1
|
Zhang H, Tang M, Liu Q, Wu D, Sun B, Dong J, Guan L, Luo J, Zeng M. PAT exposure caused human hepatocytes apoptosis and induced mice subacute liver injury by activating oxidative stress and the ERS-associated PERK pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:177003. [PMID: 39433224 DOI: 10.1016/j.scitotenv.2024.177003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/06/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024]
Abstract
With the widespread use of antimony compounds in synthetic materials and processing, the occupational exposure and environmental pollution caused by antimony have attracted the attention of researchers. Studies have shown that antimony compounds can cause liver damage, but the mechanism has not yet been elucidated. In this study, we used the trivalent potassium antimony tartrate (PAT) to infect L02 hepatocytes and Kunming (KM) mice to establish an antimony-induced apoptosis model of L02 cells and a subacute liver injury model of KM mice. We found that PAT exposure caused hepatocyte apoptosis and was accompanied by oxidative stress and endoplasmic reticulum stress (ERS), and the ERS-associated PERK pathway was activated. Further experimental results showed that N-acetyl-l-cysteine (NAC) pretreatment or silencing of the PERK gene in L02 cells reduced PAT-induced apoptosis. The activity of SOD and CAT in treated L02 cells was increased, the malondialdehyde content in L02 cells and liver tissues was decreased, and the content of ERS-related proteins GRP78 and CHOP, as well as the content of PERK-pathway-related proteins p-PERK/PERK, p-eif2α/eif2α and ATF4 protein were significantly reduced. Overall, PAT exposure triggered hepatocyte apoptosis and liver injury by inducing oxidative stress and activating the ERS-associated PERK pathway; however, this effect could be alleviated by NAC intervention or silencing of PERK in hepatocytes.
Collapse
Affiliation(s)
- Hualing Zhang
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Meng Tang
- Center for Disease Control and Prevention, Jiulongpo District, Chongqing 400050, PR China
| | - Qin Liu
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Die Wu
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Bing Sun
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Jingbang Dong
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Lan Guan
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Jianlan Luo
- Institute of Geophysical & Geochemical Exploration of Hunan, Changsha 411100, PR China
| | - Ming Zeng
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China.
| |
Collapse
|
2
|
Hellmuth S, Stemmann O. Requirement of Nek2a and cyclin A2 for Wapl-dependent removal of cohesin from prophase chromatin. EMBO J 2024; 43:5237-5259. [PMID: 39271794 PMCID: PMC11535040 DOI: 10.1038/s44318-024-00228-9] [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: 03/08/2024] [Revised: 08/14/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Sister chromatid cohesion is mediated by the cohesin complex. In mitotic prophase cohesin is removed from chromosome arms in a Wapl- and phosphorylation-dependent manner. Sgo1-PP2A protects pericentromeric cohesion by dephosphorylation of cohesin and its associated Wapl antagonist sororin. However, Sgo1-PP2A relocates to inner kinetochores well before sister chromatids are separated by separase, leaving pericentromeric regions unprotected. Why deprotected cohesin is not removed by Wapl remains enigmatic. By reconstituting Wapl-dependent cohesin removal from chromatin in vitro, we discovered a requirement for Nek2a and Cdk1/2-cyclin A2. These kinases phosphorylate cohesin-bound Pds5b, thereby converting it from a sororin- to a Wapl-interactor. Replacement of endogenous Pds5b by a phosphorylation mimetic variant causes premature sister chromatid separation (PCS). Conversely, phosphorylation-resistant Pds5b impairs chromosome arm separation in prometaphase-arrested cells and suppresses PCS in the absence of Sgo1. Early mitotic degradation of Nek2a and cyclin A2 may therefore explain why only separase, but not Wapl, can trigger anaphase.
Collapse
Affiliation(s)
- Susanne Hellmuth
- Chair of Genetics, University of Bayreuth, 95440, Bayreuth, Germany.
| | - Olaf Stemmann
- Chair of Genetics, University of Bayreuth, 95440, Bayreuth, Germany
| |
Collapse
|
3
|
Chen ZL, Xie C, Zeng W, Huang RQ, Yang JE, Liu JY, Chen YJ, Zhuang SM. Synergistic induction of mitotic pyroptosis and tumor remission by inhibiting proteasome and WEE family kinases. Signal Transduct Target Ther 2024; 9:181. [PMID: 38992067 PMCID: PMC11239683 DOI: 10.1038/s41392-024-01896-z] [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: 12/30/2023] [Revised: 05/26/2024] [Accepted: 06/13/2024] [Indexed: 07/13/2024] Open
Abstract
Mitotic catastrophe (MC), which occurs under dysregulated mitosis, represents a fascinating tactic to specifically eradicate tumor cells. Whether pyroptosis can be a death form of MC remains unknown. Proteasome-mediated protein degradation is crucial for M-phase. Bortezomib (BTZ), which inhibits the 20S catalytic particle of proteasome, is approved to treat multiple myeloma and mantle cell lymphoma, but not solid tumors due to primary resistance. To date, whether and how proteasome inhibitor affected the fates of cells in M-phase remains unexplored. Here, we show that BTZ treatment, or silencing of PSMC5, a subunit of 19S regulatory particle of proteasome, causes G2- and M-phase arrest, multi-polar spindle formation, and consequent caspase-3/GSDME-mediated pyroptosis in M-phase (designated as mitotic pyroptosis). Further investigations reveal that inhibitor of WEE1/PKMYT1 (PD0166285), but not inhibitor of ATR, CHK1 or CHK2, abrogates the BTZ-induced G2-phase arrest, thus exacerbates the BTZ-induced mitotic arrest and pyroptosis. Combined BTZ and PD0166285 treatment (named BP-Combo) selectively kills various types of solid tumor cells, and significantly lessens the IC50 of both BTZ and PD0166285 compared to BTZ or PD0166285 monotreatment. Studies using various mouse models show that BP-Combo has much stronger inhibition on tumor growth and metastasis than BTZ or PD0166285 monotreatment, and no obvious toxicity is observed in BP-Combo-treated mice. These findings disclose the effect of proteasome inhibitors in inducing pyroptosis in M-phase, characterize pyroptosis as a new death form of mitotic catastrophe, and identify dual inhibition of proteasome and WEE family kinases as a promising anti-cancer strategy to selectively kill solid tumor cells.
Collapse
Affiliation(s)
- Zhan-Li Chen
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou, PR China
| | - Chen Xie
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou, PR China
| | - Wei Zeng
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou, PR China
| | - Rui-Qi Huang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou, PR China
| | - Jin-E Yang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou, PR China
| | - Jin-Yu Liu
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou, PR China
| | - Ya-Jing Chen
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou, PR China
| | - Shi-Mei Zhuang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou, PR China.
- Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, PR China.
| |
Collapse
|
4
|
Yamada Y, Zheng Z, Jad AK, Yamashita M. Lethal and sublethal effects of programmed cell death pathways on hematopoietic stem cells. Exp Hematol 2024; 134:104214. [PMID: 38582294 DOI: 10.1016/j.exphem.2024.104214] [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: 01/31/2024] [Revised: 03/26/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024]
Abstract
Programmed cell death is an evolutionally conserved cellular process in multicellular organisms that eliminates unnecessary or rogue cells during development, infection, and carcinogenesis. Hematopoietic stem cells (HSCs) are a rare, self-renewing, and multipotent cell population necessary for the establishment and regeneration of the hematopoietic system. Counterintuitively, key components necessary for programmed cell death induction are abundantly expressed in long-lived HSCs, which often survive myeloablative stress by engaging a prosurvival response that counteracts cell death-inducing stimuli. Although HSCs are well known for their apoptosis resistance, recent studies have revealed their unique vulnerability to certain types of programmed necrosis, such as necroptosis and ferroptosis. Moreover, emerging evidence has shown that programmed cell death pathways can be sublethally activated to cause nonlethal consequences such as innate immune response, organelle dysfunction, and mutagenesis. In this review, we summarized recent findings on how divergent cell death programs are molecularly regulated in HSCs. We then discussed potential side effects caused by sublethal activation of programmed cell death pathways on the functionality of surviving HSCs.
Collapse
Affiliation(s)
- Yuta Yamada
- Division of Stem Cell and Molecular Medicine, Centre for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Zhiqian Zheng
- Division of Stem Cell and Molecular Medicine, Centre for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Division of Experimental Hematology, Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Alaa K Jad
- Division of Stem Cell and Molecular Medicine, Centre for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masayuki Yamashita
- Division of Stem Cell and Molecular Medicine, Centre for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Division of Experimental Hematology, Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
5
|
Jenner A, Garcia-Saez AJ. The regulation of the apoptotic pore-An immunological tightrope walk. Adv Immunol 2024; 162:59-108. [PMID: 38866439 DOI: 10.1016/bs.ai.2024.02.004] [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] [Indexed: 06/14/2024]
Abstract
Apoptotic pore formation in mitochondria is the pivotal point for cell death during mitochondrial apoptosis. It is regulated by BCL-2 family proteins in response to various cellular stress triggers and mediates mitochondrial outer membrane permeabilization (MOMP). This allows the release of mitochondrial contents into the cytosol, which triggers rapid cell death and clearance through the activation of caspases. However, under conditions of low caspase activity, the mitochondrial contents released into the cytosol through apoptotic pores serve as inflammatory signals and activate various inflammatory responses. In this chapter, we discuss how the formation of the apoptotic pore is regulated by BCL-2 proteins as well as other cellular or mitochondrial proteins and membrane lipids. Moreover, we highlight the importance of sublethal MOMP in the regulation of mitochondrial-activated inflammation and discuss its physiological consequences in the context of pathogen infection and disease and how it can potentially be exploited therapeutically, for example to improve cancer treatment.
Collapse
Affiliation(s)
- Andreas Jenner
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Ana J Garcia-Saez
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
| |
Collapse
|
6
|
Rahman MS, Shindo Y, Oka K, Ikeda W, Suzuki M. Live Cell Monitoring of Separase Activity, a Key Enzymatic Reaction for Chromosome Segregation, with Chimeric FRET-Based Molecular Sensor upon Cell Cycle Progression. BIOSENSORS 2024; 14:192. [PMID: 38667185 PMCID: PMC11048197 DOI: 10.3390/bios14040192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/29/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024]
Abstract
Separase is a key cysteine protease in the separation of sister chromatids through the digestion of the cohesin ring that inhibits chromosome segregation as a trigger of the metaphase-anaphase transition in eukaryotes. Its activity is highly regulated by binding with securin and cyclinB-CDK1 complex. These bindings prevent the proteolytic activity of separase until the onset of anaphase. Chromosome missegregation and aneuploidy are frequently observed in malignancies. However, there are some difficulties in biochemical examinations due to the instability of separase in vitro and the fact that few spatiotemporal resolution approaches exist for monitoring live separase activity throughout mitotic processes. Here, we have developed FRET-based molecular sensors, including GFP variants, with separase-cleavable sequences as donors and covalently attached fluorescent dyes as acceptor molecules. These are applicable to conventional live cell imaging and flow cytometric analysis because of efficient live cell uptake. We investigated the performance of equivalent molecular sensors, either localized or not localized inside the nucleus under cell cycle control, using flow cytometry. Synchronized cell cycle progression rendered significant separase activity detections in both molecular sensors. We obtained consistent outcomes with localized molecular sensor introduction and cell cycle control by fluorescent microscopic observations. We thus established live cell separase activity monitoring systems that can be used specifically or statistically, which could lead to the elucidation of separase properties in detail.
Collapse
Affiliation(s)
- Md. Shazadur Rahman
- Graduate School of Science and Engineering, Saitama University, 255 Shimo-Okubo, Sakura-ku, Saitama 338-8570, Japan; (M.S.R.); (W.I.)
- Department of Agricultural Chemistry, Hajee Mohammad Danesh Science and Technology University, Dinajpur 5200, Bangladesh
| | - Yutaka Shindo
- Department of Bioscience and informatics, Faculty of Science and Technology, Keio University, Yokohama 223-0061, Japan; (Y.S.); (K.O.)
| | - Kotaro Oka
- Department of Bioscience and informatics, Faculty of Science and Technology, Keio University, Yokohama 223-0061, Japan; (Y.S.); (K.O.)
- School of Frontier Engineering, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0373, Japan
| | - Wataru Ikeda
- Graduate School of Science and Engineering, Saitama University, 255 Shimo-Okubo, Sakura-ku, Saitama 338-8570, Japan; (M.S.R.); (W.I.)
| | - Miho Suzuki
- Graduate School of Science and Engineering, Saitama University, 255 Shimo-Okubo, Sakura-ku, Saitama 338-8570, Japan; (M.S.R.); (W.I.)
| |
Collapse
|
7
|
Czabotar PE, Garcia-Saez AJ. Mechanisms of BCL-2 family proteins in mitochondrial apoptosis. Nat Rev Mol Cell Biol 2023; 24:732-748. [PMID: 37438560 DOI: 10.1038/s41580-023-00629-4] [Citation(s) in RCA: 223] [Impact Index Per Article: 111.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2023] [Indexed: 07/14/2023]
Abstract
The proteins of the BCL-2 family are key regulators of mitochondrial apoptosis, acting as either promoters or inhibitors of cell death. The functional interplay and balance between the opposing BCL-2 family members control permeabilization of the outer mitochondrial membrane, leading to the release of activators of the caspase cascade into the cytosol and ultimately resulting in cell death. Despite considerable research, our knowledge about the mechanisms of the BCL-2 family of proteins remains insufficient, which complicates cell fate predictions and does not allow us to fully exploit these proteins as targets for drug discovery. Detailed understanding of the formation and molecular architecture of the apoptotic pore in the outer mitochondrial membrane remains a holy grail in the field, but new studies allow us to begin constructing a structural model of its arrangement. Recent literature has also revealed unexpected activities for several BCL-2 family members that challenge established concepts of how they regulate mitochondrial permeabilization. In this Review, we revisit the most important advances in the field and integrate them into a new structure-function-based classification of the BCL-2 family members that intends to provide a comprehensive model for BCL-2 action in apoptosis. We close this Review by discussing the potential of drugging the BCL-2 family in diseases characterized by aberrant apoptosis.
Collapse
Affiliation(s)
- Peter E Czabotar
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia.
| | - Ana J Garcia-Saez
- Membrane Biophysics, Institute of Genetics, CECAD, University of Cologne, Cologne, Germany.
| |
Collapse
|
8
|
Liu C, Mentzelopoulou A, Papagavriil F, Ramachandran P, Perraki A, Claus L, Barg S, Dörmann P, Jaillais Y, Johnen P, Russinova E, Gizeli E, Schaaf G, Moschou PN. SEC14-like condensate phase transitions at plasma membranes regulate root growth in Arabidopsis. PLoS Biol 2023; 21:e3002305. [PMID: 37721949 PMCID: PMC10538751 DOI: 10.1371/journal.pbio.3002305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 09/28/2023] [Accepted: 08/20/2023] [Indexed: 09/20/2023] Open
Abstract
Protein function can be modulated by phase transitions in their material properties, which can range from liquid- to solid-like; yet, the mechanisms that drive these transitions and whether they are important for physiology are still unknown. In the model plant Arabidopsis, we show that developmental robustness is reinforced by phase transitions of the plasma membrane-bound lipid-binding protein SEC14-like. Using imaging, genetics, and in vitro reconstitution experiments, we show that SEC14-like undergoes liquid-like phase separation in the root stem cells. Outside the stem cell niche, SEC14-like associates with the caspase-like protease separase and conserved microtubule motors at unique polar plasma membrane interfaces. In these interfaces, SEC14-like undergoes processing by separase, which promotes its liquid-to-solid transition. This transition is important for root development, as lines expressing an uncleavable SEC14-like variant or mutants of separase and associated microtubule motors show similar developmental phenotypes. Furthermore, the processed and solidified but not the liquid form of SEC14-like interacts with and regulates the polarity of the auxin efflux carrier PINFORMED2. This work demonstrates that robust development can involve liquid-to-solid transitions mediated by proteolysis at unique plasma membrane interfaces.
Collapse
Affiliation(s)
- Chen Liu
- Department of Biology, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Department of Plant Biology, Uppsala BioCenter, Swedish University of Agricultural Sciences and Linnean Center for Plant Biology, Uppsala, Sweden
| | - Andriani Mentzelopoulou
- Department of Biology, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Fotini Papagavriil
- Department of Biology, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Prashanth Ramachandran
- Department of Organismal Biology, Physiological Botany, Linnean Centre for Plant Biology, Uppsala University, Uppsala, Sweden
| | - Artemis Perraki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Lucas Claus
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- Center for Plant Systems Biology, Ghent, Belgium
| | - Sebastian Barg
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Peter Dörmann
- Institute of Molecular Physiology and Biotechnology of Plants, University of Bonn, Bonn, Germany
| | - Yvon Jaillais
- Laboratoire Reproduction et Développement des Plantes, ENS de Lyon, Université Claude Bernard Lyon 1, CNRS, INRAE, Lyon, France
| | - Philipp Johnen
- Department of Plant Nutrition, Institute of Crop Science and Resource Conservation, University of Bonn, Bonn, Germany
| | - Eugenia Russinova
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- Center for Plant Systems Biology, Ghent, Belgium
| | - Electra Gizeli
- Department of Biology, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Gabriel Schaaf
- Department of Plant Nutrition, Institute of Crop Science and Resource Conservation, University of Bonn, Bonn, Germany
| | - Panagiotis Nikolaou Moschou
- Department of Biology, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Department of Plant Biology, Uppsala BioCenter, Swedish University of Agricultural Sciences and Linnean Center for Plant Biology, Uppsala, Sweden
| |
Collapse
|
9
|
Zheng Z, Han L, Li Y, Chen Z, Yang W, Liu C, Tao M, Jiang Y, Ke X, Liu Y, Guo X. Phospholipase A2-activating protein induces mitophagy trough anti-apoptotic MCL1-mediated NLRX1 oligomerization. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023:119487. [PMID: 37211156 DOI: 10.1016/j.bbamcr.2023.119487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/17/2023] [Accepted: 05/11/2023] [Indexed: 05/23/2023]
Abstract
Mitochondrial protein homeostasis is fine-tuned by diverse physiological processes such as mitochondria-associated degradation (MAD), which is regulated by valosin-containing protein (VCP) and its cofactors. As a cofactor of VCP, the mutation of phospholipase A-2-activating protein (PLAA) is the genetic cause of PLAA-associated neurodevelopmental disorder (PLAAND). However, the physiological and pathological roles of PLAA in mitochondria remain unclear. Here, we demonstrate that PLAA partially associates with mitochondria. Deficiency in PLAA increases mitochondrial reactive oxygen species (ROS) production, reduces mitochondrial membrane potential, inhibits mitochondrial respiratory activity and causes excessive mitophagy. Mechanically, PLAA interacts with myeloid cell leukemia-1 (MCL1) and facilitates its retro-translocation and proteasome-dependent degradation. The upregulation of MCL1 promotes the oligomerization of NLR family member X1 (NLRX1) and activation of mitophagy. Whereas downregulating NLRX1 abolishes MCL1 induced mitophagy. In summary, our data identify PLAA as a novel mediator of mitophagy by regulating MCL1-NLRX1 axis. We propose mitophagy as a target for therapeutic intervention in PLAAND.
Collapse
Affiliation(s)
- Zhilong Zheng
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Lu Han
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China; Child Mental Health Research Center, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuanbo Li
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhen Chen
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wangju Yang
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunyue Liu
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mengdan Tao
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yueqing Jiang
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoyan Ke
- Child Mental Health Research Center, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Yan Liu
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Xing Guo
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
10
|
Yu J, Morgan DO, Boland A. The molecular mechanisms of human separase regulation. Biochem Soc Trans 2023:233012. [PMID: 37140261 DOI: 10.1042/bst20221400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/11/2023] [Accepted: 04/18/2023] [Indexed: 05/05/2023]
Abstract
Sister chromatid segregation is the final irreversible step of mitosis. It is initiated by a complex regulatory system that ultimately triggers the timely activation of a conserved cysteine protease named separase. Separase cleaves the cohesin protein ring that links the sister chromatids and thus facilitates their separation and segregation to the opposite poles of the dividing cell. Due to the irreversible nature of this process, separase activity is tightly controlled in all eukaryotic cells. In this mini-review, we summarize the latest structural and functional findings on the regulation of separase, with an emphasis on the regulation of the human enzyme by two inhibitors, the universal inhibitor securin and the vertebrate-specific inhibitor CDK1-cyclin B. We discuss the two fundamentally different inhibitory mechanisms by which these inhibitors block separase activity by occluding substrate binding. We also describe conserved mechanisms that facilitate substrate recognition and point out open research questions that will guide studies of this fascinating enzyme for years to come.
Collapse
Affiliation(s)
- Jun Yu
- Department of Molecular and Cellular Biology, University of Geneva, CH-1211 Geneva, Switzerland
| | - David O Morgan
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, U.S.A
| | - Andreas Boland
- Department of Molecular and Cellular Biology, University of Geneva, CH-1211 Geneva, Switzerland
| |
Collapse
|
11
|
Xie D, Deng T, Zhai Z, Qin T, Song C, Xu Y, Sun T. Moschus exerted protective activity against H 2O 2-induced cell injury in PC12 cells through regulating Nrf-2/ARE signaling pathways. Biomed Pharmacother 2023; 159:114290. [PMID: 36708701 DOI: 10.1016/j.biopha.2023.114290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023] Open
Abstract
The pivotal characteristics of Alzheimer's disease (AD) are irreversible memory loss and progressive cognitive decline, eventually causing death from brain failure. In the various proposed hypotheses of AD, oxidative stress is also regarded as a symbolic pathophysiologic cascade contributing to brain diseases. Using Chinese herbal medicine may be beneficial for treating and preventing AD. As a rare and valuable animal medicine, Moschus possesses antioxidant and antiapoptotic efficacy and is extensively applied for treating unconsciousness, stroke, coma, and cerebrovascular diseases. We aim to evaluate whether Moschus protects PC12 cells from hydrogen peroxide (H2O2)-induced cellular injury. The chemical constituents of Moschus are analyzed by GC-MS assay. The cell viability, reactive oxygen species (ROS) levels, mitochondrial membrane potential (MMP) levels, oxidative stress-related indicators, and apoptotic proteins are determined. Through GC-MS analysis, nineteen active contents were identified. The cell viability loss, lactate dehydrogenase releases, MMP levels, ROS productions, and Malondialdehyde (MDA) activities decreased, and BAX, Caspase-3, and Kelch-like ECH-associated protein 1 expression also significantly down-regulated and heme oxygenase 1, nuclear factor erythroid-2-related factor 2 (Nrf-2), and quinine oxidoreductase 1 expression upregulated after pretreatment of Moschus. The result indicated Moschus has neuroprotective activity in relieving H2O2-induced cellular damage, and the potential mechanism might be associated with regulating the Nrf-2/ARE signaling pathway. A more in-depth and comprehensive understanding of Moschus in the pathogenesis of AD will provide a fundamental basis for in vivo AD animal model research, which may be able to provide further insights and new targets for AD therapy.
Collapse
Affiliation(s)
- Danni Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Ting Deng
- Jintang Second People' s Hospital, Chengdu 610404, China.
| | - Zhenwei Zhai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Tao Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Caiyou Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Ying Xu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Tao Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
12
|
Flores‐Romero H, Hohorst L, John M, Albert M, King LE, Beckmann L, Szabo T, Hertlein V, Luo X, Villunger A, Frenzel LP, Kashkar H, Garcia‐Saez AJ. BCL-2-family protein tBID can act as a BAX-like effector of apoptosis. EMBO J 2022; 41:e108690. [PMID: 34931711 PMCID: PMC8762556 DOI: 10.15252/embj.2021108690] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 11/14/2021] [Accepted: 11/22/2021] [Indexed: 12/26/2022] Open
Abstract
During apoptosis, the BCL-2-family protein tBID promotes mitochondrial permeabilization by activating BAX and BAK and by blocking anti-apoptotic BCL-2 members. Here, we report that tBID can also mediate mitochondrial permeabilization by itself, resulting in release of cytochrome c and mitochondrial DNA, caspase activation and apoptosis even in absence of BAX and BAK. This previously unrecognized activity of tBID depends on helix 6, homologous to the pore-forming regions of BAX and BAK, and can be blocked by pro-survival BCL-2 proteins. Importantly, tBID-mediated mitochondrial permeabilization independent of BAX and BAK is physiologically relevant for SMAC release in the immune response against Shigella infection. Furthermore, it can be exploited to kill leukaemia cells with acquired venetoclax resistance due to lack of active BAX and BAK. Our findings define tBID as an effector of mitochondrial permeabilization in apoptosis and provide a new paradigm for BCL-2 proteins, with implications for anti-bacterial immunity and cancer therapy.
Collapse
Affiliation(s)
- Hector Flores‐Romero
- Institute for GeneticsUniversity of CologneCologneGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
- Interfaculty Institute of BiochemistryEberhard‐Karls‐Universität TübingenTübingenGermany
| | - Lisa Hohorst
- Institute for GeneticsUniversity of CologneCologneGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
| | - Malina John
- Interfaculty Institute of BiochemistryEberhard‐Karls‐Universität TübingenTübingenGermany
| | - Marie‐Christine Albert
- Institute for Molecular Immunology, and Center for Molecular Medicine Cologne (CMMC)Faculty of MedicineUniversity Hospital of CologneUniversity of CologneCologneGermany
| | - Louise E King
- Institute for GeneticsUniversity of CologneCologneGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
| | - Laura Beckmann
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
- Department I of Internal MedicineUniversity Hospital of CologneCologneGermany
- Center of Integrated Oncology ABCDUniversity Hospital of CologneCologneGermany
| | - Tamas Szabo
- Division of Developmental ImmunologyBiocenterMedical University of InnsbruckInnsbruckAustria
| | - Vanessa Hertlein
- Interfaculty Institute of BiochemistryEberhard‐Karls‐Universität TübingenTübingenGermany
- Present address:
Children Cancer Research Institute (CCRI)ViennaAustria
| | - Xu Luo
- Eppley Institute for Research in Cancer and Allied DiseasesFred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaMEUSA
- Department of Pathology and MicrobiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Andreas Villunger
- Division of Developmental ImmunologyBiocenterMedical University of InnsbruckInnsbruckAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Ludwig Boltzmann Institute for Rare and Undiagnosed DiseasesViennaAustria
| | - Lukas P Frenzel
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
- Department I of Internal MedicineUniversity Hospital of CologneCologneGermany
- Center of Integrated Oncology ABCDUniversity Hospital of CologneCologneGermany
| | - Hamid Kashkar
- Institute for Molecular Immunology, and Center for Molecular Medicine Cologne (CMMC)Faculty of MedicineUniversity Hospital of CologneUniversity of CologneCologneGermany
| | - Ana J Garcia‐Saez
- Institute for GeneticsUniversity of CologneCologneGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
- Interfaculty Institute of BiochemistryEberhard‐Karls‐Universität TübingenTübingenGermany
| |
Collapse
|
13
|
Marker-free co-selection for successive rounds of prime editing in human cells. Nat Commun 2022; 13:5909. [PMID: 36207338 PMCID: PMC9546848 DOI: 10.1038/s41467-022-33669-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 09/27/2022] [Indexed: 11/18/2022] Open
Abstract
Prime editing enables the introduction of precise point mutations, small insertions, or short deletions without requiring donor DNA templates. However, efficiency remains a key challenge in a broad range of human cell types. In this work, we design a robust co-selection strategy through coediting of the ubiquitous and essential sodium/potassium pump (Na+/K+ ATPase). We readily engineer highly modified pools of cells and clones with homozygous modifications for functional studies with minimal pegRNA optimization. This process reveals that nicking the non-edited strand stimulates multiallelic editing but often generates tandem duplications and large deletions at the target site, an outcome dictated by the relative orientation of the protospacer adjacent motifs. Our approach streamlines the production of cell lines with multiple genetic modifications to create cellular models for biological research and lays the foundation for the development of cell-type specific co-selection strategies. Prime editing enables the introduction of precise point mutations, small insertions, or short deletions without requiring donor DNA templates. Here the authors develop a co-selection strategy to facilitate prime editing in human cells and provide design principles to prevent the formation of undesired editing byproducts at the target site.
Collapse
|
14
|
Identification of a novel substrate motif of yeast separase and deciphering the recognition specificity using AlphaFold2 and molecular dynamics simulation. Biochem Biophys Res Commun 2022; 620:173-179. [DOI: 10.1016/j.bbrc.2022.06.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 06/19/2022] [Indexed: 11/19/2022]
|
15
|
Vukušić K, Tolić IM. Polar Chromosomes-Challenges of a Risky Path. Cells 2022; 11:1531. [PMID: 35563837 PMCID: PMC9101661 DOI: 10.3390/cells11091531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 12/29/2022] Open
Abstract
The process of chromosome congression and alignment is at the core of mitotic fidelity. In this review, we discuss distinct spatial routes that the chromosomes take to align during prometaphase, which are characterized by distinct biomolecular requirements. Peripheral polar chromosomes are an intriguing case as their alignment depends on the activity of kinetochore motors, polar ejection forces, and a transition from lateral to end-on attachments to microtubules, all of which can result in the delayed alignment of these chromosomes. Due to their undesirable position close to and often behind the spindle pole, these chromosomes may be particularly prone to the formation of erroneous kinetochore-microtubule interactions, such as merotelic attachments. To prevent such errors, the cell employs intricate mechanisms to preposition the spindle poles with respect to chromosomes, ensure the formation of end-on attachments in restricted spindle regions, repair faulty attachments by error correction mechanisms, and delay segregation by the spindle assembly checkpoint. Despite this protective machinery, there are several ways in which polar chromosomes can fail in alignment, mis-segregate, and lead to aneuploidy. In agreement with this, polar chromosomes are present in certain tumors and may even be involved in the process of tumorigenesis.
Collapse
Affiliation(s)
- Kruno Vukušić
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia;
| | | |
Collapse
|
16
|
Huang X, Zhang G, Tang T, Gao X, Liang T. One shoot, three birds: Targeting NEK2 orchestrates chemoradiotherapy, targeted therapy, and immunotherapy in cancer treatment. Biochim Biophys Acta Rev Cancer 2022; 1877:188696. [PMID: 35157980 DOI: 10.1016/j.bbcan.2022.188696] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/16/2022]
Abstract
Combinational therapy has improved the cancer therapeutic landscape but is associated with a concomitant increase in adverse side reactions. Emerging evidence proposes that targeting one core target with multiple critical roles in tumors can achieve combined anti-tumor effects. This review focuses on NEK2, a member of serine/threonine kinases, with broad sequence identity to the mitotic regulator NIMA of the filamentous fungus Aspergillus nidulans. Elevated expression of NEK2 was initially found to promote tumorigeneses through abnormal regulation of the cell cycle. Subsequent studies report that NEK2 is overexpressed in a broad spectrum of tumor types and is associated with tumor progression and therapeutic resistance. Intriguingly, NEK2 has recently been revealed to mediate tumor immune escape by stabilizing the expression of PD-L1. Targeting NEK2 is thus becoming a promising approach for cancer treatment by orchestrating chemoradiotherapy, targeted therapy, and immunotherapy. It represents a novel strategy for inducing combined anti-cancer effects using a mono-agent.
Collapse
Affiliation(s)
- Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| | - Gang Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Tianyu Tang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Xiang Gao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
17
|
Ho CJ, Ko HJ, Liao TS, Zheng XR, Chou PH, Wang LT, Lin RW, Chen CH, Wang C. Severe cellular stress activates apoptosis independently of p53 in osteosarcoma. Cell Death Discov 2021; 7:275. [PMID: 34608124 PMCID: PMC8490387 DOI: 10.1038/s41420-021-00658-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/25/2021] [Accepted: 09/16/2021] [Indexed: 12/30/2022] Open
Abstract
Apoptosis induced by doxorubicin, bortezomib, or paclitaxel, targeting DNA, 26S proteasome, and microtubules respectively, was assessed in two osteosarcoma cells, p53 wild-type U2OS and p53-null MG63 cells. Doxorubicin-induced apoptosis only occurred in U2OS, not in MG63. In contrast, bortezomib and paclitaxel could drive U2OS or MG63 toward apoptosis effectively, suggesting that apoptosis induced by bortezomib or paclitaxel is p53-independent. The expressions of Bcl2 family members such as Bcl2, Bcl-xl, and Puma could be seen in U2OS and MG63 cells with or without doxorubicin, bortezomib, or paclitaxel treatment. In contrast, another member, Bim, only could be observed in U2OS, not in MG63, under the same conditions. Bim knockdown did not affect the doxorubicin-induced apoptosis in U2OS, suggested that a BH3-only protein other than Bim might participate in apoptosis induced by doxorubicin. Using a BH3-mimetic, ABT-263, to inhibit Bcl2 or Bcl-xl produced a limited apoptotic response in U2OS and MG63 cells, suggesting that this BH3-mimetic cannot activate the Bax/Bak pathway efficiently. Significantly, ABT-263 enhanced doxorubicin- and bortezomib-induced apoptosis synergistically in U2OS and MG63 cells. These results implied that the severe cellular stress caused by doxorubicin or bortezomib might be mediated through a dual process to control apoptosis. Respectively, doxorubicin or bortezomib activates a BH3-only protein in one way and corresponding unknown factors in another way to affect mitochondrial outer membrane permeability, resulting in apoptosis. The combination of doxorubicin with ABT-263 could produce synergistic apoptosis in MG63 cells, which lack p53, suggesting that p53 has no role in doxorubicin-induced apoptosis in osteosarcoma. In addition, ABT-263 enhanced paclitaxel to induce moderate levels of apoptosis.
Collapse
Affiliation(s)
- Cheng-Jung Ho
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Huey-Jiun Ko
- Department of Biochemistry & Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Tzu-Shao Liao
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Xiang-Ren Zheng
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Po-Hsu Chou
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Li-Ting Wang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Ru-Wei Lin
- Department of Plant Industry, National Pingtung University of Science and Technology, Pingtung, 91201, Taiwan
| | - Chung-Hwan Chen
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
| | - Chihuei Wang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
18
|
Aouadi E, Fornier M, Gosseye A, Castillo-Ferrer C, Frachet V. [Separase, a key-player of mitosis: A new target for cancer therapy?]. Med Sci (Paris) 2021; 37:684-686. [PMID: 34180834 DOI: 10.1051/medsci/2021086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Elyes Aouadi
- Master 2 Sciences du vivant, Parcours IMaGHE, Université Paris, Sciences et Lettres (PSL), École pratique des hautes études (EPHE), 75014 Paris, France
| | - Marie Fornier
- Master 2 Sciences du vivant, Parcours IMaGHE, Université Paris, Sciences et Lettres (PSL), École pratique des hautes études (EPHE), 75014 Paris, France
| | - Axel Gosseye
- Master 2 Sciences du vivant, Parcours IMaGHE, Université Paris, Sciences et Lettres (PSL), École pratique des hautes études (EPHE), 75014 Paris, France
| | - Camila Castillo-Ferrer
- Institut pour l'avancée des biosciences, Inserm U1209, UMR CNRS 5309, université Grenoble Alpes, 38700 La Tronche, France. - EPHE, université PSL, 4-14 rue Ferrus, 75014 Paris, France
| | - Véronique Frachet
- Institut pour l'avancée des biosciences, Inserm U1209, UMR CNRS 5309, université Grenoble Alpes, 38700 La Tronche, France. - EPHE, université PSL, 4-14 rue Ferrus, 75014 Paris, France
| |
Collapse
|
19
|
Gryaznova Y, Keating L, Touati SA, Cladière D, El Yakoubi W, Buffin E, Wassmann K. Kinetochore individualization in meiosis I is required for centromeric cohesin removal in meiosis II. EMBO J 2021; 40:e106797. [PMID: 33644892 PMCID: PMC8013791 DOI: 10.15252/embj.2020106797] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022] Open
Abstract
Partitioning of the genome in meiosis occurs through two highly specialized cell divisions, named meiosis I and meiosis II. Step-wise cohesin removal is required for chromosome segregation in meiosis I, and sister chromatid segregation in meiosis II. In meiosis I, mono-oriented sister kinetochores appear as fused together when examined by high-resolution confocal microscopy, whereas they are clearly separated in meiosis II, when attachments are bipolar. It has been proposed that bipolar tension applied by the spindle is responsible for the physical separation of sister kinetochores, removal of cohesin protection, and chromatid separation in meiosis II. We show here that this is not the case, and initial separation of sister kinetochores occurs already in anaphase I independently of bipolar spindle forces applied on sister kinetochores, in mouse oocytes. This kinetochore individualization depends on separase cleavage activity. Crucially, without kinetochore individualization in meiosis I, bivalents when present in meiosis II oocytes separate into chromosomes and not sister chromatids. This shows that whether centromeric cohesin is removed or not is determined by the kinetochore structure prior to meiosis II.
Collapse
Affiliation(s)
- Yulia Gryaznova
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| | - Leonor Keating
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| | - Sandra A Touati
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| | - Damien Cladière
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| | - Warif El Yakoubi
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
- Present address:
Cell and Developmental Biology CenterNational Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Eulalie Buffin
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| | - Katja Wassmann
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| |
Collapse
|
20
|
Chen S, Wu Y, Qin X, Wen P, Liu J, Yang M. Global gene expression analysis using RNA-seq reveals the new roles of Panax notoginseng Saponins in ischemic cardiomyocytes. JOURNAL OF ETHNOPHARMACOLOGY 2021; 268:113639. [PMID: 33301914 DOI: 10.1016/j.jep.2020.113639] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 11/10/2020] [Accepted: 11/23/2020] [Indexed: 05/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax notoginseng saponins (PNS), the main active ingredients of Panax notoginseng (Burkill) F.H.Chen, have been clinically used for cardiovascular diseases treatment in China as the Traditional Chinese Medicine (TCM) (Duan et al., 2017). Evidence demonstrated that PNS protected cardiomyocytes from myocardial ischemia, but the more underlying molecular mechanisms of the protective effect are still unclear. The aims of this study are to systematically know the function of PNS and discover new roles of PNS in ischemic cardiomyocytes. MATERIALS AND METHODS To confirm PNS function on ischemic cardiomyopathy, we established in vitro myocardial ischemia model on H9C2 cardiomyocyte line, which was induced by oxygen-glucose depletion (OGD). Then RNA-seq was carried out to systematically analyze global gene expression. This study was aimed to systematically investigate the protective effect and more potential molecular mechanisms of PNS on H9C2 cardiomyocytes in vitro through whole-transcriptome analysis with total RNA sequencing (RNA-Seq). RESULTS PNS exhibited anti-apoptotic effect in H9C2 cardiomyocytes in OGD-induced myocardial ischemia model. Through RNA-seq, we found that OGD affected expression profiling of many genes, including upregulated and downregulated genes. PNS inhibited cardiomyocyte apoptosis and death through rescuing cell cycle arrest, the DNA double-strand breakage repair process and chromosome segregation. Interestingly, for the canonical signaling pathways regulation, RNA-seq showed PNS could inhibit cardiac hypertrophy, MAPK signaling pathway, and re-activate PI3K/AKT and AMPK signaling pathways. Experimental data also confirmed the PNS could protect cardiomyocytes from OGD-induced apoptosis through activating PI3K/AKT and AMPK signaling pathways. Moreover, RNA-seq demonstrated that the expression levels of many non-coding RNAs, such as miRNAs and lncRNAs, were significantly affected after PNS treatment, suggesting that PNS could protect cardiomyocytes through regulating non-coding RNAs. CONCLUSION RNA-seq systematically revealed different novel roles of Panax Notoginseng Saponins (PNS) in protecting cardiomyocytes from apoptosis, induced by myocardial ischemia, through rescuing cell cycle arrest and cardiac hypertrophy, re-activating the DNA double-strand breakage repair process, chromosome segregation, PI3K/Akt and AMPK signaling pathways and regulating non-coding RNAs.
Collapse
Affiliation(s)
- Shaoxian Chen
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China; Research Department of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Yueheng Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China; Research Department of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Xianyu Qin
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Pengju Wen
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Juli Liu
- Department of Pediatrics, Indiana University School of Medicine, 1044 W Walnut St, Indianapolis, 46202, IN, USA.
| | - Min Yang
- Research Department of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
21
|
Du X, Li Q, Yang L, Zeng Q, Wang S, Li Q. Transcriptomic Data Analyses Reveal That Sow Fertility-Related lincRNA NORFA Is Essential for the Normal States and Functions of Granulosa Cells. Front Cell Dev Biol 2021; 9:610553. [PMID: 33708768 PMCID: PMC7940361 DOI: 10.3389/fcell.2021.610553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 01/25/2021] [Indexed: 12/13/2022] Open
Abstract
NORFA, the first lincRNA associated with sow fertility, has been shown to control granulosa cell (GC) functions and follicular atresia. However, the underlying mechanism is not fully understood. In this study, RNA-seq was performed and we noticed that inhibition of NORFA led to dramatic transcriptomic alterations in porcine GCs. A total of 1,272 differentially expressed transcripts were identified, including 1167 DEmRNAs and 105 DEmiRNAs. Furthermore, protein-protein interaction, gene-pathway function, and TF-miRNA-mRNA regulatory networks were established and yielded four regulatory modules with multiple hub genes, such as AR, ATG5, BAK1, CENPE, NR5A1, NFIX, WNT5B, ssc-miR-27b, and ssc-miR-126. Functional assessment showed that these hub DEGs were mainly enriched in TGF-β, PI3K-Akt, FoxO, Wnt, MAPK, and ubiquitin pathways that are essential for GC states (apoptosis and proliferation) and functions (hormone secretion). In vitro, we also found that knockdown of NORFA in porcine GCs significantly induced cell apoptosis, impaired cell viability, and suppressed 17β-estradiol (E2) synthesis. Notably, four candidate genes for sow reproductive traits (INHBA, NCOA1, TGFβ-1, and TGFBR2) were also identified as potential targets of NORFA. These findings present a panoramic view of the transcriptome in NORFA-reduced GCs, highlighting that NORFA, a candidate lincRNA for sow fertility, is crucial for the normal states and functions of GCs.
Collapse
Affiliation(s)
- Xing Du
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | | | | | | | | | - Qifa Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
22
|
Abstract
Separase is a large cysteine protease in eukaryotes and has crucial roles in many cellular processes, especially chromosome segregation during mitosis and meiosis, apoptosis, DNA damage repair, centrosome disengagement and duplication, spindle stabilization and elongation. It dissolves the cohesion between sister chromatids by cleaving one of the subunits of the cohesin ring for chromosome segregation. The activity of separase is tightly controlled at many levels, through direct binding of inhibitory proteins as well as posttranslational modification. Dysregulation of separase activity is linked to cancer and genome instability, making it a target for drug discovery. One of the best-known inhibitors of separase is securin, which has been identified in yeast, plants, and animals. Securin forms a tight complex with separase and potently inhibits its catalytic activity. Recent structures of the separase-securin complex have revealed the molecular mechanism for the inhibitory activity of securin. A segment of securin is bound in the active site of separase, thereby blocking substrate binding. Securin itself is not cleaved by separase as its binding mode is not compatible with catalysis. Securin also has extensive interactions with separase outside the active site, consistent with its function as a chaperone to stabilize this enzyme.
Collapse
Affiliation(s)
- Shukun Luo
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Liang Tong
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
23
|
Gliech CR, Holland AJ. Keeping track of time: The fundamentals of cellular clocks. J Cell Biol 2020; 219:e202005136. [PMID: 32902596 PMCID: PMC7594491 DOI: 10.1083/jcb.202005136] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
Biological timekeeping enables the coordination and execution of complex cellular processes such as developmental programs, day/night organismal changes, intercellular signaling, and proliferative safeguards. While these systems are often considered separately owing to a wide variety of mechanisms, time frames, and outputs, all clocks are built by calibrating or delaying the rate of biochemical reactions and processes. In this review, we explore the common themes and core design principles of cellular clocks, giving special consideration to the challenges associated with building timers from biochemical components. We also outline how evolution has coopted time to increase the reliability of a diverse range of biological systems.
Collapse
Affiliation(s)
| | - Andrew J. Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
24
|
Mcl-1 and Bok transmembrane domains: Unexpected players in the modulation of apoptosis. Proc Natl Acad Sci U S A 2020; 117:27980-27988. [PMID: 33093207 DOI: 10.1073/pnas.2008885117] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The Bcl-2 protein family comprises both pro- and antiapoptotic members that control the permeabilization of the mitochondrial outer membrane, a crucial step in the modulation of apoptosis. Recent research has demonstrated that the carboxyl-terminal transmembrane domain (TMD) of some Bcl-2 protein family members can modulate apoptosis; however, the transmembrane interactome of the antiapoptotic protein Mcl-1 remains largely unexplored. Here, we demonstrate that the Mcl-1 TMD forms homooligomers in the mitochondrial membrane, competes with full-length Mcl-1 protein with regards to its antiapoptotic function, and induces cell death in a Bok-dependent manner. While the Bok TMD oligomers locate preferentially to the endoplasmic reticulum (ER), heterooligomerization between the TMDs of Mcl-1 and Bok predominantly takes place at the mitochondrial membrane. Strikingly, the coexpression of Mcl-1 and Bok TMDs produces an increase in ER mitochondrial-associated membranes, suggesting an active role of Mcl-1 in the induced mitochondrial targeting of Bok. Finally, the introduction of Mcl-1 TMD somatic mutations detected in cancer patients alters the TMD interaction pattern to provide the Mcl-1 protein with enhanced antiapoptotic activity, thereby highlighting the clinical relevance of Mcl-1 TMD interactions.
Collapse
|
25
|
Trotter EW, Hagan IM. Release from cell cycle arrest with Cdk4/6 inhibitors generates highly synchronized cell cycle progression in human cell culture. Open Biol 2020; 10:200200. [PMID: 33052073 PMCID: PMC7653349 DOI: 10.1098/rsob.200200] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 10/02/2020] [Indexed: 12/18/2022] Open
Abstract
Each approach used to synchronize cell cycle progression of human cell lines presents a unique set of challenges. Induction synchrony with agents that transiently block progression through key cell cycle stages are popular, but change stoichiometries of cell cycle regulators, invoke compensatory changes in growth rate and, for DNA replication inhibitors, damage DNA. The production, replacement or manipulation of a target molecule must be exceptionally rapid if the interpretation of phenotypes in the cycle under study is to remain independent of impacts upon progression through the preceding cycle. We show how these challenges are avoided by exploiting the ability of the Cdk4/6 inhibitors, palbociclib, ribociclib and abemaciclib to arrest cell cycle progression at the natural control point for cell cycle commitment: the restriction point. After previous work found no change in the coupling of growth and division during recovery from CDK4/6 inhibition, we find high degrees of synchrony in cell cycle progression. Although we validate CDK4/6 induction synchronization with hTERT-RPE-1, A549, THP1 and H1299, it is effective in other lines and avoids the DNA damage that accompanies synchronization by thymidine block/release. Competence to return to cycle after 72 h arrest enables out of cycle target induction/manipulation, without impacting upon preceding cycles.
Collapse
Affiliation(s)
| | - Iain Michael Hagan
- Cell Division Group, CRUK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, UK
| |
Collapse
|