Kuster B. Serendipitous and Systematic Chemoproteomic Discovery of MBLAC2, HINT1, and NME1-4 Inhibitors from Histone Deacetylase-Targeting Pharmacophores. ACS Chem Biol 2025. [PMID: 40340313 DOI: 10.1021/acschembio.5c00108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Metalloenzyme inhibitors often incorporate a hydroxamic acid moiety to bind the bivalent metal ion cofactor within the enzyme's active site. Recently, inhibitors of Zn2+-dependent histone deacetylases (HDACs), including clinically advanced drugs, have been identified as potent inhibitors of the metalloenzyme MBLAC2. However, selective chemical probes for MBLAC2, which are essential for studying its inhibitory effects, have not yet been reported. To discover highly selective MBLAC2 inhibitors, we conducted chemoproteomic target deconvolution and selectivity profiling of a library of hydroxamic acid-type molecules and other metal-chelating compounds. This screen revealed MBLAC2 as a frequent off-target of supposedly selective HDAC inhibitors, including the HDAC6 inhibitor SW-100. Profiling a focused library of SW-100-related phenylhydroxamic acids led to identifying two compounds, KV-65 and KV-79, which exhibit nanomolar binding affinity for MBLAC2 and over 60-fold selectivity compared to HDACs. Interestingly, some phenylhydroxamic acids were found to bind additional off-targets. We identified KV-30 as the first drug-like inhibitor of the histidine triad nucleotide-binding protein HINT1 and confirmed its mode of inhibition through a cocrystal structure analysis. Furthermore, we report the discovery of the first inhibitors for the undrugged nucleoside diphosphate kinases NME1, NME2, NME3, and NME4. Overall, this study maps the target and off-target landscape of 53 metalloenzyme inhibitors, providing the first selective MBLAC2 inhibitors. Additionally, the discovery of pharmacophores for NME1-4 and HINT1 establishes a foundation for the future design of potent and selective inhibitors for these targets.
Collapse
Affiliation(s)
- Severin Lechner
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| | - Shuyao Sha
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| | - Jigar Paras Sethiya
- Department of Medicinal Chemistry, University of Minnesota College of Pharmacy, Minneapolis, Minnesota 55414, United States
| | - Patrycja Szczupak
- Division of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Łódź 90-363, Poland
| | - Rafal Dolot
- Division of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Łódź 90-363, Poland
| | - Santosh Lomada
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, Mannheim 68167, Germany
| | - Amirhossein Sakhteman
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| | - Johanna Tushaus
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| | - Polina Prokofeva
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| | - Michael Krauss
- Department of Biology, Chemistry, Pharmacy, Leibniz Institute fur Molecular Pharmacologie, Robert-Roessle-Strasse 10, Berlin 13125, Germany
| | - Ferdinand Breu
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians University Munich, Munich 81377, Germany
| | - Katharina Vögerl
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians University Munich, Munich 81377, Germany
| | - Martin Morgenstern
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians University Munich, Munich 81377, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Experimental Genetics, Neuherberg 85764, Germany
- Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Freising 85354, Germany
- German Center for Diabetes Research (DZD), Neuherberg 85764, Germany
| | - Volker Haucke
- Department of Biology, Chemistry, Pharmacy, Leibniz Institute fur Molecular Pharmacologie, Robert-Roessle-Strasse 10, Berlin 13125, Germany
| | - Thomas Wieland
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, Mannheim 68167, Germany
| | - Carston Wagner
- Department of Medicinal Chemistry, University of Minnesota College of Pharmacy, Minneapolis, Minnesota 55414, United States
| | - Guillaume Médard
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| | - Franz Bracher
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians University Munich, Munich 81377, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| |
Collapse
|
3
|
Fan Z, Mao X, Zhu M, Hu X, Li M, Huang L, Li J, Maimaiti T, Zuo X, Fan C, Li Q, Liu M, Tian Y. Probing Twist-Induced Endocytotic Membrane Fission using Anisotropic Gold Homodimers. Angew Chem Int Ed Engl 2025; 64:e202413244. [PMID: 39227862 DOI: 10.1002/anie.202413244] [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: 07/14/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/05/2024]
Abstract
Membrane fission involves a crucial step of lipid remodeling, in which the dynamin collar constricts and severs the tubulated lipid membrane at the neck of budding vesicles. Nevertheless, the difficulty in accurately determining the rotational dynamics of live endocytotic vesicles poses a limit on the elucidation of dynamin-induced membrane remodeling for endocytotic vesicle scission. Herein, we designed a DNA-modified gold homodimer (AuHD)-based anisotropic plasmonic probe with uniform surface chemistry, minimizing orientational fluctuation within vesicle encapsulation. Using AuHDs as cargos to image the dynamics of cargo-containing vesicles during endocytosis, we showed that, prior to detachment from plasma membrane, the cargo-containing vesicles underwent multiple intermittent twists of ~4° angular orientation relative to plasma membrane with a ~0.2 s dwell time. These findings suggest that the membrane torques resulting from dynamin actions in vivo constitute the pathway to membrane fission, potentially shedding light on how dynamin-mediated lipid remodeling orchestrates membrane fission.
Collapse
Affiliation(s)
- Zhiying Fan
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 200241, Shanghai, China
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Xiuhai Mao
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Meng Zhu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 200241, Shanghai, China
| | - Xingjie Hu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Mingqiang Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Institute of Translational Medicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| | - Lulu Huang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 200241, Shanghai, China
| | - Jie Li
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 200241, Shanghai, China
| | - Tumala Maimaiti
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 200241, Shanghai, China
| | - Xiaolei Zuo
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Institute of Translational Medicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Institute of Translational Medicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| | - Mengmeng Liu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 200241, Shanghai, China
| | - Yang Tian
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 200241, Shanghai, China
| |
Collapse
|
4
|
Houles T, Yoon SO, Roux PP. The expanding landscape of canonical and non-canonical protein phosphorylation. Trends Biochem Sci 2024; 49:986-999. [PMID: 39266329 DOI: 10.1016/j.tibs.2024.08.004] [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: 03/29/2024] [Revised: 08/01/2024] [Accepted: 08/14/2024] [Indexed: 09/14/2024]
Abstract
Protein phosphorylation is a crucial regulatory mechanism in cell signaling, acting as a molecular switch that modulates protein function. Catalyzed by protein kinases and reversed by phosphoprotein phosphatases, it is essential in both normal physiological and pathological states. Recent advances have uncovered a vast and intricate landscape of protein phosphorylation that include histidine phosphorylation and more unconventional events, such as pyrophosphorylation and polyphosphorylation. Many questions remain about the true size of the phosphoproteome and, more importantly, its site-specific functional relevance. The involvement of unconventional actors such as pseudokinases and pseudophosphatases adds further complexity to be resolved. This review explores recent discoveries and ongoing challenges, highlighting the need for continued research to fully elucidate the roles and regulation of protein phosphorylation.
Collapse
Affiliation(s)
- Thibault Houles
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, Canada; Institute of Molecular Genetics of Montpellier (IGMM), Université de Montpellier, CNRS, Montpellier, France.
| | - Sang-Oh Yoon
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Philippe P Roux
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, Canada; Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
5
|
Musliha A, Dermawan D, Rahayu P, Tjandrawinata RR. Unraveling modulation effects on albumin synthesis and inflammation by Striatin, a bioactive protein fraction isolated from Channa striata: In silico proteomics and in vitro approaches. Heliyon 2024; 10:e38386. [PMID: 39398063 PMCID: PMC11467539 DOI: 10.1016/j.heliyon.2024.e38386] [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: 11/22/2023] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024] Open
Abstract
Hypoalbuminemia, associated with inflammation in severely ill patients, can emerge due to decreased albumin production. Transforming growth factor-beta (TGF-β) and nuclear factor-kappa B (NF-κB) are critical signaling pathways responsible for decreased albumin expression. This study explores the protein content and modulation effects of Striatin on albumin synthesis and inflammation, employing in silico proteomics and in vitro investigations. In the in silico proteomics realm, LC/MS-MS protein sequencing, 3D modeling, protein-protein docking simulations, 100 ns molecular dynamics (MD) simulations, and MM/PBSA binding free energy calculations were carried out. Complementing this, in vitro studies examined Albumin gene expression and extracellular secretion in HepG2 cells subjected to lipopolysaccharides-induced hypoalbuminemia. Furthermore, the study probed Striatin's influence on the NF-ᴋB expression, given albumin's role as a negative acute-phase protein. The results unveiled nucleoside diphosphate kinase (NdK) and parvalbumin (PV) as the prominent constituents within Striatin. Notably, NdK and PV exhibited the ability to disrupt hydrogen bonds with specific residues in both TGF-β and NF-κB complexes, thereby enhancing their flexibility, akin to the action of the FKBP12 complex (antagonist complex). In the in vitro experiments, Striatin demonstrated a dose and time-dependent inhibition of hypoalbuminemia, with peak efficacy observed at a concentration of 20 μg/mL. At this concentration, Striatin also suppressed NF-κB expression when co-incubated with lipopolysaccharides. While these findings suggest potential inhibitory effects of Striatin on TGF-β and NF-κB activities, they are preliminary and warrant further investigation. This study highlights Striatin's potential as a therapeutic agent for inflammation-related hypoalbuminemia, though additional research is needed to fully validate these results.
Collapse
Affiliation(s)
- Affina Musliha
- Dexa Laboratories of Biomolecular Sciences, PT Dexa Medica, Jababeka Industrial Estate II, Jl. Industri Selatan V Blok PP No. 7 Cikarang, 17550, Indonesia
| | - Doni Dermawan
- Dexa Laboratories of Biomolecular Sciences, PT Dexa Medica, Jababeka Industrial Estate II, Jl. Industri Selatan V Blok PP No. 7 Cikarang, 17550, Indonesia
| | - Puji Rahayu
- Dexa Laboratories of Biomolecular Sciences, PT Dexa Medica, Jababeka Industrial Estate II, Jl. Industri Selatan V Blok PP No. 7 Cikarang, 17550, Indonesia
| | - Raymond R. Tjandrawinata
- Dexa Laboratories of Biomolecular Sciences, PT Dexa Medica, Jababeka Industrial Estate II, Jl. Industri Selatan V Blok PP No. 7 Cikarang, 17550, Indonesia
- Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, South Jakarta 12930, Indonesia
| |
Collapse
|
6
|
Khatun J, Gelles JD, Chipuk JE. Dynamic death decisions: How mitochondrial dynamics shape cellular commitment to apoptosis and ferroptosis. Dev Cell 2024; 59:2549-2565. [PMID: 39378840 PMCID: PMC11469553 DOI: 10.1016/j.devcel.2024.09.004] [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: 06/03/2024] [Revised: 08/15/2024] [Accepted: 09/03/2024] [Indexed: 10/10/2024]
Abstract
The incorporation of mitochondria into early eukaryotes established organelle-based biochemistry and enabled metazoan development. Diverse mitochondrial biochemistry is essential for life, and its homeostatic control via mitochondrial dynamics supports organelle quality and function. Mitochondrial crosstalk with numerous regulated cell death (RCD) pathways controls the decision to die. In this review, we will focus on apoptosis and ferroptosis, two distinct forms of RCD that utilize divergent signaling to kill a targeted cell. We will highlight how proteins and processes involved in mitochondrial dynamics maintain biochemically diverse subcellular compartments to support apoptosis and ferroptosis machinery, as well as unite disparate RCD pathways through dual control of organelle biochemistry and the decision to die.
Collapse
Affiliation(s)
- Jesminara Khatun
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Jesse D Gelles
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Jerry Edward Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA.
| |
Collapse
|
7
|
Tseng WW, Chu CH, Lee YJ, Zhao S, Chang C, Ho YP, Wei AC. Metabolic regulation of mitochondrial morphologies in pancreatic beta cells: coupling of bioenergetics and mitochondrial dynamics. Commun Biol 2024; 7:1267. [PMID: 39369076 PMCID: PMC11455970 DOI: 10.1038/s42003-024-06955-3] [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: 12/02/2021] [Accepted: 09/24/2024] [Indexed: 10/07/2024] Open
Abstract
Cellular bioenergetics and mitochondrial dynamics are crucial for the secretion of insulin by pancreatic beta cells in response to elevated levels of blood glucose. To elucidate the interactions between energy production and mitochondrial fission/fusion dynamics, we combine live-cell mitochondria imaging with biophysical-based modeling and graph-based network analysis. The aim is to determine the mechanism that regulates mitochondrial morphology and balances metabolic demands in pancreatic beta cells. A minimalistic differential equation-based model for beta cells is constructed that includes glycolysis, oxidative phosphorylation, calcium dynamics, and fission/fusion dynamics, with ATP synthase flux and proton leak flux as main regulators of mitochondrial dynamics. The model shows that mitochondrial fission occurs in response to hyperglycemia, starvation, ATP synthase inhibition, uncoupling, and diabetic conditions, in which the rate of proton leakage exceeds the rate of mitochondrial ATP synthesis. Under these metabolic challenges, the propensities of tip-to-tip fusion events simulated from the microscopy images of the mitochondrial networks are lower than those in the control group and prevent the formation of mitochondrial networks. The study provides a quantitative framework that couples bioenergetic regulation with mitochondrial dynamics, offering insights into how mitochondria adapt to metabolic challenges.
Collapse
Affiliation(s)
- Wen-Wei Tseng
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Ching-Hsiang Chu
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Yi-Ju Lee
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Shirui Zhao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Centre for Novel Biomaterials, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Hong Kong Branch of the CAS Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- The Ministry of Education Key Laboratory of Regeneration Medicine, Shatin, New Territories, Hong Kong SAR, China
| | - Chen Chang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Yi-Ping Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Centre for Novel Biomaterials, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Hong Kong Branch of the CAS Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- The Ministry of Education Key Laboratory of Regeneration Medicine, Shatin, New Territories, Hong Kong SAR, China
| | - An-Chi Wei
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan.
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
8
|
Amjadi R, Werten S, Lomada SK, Baldin C, Scheffzek K, Dunzendorfer-Matt T, Wieland T. Mechanistic Insights into Substrate Recognition of Human Nucleoside Diphosphate Kinase C Based on Nucleotide-Induced Structural Changes. Int J Mol Sci 2024; 25:9768. [PMID: 39337255 PMCID: PMC11431768 DOI: 10.3390/ijms25189768] [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: 08/08/2024] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
Nucleoside diphosphate kinases (NDPKs) are encoded by nme genes and exist in various isoforms. Based on interactions with other proteins, they are involved in signal transduction, development and pathological processes such as tumorigenesis, metastasis and heart failure. In this study, we report a 1.25 Å resolution structure of human homohexameric NDPK-C bound to ADP and describe the yet unknown complexes formed with GDP, UDP and cAMP, all obtained at a high resolution via X-ray crystallography. Each nucleotide represents a distinct group of mono- or diphosphate purine or pyrimidine bases. We analyzed different NDPK-C nucleotide complexes in the presence and absence of Mg2+ and explain how this ion plays an essential role in NDPKs' phosphotransferase activity. By analyzing a nucleotide-depleted NDPK-C structure, we detected conformational changes upon substrate binding and identify flexible regions in the substrate binding site. A comparison of NDPK-C with other human isoforms revealed a strong similarity in the overall composition with regard to the 3D structure, but significant differences in the charge and hydrophobicity of the isoforms' surfaces. This may play a role in isoform-specific NDPK interactions with ligands and/or important complex partners like other NDPK isoforms, as well as monomeric and heterotrimeric G proteins. Considering the recently discovered role of NDPK-C in different pathologies, these high-resolution structures thus might provide a basis for interaction studies with other proteins or small ligands, like activators or inhibitors.
Collapse
Affiliation(s)
- Rezan Amjadi
- Institute of Molecular Biochemistry, Medical University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (R.A.); (K.S.)
| | - Sebastiaan Werten
- Department of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria;
| | - Santosh Kumar Lomada
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13–17, 68167 Mannheim, Germany;
| | - Clara Baldin
- Department of Microbiology, University of Innsbruck, Technikerstraße 25, 6020 Innsbruck, Austria;
| | - Klaus Scheffzek
- Institute of Molecular Biochemistry, Medical University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (R.A.); (K.S.)
| | - Theresia Dunzendorfer-Matt
- Institute of Molecular Biochemistry, Medical University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (R.A.); (K.S.)
| | - Thomas Wieland
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13–17, 68167 Mannheim, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 68167 Mannheim, Germany
| |
Collapse
|
9
|
Proust B, Horvat A, Tadijan A, Vlašić I, Herak Bosnar M. Mitochondrial NME6 Influences Basic Cellular Processes in Tumor Cells In Vitro. Int J Mol Sci 2024; 25:9580. [PMID: 39273527 PMCID: PMC11395177 DOI: 10.3390/ijms25179580] [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: 06/10/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
NME6 belongs to the family of nucleoside diphosphate kinase enzymes, whose major role is to transfer the terminal phosphate from NTPs, mostly ATP, to other (d)NDPs via a high-energy intermediate. Beside this basic enzymatic activity, the family, comprising 10 genes/proteins in humans, executes a number of diverse biochemical/biological functions in the cell. A few previous studies have reported that NME6 resides in the mitochondria and influences oxidative phosphorylation while interacting with RCC1L, a GTPase involved in mitochondrial ribosome assembly and translation. Considering the multifunctional role of NME family members, the goal of the present study was to assess the influence of the overexpression or silencing of NME6 on fundamental cellular events of MDA-MB-231T metastatic breast cancer cells. Using flow cytometry, Western blotting, and a wound-healing assay, we demonstrated that the overexpression of NME6 reduces cell migration and alters the expression of EMT (epithelial-mesenchymal transition) markers. In addition, NME6 overexpression influences cell cycle distribution exclusively upon DNA damage and impacts the MAPK/ERK signaling pathway, while it has no effect on apoptosis. To conclude, our results demonstrate that NME6 is involved in different cellular processes, providing a solid basis for future, more precise investigations of its role.
Collapse
Affiliation(s)
| | | | | | | | - Maja Herak Bosnar
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička Cesta 54, 10002 Zagreb, Croatia; (B.P.); (A.H.); (A.T.); (I.V.)
| |
Collapse
|
10
|
Iyer P, Zhang B, Liu T, Jin M, Hart K, Zhang J, Siegert V, Remke M, Wang X, Yu L, Song J, Venkataraman G, Chan WC, Jia Z, Buchner M, Siddiqi T, Rosen ST, Danilov A, Wang L. MGA deletion leads to Richter's transformation by modulating mitochondrial OXPHOS. Sci Transl Med 2024; 16:eadg7915. [PMID: 39083585 DOI: 10.1126/scitranslmed.adg7915] [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: 01/20/2023] [Revised: 03/08/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024]
Abstract
Richter's transformation (RT) is a progression of chronic lymphocytic leukemia (CLL) to aggressive lymphoma. MGA (Max gene associated), a functional MYC suppressor, is mutated at 3% in CLL and 36% in RT. However, genetic models and molecular mechanisms of MGA deletion that drive CLL to RT remain elusive. We established an RT mouse model by knockout of Mga in the Sf3b1/Mdr CLL model using CRISPR-Cas9 to determine the role of Mga in RT. Murine RT cells exhibited mitochondrial aberrations with elevated oxidative phosphorylation (OXPHOS). Through RNA sequencing and functional characterization, we identified Nme1 (nucleoside diphosphate kinase) as an Mga target, which drives RT by modulating OXPHOS. Given that NME1 is also a known MYC target without targetable compounds, we found that concurrent inhibition of MYC and electron transport chain complex II substantially prolongs the survival of RT mice in vivo. Our results suggest that the Mga-Nme1 axis drives murine CLL-to-RT transition via modulating OXPHOS, highlighting a potential therapeutic avenue for RT.
Collapse
MESH Headings
- Animals
- Oxidative Phosphorylation
- Mitochondria/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Mice
- Gene Deletion
- Humans
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Proto-Oncogene Proteins c-myc/metabolism
- Proto-Oncogene Proteins c-myc/genetics
- Disease Models, Animal
Collapse
Affiliation(s)
- Prajish Iyer
- Department of Systems Biology, Beckman Research Institute, City of Hope National Comprehensive Cancer Center, Monrovia, CA 91016, USA
| | - Bo Zhang
- Department of Systems Biology, Beckman Research Institute, City of Hope National Comprehensive Cancer Center, Monrovia, CA 91016, USA
| | - Tingting Liu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Meiling Jin
- Department of Systems Biology, Beckman Research Institute, City of Hope National Comprehensive Cancer Center, Monrovia, CA 91016, USA
| | - Kevyn Hart
- Department of Systems Biology, Beckman Research Institute, City of Hope National Comprehensive Cancer Center, Monrovia, CA 91016, USA
| | - Jibin Zhang
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
- Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Viola Siegert
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine and Health, Technical University of Munich, Munich 81675, Germany
- Central Institute for Translational Cancer Research, Technische Universität München, Munich 81675, Germany
| | - Marianne Remke
- Institute of Pathology, TUM School of Medicine and Health, Technical University of Munich, Munich 81675, Germany
| | - Xuesong Wang
- Department of Botany and Plant Sciences, University of California, Riverside, CA 92507, USA
- Graduate Program in Genetics, Genomics, and Bioinformatics, University of California, Riverside, CA 92507, USA
| | - Lei Yu
- Department of Botany and Plant Sciences, University of California, Riverside, CA 92507, USA
- Graduate Program in Genetics, Genomics, and Bioinformatics, University of California, Riverside, CA 92507, USA
| | - Joo Song
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
- Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | | | - Wing C Chan
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
- Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Zhenyu Jia
- Department of Botany and Plant Sciences, University of California, Riverside, CA 92507, USA
| | - Maike Buchner
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine and Health, Technical University of Munich, Munich 81675, Germany
- Central Institute for Translational Cancer Research, Technische Universität München, Munich 81675, Germany
| | - Tanya Siddiqi
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
- Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Steven T Rosen
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
- Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Alexey Danilov
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
- Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Lili Wang
- Department of Systems Biology, Beckman Research Institute, City of Hope National Comprehensive Cancer Center, Monrovia, CA 91016, USA
- Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
11
|
Proust B, Herak Bosnar M, Ćetković H, Tokarska-Schlattner M, Schlattner U. Mitochondrial NME6: A Paradigm Change within the NME/NDP Kinase Protein Family? Cells 2024; 13:1278. [PMID: 39120309 PMCID: PMC11312278 DOI: 10.3390/cells13151278] [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: 07/02/2024] [Revised: 07/27/2024] [Accepted: 07/27/2024] [Indexed: 08/10/2024] Open
Abstract
Eukaryotic NMEs/NDP kinases are a family of 10 multifunctional proteins that occur in different cellular compartments and interact with various cellular components (proteins, membranes, and DNA). In contrast to the well-studied Group I NMEs (NME1-4), little is known about the more divergent Group II NMEs (NME5-9). Three recent publications now shed new light on NME6. First, NME6 is a third mitochondrial NME, largely localized in the matrix space, associated with the mitochondrial inner membrane. Second, while its monomeric form is inactive, NME6 gains NDP kinase activity through interaction with mitochondrial RCC1L. This challenges the current notion that mammalian NMEs require the formation of hexamers to become active. The formation of complexes between NME6 and RCC1L, likely heterodimers, seemingly obviates the necessity for hexamer formation, stabilizing a NDP kinase-competent conformation. Third, NME6 is involved in mitochondrial gene maintenance and expression by providing (d)NTPs for replication and transcription (in particular the pyrimidine nucleotides) and by a less characterized mechanism that supports mitoribosome function. This review offers an overview of NME evolution and structure and highlights the new insight into NME6. The new findings position NME6 as the most comprehensively studied protein in NME Group II and may even suggest it as a new paradigm for related family members.
Collapse
Affiliation(s)
- Bastien Proust
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia;
| | - Maja Herak Bosnar
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia;
| | - Helena Ćetković
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia;
| | | | - Uwe Schlattner
- Univ. Grenoble Alpes, Inserm U1055, Lab. of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France;
- Institut Universitaire de France (IUF), 75231 Paris, France
| |
Collapse
|
12
|
Ning J, Sala M, Reina J, Kalagiri R, Hunter T, McCullough BS. Histidine Phosphorylation: Protein Kinases and Phosphatases. Int J Mol Sci 2024; 25:7975. [PMID: 39063217 PMCID: PMC11277029 DOI: 10.3390/ijms25147975] [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: 06/07/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Phosphohistidine (pHis) is a reversible protein post-translational modification (PTM) that is currently poorly understood. The P-N bond in pHis is heat and acid-sensitive, making it more challenging to study than the canonical phosphoamino acids pSer, pThr, and pTyr. As advancements in the development of tools to study pHis have been made, the roles of pHis in cells are slowly being revealed. To date, a handful of enzymes responsible for controlling this modification have been identified, including the histidine kinases NME1 and NME2, as well as the phosphohistidine phosphatases PHPT1, LHPP, and PGAM5. These tools have also identified the substrates of these enzymes, granting new insights into previously unknown regulatory mechanisms. Here, we discuss the cellular function of pHis and how it is regulated on known pHis-containing proteins, as well as cellular mechanisms that regulate the activity of the pHis kinases and phosphatases themselves. We further discuss the role of the pHis kinases and phosphatases as potential tumor promoters or suppressors. Finally, we give an overview of various tools and methods currently used to study pHis biology. Given their breadth of functions, unraveling the role of pHis in mammalian systems promises radical new insights into existing and unexplored areas of cell biology.
Collapse
Affiliation(s)
- Jia Ning
- Correspondence: (J.N.); (B.S.M.)
| | | | | | | | | | - Brandon S. McCullough
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; (M.S.); (J.R.); (R.K.); (T.H.)
| |
Collapse
|
13
|
Scott KL, Halfmann CT, Hoefakker AD, Purkayastha P, Wang TC, Lele TP, Roux KJ. Nucleocytoplasmic transport rates are regulated by cellular processes that modulate GTP availability. J Cell Biol 2024; 223:e202308152. [PMID: 38683248 PMCID: PMC11059771 DOI: 10.1083/jcb.202308152] [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: 09/01/2023] [Revised: 03/08/2024] [Accepted: 04/01/2024] [Indexed: 05/01/2024] Open
Abstract
Nucleocytoplasmic transport (NCT), the facilitated diffusion of cargo molecules between the nucleus and cytoplasm through nuclear pore complexes (NPCs), enables numerous fundamental eukaryotic cellular processes. Ran GTPase uses cellular energy in the direct form of GTP to create a gradient across the nuclear envelope (NE) that drives the majority of NCT. We report here that changes in GTP availability resulting from altered cellular physiology modulate the rate of NCT, as monitored using synthetic and natural cargo, and the dynamics of Ran itself. Cell migration, cell spreading, and/or modulation of the cytoskeleton or its connection to the nucleus alter GTP availability and thus rates of NCT, regulating RNA export and protein synthesis. These findings support a model in which changes in cellular physiology that alter GTP availability can regulate the rate of NCT, impacting fundamental cellular processes that extensively utilize NCT.
Collapse
Affiliation(s)
- Kelsey L. Scott
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD, USA
| | | | - Allison D. Hoefakker
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD, USA
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Purboja Purkayastha
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Ting Ching Wang
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Tanmay P. Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
- Department of Translational Medical Sciences, Texas A&M University, Houston, TX, USA
| | - Kyle J. Roux
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| |
Collapse
|
14
|
Ikeda A, Iijima M, Sesaki H. Systemic phospho-defective and phospho-mimetic Drp1 mice exhibit normal growth and development with altered anxiety-like behavior. iScience 2024; 27:109874. [PMID: 38784001 PMCID: PMC11112374 DOI: 10.1016/j.isci.2024.109874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/21/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Mitochondrial division controls the size, distribution, and turnover of this essential organelle. A dynamin-related GTPase, Drp1, drives membrane division as a force-generating mechano-chemical enzyme. Drp1 is regulated by multiple mechanisms, including phosphorylation at two primary sites: serine 579 and serine 600. While previous studies in cell culture systems have shown that Drp1 S579 phosphorylation promotes mitochondrial division, its physiological functions remained unclear. Here, we generated phospho-mimetic Drp1 S579D and phospho-defective Drp1 S579R mice using the CRISPR-Cas system. Both mouse models exhibited normal growth, development, and breeding. We found that Drp1 is highly phosphorylated at S579 in brain neurons. Notably, the Drp1 S579D mice showed decreased anxiety-like behaviors, whereas the Drp1 S579R mice displayed increased anxiety-like behaviors. These findings suggest a critical role for Drp1 S579 phosphorylation in brain function. The Drp1 S579D and S579R mice thus offer valuable in vivo models for specific analysis of Drp1 S579 phosphorylation.
Collapse
Affiliation(s)
- Arisa Ikeda
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21212, USA
| | - Miho Iijima
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21212, USA
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21212, USA
| |
Collapse
|
15
|
Ferrucci V, Lomada S, Wieland T, Zollo M. PRUNE1 and NME/NDPK family proteins influence energy metabolism and signaling in cancer metastases. Cancer Metastasis Rev 2024; 43:755-775. [PMID: 38180572 PMCID: PMC11156750 DOI: 10.1007/s10555-023-10165-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/19/2023] [Indexed: 01/06/2024]
Abstract
We describe here the molecular basis of the complex formation of PRUNE1 with the tumor metastasis suppressors NME1 and NME2, two isoforms appertaining to the nucleoside diphosphate kinase (NDPK) enzyme family, and how this complex regulates signaling the immune system and energy metabolism, thereby shaping the tumor microenvironment (TME). Disrupting the interaction between NME1/2 and PRUNE1, as suggested, holds the potential to be an excellent therapeutic target for the treatment of cancer and the inhibition of metastasis dissemination. Furthermore, we postulate an interaction and regulation of the other Class I NME proteins, NME3 and NME4 proteins, with PRUNE1 and discuss potential functions. Class I NME1-4 proteins are NTP/NDP transphosphorylases required for balancing the intracellular pools of nucleotide diphosphates and triphosphates. They regulate different cellular functions by interacting with a large variety of other proteins, and in cancer and metastasis processes, they can exert pro- and anti-oncogenic properties depending on the cellular context. In this review, we therefore additionally discuss general aspects of class1 NME and PRUNE1 molecular structures as well as their posttranslational modifications and subcellular localization. The current knowledge on the contributions of PRUNE1 as well as NME proteins to signaling cascades is summarized with a special regard to cancer and metastasis.
Collapse
Affiliation(s)
- Veronica Ferrucci
- Department of Molecular Medicine and Medical Biotechnology, DMMBM, University of Naples, Federico II, Via Pansini 5, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate "Franco Salvatore", Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Santosh Lomada
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
- DZHK, German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, 68167, Mannheim, Germany
| | - Thomas Wieland
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany.
- DZHK, German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, 68167, Mannheim, Germany.
- Medical Faculty Mannheim, Ludolf Krehl-Str. 13-17, 68167, Mannheim, Germany.
| | - Massimo Zollo
- Department of Molecular Medicine and Medical Biotechnology, DMMBM, University of Naples, Federico II, Via Pansini 5, 80131, Naples, Italy.
- CEINGE Biotecnologie Avanzate "Franco Salvatore", Via Gaetano Salvatore 486, 80145, Naples, Italy.
- DAI Medicina di Laboratorio e Trasfusionale, 'AOU' Federico II Policlinico, 80131, Naples, Italy.
| |
Collapse
|
16
|
Mao X, Li L, Abubakar YS, Li Y, Luo Z, Chen M, Zheng W, Wang Z, Zheng H. Nucleoside Diphosphate Kinase FgNdpk Is Required for DON Production and Pathogenicity by Regulating the Growth and Toxisome Formation of Fusarium graminearum. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:9637-9646. [PMID: 38642053 DOI: 10.1021/acs.jafc.4c00593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/22/2024]
Abstract
Nucleoside diphosphate kinases (NDPKs) are nucleotide metabolism enzymes that play different physiological functions in different species. However, the roles of NDPK in phytopathogen and mycotoxin production are not well understood. In this study, we showed that Fusarium graminearum FgNdpk is important for vegetative growth, conidiation, sexual development, and pathogenicity. Furthermore, FgNdpk is required for deoxynivalenol (DON) production; deletion of FgNDPK downregulates the expression of DON biosynthesis genes and disrupts the formation of FgTri4-GFP-labeled toxisomes, while overexpression of FgNDPK significantly increases DON production. Interestingly, FgNdpk colocalizes with the DON biosynthesis proteins FgTri1 and FgTri4 in the toxisome, and coimmunoprecipitation (Co-IP) assays show that FgNdpk associates with FgTri1 and FgTri4 in vivo and regulates their localizations and expressions, respectively. Taken together, these data demonstrate that FgNdpk is important for vegetative growth, conidiation, and pathogenicity and acts as a key protein that regulates toxisome formation and DON biosynthesis in F. graminearum.
Collapse
Affiliation(s)
- Xuzhao Mao
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou 350108, China
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Lingping Li
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yakubu Saddeeq Abubakar
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria 810281, Nigeria
| | - Yulong Li
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou 350108, China
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Zenghong Luo
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Meilian Chen
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou 350108, China
| | - Wenhui Zheng
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Zonghua Wang
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou 350108, China
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Huawei Zheng
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou 350108, China
| |
Collapse
|
17
|
Chen CW, Su C, Huang CY, Huang XR, Cuili X, Chao T, Fan CH, Ting CW, Tsai YW, Yang KC, Yeh TY, Hsieh ST, Chen YJ, Feng Y, Hunter T, Chang ZF. NME3 is a gatekeeper for DRP1-dependent mitophagy in hypoxia. Nat Commun 2024; 15:2264. [PMID: 38480688 PMCID: PMC10938004 DOI: 10.1038/s41467-024-46385-7] [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: 07/11/2023] [Accepted: 02/23/2024] [Indexed: 03/17/2024] Open
Abstract
NME3 is a member of the nucleoside diphosphate kinase (NDPK) family localized on the mitochondrial outer membrane (MOM). Here, we report a role of NME3 in hypoxia-induced mitophagy dependent on its active site phosphohistidine but not the NDPK function. Mice carrying a knock-in mutation in the Nme3 gene disrupting NME3 active site histidine phosphorylation are vulnerable to ischemia/reperfusion-induced infarction and develop abnormalities in cerebellar function. Our mechanistic analysis reveals that hypoxia-induced phosphatidic acid (PA) on mitochondria is essential for mitophagy and the interaction of DRP1 with NME3. The PA binding function of MOM-localized NME3 is required for hypoxia-induced mitophagy. Further investigation demonstrates that the interaction with active NME3 prevents DRP1 susceptibility to MUL1-mediated ubiquitination, thereby allowing a sufficient amount of active DRP1 to mediate mitophagy. Furthermore, MUL1 overexpression suppresses hypoxia-induced mitophagy, which is reversed by co-expression of ubiquitin-resistant DRP1 mutant or histidine phosphorylatable NME3. Thus, the site-specific interaction with active NME3 provides DRP1 a microenvironment for stabilization to proceed the segregation process in mitophagy.
Collapse
Affiliation(s)
- Chih-Wei Chen
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, 10002, Taipei, Taiwan
| | - Chi Su
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, 10002, Taipei, Taiwan
| | - Chang-Yu Huang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, 10002, Taipei, Taiwan
| | - Xuan-Rong Huang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, 10002, Taipei, Taiwan
| | - Xiaojing Cuili
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, 10002, Taipei, Taiwan
| | - Tung Chao
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, 10002, Taipei, Taiwan
| | - Chun-Hsiang Fan
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, 10002, Taipei, Taiwan
| | - Cheng-Wei Ting
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, 10002, Taipei, Taiwan
| | - Yi-Wei Tsai
- Institute of Pharmacology, College of Medicine, National Taiwan University, 10002, Taipei, Taiwan
- Department of Medical Research, National Taiwan University Hospital, 10002, Taipei, Taiwan
| | - Kai-Chien Yang
- Institute of Pharmacology, College of Medicine, National Taiwan University, 10002, Taipei, Taiwan
| | - Ti-Yen Yeh
- Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, 10002, Taipei, Taiwan
| | - Sung-Tsang Hsieh
- Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, 10002, Taipei, Taiwan
| | - Yi-Ju Chen
- Institute of Chemistry, Academia Sinica, 11529, Taipei, Taiwan
| | - Yuxi Feng
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute, La Jolla, CA, 92037-1002, USA
| | - Zee-Fen Chang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, 10002, Taipei, Taiwan.
- Center of Precision Medicine, College of Medicine, National Taiwan University, 10002, Taipei, Taiwan.
| |
Collapse
|
18
|
Lu Y, Xu J, Li Y, Wang R, Dai C, Zhang B, Zhang X, Xu L, Tao Y, Han M, Guo R, Wu Q, Wu L, Meng Z, Tan M, Li J. DRAK2 suppresses autophagy by phosphorylating ULK1 at Ser 56 to diminish pancreatic β cell function upon overnutrition. Sci Transl Med 2024; 16:eade8647. [PMID: 38324636 DOI: 10.1126/scitranslmed.ade8647] [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: 09/23/2022] [Accepted: 01/12/2024] [Indexed: 02/09/2024]
Abstract
Impeded autophagy can impair pancreatic β cell function by causing apoptosis, of which DAP-related apoptosis-inducing kinase-2 (DRAK2) is a critical regulator. Here, we identified a marked up-regulation of DRAK2 in pancreatic tissue across humans, macaques, and mice with type 2 diabetes (T2D). Further studies in mice showed that conditional knockout (cKO) of DRAK2 in pancreatic β cells protected β cell function against high-fat diet feeding along with sustained autophagy and mitochondrial function. Phosphoproteome analysis in isolated mouse primary islets revealed that DRAK2 directly phosphorylated unc-51-like autophagy activating kinase 1 (ULK1) at Ser56, which was subsequently found to induce ULK1 ubiquitylation and suppress autophagy. ULK1-S56A mutation or pharmacological inhibition of DRAK2 preserved mitochondrial function and insulin secretion against lipotoxicity in mouse primary islets, Min6 cells, or INS-1E cells. In conclusion, these findings together indicate an indispensable role of the DRAK2-ULK1 axis in pancreatic β cells upon metabolic challenge, which offers a potential target to protect β cell function in T2D.
Collapse
Affiliation(s)
- Yuting Lu
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
| | - Junyu Xu
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong 528400, P. R. China
| | - Yufeng Li
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
| | - Ruoran Wang
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P. R. China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China
| | - Chengqiu Dai
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Bingqian Zhang
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xinwen Zhang
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
| | - Lei Xu
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong 528400, P. R. China
| | - Yunhua Tao
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
| | - Ming Han
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
| | - Ren Guo
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
| | - Qingqian Wu
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P. R. China
| | - Linshi Wu
- Shanghai Jiaotong University, School of Medicine, Renji Hospital, Shanghai, 201112, P. R. China
| | - Zhuoxian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P. R. China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P. R. China
| | - Minjia Tan
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong 528400, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jingya Li
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
19
|
Liu A, Hatch AL, Higgs HN. Effects of phosphorylation on Drp1 activation by its receptors, actin, and cardiolipin. Mol Biol Cell 2024; 35:ar16. [PMID: 38019609 PMCID: PMC10881151 DOI: 10.1091/mbc.e23-11-0427] [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: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 12/01/2023] Open
Abstract
Drp1 is a dynamin family GTPase required for mitochondrial and peroxisomal division. Oligomerization increases Drp1 GTPase activity through interactions between neighboring GTPase domains. In cells, Drp1 is regulated by several factors including Drp1 receptors, actin filaments, cardiolipin, and phosphorylation at two sites: S579 and S600. Commonly, phosphorylation of S579 is considered activating, while S600 phosphorylation is considered inhibiting. However, direct effects of phosphorylation on Drp1 GTPase activity have not been investigated in detail. Here, we compare effects of S579 and S600 phosphorylation on purified Drp1, using phosphomimetic mutants and in vitro phosphorylation. Both phosphomimetic mutants are shifted toward smaller oligomers. Both phosphomimetic mutations maintain basal GTPase activity, but eliminate GTPase stimulation by actin and decrease GTPase stimulation by cardiolipin, Mff, and MiD49. Phosphorylation of S579 by Erk2 produces similar effects. When mixed with wildtype Drp1, both S579D and S600D phosphomimetic mutants reduce the actin-stimulated GTPase activity of Drp1-WT. Conversely, a Drp1 mutant (K38A) lacking GTPase activity stimulates Drp1-WT GTPase activity under both basal and actin-stimulated conditions. These results suggest that the effect of S579 phosphorylation is not to activate Drp1 directly. In addition, our results suggest that nearest neighbor interactions within the Drp1 oligomer affect catalytic activity.
Collapse
Affiliation(s)
- Ao Liu
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755
| | - Anna L. Hatch
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755
| | - Henry N. Higgs
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755
| |
Collapse
|
20
|
Tiwari A, Hashemiaghdam A, Laramie MA, Maschi D, Haddad T, Stunault MI, Bergom C, Javaheri A, Klyachko V, Ashrafi G. Sirtuin3 ensures the metabolic plasticity of neurotransmission during glucose deprivation. J Cell Biol 2024; 223:e202305048. [PMID: 37988067 PMCID: PMC10660140 DOI: 10.1083/jcb.202305048] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/18/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023] Open
Abstract
Neurotransmission is an energetically expensive process that underlies cognition. During intense electrical activity or dietary restrictions, the glucose level in the brain plummets, forcing neurons to utilize alternative fuels. However, the molecular mechanisms of neuronal metabolic plasticity remain poorly understood. Here, we demonstrate that glucose-deprived neurons activate the CREB and PGC1α transcriptional program, which induces expression of the mitochondrial deacetylase Sirtuin 3 (Sirt3) both in vitro and in vivo. We show that Sirt3 localizes to axonal mitochondria and stimulates mitochondrial oxidative capacity in hippocampal nerve terminals. Sirt3 plays an essential role in sustaining synaptic transmission in the absence of glucose by providing metabolic support for the retrieval of synaptic vesicles after release. These results demonstrate that the transcriptional induction of Sirt3 facilitates the metabolic plasticity of synaptic transmission.
Collapse
Affiliation(s)
- Anupama Tiwari
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Arsalan Hashemiaghdam
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marissa A. Laramie
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Dario Maschi
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tristaan Haddad
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marion I. Stunault
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Carmen Bergom
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Ali Javaheri
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- John Cochran VA Hospital, St. Louis, MO, USA
| | - Vitaly Klyachko
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ghazaleh Ashrafi
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
21
|
Scott KL, Halfmann CT, Hoefakker AD, Purkayastha P, Wang TC, Lele TP, Roux KJ. Nucleocytoplasmic transport rates are regulated by cellular processes that modulate GTP availability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.29.573651. [PMID: 38234722 PMCID: PMC10793428 DOI: 10.1101/2023.12.29.573651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Nucleocytoplasmic transport (NCT), the facilitated diffusion of cargo molecules between the nucleus and cytoplasm through nuclear pore complexes (NPCs), enables numerous fundamental eukaryotic cellular processes. Ran GTPase uses cellular energy in the direct form of GTP to create a gradient across the nuclear envelope (NE) that drives the majority of NCT. We report here that changes in GTP availability resulting from altered cellular physiology modulate the rate of NCT, as monitored using synthetic and natural cargo, and the dynamics of Ran itself. Cell migration, cell spreading and/or modulation of the cytoskeleton or its connection to the nucleus alter GTP availability and thus rates of NCT, regulating RNA export and protein synthesis. These findings support a model in which changes in cellular physiology that alter GTP availability can regulate the rate of NCT, impacting fundamental cellular processes that extensively utilize NCT.
Collapse
Affiliation(s)
- Kelsey L. Scott
- Enabling Technologies Group, Sanford Research, Sioux Falls SD
| | | | - Allison D. Hoefakker
- Enabling Technologies Group, Sanford Research, Sioux Falls SD
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD
| | - Purboja Purkayastha
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas
| | - Ting Ching Wang
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas
| | - Tanmay P. Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas
- Department of Translational Medical Sciences, Texas A&M University, Houston, Texas
| | - Kyle J. Roux
- Enabling Technologies Group, Sanford Research, Sioux Falls SD
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD
| |
Collapse
|
22
|
Lee YT, Savini M, Chen T, Yang J, Zhao Q, Ding L, Gao SM, Senturk M, Sowa JN, Wang JD, Wang MC. Mitochondrial GTP metabolism controls reproductive aging in C. elegans. Dev Cell 2023; 58:2718-2731.e7. [PMID: 37708895 PMCID: PMC10842941 DOI: 10.1016/j.devcel.2023.08.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/17/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023]
Abstract
Healthy mitochondria are critical for reproduction. During aging, both reproductive fitness and mitochondrial homeostasis decline. Mitochondrial metabolism and dynamics are key factors in supporting mitochondrial homeostasis. However, how they are coupled to control reproductive health remains unclear. We report that mitochondrial GTP (mtGTP) metabolism acts through mitochondrial dynamics factors to regulate reproductive aging. We discovered that germline-only inactivation of GTP- but not ATP-specific succinyl-CoA synthetase (SCS) promotes reproductive longevity in Caenorhabditis elegans. We further identified an age-associated increase in mitochondrial clustering surrounding oocyte nuclei, which is attenuated by GTP-specific SCS inactivation. Germline-only induction of mitochondrial fission factors sufficiently promotes mitochondrial dispersion and reproductive longevity. Moreover, we discovered that bacterial inputs affect mtGTP levels and dynamics factors to modulate reproductive aging. These results demonstrate the significance of mtGTP metabolism in regulating oocyte mitochondrial homeostasis and reproductive longevity and identify mitochondrial fission induction as an effective strategy to improve reproductive health.
Collapse
Affiliation(s)
- Yi-Tang Lee
- Integrative Program of Molecular and Biochemical Sciences, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Marzia Savini
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tao Chen
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20147, USA
| | - Jin Yang
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Qian Zhao
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20147, USA
| | - Lang Ding
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20147, USA; Graduate Program in Chemical, Physical & Structural Biology, Graduate School of Biomedical Science, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shihong Max Gao
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20147, USA
| | - Mumine Senturk
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jessica N Sowa
- Department of Biology, West Chester University, West Chester, PA 19383, USA
| | - Jue D Wang
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Meng C Wang
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20147, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
23
|
Khan I, Steeg PS. A perspective on the metastasis suppressor field. Cancer Metastasis Rev 2023; 42:1061-1063. [PMID: 37581870 DOI: 10.1007/s10555-023-10131-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
Metastasis is the leading cause of cancer patient mortality. Metastasis suppressors are genes that, upon reexpression in metastatic tumor cells to levels observed in their nonmetastatic counterparts, significantly reduce metastasis without affecting the growth of the primary tumor. Analysis of > 30 metastasis suppressors revealed complex mechanisms of action that include multiple signaling pathways, transcriptional patterns, posttranscriptional regulatory mechanisms, and potential contributions of genomic stability. Clinical testing of strategies to re-establish a validated metastasis suppressor pathway in tumors is best directed to the adjuvant setting, with the goal of inhibiting the outgrowth of occult micrometastases.
Collapse
Affiliation(s)
- Imran Khan
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Building 37, Room 1126, NCI, NIH, Bethesda, MD, 20892, USA
| | - Patricia S Steeg
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Building 37, Room 1126, NCI, NIH, Bethesda, MD, 20892, USA.
| |
Collapse
|
24
|
Affiliation(s)
- Danny R Welch
- Departments of Cancer Biology, The University of Kansas, Medical Center, Kansas City, KS, 66160, USA.
- Pathology & Laboratory Medicine, The University of Kansas, Medical Center, Kansas City, KS, 66160, USA.
- Internal Medicine - Hematology/Oncology, The University of Kansas, Medical Center, Kansas City, KS, 66160, USA.
- The University of Kansas Cancer Center, The University of Kansas, Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
25
|
Yang Z, Chan DC. Control of mitochondrial dynamics by a fusogenic lipid. Trends Cell Biol 2023; 33:1005-1006. [PMID: 37858503 PMCID: PMC10841390 DOI: 10.1016/j.tcb.2023.10.006] [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: 09/26/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/21/2023]
Abstract
Mitochondrial fusion enables cooperation between the mitochondrial population and is critical for mitochondrial function. Phosphatidic acid (PA) on the mitochondrial surface has a key role in mitochondrial fusion. A recent study by Su et al. shows that the nucleoside diphosphate (NDP) kinase NME3 recognizes PA and mediates its effects on mitochondrial dynamics.
Collapse
Affiliation(s)
- Zheng Yang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
26
|
Prunier C, Chavrier P, Boissan M. Mechanisms of action of NME metastasis suppressors - a family affair. Cancer Metastasis Rev 2023; 42:1155-1167. [PMID: 37353690 PMCID: PMC10713741 DOI: 10.1007/s10555-023-10118-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/09/2023] [Indexed: 06/25/2023]
Abstract
Metastatic progression is regulated by metastasis promoter and suppressor genes. NME1, the prototypic and first described metastasis suppressor gene, encodes a nucleoside diphosphate kinase (NDPK) involved in nucleotide metabolism; two related family members, NME2 and NME4, are also reported as metastasis suppressors. These proteins physically interact with members of the GTPase dynamin family, which have key functions in membrane fission and fusion reactions necessary for endocytosis and mitochondrial dynamics. Evidence supports a model in which NDPKs provide GTP to dynamins to maintain a high local GTP concentration for optimal dynamin function. NME1 and NME2 are cytosolic enzymes that provide GTP to dynamins at the plasma membrane, which drive endocytosis, suggesting that these NMEs are necessary to attenuate signaling by receptors on the cell surface. Disruption of NDPK activity in NME-deficient tumors may thus drive metastasis by prolonging signaling. NME4 is a mitochondrial enzyme that interacts with the dynamin OPA1 at the mitochondria inner membrane to drive inner membrane fusion and maintain a fused mitochondrial network. This function is consistent with the current view that mitochondrial fusion inhibits the metastatic potential of tumor cells whereas mitochondrial fission promotes metastasis progression. The roles of NME family members in dynamin-mediated endocytosis and mitochondrial dynamics and the intimate link between these processes and metastasis provide a new framework to understand the metastasis suppressor functions of NME proteins.
Collapse
Affiliation(s)
- Céline Prunier
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, CRSA, Paris, France
| | - Philippe Chavrier
- Actin and Membrane Dynamics Laboratory, Institut Curie - Research Center, CNRS UMR144, PSL Research University, Paris, France
| | - Mathieu Boissan
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, CRSA, Paris, France.
- Laboratoire de Biochimie Endocrinienne Et Oncologique, Oncobiologie Cellulaire Et Moléculaire, APHP, Hôpitaux Universitaires Pitié-Salpêtrière-Charles Foix, Paris, France.
| |
Collapse
|
27
|
Su YA, Chiu HY, Chang YC, Sung CJ, Chen CW, Tei R, Huang XR, Hsu SC, Lin SS, Wang HC, Lin YC, Hsu JC, Bauer H, Feng Y, Baskin JM, Chang ZF, Liu YW. NME3 binds to phosphatidic acid and mediates PLD6-induced mitochondrial tethering. J Cell Biol 2023; 222:e202301091. [PMID: 37584589 PMCID: PMC10432850 DOI: 10.1083/jcb.202301091] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/10/2023] [Accepted: 07/26/2023] [Indexed: 08/17/2023] Open
Abstract
Mitochondria are dynamic organelles regulated by fission and fusion processes. The fusion of membranes requires elaborative coordination of proteins and lipids and is particularly crucial for the function and quality control of mitochondria. Phosphatidic acid (PA) on the mitochondrial outer membrane generated by PLD6 facilitates the fusion of mitochondria. However, how PA promotes mitochondrial fusion remains unclear. Here, we show that a mitochondrial outer membrane protein, NME3, is required for PLD6-induced mitochondrial tethering or clustering. NME3 is enriched at the contact interface of two closely positioned mitochondria depending on PLD6, and NME3 binds directly to PA-exposed lipid packing defects via its N-terminal amphipathic helix. The PA binding function and hexamerization confer NME3 mitochondrial tethering activity. Importantly, nutrient starvation enhances the enrichment efficiency of NME3 at the mitochondrial contact interface, and the tethering ability of NME3 contributes to fusion efficiency. Together, our findings demonstrate NME3 as a tethering protein promoting selective fusion between PLD6-remodeled mitochondria for quality control.
Collapse
Affiliation(s)
- You-An Su
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Yi Chiu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Chen Chang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chieh-Ju Sung
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chih-Wei Chen
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Reika Tei
- Department of Chemistry and Chemical Biology and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Xuang-Rong Huang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shao-Chun Hsu
- Imaging Core, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shan-Shan Lin
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsien-Chu Wang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Chun Lin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Jui-Cheng Hsu
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Hermann Bauer
- Department of Developmental Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Yuxi Feng
- Department of Experimental Pharmacology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Jeremy M. Baskin
- Department of Chemistry and Chemical Biology and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Zee-Fen Chang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Wen Liu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
28
|
Ikeda A, Iijima M, Sesaki H. A nucleotide diphosphate kinase mediates tethering between mitochondria prior to fusion. J Cell Biol 2023; 222:e202309037. [PMID: 37707790 PMCID: PMC10501386 DOI: 10.1083/jcb.202309037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023] Open
Abstract
Mitochondrial fusion plays an important role in both their structure and function. In this issue, Su et al. (2023. J. Cell Biol.https://doi.org/10.1083/jcb.202301091) report that a nucleoside diphosphate kinase, NME3, facilitates mitochondrial tethering prior to fusion through its direct membrane-binding and hexamerization but not its kinase activity.
Collapse
Affiliation(s)
- Arisa Ikeda
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Miho Iijima
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Liu A, Hatch AL, Higgs HN. Effects of phosphorylation on Drp1 activation by its receptors, actin, and cardiolipin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.20.554022. [PMID: 37645886 PMCID: PMC10462108 DOI: 10.1101/2023.08.20.554022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Drp1 is a dynamin family GTPase that is required for mitochondrial and peroxisomal division, in which it oligomerizes into a ring and constricts the underlying membrane in a GTP hydrolysis-dependent manner. Oligomerization increases Drp1 GTPase activity through interactions between neighboring GTPase domains. In cells, Drp1 is regulated by several factors including Drp1 receptors, actin filaments, cardiolipin, and phosphorylation at two sites: S579 and S600. Phosphorylation of S579 is widely regarded as activating, while S600 phosphorylation is commonly considered inhibiting. However, the direct effects of phosphorylation on Drp1 GTPase activity have not been investigated in detail. In this study, we compare the effects of S579 and S600 phosphorylation on purified Drp1, using phospho-mimetic mutants and in vitro phosphorylation. The oligomerization state of both phospho-mimetic mutants is shifted toward smaller oligomers. Both phospho-mimetic mutations maintain basal GTPase activity, but eliminate GTPase stimulation by actin and decrease GTPase stimulation by cardiolipin, Mff, and MiD49. Phosphorylation of S579 by Erk2 produces similar effects. When mixed with wild-type Drp1, both S579D and S600D phospho-mimetic mutants reduce the actin-stimulated GTPase activity of Drp1-WT. Conversely, a Drp1 mutant that lacks GTPase activity, the K38A mutant, stimulates Drp1-WT GTPase activity under both basal and actin-stimulated conditions, similar to previous results for dynamin-1. These results suggest that the effect of S579 phosphorylation is not to activate Drp1 directly, and likely requires additional factors for stimulation of mitochondrial fission in cells. In addition, our results suggest that nearest neighbor interactions within the Drp1 oligomer affect catalytic activity.
Collapse
Affiliation(s)
| | | | - Henry N. Higgs
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755
| |
Collapse
|
30
|
Chapa-Dubocq XR, Rodríguez-Graciani KM, García-Báez J, Vadovsky A, Bazil JN, Javadov S. The Role of Swelling in the Regulation of OPA1-Mediated Mitochondrial Function in the Heart In Vitro. Cells 2023; 12:2017. [PMID: 37626827 PMCID: PMC10453793 DOI: 10.3390/cells12162017] [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: 06/18/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Optic atrophy-1 (OPA1) plays a crucial role in the regulation of mitochondria fusion and participates in maintaining the structural integrity of mitochondrial cristae. Here we elucidate the role of OPA1 cleavage induced by calcium swelling in the presence of Myls22 (an OPA1 GTPase activity inhibitor) and TPEN (an OMA1 inhibitor). The rate of ADP-stimulated respiration was found diminished by both inhibitors, and they did not prevent Ca2+-induced mitochondrial respiratory dysfunction, membrane depolarization, or swelling. L-OPA1 cleavage was stimulated at state 3 respiration; therefore, our data suggest that L-OPA1 cleavage produces S-OPA1 to maintain mitochondrial bioenergetics in response to stress.
Collapse
Affiliation(s)
- Xavier R. Chapa-Dubocq
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR 00936-5067, USA; (X.R.C.-D.); (K.M.R.-G.); (J.G.-B.)
| | - Keishla M. Rodríguez-Graciani
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR 00936-5067, USA; (X.R.C.-D.); (K.M.R.-G.); (J.G.-B.)
| | - Jorge García-Báez
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR 00936-5067, USA; (X.R.C.-D.); (K.M.R.-G.); (J.G.-B.)
| | - Alyssa Vadovsky
- Department of Physiology, Michigan State University, East Lansing, MI 48824-1046, USA; (A.V.); (J.N.B.)
| | - Jason N. Bazil
- Department of Physiology, Michigan State University, East Lansing, MI 48824-1046, USA; (A.V.); (J.N.B.)
| | - Sabzali Javadov
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR 00936-5067, USA; (X.R.C.-D.); (K.M.R.-G.); (J.G.-B.)
| |
Collapse
|
31
|
Bouin AP, Kyumurkov A, Planus E, Albiges-Rizo C. [Cellular tension and integrin trafficking]. Med Sci (Paris) 2023; 39:597-599. [PMID: 37695144 DOI: 10.1051/medsci/2023089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023] Open
Affiliation(s)
- Anne-Pascale Bouin
- Université Grenoble Alpes, Inserm 1209, CNRS UMR5309, Institut pour l'avancée des biosciences, Grenoble, France
| | - Alexander Kyumurkov
- Université Grenoble Alpes, Inserm 1209, CNRS UMR5309, Institut pour l'avancée des biosciences, Grenoble, France
| | - Emmanuelle Planus
- Université Grenoble Alpes, Inserm 1209, CNRS UMR5309, Institut pour l'avancée des biosciences, Grenoble, France
| | - Corinne Albiges-Rizo
- Université Grenoble Alpes, Inserm 1209, CNRS UMR5309, Institut pour l'avancée des biosciences, Grenoble, France
| |
Collapse
|
32
|
Pagano A, Kunz L, Dittmann A, Araújo SDS, Macovei A, Shridhar Gaonkar S, Sincinelli F, Wazeer H, Balestrazzi A. Changes in Medicago truncatula seed proteome along the rehydration-dehydration cycle highlight new players in the genotoxic stress response. FRONTIERS IN PLANT SCIENCE 2023; 14:1188546. [PMID: 37409306 PMCID: PMC10319343 DOI: 10.3389/fpls.2023.1188546] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/24/2023] [Indexed: 07/07/2023]
Abstract
Introduction Several molecular aspects underlying the seed response to priming and the resulting vigor profile are still poorly understood. Mechanisms involved in genome maintenance deserve attention since the balance between stimulation of germination and DNA damage accumulation versus active repair is a key determinant for designing successful seed priming protocols. Methods Changes in the Medicago truncatula seed proteome were investigated in this study, using discovery mass spectrometry and label-free quantification, along the rehydration-dehydration cycle of a standard vigorization treatment (hydropriming plus dry-back), and during post-priming imbibition. Resuts and discussion From 2056 to 2190 proteins were detected in each pairwise comparison, among which six were differentially accumulated and 36 were detected only in one condition. The following proteins were selected for further investigation: MtDRP2B (DYNAMIN-RELATED PROTEIN), MtTRXm4 (THIOREDOXIN m4), and MtASPG1 (ASPARTIC PROTEASE IN GUARD CELL 1) showing changes in seeds under dehydration stress; MtITPA (INOSINE TRIPHOSPHATE PYROPHOSPHORYLASE), MtABA2 (ABSCISIC ACID DEFICIENT 2), MtRS2Z32 (SERINE/ARGININE-RICH SPLICING FACTOR RS2Z32), and MtAQR (RNA HELICASE AQUARIUS) that were differentially regulated during post-priming imbibition. Changes in the corresponding transcript levels were assessed by qRT-PCR. In animal cells, ITPA hydrolyses 2'-deoxyinosine triphosphate and other inosine nucleotides, preventing genotoxic damage. A proof of concept was performed by imbibing primed and control M. truncatula seeds in presence/absence of 20 mM 2'-deoxyinosine (dI). Results from comet assay highlighted the ability of primed seeds to cope with dI-induced genotoxic damage. The seed repair response was assessed by monitoring the expression profiles of MtAAG (ALKYL-ADENINE DNA GLYCOSILASE) and MtEndoV (ENDONUCLEASE V) genes that participate in the repair of the mismatched I:T pair in BER (base excision repair) and AER (alternative excision repair) pathways, respectively.
Collapse
Affiliation(s)
- Andrea Pagano
- Department of Biology and Biotechnology ‘L. Spallanzani’, University of Pavia, Pavia, Italy
| | - Laura Kunz
- Functional Genomics Center Zurich (FGCZ), University of Zurich/Eidgenossische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Antje Dittmann
- Functional Genomics Center Zurich (FGCZ), University of Zurich/Eidgenossische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Susana De Sousa Araújo
- Association BLC3 - Campus of Technology and Innovation, Centre BIO R&D Unit | North Delegation, Macedo de Cavaleiros, Portugal
| | - Anca Macovei
- Department of Biology and Biotechnology ‘L. Spallanzani’, University of Pavia, Pavia, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
| | | | - Federico Sincinelli
- Department of Biology and Biotechnology ‘L. Spallanzani’, University of Pavia, Pavia, Italy
| | - Hisham Wazeer
- Department of Biology and Biotechnology ‘L. Spallanzani’, University of Pavia, Pavia, Italy
| | - Alma Balestrazzi
- Department of Biology and Biotechnology ‘L. Spallanzani’, University of Pavia, Pavia, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
| |
Collapse
|
33
|
Banushi B, Joseph SR, Lum B, Lee JJ, Simpson F. Endocytosis in cancer and cancer therapy. Nat Rev Cancer 2023:10.1038/s41568-023-00574-6. [PMID: 37217781 DOI: 10.1038/s41568-023-00574-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/24/2023]
Abstract
Endocytosis is a complex process whereby cell surface proteins, lipids and fluid from the extracellular environment are packaged, sorted and internalized into cells. Endocytosis is also a mechanism of drug internalization into cells. There are multiple routes of endocytosis that determine the fate of molecules, from degradation in the lysosomes to recycling back to the plasma membrane. The overall rates of endocytosis and temporal regulation of molecules transiting through endocytic pathways are also intricately linked with signalling outcomes. This process relies on an array of factors, such as intrinsic amino acid motifs and post-translational modifications. Endocytosis is frequently disrupted in cancer. These disruptions lead to inappropriate retention of receptor tyrosine kinases on the tumour cell membrane, changes in the recycling of oncogenic molecules, defective signalling feedback loops and loss of cell polarity. In the past decade, endocytosis has emerged as a pivotal regulator of nutrient scavenging, response to and regulation of immune surveillance and tumour immune evasion, tumour metastasis and therapeutic drug delivery. This Review summarizes and integrates these advances into the understanding of endocytosis in cancer. The potential to regulate these pathways in the clinic to improve cancer therapy is also discussed.
Collapse
Affiliation(s)
- Blerida Banushi
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Shannon R Joseph
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Benedict Lum
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Jason J Lee
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Fiona Simpson
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia.
| |
Collapse
|
34
|
Qi L, Li Y, Dong Y, Ma S, Li G. Integrated metabolomics and transcriptomics reveal glyphosate based-herbicide induced reproductive toxicity through disturbing energy and nucleotide metabolism in mice testes. ENVIRONMENTAL TOXICOLOGY 2023. [PMID: 37087751 DOI: 10.1002/tox.23808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 05/03/2023]
Abstract
Glyphosate is a widely used herbicide that has deleterious effects on animal reproduction. However, details regarding the systematic mechanisms of glyphosate-induced reproductive toxicity are limited. This study aimed to investigate the toxic effects of glyphosate-based herbicide (GBH) on reproduction in mice exposed to 0 (control group), 50 (low-dose group), 250 (middle-dose group), and 500 (high-dose group) mg/kg/day GBH for 30 days. Toxicological parameters, metabolomics, and transcriptomics were performed to reveal GBH-induced reproductive toxicity. Our findings demonstrated that GBH exposure damaged mitochondrial pyknosis and the nuclear membrane of spermatogonia. GBH triggered a significant increase in sperm malformations in the high-dose group. Omics data showed that GBH impaired the Krebs cycle and respiratory chain, blocked pyruvate metabolism and glycolysis/gluconeogenesis, and influenced the pentose phosphate pathway and nucleotide synthesis and metabolism. Overall, the multi-omics results revealed systematic and comprehensive evidence of the adverse effects of GBH exposure, providing new insights into the reproductive toxicity of organophosphorus pesticides.
Collapse
Affiliation(s)
- Lei Qi
- Department of Nutrition and Food Hygiene, Public Health College, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Yupeng Li
- Physical Examination Center, the Third Affiliated Hospital, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Yanmei Dong
- Department of Nutrition and Food Hygiene, Public Health College, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Shuli Ma
- Public Health Experimental Center, Public Health College, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Gang Li
- Department of Preventive Medicine, Public Health College, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| |
Collapse
|
35
|
Lee YT, Savini M, Chen T, Yang J, Zhao Q, Ding L, Gao SM, Senturk M, Sowa J, Wang JD, Wang MC. Mitochondrial GTP Metabolism Regulates Reproductive Aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.02.535296. [PMID: 37066227 PMCID: PMC10103970 DOI: 10.1101/2023.04.02.535296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Healthy mitochondria are critical for reproduction. During aging, both reproductive fitness and mitochondrial homeostasis decline. Mitochondrial metabolism and dynamics are key factors in supporting mitochondrial homeostasis. However, how they are coupled to control reproductive health remains unclear. We report that mitochondrial GTP metabolism acts through mitochondrial dynamics factors to regulate reproductive aging. We discovered that germline-only inactivation of GTP- but not ATP-specific succinyl-CoA synthetase (SCS), promotes reproductive longevity in Caenorhabditis elegans. We further revealed an age-associated increase in mitochondrial clustering surrounding oocyte nuclei, which is attenuated by the GTP-specific SCS inactivation. Germline-only induction of mitochondrial fission factors sufficiently promotes mitochondrial dispersion and reproductive longevity. Moreover, we discovered that bacterial inputs affect mitochondrial GTP and dynamics factors to modulate reproductive aging. These results demonstrate the significance of mitochondrial GTP metabolism in regulating oocyte mitochondrial homeostasis and reproductive longevity and reveal mitochondrial fission induction as an effective strategy to improve reproductive health.
Collapse
|
36
|
Martínez RAS, Pinky PD, Harlan BA, Brewer GJ. GTP energy dependence of endocytosis and autophagy in the aging brain and Alzheimer's disease. GeroScience 2023; 45:757-780. [PMID: 36622562 PMCID: PMC9886713 DOI: 10.1007/s11357-022-00717-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/15/2022] [Indexed: 01/10/2023] Open
Abstract
Increased interest in the aging and Alzheimer's disease (AD)-related impairments in autophagy in the brain raise important questions about regulation and treatment. Since many steps in endocytosis and autophagy depend on GTPases, new measures of cellular GTP levels are needed to evaluate energy regulation in aging and AD. The recent development of ratiometric GTP sensors (GEVALS) and findings that GTP levels are not homogenous inside cells raise new issues of regulation of GTPases by the local availability of GTP. In this review, we highlight the metabolism of GTP in relation to the Rab GTPases involved in formation of early endosomes, late endosomes, and lysosomal transport to execute the autophagic degradation of damaged cargo. Specific GTPases control macroautophagy (mitophagy), microautophagy, and chaperone-mediated autophagy (CMA). By inference, local GTP levels would control autophagy, if not in excess. Additional levels of control are imposed by the redox state of the cell, including thioredoxin involvement. Throughout this review, we emphasize the age-related changes that could contribute to deficits in GTP and AD. We conclude with prospects for boosting GTP levels and reversing age-related oxidative redox shift to restore autophagy. Therefore, GTP levels could regulate the numerous GTPases involved in endocytosis, autophagy, and vesicular trafficking. In aging, metabolic adaptation to a sedentary lifestyle could impair mitochondrial function generating less GTP and redox energy for healthy management of amyloid and tau proteostasis, synaptic function, and inflammation.
Collapse
Affiliation(s)
| | - Priyanka D. Pinky
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697 USA
| | - Benjamin A. Harlan
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697 USA
| | - Gregory J. Brewer
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697 USA
- Center for Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA 92697 USA
- MIND Institute, University of California Irvine, Irvine, CA 92697 USA
| |
Collapse
|
37
|
Tiwari A, Hashemiaghdam A, Laramie MA, Maschi D, Haddad T, Stunault MI, Bergom C, Javaheri A, Klyachko V, Ashrafi G. Sirtuin3 ensures the metabolic plasticity of neurotransmission during glucose deprivation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.08.531724. [PMID: 36945567 PMCID: PMC10028948 DOI: 10.1101/2023.03.08.531724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Neurotransmission is an energetically expensive process that underlies cognition. During intense electrical activity or dietary restrictions, glucose levels in the brain plummet, forcing neurons to utilize alternative fuels. However, the molecular mechanisms of neuronal metabolic plasticity remain poorly understood. Here, we demonstrate that glucose-deprived neurons activate the CREB and PGC1α transcriptional program that induces the expression of the mitochondrial deacetylase Sirtuin 3 (Sirt3) both in vitro and in vivo . We show that Sirt3 localizes to axonal mitochondria and stimulates mitochondrial oxidative capacity in hippocampal nerve terminals. Sirt3 plays an essential role in sustaining synaptic transmission in the absence of glucose by powering the retrieval of synaptic vesicles after release. These results demonstrate that the transcriptional induction of Sirt3 ensures the metabolic plasticity of synaptic transmission. Highlights Glucose deprivation drives transcriptional reprogramming of neuronal metabolism via CREB and PGC1α. Glucose or food deprivation trigger the neuronal expression of mitochondrial deacetylase sirtuin 3 (Sirt3) both in vitro and in vivo . Sirt3 stimulates oxidative ATP synthesis in nerve terminals.Sirt3 sustains the synaptic vesicle cycle in the absence of glucose.
Collapse
|
38
|
Cagnin S, Knedlik T, Vianello C, Magalhães Rebelo AP, De Mario A, Giacomello M. Comparison among Neuroblastoma Stages Suggests the Involvement of Mitochondria in Tumor Progression. Biomedicines 2023; 11:biomedicines11020596. [PMID: 36831133 PMCID: PMC9953471 DOI: 10.3390/biomedicines11020596] [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: 01/27/2023] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Neuroblastoma (NB) is the most common extracranial tumor of early childhood and accounts for 15% of all pediatric cancer mortalities. However, the precise pathways and genes underlying its progression are unknown. Therefore, we performed a differential gene expression analysis of neuroblastoma stage 1 and stage 4 + 4S to discover biological processes associated with NB progression. From this preliminary analysis, we found that NB samples (stage 4 + 4S) are characterized by altered expression of some proteins involved in mitochondria function and mitochondria-ER contact sites (MERCS). Although further analyses remain necessary, this review may provide new hints to better understand NB molecular etiopathogenesis, by suggesting that MERCS alterations could be involved in the progression of NB.
Collapse
Affiliation(s)
- Stefano Cagnin
- Department of Biology, University of Padua, Via U. Bassi 58/B, 35121 Padua, Italy
- CIR-Myo Myology Center, University of Padova, 35121 Padua, Italy
| | - Tomas Knedlik
- Department of Biology, University of Padua, Via U. Bassi 58/B, 35121 Padua, Italy
| | - Caterina Vianello
- Department of Biology, University of Padua, Via U. Bassi 58/B, 35121 Padua, Italy
| | | | - Agnese De Mario
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35121 Padua, Italy
- Correspondence: (A.D.M.); (M.G.)
| | - Marta Giacomello
- Department of Biology, University of Padua, Via U. Bassi 58/B, 35121 Padua, Italy
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35121 Padua, Italy
- Correspondence: (A.D.M.); (M.G.)
| |
Collapse
|
39
|
Iyer P, Zhang B, Liu T, Jin M, Hart K, Zhang J, Song J, Chan WC, Siddiqi T, Rosen ST, Danilov A, Wang L. MGA deletion leads to Richter's transformation via modulation of mitochondrial OXPHOS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.07.527502. [PMID: 36798339 PMCID: PMC9934534 DOI: 10.1101/2023.02.07.527502] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Richter's transformation (RT) is a progression of chronic lymphocytic leukemia (CLL) to aggressive lymphoma. MGA ( Max gene associated ), a functional MYC suppressor, is mutated at 3% in CLL and 36% in RT. However, genetic models and molecular mechanisms of MGA deletion driving CLL to RT remain elusive. We established a novel RT mouse model by knockout of Mga in the Sf3b1 / Mdr CLL model via CRISPR-Cas9 to determine the role of Mga in RT. Murine RT cells exhibit mitochondrial aberrations with elevated oxidative phosphorylation (OXPHOS). We identified Nme1 (Nucleoside diphosphate kinase) as a Mga target through RNA sequencing and functional characterization, which drives RT by modulating OXPHOS. As NME1 is also a known MYC target without targetable compounds, we found that concurrent inhibition of MYC and ETC complex II significantly prolongs the survival of RT mice in vivo . Our results suggest that Mga-Nme1 axis drives murine CLL-to-RT transition via modulating OXPHOS, highlighting a novel therapeutic avenue for RT. Statement of Significance We established a murine RT model through knockout of Mga in an existing CLL model based on co-expression of Sf3b1 -K700E and del ( 13q ). We determined that the MGA/NME1 regulatory axis is essential to the CLL-to-RT transition via modulation of mitochondrial OXPHOS, highlighting this pathway as a novel target for RT treatment.
Collapse
|
40
|
Kyumurkov A, Bouin AP, Boissan M, Manet S, Baschieri F, Proponnet-Guerault M, Balland M, Destaing O, Régent-Kloeckner M, Calmel C, Nicolas A, Waharte F, Chavrier P, Montagnac G, Planus E, Albiges-Rizo C. Force tuning through regulation of clathrin-dependent integrin endocytosis. J Cell Biol 2022; 222:213549. [PMID: 36250940 PMCID: PMC9579986 DOI: 10.1083/jcb.202004025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/22/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022] Open
Abstract
Integrin endocytosis is essential for many fundamental cellular processes. Whether and how the internalization impacts cellular mechanics remains elusive. Whereas previous studies reported the contribution of the integrin activator, talin, in force development, the involvement of inhibitors is less documented. We identified ICAP-1 as an integrin inhibitor involved in mechanotransduction by co-working with NME2 to control clathrin-mediated endocytosis of integrins at the edge of focal adhesions (FA). Loss of ICAP-1 enables β3-integrin-mediated force generation independently of β1 integrin. β3-integrin-mediated forces were associated with a decrease in β3 integrin dynamics stemming from their reduced diffusion within adhesion sites and slow turnover of FA. The decrease in β3 integrin dynamics correlated with a defect in integrin endocytosis. ICAP-1 acts as an adaptor for clathrin-dependent endocytosis of integrins. ICAP-1 controls integrin endocytosis by interacting with NME2, a key regulator of dynamin-dependent clathrin-coated pits fission. Control of clathrin-mediated integrin endocytosis by an inhibitor is an unprecedented mechanism to tune forces at FA.
Collapse
Affiliation(s)
- Alexander Kyumurkov
- University Grenoble Alpes, INSERM 1209, CNRS UMR5309, Institute for Advanced Biosciences, Grenoble, France
| | - Anne-Pascale Bouin
- University Grenoble Alpes, INSERM 1209, CNRS UMR5309, Institute for Advanced Biosciences, Grenoble, France
| | - Mathieu Boissan
- University Sorbonne, INSERM UMR_S 938, Saint-Antoine Research Center, CRSA, Paris, France,Laboratory of Biochemistry and Hormonology, Tenon Hospital, AP-HP, Paris, France
| | - Sandra Manet
- University Grenoble Alpes, INSERM 1209, CNRS UMR5309, Institute for Advanced Biosciences, Grenoble, France
| | - Francesco Baschieri
- Inserm U1279, Gustave Roussy Institute, Université Paris-Saclay, Villejuif, France
| | | | - Martial Balland
- Laboratoire Interdisciplinaire de Physique, UMR CNRS 5588, University Grenoble Alpes, Grenoble, France
| | - Olivier Destaing
- University Grenoble Alpes, INSERM 1209, CNRS UMR5309, Institute for Advanced Biosciences, Grenoble, France
| | - Myriam Régent-Kloeckner
- University Grenoble Alpes, INSERM 1209, CNRS UMR5309, Institute for Advanced Biosciences, Grenoble, France
| | - Claire Calmel
- University Sorbonne, INSERM UMR_S 938, Saint-Antoine Research Center, CRSA, Paris, France,Laboratory of Biochemistry and Hormonology, Tenon Hospital, AP-HP, Paris, France
| | - Alice Nicolas
- University Grenoble Alpes, CNRS, CEA/LETIMinatec, Grenoble Institute of Technology, Microelectronics Technology Laboratory, Grenoble, France
| | - François Waharte
- University Sorbonne, INSERM UMR_S 938, Saint-Antoine Research Center, CRSA, Paris, France,Laboratory of Biochemistry and Hormonology, Tenon Hospital, AP-HP, Paris, France
| | - Philippe Chavrier
- Institut Curie, UMR144, Université de Recherche Paris Sciences et Lettres, Centre Universitaire, Paris, France
| | - Guillaume Montagnac
- Inserm U1279, Gustave Roussy Institute, Université Paris-Saclay, Villejuif, France
| | - Emmanuelle Planus
- University Grenoble Alpes, INSERM 1209, CNRS UMR5309, Institute for Advanced Biosciences, Grenoble, France,Correspondence to Emmanuelle Planus: mailto:
| | - Corinne Albiges-Rizo
- University Grenoble Alpes, INSERM 1209, CNRS UMR5309, Institute for Advanced Biosciences, Grenoble, France,Corinne Albiges-Rizo:
| |
Collapse
|
41
|
Khan I, Gril B, Hoshino A, Yang HH, Lee MP, Difilippantonio S, Lyden DC, Steeg PS. Metastasis suppressor NME1 in exosomes or liposomes conveys motility and migration inhibition in breast cancer model systems. Clin Exp Metastasis 2022; 39:815-831. [PMID: 35939247 PMCID: PMC10642714 DOI: 10.1007/s10585-022-10182-7] [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: 04/15/2022] [Accepted: 07/27/2022] [Indexed: 11/03/2022]
Abstract
Tumor-derived exosomes have documented roles in accelerating the initiation and outgrowth of metastases, as well as in therapy resistance. Little information supports the converse, that exosomes or similar vesicles can suppress metastasis. We investigated the NME1 (Nm23-H1) metastasis suppressor as a candidate for metastasis suppression by extracellular vesicles. Exosomes derived from two cancer cell lines (MDA-MB-231T and MDA-MB-435), when transfected with the NME1 (Nm23-H1) metastasis suppressor, secreted exosomes with NME1 as the predominant constituent. These exosomes entered recipient tumor cells, altered their endocytic patterns in agreement with NME1 function, and suppressed in vitro tumor cell motility and migration compared to exosomes from control transfectants. Proteomic analysis of exosomes revealed multiple differentially expressed proteins that could exert biological functions. Therefore, we also prepared and investigated liposomes, empty or containing partially purified rNME1. rNME1 containing liposomes recapitulated the effects of exosomes from NME1 transfectants in vitro. In an experimental lung metastasis assay the median lung metastases per histologic section was 158 using control liposomes and 15 in the rNME1 liposome group, 90.5% lower than the control liposome group (P = 0.016). The data expand the exosome/liposome field to include metastasis suppressive functions and describe a new translational approach to prevent metastasis.
Collapse
Affiliation(s)
- Imran Khan
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 37, Convent Drive, Room 1126, Bethesda, MD, 20892, USA.
| | - Brunilde Gril
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 37, Convent Drive, Room 1126, Bethesda, MD, 20892, USA
| | - Ayuko Hoshino
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Howard H Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, USA
| | - Maxwell P Lee
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, USA
| | - Simone Difilippantonio
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - David C Lyden
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Patricia S Steeg
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 37, Convent Drive, Room 1126, Bethesda, MD, 20892, USA
| |
Collapse
|
42
|
Zhao J, Zhuang M, Liu J, Zhang M, Zeng C, Jiang B, Wu J, Song X. pHisPred: a tool for the identification of histidine phosphorylation sites by integrating amino acid patterns and properties. BMC Bioinformatics 2022; 23:399. [PMID: 36171552 PMCID: PMC9520798 DOI: 10.1186/s12859-022-04938-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Background Protein histidine phosphorylation (pHis) plays critical roles in prokaryotic signal transduction pathways and various eukaryotic cellular processes. It is estimated to account for 6–10% of the phosphoproteome, however only hundreds of pHis sites have been discovered to date. Due to the inherent disadvantages of experimental methods, it is an urgent task for developing efficient computational approaches to identify pHis sites. Results Here, we present a novel tool, pHisPred, for accurately identifying pHis sites from protein sequences. We manually collected the largest number of experimental validated pHis sites to build benchmark datasets. Using randomized tenfold CV, the weighted SVM-RBF model shows the best performance than other four commonly used classification models (LR, KNN, RF, and MLP). From ten thousands of features, 140 and 150 most informative features were individually selected out for eukaryotic and prokaryotic models. The average AUC and F1-score values of pHisPred were (0.81, 0.40) and (0.78, 0.46) for tenfold CV on the eukaryotic and prokaryotic training datasets, respectively. In addition, pHisPred significantly outperforms other tools on testing datasets, in particular on the eukaryotic one. Conclusion We implemented a python program of pHisPred, which is freely available for non-commercial use at https://github.com/xiaofengsong/pHisPred. Moreover, users can use it to train new models with their own data. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-022-04938-x.
Collapse
Affiliation(s)
- Jian Zhao
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Minhui Zhuang
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Jingjing Liu
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Meng Zhang
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Cong Zeng
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Bin Jiang
- College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 211106, China
| | - Jing Wu
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211166, China.
| | - Xiaofeng Song
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China.
| |
Collapse
|
43
|
Wolff DW, Bianchi-Smiraglia A, Nikiforov MA. Compartmentalization and regulation of GTP in control of cellular phenotypes. Trends Mol Med 2022; 28:758-769. [PMID: 35718686 PMCID: PMC9420775 DOI: 10.1016/j.molmed.2022.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 10/18/2022]
Abstract
Genetic or pharmacological inhibition of enzymes involved in GTP biosynthesis has substantial biological effects, underlining the need to better understand the function of GTP levels in regulation of cellular processes and the significance of targeting GTP biosynthesis enzymes for therapeutic intervention. Our current understanding of spatiotemporal regulation of GTP metabolism and its role in physiological and pathological cellular processes is far from complete. Novel methodologies such as genetically encoded sensors of free GTP offered insights into intracellular distribution and function of GTP molecules. In the current Review, we provide analysis of recent discoveries in the field of GTP metabolism and evaluate the key enzymes as molecular targets.
Collapse
Affiliation(s)
- David W Wolff
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA.
| | - Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Mikhail A Nikiforov
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA; Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
44
|
Abstract
Adenosine is an evolutionary ancient metabolic regulator linking energy state to physiologic processes, including immunomodulation and cell proliferation. Tumors create an adenosine-rich immunosuppressive microenvironment through the increased release of ATP from dying and stressed cells and its ectoenzymatic conversion into adenosine. Therefore, the adenosine pathway becomes an important therapeutic target to improve the effectiveness of immune therapies. Prior research has focused largely on the two major ectonucleotidases, ectonucleoside triphosphate diphosphohydrolase 1/cluster of differentiation (CD)39 and ecto-5'-nucleotidase/CD73, which catalyze the breakdown of extracellular ATP into adenosine, and on the subsequent activation of different subtypes of adenosine receptors with mixed findings of antitumor and protumor effects. New findings, needed for more effective therapeutic approaches, require consideration of redundant pathways controlling intratumoral adenosine levels, including the alternative NAD-inactivating pathway through the CD38-ectonucleotide pyrophosphatase phosphodiesterase (ENPP)1-CD73 axis, the counteracting ATP-regenerating ectoenzymatic pathway, and cellular adenosine uptake and its phosphorylation by adenosine kinase. This review provides a holistic view of extracellular and intracellular adenosine metabolism as an integrated complex network and summarizes recent data on the underlying mechanisms through which adenosine and its precursors ATP and ADP control cancer immunosurveillance, tumor angiogenesis, lymphangiogenesis, cancer-associated thrombosis, blood flow, and tumor perfusion. Special attention is given to differences and commonalities in the purinome of different cancers, heterogeneity of the tumor microenvironment, subcellular compartmentalization of the adenosine system, and novel roles of purine-converting enzymes as targets for cancer therapy. SIGNIFICANCE STATEMENT: The discovery of the role of adenosine as immune checkpoint regulator in cancer has led to the development of novel therapeutic strategies targeting extracellular adenosine metabolism and signaling in multiple clinical trials and preclinical models. Here we identify major gaps in knowledge that need to be filled to improve the therapeutic gain from agents targeting key components of the adenosine metabolic network and, on this basis, provide a holistic view of the cancer purinome as a complex and integrated network.
Collapse
Affiliation(s)
- Gennady G Yegutkin
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland (G.G.Y.); Department of Neurosurgery, Robert Wood Johnson and New Jersey Medical Schools, Rutgers University, Piscataway, New Jersey (D.B.); and Rutgers Brain Health Institute, Piscataway, New Jersey (D.B.)
| | - Detlev Boison
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland (G.G.Y.); Department of Neurosurgery, Robert Wood Johnson and New Jersey Medical Schools, Rutgers University, Piscataway, New Jersey (D.B.); and Rutgers Brain Health Institute, Piscataway, New Jersey (D.B.)
| |
Collapse
|
45
|
Borsley S, Leigh DA, Roberts BMW. Chemical fuels for molecular machinery. Nat Chem 2022; 14:728-738. [PMID: 35778564 DOI: 10.1038/s41557-022-00970-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 05/10/2022] [Indexed: 12/11/2022]
Abstract
Chemical reaction networks that transform out-of-equilibrium 'fuel' to 'waste' are the engines that power the biomolecular machinery of the cell. Inspired by such systems, autonomous artificial molecular machinery is being developed that functions by catalysing the decomposition of chemical fuels, exploiting kinetic asymmetry to harness energy released from the fuel-to-waste reaction to drive non-equilibrium structures and dynamics. Different aspects of chemical fuels profoundly influence their ability to power molecular machines. Here we consider the structure and properties of the fuels that biology has evolved and compare their features with those of the rudimentary synthetic chemical fuels that have so far been used to drive autonomous non-equilibrium molecular-level dynamics. We identify desirable, but context-specific, traits for chemical fuels together with challenges and opportunities for the design and invention of new chemical fuels to power synthetic molecular machinery and other dissipative nanoscale processes.
Collapse
Affiliation(s)
- Stefan Borsley
- Department of Chemistry, University of Manchester, Manchester, UK
| | - David A Leigh
- Department of Chemistry, University of Manchester, Manchester, UK.
| | | |
Collapse
|
46
|
Abstract
SignificanceThe study provided a long-sought molecular mechanism that could explain the link between fatty acid metabolism and cancer metastasis. Further understanding may lead to new strategies to inhibit cancer metastasis. The chemical proteomic approach developed here will be useful for discovering other regulatory mechanisms of protein function by small molecule metabolites.
Collapse
|
47
|
Schlattner U. The Complex Functions of the NME Family-A Matter of Location and Molecular Activity. Int J Mol Sci 2021; 22:13083. [PMID: 34884887 PMCID: PMC8658066 DOI: 10.3390/ijms222313083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022] Open
Abstract
The family of NME proteins represents a quite complex group of multifunctional enzymes [...].
Collapse
Affiliation(s)
- Uwe Schlattner
- University Grenoble Alpes and Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France;
- Institut Universitaire de France (IUF), 75231 Paris, France
| |
Collapse
|
48
|
Proust B, Radić M, Vidaček NŠ, Cottet C, Attia S, Lamarche F, Ačkar L, Mikulčić VG, Tokarska-Schlattner M, Ćetković H, Schlattner U, Bosnar MH. NME6 is a phosphotransfer-inactive, monomeric NME/NDPK family member and functions in complexes at the interface of mitochondrial inner membrane and matrix. Cell Biosci 2021; 11:195. [PMID: 34789336 PMCID: PMC8597243 DOI: 10.1186/s13578-021-00707-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 11/01/2021] [Indexed: 11/10/2022] Open
Abstract
Background NME6 is a member of the nucleoside diphosphate kinase (NDPK/NME/Nm23) family which has key roles in nucleotide homeostasis, signal transduction, membrane remodeling and metastasis suppression. The well-studied NME1-NME4 proteins are hexameric and catalyze, via a phospho-histidine intermediate, the transfer of the terminal phosphate from (d)NTPs to (d)NDPs (NDP kinase) or proteins (protein histidine kinase). For the NME6, a gene/protein that emerged early in eukaryotic evolution, only scarce and partially inconsistent data are available. Here we aim to clarify and extend our knowledge on the human NME6. Results We show that NME6 is mostly expressed as a 186 amino acid protein, but that a second albeit much less abundant isoform exists. The recombinant NME6 remains monomeric, and does not assemble into homo-oligomers or hetero-oligomers with NME1-NME4. Consequently, NME6 is unable to catalyze phosphotransfer: it does not generate the phospho-histidine intermediate, and no NDPK activity can be detected. In cells, we could resolve and extend existing contradictory reports by localizing NME6 within mitochondria, largely associated with the mitochondrial inner membrane and matrix space. Overexpressing NME6 reduces ADP-stimulated mitochondrial respiration and complex III abundance, thus linking NME6 to dysfunctional oxidative phosphorylation. However, it did not alter mitochondrial membrane potential, mass, or network characteristics. Our screen for NME6 protein partners revealed its association with NME4 and OPA1, but a direct interaction was observed only with RCC1L, a protein involved in mitochondrial ribosome assembly and mitochondrial translation, and identified as essential for oxidative phosphorylation. Conclusions NME6, RCC1L and mitoribosomes localize together at the inner membrane/matrix space where NME6, in concert with RCC1L, may be involved in regulation of the mitochondrial translation of essential oxidative phosphorylation subunits. Our findings suggest new functions for NME6, independent of the classical phosphotransfer activity associated with NME proteins. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00707-0.
Collapse
Affiliation(s)
- Bastien Proust
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000, Zagreb, Croatia
| | - Martina Radić
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000, Zagreb, Croatia
| | - Nikolina Škrobot Vidaček
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000, Zagreb, Croatia.,Division of Molecular Biology, Ruđer Bošković Institute, 10000, Zagreb, Croatia
| | - Cécile Cottet
- Laboratory of Fundamental and Applied Bioenergetics, Univ. Grenoble Alpes and Inserm U1055, Grenoble, France
| | - Stéphane Attia
- Laboratory of Fundamental and Applied Bioenergetics, Univ. Grenoble Alpes and Inserm U1055, Grenoble, France
| | - Frédéric Lamarche
- Laboratory of Fundamental and Applied Bioenergetics, Univ. Grenoble Alpes and Inserm U1055, Grenoble, France
| | - Lucija Ačkar
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000, Zagreb, Croatia
| | - Vlatka Godinić Mikulčić
- The Miroslav Krleža Institute of Lexicography, 10000, Zagreb, Croatia.,Division of Molecular Biology, Ruđer Bošković Institute, 10000, Zagreb, Croatia
| | | | - Helena Ćetković
- Division of Molecular Biology, Ruđer Bošković Institute, 10000, Zagreb, Croatia
| | - Uwe Schlattner
- Univ. Grenoble Alpes and Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France, and Institut Universitaire de France (IUF), Paris, France
| | - Maja Herak Bosnar
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000, Zagreb, Croatia.
| |
Collapse
|
49
|
Lacombe ML, Lamarche F, De Wever O, Padilla-Benavides T, Carlson A, Khan I, Huna A, Vacher S, Calmel C, Desbourdes C, Cottet-Rousselle C, Hininger-Favier I, Attia S, Nawrocki-Raby B, Raingeaud J, Machon C, Guitton J, Le Gall M, Clary G, Broussard C, Chafey P, Thérond P, Bernard D, Fontaine E, Tokarska-Schlattner M, Steeg P, Bièche I, Schlattner U, Boissan M. The mitochondrially-localized nucleoside diphosphate kinase D (NME4) is a novel metastasis suppressor. BMC Biol 2021; 19:228. [PMID: 34674701 PMCID: PMC8529772 DOI: 10.1186/s12915-021-01155-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 09/17/2021] [Indexed: 12/11/2022] Open
Abstract
Background Mitochondrial nucleoside diphosphate kinase (NDPK-D, NME4, NM23-H4) is a multifunctional enzyme mainly localized in the intermembrane space, bound to the inner membrane. Results We constructed loss-of-function mutants of NDPK-D, lacking either NDP kinase activity or membrane interaction and expressed mutants or wild-type protein in cancer cells. In a complementary approach, we performed depletion of NDPK-D by RNA interference. Both loss-of-function mutations and NDPK-D depletion promoted epithelial-mesenchymal transition and increased migratory and invasive potential. Immunocompromised mice developed more metastases when injected with cells expressing mutant NDPK-D as compared to wild-type. This metastatic reprogramming is a consequence of mitochondrial alterations, including fragmentation and loss of mitochondria, a metabolic switch from respiration to glycolysis, increased ROS generation, and further metabolic changes in mitochondria, all of which can trigger pro-metastatic protein expression and signaling cascades. In human cancer, NME4 expression is negatively associated with markers of epithelial-mesenchymal transition and tumor aggressiveness and a good prognosis factor for beneficial clinical outcome. Conclusions These data demonstrate NME4 as a novel metastasis suppressor gene, the first localizing to mitochondria, pointing to a role of mitochondria in metastatic dissemination. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01155-5.
Collapse
Affiliation(s)
- Marie-Lise Lacombe
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Paris, France
| | - Frederic Lamarche
- Université Grenoble Alpes, INSERM U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), and SFR Environmental and Systems Biology (BEeSy), Grenoble, France
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | | | - Alyssa Carlson
- Molecular Biology and Biochemistry Department, Wesleyan University, Middletown, USA
| | - Imran Khan
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Anda Huna
- Cancer Research Center of Lyon, INSERM U1052, CNRS UMR 5286, Léon Bérard Center, Lyon University, Lyon, France
| | - Sophie Vacher
- Unit of Pharmacogenetics, Department of Genetics, Curie Institute, Paris, France
| | - Claire Calmel
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Paris, France
| | - Céline Desbourdes
- Université Grenoble Alpes, INSERM U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), and SFR Environmental and Systems Biology (BEeSy), Grenoble, France
| | - Cécile Cottet-Rousselle
- Université Grenoble Alpes, INSERM U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), and SFR Environmental and Systems Biology (BEeSy), Grenoble, France
| | - Isabelle Hininger-Favier
- Université Grenoble Alpes, INSERM U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), and SFR Environmental and Systems Biology (BEeSy), Grenoble, France
| | - Stéphane Attia
- Université Grenoble Alpes, INSERM U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), and SFR Environmental and Systems Biology (BEeSy), Grenoble, France
| | - Béatrice Nawrocki-Raby
- Reims Champagne Ardenne University, INSERM, P3Cell UMR-S 1250, SFR CAP-SANTE, Reims, France
| | - Joël Raingeaud
- INSERM U1279, Gustave Roussy Institute, Villejuif, France
| | - Christelle Machon
- Cancer Research Center of Lyon, INSERM U1052, CNRS UMR 5286, Léon Bérard Center, Lyon University, Lyon, France
| | - Jérôme Guitton
- Cancer Research Center of Lyon, INSERM U1052, CNRS UMR 5286, Léon Bérard Center, Lyon University, Lyon, France
| | - Morgane Le Gall
- Proteomics Platform 3P5, Paris University, Cochin Institute, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - Guilhem Clary
- Proteomics Platform 3P5, Paris University, Cochin Institute, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - Cedric Broussard
- Proteomics Platform 3P5, Paris University, Cochin Institute, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - Philippe Chafey
- Proteomics Platform 3P5, Paris University, Cochin Institute, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - Patrice Thérond
- AP-HP, CHU Bicêtre, Laboratory of Biochemistry, Le Kremlin-Bicêtre Hospital, Le Kremlin-Bicêtre, France.,EA7537, Paris Saclay University, Châtenay-Malabry, France
| | - David Bernard
- Cancer Research Center of Lyon, INSERM U1052, CNRS UMR 5286, Léon Bérard Center, Lyon University, Lyon, France
| | - Eric Fontaine
- Université Grenoble Alpes, INSERM U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), and SFR Environmental and Systems Biology (BEeSy), Grenoble, France
| | - Malgorzata Tokarska-Schlattner
- Université Grenoble Alpes, INSERM U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), and SFR Environmental and Systems Biology (BEeSy), Grenoble, France
| | - Patricia Steeg
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Ivan Bièche
- Unit of Pharmacogenetics, Department of Genetics, Curie Institute, Paris, France
| | - Uwe Schlattner
- Université Grenoble Alpes, INSERM U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), Institut Universitaire de France (IUF), Grenoble, France.
| | - Mathieu Boissan
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Paris, France. .,AP-HP, Laboratory of Biochemistry and Hormonology, Tenon Hospital, Paris, France.
| |
Collapse
|
50
|
Murali Mahadevan H, Hashemiaghdam A, Ashrafi G, Harbauer AB. Mitochondria in Neuronal Health: From Energy Metabolism to Parkinson's Disease. Adv Biol (Weinh) 2021; 5:e2100663. [PMID: 34382382 DOI: 10.1002/adbi.202100663] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/30/2021] [Indexed: 01/01/2023]
Abstract
Mitochondria are the main suppliers of neuronal adenosine triphosphate and play a critical role in brain energy metabolism. Mitochondria also serve as Ca2+ sinks and anabolic factories and are therefore essential for neuronal function and survival. Dysregulation of neuronal bioenergetics is increasingly implicated in neurodegenerative disorders, particularly Parkinson's disease. This review describes the role of mitochondria in energy metabolism under resting conditions and during synaptic transmission, and presents evidence for the contribution of neuronal mitochondrial dysfunction to Parkinson's disease.
Collapse
Affiliation(s)
| | - Arsalan Hashemiaghdam
- Department of Cell Biology and Physiology, Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Ghazaleh Ashrafi
- Department of Cell Biology and Physiology, Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Angelika Bettina Harbauer
- Max-Planck-Institute for Neurobiology, 82152, Martinsried, Germany.,Technical University of Munich, Institute of Neuronal Cell Biology, 80333, Munich, Germany.,Munich Cluster for Systems Neurology, Munich, Germany
| |
Collapse
|