1
|
Kong ASY, Tan YC, Thew HY, Lai KS, Lim SHE, Maran S, Loh HS. In-silico analysis of nsSNPs in BCL-2 family proteins: Implications for colorectal cancer pathogenesis and therapeutics. Biochem Biophys Rep 2025; 42:101957. [PMID: 40207085 PMCID: PMC11979393 DOI: 10.1016/j.bbrep.2025.101957] [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: 12/09/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 04/11/2025] Open
Abstract
Colorectal cancer (CRC) is a multifaceted disease characterized by abnormal cell proliferation in the colon and rectum. The BCL-2 family proteins are implicated in CRC pathogenesis, yet the impacts of genetic variations within these proteins remains elusive. This in-silico study employs diverse sequence- and structure-based bioinformatics tools to identify potentially pathogenic nonsynonymous single nucleotide polymorphisms (nsSNPs) in BCL-2 family proteins. Leveraging computational tools including SIFT, PolyPhen-2, SNPs&GO, PhD-SNP, PANTHER, and Condel, 94 nsSNPs were predicted as deleterious, damaging, and disease-associated by at least five tools. Stability analysis with I-Mutant2.0, MutPred, and PredictSNP further identified 31 nsSNPs that reduce protein stability. Conservation analysis highlighted highly functional, exposed variants (rs960653284, rs758817904, rs1466732626, rs569276903, rs746711568, rs764437421, rs779690846, and rs2038330314) and structural, buried variants (rs376149674, rs1375767408, rs1582066443, rs367558446, rs367558446, rs1319541919, and rs1370070128). To explore the functional effects of these mutations, molecular docking and molecular dynamics simulations were conducted. G233D (rs376149674) and R12G (rs960653284) mutations in the BCL2 protein exhibited the greatest differences in docking scores with d-α-Tocopherol and Tocotrienol, suggesting enhanced protein-ligand interactions. The simulations revealed that d-α-Tocopherol and Tocotrienol (strong binders) contributed to greater stability of BCL-2 family proteins, while Fluorouracil, though weaker, still demonstrated selective binding stability. This work represents the first comprehensive computational analysis of functional nsSNPs in BCL-2 family proteins, providing insights into their roles in CRC pathogenesis. While these findings demand experimental validation, they hold great promise for guiding future large-scale population studies, facilitating drug repurposing efforts, and advancing the development of targeted diagnostic and therapeutic modalities for CRC.
Collapse
Affiliation(s)
- Amanda Shen-Yee Kong
- School of Biosciences, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor Darul Ehsan, Malaysia
| | - Yong Chiang Tan
- International Medical University, 57000, Kuala Lumpur, Federal Territory of Kuala Lumpur, Malaysia
| | - Hin-Yee Thew
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Kok-Song Lai
- Health Sciences Division, Abu Dhabi Women's College, Higher Colleges of Technology, Abu Dhabi, 41012, United Arab Emirates
| | - Swee-Hua Erin Lim
- Health Sciences Division, Abu Dhabi Women's College, Higher Colleges of Technology, Abu Dhabi, 41012, United Arab Emirates
| | - Sathiya Maran
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Hwei-San Loh
- School of Biosciences, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
2
|
Horkan HR, Popgeorgiev N, Vervoort M, Gazave E, Krasovec G. Evolution of Apoptotic Signaling Pathways Within Lophotrochozoans. Genome Biol Evol 2024; 16:evae204. [PMID: 39318156 PMCID: PMC11463336 DOI: 10.1093/gbe/evae204] [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: 12/13/2023] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/26/2024] Open
Abstract
Apoptosis is the main form of regulated cell death in metazoans. Apoptotic pathways are well characterized in nematodes, flies, and mammals, leading to a vision of the conservation of apoptotic pathways in metazoans. However, we recently showed that intrinsic apoptosis is in fact divergent among metazoans. In addition, extrinsic apoptosis is poorly studied in non-mammalian animals, making its evolution unclear. Consequently, our understanding of apoptotic signaling pathways evolution is a black box which must be illuminated by extending research to new biological systems. Lophotrochozoans are a major clade of metazoans which, despite their considerable biological diversity and key phylogenetic position as sister group of ecdysozoans (i.e. flies and nematodes), are poorly explored, especially regarding apoptosis mechanisms. Traditionally, each apoptotic signaling pathway was considered to rely on a specific initiator caspase, associated with an activator. To shed light on apoptosis evolution in animals, we explored the evolutionary history of initiator caspases, caspase activators, and the BCL-2 family (which control mitochondrial apoptotic pathway) in lophotrochozoans using phylogenetic analysis and protein interaction predictions. We discovered a diversification of initiator caspases in molluscs, annelids, and brachiopods, and the loss of key extrinsic apoptosis components in platyhelminths, along with the emergence of a clade-specific caspase with an ankyrin pro-domain. Taken together, our data show a specific history of apoptotic actors' evolution in lophotrochozoans, further demonstrating the appearance of distinct apoptotic signaling pathways during metazoan evolution.
Collapse
Affiliation(s)
- Helen R Horkan
- Centre for Chromosome Biology, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Nikolay Popgeorgiev
- Centre de Recherche en Cancérologie de Lyon, U1052 INSERM, UMR CNRS 5286, Centre Léon Bérard, Université Claude Bernard Lyon 1, Lyon, France
- Institut Universitaire de France (IUF), Paris, France
| | - Michel Vervoort
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Eve Gazave
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Gabriel Krasovec
- Centre for Chromosome Biology, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| |
Collapse
|
3
|
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
|
4
|
Dehghan S, Kheshtchin N, Hassannezhad S, Soleimani M. Cell death classification: A new insight based on molecular mechanisms. Exp Cell Res 2023; 433:113860. [PMID: 38013091 DOI: 10.1016/j.yexcr.2023.113860] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/17/2023] [Accepted: 11/18/2023] [Indexed: 11/29/2023]
Abstract
Cells tend to disintegrate themselves or are forced to undergo such destructive processes in critical circumstances. This complex cellular function necessitates various mechanisms and molecular pathways in order to be executed. The very nature of cell death is essentially important and vital for maintaining homeostasis, thus any type of disturbing occurrence might lead to different sorts of diseases and dysfunctions. Cell death has various modalities and yet, every now and then, a new type of this elegant procedure gets to be discovered. The diversity of cell death compels the need for a universal organizing system in order to facilitate further studies, therapeutic strategies and the invention of new methods of research. Considering all that, we attempted to review most of the known cell death mechanisms and sort them all into one arranging system that operates under a simple but subtle decision-making (If \ Else) order as a sorting algorithm, in which it decides to place and sort an input data (a type of cell death) into its proper set, then a subset and finally a group of cell death. By proposing this algorithm, the authors hope it may solve the problems regarding newer and/or undiscovered types of cell death and facilitate research and therapeutic applications of cell death.
Collapse
Affiliation(s)
- Sepehr Dehghan
- Department of Medical Basic Sciences, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Nasim Kheshtchin
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Maryam Soleimani
- Department of Medical Basic Sciences, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Kulkarni M, Hardwick JM. Programmed Cell Death in Unicellular Versus Multicellular Organisms. Annu Rev Genet 2023; 57:435-459. [PMID: 37722687 PMCID: PMC11491101 DOI: 10.1146/annurev-genet-033123-095833] [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] [Indexed: 09/20/2023]
Abstract
Programmed cell death (self-induced) is intrinsic to all cellular life forms, including unicellular organisms. However, cell death research has focused on animal models to understand cancer, degenerative disorders, and developmental processes. Recently delineated suicidal death mechanisms in bacteria and fungi have revealed ancient origins of animal cell death that are intertwined with immune mechanisms, allaying earlier doubts that self-inflicted cell death pathways exist in microorganisms. Approximately 20 mammalian death pathways have been partially characterized over the last 35 years. By contrast, more than 100 death mechanisms have been identified in bacteria and a few fungi in recent years. However, cell death is nearly unstudied in most human pathogenic microbes that cause major public health burdens. Here, we consider how the current understanding of programmed cell death arose through animal studies and how recently uncovered microbial cell death mechanisms in fungi and bacteria resemble and differ from mechanisms of mammalian cell death.
Collapse
Affiliation(s)
- Madhura Kulkarni
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA; ,
| | - J Marie Hardwick
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA; ,
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
6
|
Gao P, Zhang Z, Wang R, Huang L, Wu H, Qiao Z, Wang X, Jin H, Peng J, Liu L, Chen Q, Lin J. Structure-destabilizing mutations unleash an intrinsic perforation activity of antiapoptotic Bcl-2 in the mitochondrial membrane enabling apoptotic cell death. MITOCHONDRIAL COMMUNICATIONS 2023; 1:48-61. [PMID: 39239250 PMCID: PMC11375749 DOI: 10.1016/j.mitoco.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Bcl-2 and Bax share a similar structural fold in solution, yet function oppositely in the mitochondrial outer membrane (MOM) during apoptosis. The proapoptotic Bax forms pores in the MOM to trigger cell death, whereas Bcl-2 inhibits the Bax pore formation to prevent cell death. Intriguingly both proteins can switch to a similar conformation after activation by BH3-only proteins, with multiple regions embedded in the MOM. Here we tested a hypothesis that destabilization of the Bcl-2 structure might convert Bcl-2 to a Bax-like perforator. We discovered that mutations of glutamate 152 which eliminate hydrogen bonds in the protein core and thereby reduce the Bcl-2 structural stability. These Bcl-2 mutants induced apoptosis by releasing cytochrome c from the mitochondria in the cells that lack Bax and Bak, the other proapoptotic perforator. Using liposomal membranes made with typical mitochondrial lipids and reconstituted with purified proteins we revealed this perforation activity was intrinsic to Bcl-2 and could be unleashed by a BH3-only protein, similar to the perforation activity of Bax. Our study thus demonstrated a structural conversion of antiapoptotic Bcl-2 to a proapoptotic perforator through a simple molecular manipulation or interaction that is worthy to explore further for eradicating cancer cells that are resistant to a current Bcl-2-targeting drug.
Collapse
Affiliation(s)
- Ping Gao
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhi Zhang
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73126, USA
| | - Rui Wang
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Li Huang
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hao Wu
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhenzhen Qiao
- College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xiaohui Wang
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Haijing Jin
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jun Peng
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73126, USA
| | - Lei Liu
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Quan Chen
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jialing Lin
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73126, USA
- Stephenson Cancer Center, Oklahoma City, OK, 73104, USA
| |
Collapse
|
7
|
Mentel M, Illová M, Krajčovičová V, Kroupová G, Mannová Z, Chovančíková P, Polčic P. Yeast Bax Inhibitor (Bxi1p/Ybh3p) Is Not Required for the Action of Bcl-2 Family Proteins on Cell Viability. Int J Mol Sci 2023; 24:12011. [PMID: 37569387 PMCID: PMC10419234 DOI: 10.3390/ijms241512011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Permeabilization of mitochondrial membrane by proteins of the BCL-2 family is a key decisive event in the induction of apoptosis in mammalian cells. Although yeast does not have homologs of the BCL-2 family, when these are expressed in yeast, they modulate the survival of cells in a way that corresponds to their activity in mammalian cells. The yeast gene, alternatively referred to as BXI1 or YBH3, encodes for membrane protein in the endoplasmic reticulum that was, contradictorily, shown to either inhibit Bax or to be required for Bax activity. We have tested the effect of the deletion of this gene on the pro-apoptotic activity of Bax and Bak and the anti-apoptotic activity of Bcl-XL and Bcl-2, as well on survival after treatment with inducers of regulated cell death in yeast, hydrogen peroxide and acetic acid. While deletion resulted in increased sensitivity to acetic acid, it did not affect the sensitivity to hydrogen peroxide nor to BCL-2 family members. Thus, our results do not support any model in which the activity of BCL-2 family members is directly affected by BXI1 but rather indicate that it may participate in modulating survival in response to some specific forms of stress.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Peter Polčic
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava, Mlynská Dolina CH1, Ilkovičova 6, 84215 Bratislava, Slovakia
| |
Collapse
|
8
|
Hartman ML, Czyz M. BCL-G: 20 years of research on a non-typical protein from the BCL-2 family. Cell Death Differ 2023:10.1038/s41418-023-01158-5. [PMID: 37031274 DOI: 10.1038/s41418-023-01158-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/10/2023] Open
Abstract
Proteins from the BCL-2 family control cell survival and apoptosis in health and disease, and regulate apoptosis-unrelated cellular processes. BCL-Gonad (BCL-G, also known as BCL2-like 14) is a non-typical protein of the family as its long isoform (BCL-GL) consists of BH2 and BH3 domains without the BH1 motif. BCL-G is predominantly expressed in normal testes and different organs of the gastrointestinal tract. The complexity of regulatory mechanisms of BCL-G expression and post-translational modifications suggests that BCL-G may play distinct roles in different types of cells and disorders. While several genetic alterations of BCL2L14 have been reported, gene deletions and amplifications prevail, which is also confirmed by the analysis of sequencing data for different types of cancer. Although the studies validating the phenotypic consequences of genetic manipulations of BCL-G are limited, the role of BCL-G in apoptosis has been undermined. Recent studies using gene-perturbation approaches have revealed apoptosis-unrelated functions of BCL-G in intracellular trafficking, immunomodulation, and regulation of the mucin scaffolding network. These studies were, however, limited mainly to the role of BCL-G in the gastrointestinal tract. Therefore, further efforts using state-of-the-art methods and various types of cells are required to find out more about BCL-G activities. Deciphering the isoform-specific functions of BCL-G and the BCL-G interactome may result in the designing of novel therapeutic approaches, in which BCL-G activity will be either imitated using small-molecule BH3 mimetics or inhibited to counteract BCL-G upregulation. This review summarizes two decades of research on BCL-G.
Collapse
Affiliation(s)
- Mariusz L Hartman
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215, Lodz, Poland.
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215, Lodz, Poland
| |
Collapse
|
9
|
Aguilar F, Yu S, Grant RA, Swanson S, Ghose D, Su BG, Sarosiek KA, Keating AE. Peptides from human BNIP5 and PXT1 and non-native binders of pro-apoptotic BAK can directly activate or inhibit BAK-mediated membrane permeabilization. Structure 2023; 31:265-281.e7. [PMID: 36706751 PMCID: PMC9992319 DOI: 10.1016/j.str.2023.01.001] [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: 08/30/2022] [Revised: 11/24/2022] [Accepted: 01/02/2023] [Indexed: 01/27/2023]
Abstract
Apoptosis is important for development and tissue homeostasis, and its dysregulation can lead to diseases, including cancer. As an apoptotic effector, BAK undergoes conformational changes that promote mitochondrial outer membrane disruption, leading to cell death. This is termed "activation" and can be induced by peptides from the human proteins BID, BIM, and PUMA. To identify additional peptides that can regulate BAK, we used computational protein design, yeast surface display screening, and structure-based energy scoring to identify 10 diverse new binders. We discovered peptides from the human proteins BNIP5 and PXT1 and three non-native peptides that activate BAK in liposome assays and induce cytochrome c release from mitochondria. Crystal structures and binding studies reveal a high degree of similarity among peptide activators and inhibitors, ruling out a simple function-determining property. Our results shed light on the vast peptide sequence space that can regulate BAK function and will guide the design of BAK-modulating tools and therapeutics.
Collapse
Affiliation(s)
- Fiona Aguilar
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Stacey Yu
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Program in Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, Boston, MA, USA; John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Robert A Grant
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sebastian Swanson
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dia Ghose
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bonnie G Su
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kristopher A Sarosiek
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Program in Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, Boston, MA, USA; John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Amy E Keating
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
10
|
Moldoveanu T. Apoptotic mitochondrial poration by a growing list of pore-forming BCL-2 family proteins. Bioessays 2023; 45:e2200221. [PMID: 36650950 PMCID: PMC9975053 DOI: 10.1002/bies.202200221] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023]
Abstract
The pore-forming BCL-2 family proteins are effectors of mitochondrial poration in apoptosis initiation. Two atypical effectors-BOK and truncated BID (tBID)-join the canonical effectors BAK and BAX. Gene knockout revealed developmental phenotypes in the absence the effectors, supporting their roles in vivo. During apoptosis effectors are activated and change shape from dormant monomers to dynamic oligomers that associate with and permeabilize mitochondria. BID is activated by proteolysis, BOK accumulates on inhibition of its degradation by the E3 ligase gp78, while BAK and BAX undergo direct activation by BH3-only initiators, autoactivation, and crossactivation. Except tBID, effector oligomers on the mitochondria appear as arcs and rings in super-resolution microscopy images. The BH3-in-groove dimers of BAK and BAX, the tBID monomers, and uncharacterized BOK species are the putative building blocks of apoptotic pores. Effectors interact with lipids and bilayers but the mechanism of membrane poration remains elusive. I discuss effector-mediated mitochondrial poration.
Collapse
Affiliation(s)
- Tudor Moldoveanu
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences,Correspondence:
| |
Collapse
|
11
|
Ikegawa Y, Combet C, Groussin M, Navratil V, Safar-Remali S, Shiota T, Aouacheria A, Yoo SK. Evidence for existence of an apoptosis-inducing BH3-only protein, sayonara, in Drosophila. EMBO J 2023; 42:e110454. [PMID: 36727601 PMCID: PMC10107002 DOI: 10.15252/embj.2021110454] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 02/03/2023] Open
Abstract
Cells need to sense stresses to initiate the execution of the dormant cell death program. Since the discovery of the first BH3-only protein Bad, BH3-only proteins have been recognized as indispensable stress sensors that induce apoptosis. BH3-only proteins have so far not been identified in Drosophila despite their importance in other organisms. Here, we identify the first Drosophila BH3-only protein and name it sayonara. Sayonara induces apoptosis in a BH3 motif-dependent manner and interacts genetically and biochemically with the BCL-2 homologous proteins, Buffy and Debcl. There is a positive feedback loop between Sayonara-mediated caspase activation and autophagy. The BH3 motif of sayonara phylogenetically appeared at the time of the ancestral gene duplication that led to the formation of Buffy and Debcl in the dipteran lineage. To our knowledge, this is the first identification of a bona fide BH3-only protein in Drosophila, thus providing a unique example of how cell death mechanisms can evolve both through time and across taxa.
Collapse
Affiliation(s)
- Yuko Ikegawa
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Christophe Combet
- Centre de Recherche en Cancérologie de Lyon, UMR Inserm U1052, CNRS 5286, Université Claude Bernard Lyon 1, Centre Léon Bérard, Lyon, France
| | - Mathieu Groussin
- Laboratoire de Biométrie et Biologie Evolutive, Université de Lyon, Université Claude Bernard Lyon 1, CNRS, Villeurbanne, France.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Vincent Navratil
- PRABI, Rhône-Alpes Bioinformatics Center, Université Lyon 1, Villeurbanne, France.,UMS 3601, Institut Français de Bioinformatique, IFB-Core, Évry, France
| | - Sabrina Safar-Remali
- Centre de Recherche en Cancérologie de Lyon, UMR Inserm U1052, CNRS 5286, Université Claude Bernard Lyon 1, Centre Léon Bérard, Lyon, France
| | - Takuya Shiota
- Organization for Promotion of Tenure Track, University of Miyazaki, Miyazaki, Japan.,Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| | - Abdel Aouacheria
- ISEM, Institut des Sciences de l'Evolution de Montpellier, UMR 5554, CNRS, IRD, EPHE, Université de Montpellier, Montpellier, France
| | - Sa Kan Yoo
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, Japan.,Physiological Genetics Laboratory, RIKEN CPR, Kobe, Japan
| |
Collapse
|
12
|
McGriff A, Placzek WJ. Phylogenetic analysis of the MCL1 BH3 binding groove and rBH3 sequence motifs in the p53 and INK4 protein families. PLoS One 2023; 18:e0277726. [PMID: 36696417 PMCID: PMC9876281 DOI: 10.1371/journal.pone.0277726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/10/2023] [Indexed: 01/26/2023] Open
Abstract
B-cell lymphoma 2 (Bcl-2) proteins are central, conserved regulators of apoptosis. Bcl-2 family function is regulated by binding interactions between the Bcl-2 homology 3 (BH3) motif in pro-apoptotic family members and the BH3 binding groove found in both the pro-apoptotic effector and anti-apoptotic Bcl-2 family members. A novel motif, the reverse BH3 (rBH3), has been shown to interact with the anti-apoptotic Bcl-2 homolog MCL1 (Myeloid cell leukemia 1) and have been identified in the p53 homolog p73, and the CDK4/6 (cyclin dependent kinase 4/6) inhibitor p18INK4c, (p18, cyclin-dependent kinase 4 inhibitor c). To determine the conservation of rBH3 motif, we first assessed conservation of MCL1's BH3 binding groove, where the motif binds. We then constructed neighbor-joining phylogenetic trees of the INK4 and p53 protein families and analyzed sequence conservation using sequence logos of the rBH3 locus. This showed the rBH3 motif is conserved throughout jawed vertebrates p63 and p73 sequences and in chondrichthyans, amphibians, mammals, and some reptiles in p18. Finally, a potential rBH3 motif was identified in mammalian and osteichthyan p19INK4d (p19, cyclin dependent kinase 4 inhibitor d). These findings demonstrate that the interaction between MCL1 and other cellular proteins mediated by the rBH3 motif may be conserved throughout jawed vertebrates.
Collapse
Affiliation(s)
- Anna McGriff
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - William J. Placzek
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
13
|
Simonyan L, Gonin M, Hanks J, Friedlein J, Dutrec K, Arokium H, Rouchidane Eyitayo A, Doudy TM, Chaignepain S, Manon S, Dejean L. Non-phosphorylatable mutants of Ser184 lead to incomplete activation of Bax. Front Oncol 2023; 12:1068994. [PMID: 36741728 PMCID: PMC9892840 DOI: 10.3389/fonc.2022.1068994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/15/2022] [Indexed: 01/20/2023] Open
Abstract
The S184 residue of Bax is the target of several protein kinases regulating cell fate, including AKT. It is well-established that, in cellulo, the substitution of S184 by a non-phosphorylatable residue stimulates both the mitochondrial localization of Bax, cytochrome c release, and apoptosis. However, in in vitro experiments, substituted mutants did not exhibit any increase in their binding capacity to isolated mitochondria or liposomes. Despite exhibiting a significant increase of the 6A7 epitope exposure, substituted mutants remain limited in their ability to form large oligomers, suggesting that they high capacity to promote apoptosis in cells was more related to a high content than to an increased ability to form large pores in the outer mitochondrial membranes.
Collapse
Affiliation(s)
- Lilit Simonyan
- Université de Bordeaux, Centre National de la Recherche Scientifique (CNRS), Institut de Biochimie et de Génétique Cellulaires (IBGC), Bordeaux, France
| | - Mathilde Gonin
- Université de Bordeaux, Centre National de la Recherche Scientifique (CNRS), Institut de Biochimie et de Génétique Cellulaires (IBGC), Bordeaux, France
| | - James Hanks
- California State University of Fresno, Department of Chemistry and Biochemistry, Fresno, CA, United States
| | - Jordan Friedlein
- California State University of Fresno, Department of Chemistry and Biochemistry, Fresno, CA, United States
| | - Kevin Dutrec
- Université de Bordeaux, Centre National de la Recherche Scientifique (CNRS), Institut de Biochimie et de Génétique Cellulaires (IBGC), Bordeaux, France
| | - Hubert Arokium
- Université de Bordeaux, Centre National de la Recherche Scientifique (CNRS), Institut de Biochimie et de Génétique Cellulaires (IBGC), Bordeaux, France
| | - Akandé Rouchidane Eyitayo
- Université de Bordeaux, Centre National de la Recherche Scientifique (CNRS), Institut de Biochimie et de Génétique Cellulaires (IBGC), Bordeaux, France
| | - Toukounou Megann Doudy
- Université de Bordeaux, CNRS, Centre de Génomique Fonctionnelle Bordeaux (CGFB), Bordeaux, France
| | - Stéphane Chaignepain
- Université de Bordeaux, CNRS, Centre de Génomique Fonctionnelle Bordeaux (CGFB), Bordeaux, France
| | - Stéphen Manon
- Université de Bordeaux, Centre National de la Recherche Scientifique (CNRS), Institut de Biochimie et de Génétique Cellulaires (IBGC), Bordeaux, France,*Correspondence: Stéphen Manon, ; Laurent Dejean,
| | - Laurent Dejean
- California State University of Fresno, Department of Chemistry and Biochemistry, Fresno, CA, United States,*Correspondence: Stéphen Manon, ; Laurent Dejean,
| |
Collapse
|
14
|
Fasano C, Grossi V, Forte G, Simone C. Short Linear Motifs in Colorectal Cancer Interactome and Tumorigenesis. Cells 2022; 11:3739. [PMID: 36496998 PMCID: PMC9737320 DOI: 10.3390/cells11233739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Colorectal tumorigenesis is driven by alterations in genes and proteins responsible for cancer initiation, progression, and invasion. This multistage process is based on a dense network of protein-protein interactions (PPIs) that become dysregulated as a result of changes in various cell signaling effectors. PPIs in signaling and regulatory networks are known to be mediated by short linear motifs (SLiMs), which are conserved contiguous regions of 3-10 amino acids within interacting protein domains. SLiMs are the minimum sequences required for modulating cellular PPI networks. Thus, several in silico approaches have been developed to predict and analyze SLiM-mediated PPIs. In this review, we focus on emerging evidence supporting a crucial role for SLiMs in driver pathways that are disrupted in colorectal cancer (CRC) tumorigenesis and related PPI network alterations. As a result, SLiMs, along with short peptides, are attracting the interest of researchers to devise small molecules amenable to be used as novel anti-CRC targeted therapies. Overall, the characterization of SLiMs mediating crucial PPIs in CRC may foster the development of more specific combined pharmacological approaches.
Collapse
Affiliation(s)
- Candida Fasano
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (V.G.); (G.F.)
| | - Valentina Grossi
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (V.G.); (G.F.)
| | - Giovanna Forte
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (V.G.); (G.F.)
| | - Cristiano Simone
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (V.G.); (G.F.)
- Medical Genetics, Department of Precision and Regenerative Medicine and Jonic Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
15
|
Sora V, Papaleo E. Structural Details of BH3 Motifs and BH3-Mediated Interactions: an Updated Perspective. Front Mol Biosci 2022; 9:864874. [PMID: 35685242 PMCID: PMC9171138 DOI: 10.3389/fmolb.2022.864874] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/13/2022] [Indexed: 11/18/2022] Open
Abstract
Apoptosis is a mechanism of programmed cell death crucial in organism development, maintenance of tissue homeostasis, and several pathogenic processes. The B cell lymphoma 2 (BCL2) protein family lies at the core of the apoptotic process, and the delicate balance between its pro- and anti-apoptotic members ultimately decides the cell fate. BCL2 proteins can bind with each other and several other biological partners through the BCL2 homology domain 3 (BH3), which has been also classified as a possible Short Linear Motif and whose distinctive features remain elusive even after decades of studies. Here, we aim to provide an updated overview of the structural features characterizing BH3s and BH3-mediated interactions (with a focus on human proteins), elaborating on the plasticity of BCL2 proteins and the motif properties. We also discussed the implication of these findings for the discovery of interactors of the BH3-binding groove of BCL2 proteins and the design of mimetics for therapeutic purposes.
Collapse
Affiliation(s)
- Valentina Sora
- Cancer Structural Biology, Danish Cancer Society Research Center, Copenhagen, Denmark
- Cancer Systems Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Elena Papaleo
- Cancer Structural Biology, Danish Cancer Society Research Center, Copenhagen, Denmark
- Cancer Systems Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, Kongens Lyngby, Denmark
- *Correspondence: Elena Papaleo, ,
| |
Collapse
|
16
|
Bcl-2 Family Members and the Mitochondrial Import Machineries: The Roads to Death. Biomolecules 2022; 12:biom12020162. [PMID: 35204663 PMCID: PMC8961529 DOI: 10.3390/biom12020162] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 01/27/2023] Open
Abstract
The localization of Bcl-2 family members at the mitochondrial outer membrane (MOM) is a crucial step in the implementation of apoptosis. We review evidence showing the role of the components of the mitochondrial import machineries (translocase of the outer membrane (TOM) and the sorting and assembly machinery (SAM)) in the mitochondrial localization of Bcl-2 family members and how these machineries regulate the function of pro- and anti-apoptotic proteins in resting cells and in cells committed into apoptosis.
Collapse
|
17
|
Pihán P, Lisbona F, Borgonovo J, Edwards-Jorquera S, Nunes-Hasler P, Castillo K, Kepp O, Urra H, Saarnio S, Vihinen H, Carreras-Sureda A, Forveille S, Sauvat A, De Giorgis D, Pupo A, Rodríguez DA, Quarato G, Sagredo A, Lourido F, Letai A, Latorre R, Kroemer G, Demaurex N, Jokitalo E, Concha ML, Glavic Á, Green DR, Hetz C. Control of lysosomal-mediated cell death by the pH-dependent calcium channel RECS1. SCIENCE ADVANCES 2021; 7:eabe5469. [PMID: 34767445 PMCID: PMC8589314 DOI: 10.1126/sciadv.abe5469] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 09/24/2021] [Indexed: 05/27/2023]
Abstract
Programmed cell death is regulated by the balance between activating and inhibitory signals. Here, we have identified RECS1 (responsive to centrifugal force and shear stress 1) [also known as TMBIM1 (transmembrane BAX inhibitor motif containing 1)] as a proapoptotic member of the TMBIM family. In contrast to other proteins of the TMBIM family, RECS1 expression induces cell death through the canonical mitochondrial apoptosis pathway. Unbiased screening indicated that RECS1 sensitizes cells to lysosomal perturbations. RECS1 localizes to lysosomes, where it regulates their acidification and calcium content, triggering lysosomal membrane permeabilization. Structural modeling and electrophysiological studies indicated that RECS1 is a pH-regulated calcium channel, an activity that is essential to trigger cell death. RECS1 also sensitizes whole animals to stress in vivo in Drosophila melanogaster and zebrafish models. Our results unveil an unanticipated function for RECS1 as a proapoptotic component of the TMBIM family that ignites cell death programs at lysosomes.
Collapse
Affiliation(s)
- Philippe Pihán
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Fernanda Lisbona
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Center for Genome Regulation, Faculty of Sciences, University of Chile, Santiago, Chile
| | - Janina Borgonovo
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile
- Program of Integrative Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | | | - Paula Nunes-Hasler
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Hery Urra
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Suvi Saarnio
- Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Helena Vihinen
- Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Amado Carreras-Sureda
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Sabrina Forveille
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Allan Sauvat
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Daniela De Giorgis
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Amaury Pupo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Diego A. Rodríguez
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Giovanni Quarato
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Alfredo Sagredo
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Fernanda Lourido
- Center for Genome Regulation, Faculty of Sciences, University of Chile, Santiago, Chile
| | - Anthony Letai
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Dana Building, Room DA-520, Boston, MA 02215-02115, USA
- Harvard Medical School, Boston, MA 02215, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02215, USA
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
- Karolinska Institutet, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Nicolas Demaurex
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Centro de Investigación de Estudios Avanzados, Universidad Católica del Maule, Talca, Chile
| | - Eija Jokitalo
- Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Miguel L. Concha
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile
- Program of Integrative Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Álvaro Glavic
- Center for Genome Regulation, Faculty of Sciences, University of Chile, Santiago, Chile
| | - Douglas R. Green
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| |
Collapse
|
18
|
Lipids modulate the BH3-independent membrane targeting and activation of BAX and Bcl-xL. Proc Natl Acad Sci U S A 2021; 118:2025834118. [PMID: 34493661 DOI: 10.1073/pnas.2025834118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 07/21/2021] [Indexed: 12/13/2022] Open
Abstract
Regulation of apoptosis is tightly linked with the targeting of numerous Bcl-2 proteins to the mitochondrial outer membrane (MOM), where their activation or inhibition dictates cell death or survival. According to the traditional view of apoptotic regulation, BH3-effector proteins are indispensable for the cytosol-to-MOM targeting and activation of proapoptotic and antiapoptotic members of the Bcl-2 protein family. This view is challenged by recent studies showing that these processes can occur in cells lacking BH3 effectors by as yet to be determined mechanism(s). Here, we exploit a model membrane system that recapitulates key features of MOM to demonstrate that the proapoptotic Bcl-2 protein BAX and antiapoptotic Bcl-xL have an inherent ability to interact with membranes in the absence of BH3 effectors, but only in the presence of cellular concentrations of Mg2+/Ca2+ Under these conditions, BAX and Bcl-xL are selectively targeted to membranes, refolded, and activated in the presence of anionic lipids especially the mitochondrial-specific lipid cardiolipin. These results provide a mechanistic explanation for the mitochondrial targeting and activation of Bcl-2 proteins in cells lacking BH3 effectors. At cytosolic Mg2+ levels, the BH3-independent activation of BAX could provide localized amplification of apoptotic signaling at regions enriched in cardiolipin (e.g., contact sites between MOM and mitochondrial inner membrane). Increases in MOM cardiolipin, as well as cytosolic [Ca2+] during apoptosis could further contribute to its MOM targeting and activity. Meanwhile, the BH3-independent targeting and activation of Bcl-xL to the MOM is expected to counter the action of proapoptotic BAX, thereby preventing premature commitment to apoptosis.
Collapse
|
19
|
Wynen H, Heyland A. Hormonal Regulation of Programmed Cell Death in Sea Urchin Metamorphosis. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.733787] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Programmed cell death (PCD) has been identified as a key process in the metamorphic transition of indirectly developing organisms such as frogs and insects. Many marine invertebrate species with indirect development and biphasic life cycles face the challenge of completing the metamorphic transition of the larval body into a juvenile when they settle into the benthic habitat. Some key characteristics stand out during this transition in comparison to frogs and insects: (1) the transition is often remarkably fast and (2) the larval body is largely abandoned and few structures transition into the juvenile stage. In sea urchins, a group with a drastic and fast metamorphosis, development and destruction of the larval body is regulated by endocrine signals. Here we provide a brief review of the basic regulatory mechanisms of PCD in animals. We then narrow our discussion to metamorphosis with a specific emphasis on sea urchins with indirect life histories and discuss the function of thyroid hormones and histamine in larval development, metamorphosis and settlement of the sea urchin Strongylocentrotus purpuratus. We were able to annotate the large majority of PCD related genes in the sea urchin S. purpuratus and ongoing studies on sea urchin metamorphosis will shed light on the regulatory architecture underlying this dramatic life history transition. While we find overwhelming evidence for hormonal regulation of PCD in animals, especially in the context of metamorphosis, the mechanisms in many marine invertebrate groups with indirect life histories requires more work. Hence, we propose that studies of PCD in animals requires functional studies in whole organisms rather than isolated cells. We predict that future work, targeting a broader array of organisms will not only help to reveal important new functions of PCD but provide a fundamentally new perspective on its use in a diversity of taxonomic, developmental, and ecological contexts.
Collapse
|
20
|
Contribution of Yeast Studies to the Understanding of BCL-2 Family Intracellular Trafficking. Int J Mol Sci 2021; 22:ijms22084086. [PMID: 33920941 PMCID: PMC8071328 DOI: 10.3390/ijms22084086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/24/2022] Open
Abstract
BCL-2 family members are major regulators of apoptotic cell death in mammals. They form an intricate regulatory network that ultimately regulates the release of apoptogenic factors from mitochondria to the cytosol. The ectopic expression of mammalian BCL-2 family members in the yeast Saccharomyces cerevisiae, which lacks BCL-2 homologs, has been long established as a useful addition to the available models to study their function and regulation. In yeast, individual proteins can be studied independently from the whole interaction network, thus providing insight into the molecular mechanisms underlying their function in a living context. Furthermore, one can take advantage of the powerful tools available in yeast to probe intracellular trafficking processes such as mitochondrial sorting and interactions/exchanges between mitochondria and other compartments, such as the endoplasmic reticulum that are largely conserved between yeast and mammals. Yeast molecular genetics thus allows the investigation of the role of these processes on the dynamic equilibrium of BCL-2 family members between mitochondria and extramitochondrial compartments. Here we propose a model of dynamic regulation of BCL-2 family member localization, based on available evidence from ectopic expression in yeast.
Collapse
|
21
|
Schlesinger D, Elsässer SJ. Revisiting sORFs: overcoming challenges to identify and characterize functional microproteins. FEBS J 2021; 289:53-74. [PMID: 33595896 DOI: 10.1111/febs.15769] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/17/2021] [Accepted: 02/15/2021] [Indexed: 02/07/2023]
Abstract
Short ORFs (sORFs), that is, occurrences of a start and stop codon within 100 codons or less, can be found in organisms of all domains of life, outnumbering annotated protein-coding ORFs by orders of magnitude. Even though functional proteins smaller than 100 amino acids are known, the coding potential of sORFs has often been overlooked, as it is not trivial to predict and test for functionality within the large number of sORFs. Recent advances in ribosome profiling and mass spectrometry approaches, together with refined bioinformatic predictions, have enabled a huge leap forward in this field and identified thousands of likely coding sORFs. A relatively low number of small proteins or microproteins produced from these sORFs have been characterized so far on the molecular, structural, and/or mechanistic level. These however display versatile and, in some cases, essential cellular functions, allowing for the exciting possibility that many more, previously unknown small proteins might be encoded in the genome, waiting to be discovered. This review will give an overview of the steadily growing microprotein field, focusing on eukaryotic small proteins. We will discuss emerging themes in the molecular action of microproteins, as well as advances and challenges in microprotein identification and characterization.
Collapse
Affiliation(s)
- Dörte Schlesinger
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.,Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden
| | - Simon J Elsässer
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.,Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
22
|
The Mysteries around the BCL-2 Family Member BOK. Biomolecules 2020; 10:biom10121638. [PMID: 33291826 PMCID: PMC7762061 DOI: 10.3390/biom10121638] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 12/19/2022] Open
Abstract
BOK is an evolutionarily conserved BCL-2 family member that resembles the apoptotic effectors BAK and BAX in sequence and structure. Based on these similarities, BOK has traditionally been classified as a BAX-like pro-apoptotic protein. However, the mechanism of action and cellular functions of BOK remains controversial. While some studies propose that BOK could replace BAK and BAX to elicit apoptosis, others attribute to this protein an indirect way of apoptosis regulation. Adding to the debate, BOK has been associated with a plethora of non-apoptotic functions that makes this protein unpredictable when dictating cell fate. Here, we compile the current knowledge and open questions about this paradoxical protein with a special focus on its structural features as the key aspect to understand BOK biological functions.
Collapse
|
23
|
Naumova N, Šachl R. Regulation of Cell Death by Mitochondrial Transport Systems of Calcium and Bcl-2 Proteins. MEMBRANES 2020; 10:E299. [PMID: 33096926 PMCID: PMC7590060 DOI: 10.3390/membranes10100299] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022]
Abstract
Mitochondria represent the fundamental system for cellular energy metabolism, by not only supplying energy in the form of ATP, but also by affecting physiology and cell death via the regulation of calcium homeostasis and the activity of Bcl-2 proteins. A lot of research has recently been devoted to understanding the interplay between Bcl-2 proteins, the regulation of these interactions within the cell, and how these interactions lead to the changes in calcium homeostasis. However, the role of Bcl-2 proteins in the mediation of mitochondrial calcium homeostasis, and therefore the induction of cell death pathways, remain underestimated and are still not well understood. In this review, we first summarize our knowledge about calcium transport systems in mitochondria, which, when miss-regulated, can induce necrosis. We continue by reviewing and analyzing the functions of Bcl-2 proteins in apoptosis. Finally, we link these two regulatory mechanisms together, exploring the interactions between the mitochondrial Ca2+ transport systems and Bcl-2 proteins, both capable of inducing cell death, with the potential to determine the cell death pathway-either the apoptotic or the necrotic one.
Collapse
Affiliation(s)
| | - Radek Šachl
- J. Heyrovský Institute of Physical Chemistry, Czech Academy of Sciences, 182 23 Prague, Czech Republic;
| |
Collapse
|
24
|
Popgeorgiev N, Sa JD, Jabbour L, Banjara S, Nguyen TTM, Akhavan-E-Sabet A, Gadet R, Ralchev N, Manon S, Hinds MG, Osigus HJ, Schierwater B, Humbert PO, Rimokh R, Gillet G, Kvansakul M. Ancient and conserved functional interplay between Bcl-2 family proteins in the mitochondrial pathway of apoptosis. SCIENCE ADVANCES 2020; 6:6/40/eabc4149. [PMID: 32998881 PMCID: PMC7527217 DOI: 10.1126/sciadv.abc4149] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/10/2020] [Indexed: 06/10/2023]
Abstract
In metazoans, Bcl-2 family proteins are major regulators of mitochondrially mediated apoptosis; however, their evolution remains poorly understood. Here, we describe the molecular characterization of the four members of the Bcl-2 family in the most primitive metazoan, Trichoplax adhaerens All four trBcl-2 homologs are multimotif Bcl-2 group, with trBcl-2L1 and trBcl-2L2 being highly divergent antiapoptotic Bcl-2 members, whereas trBcl-2L3 and trBcl-2L4 are homologs of proapoptotic Bax and Bak, respectively. trBax expression permeabilizes the mitochondrial outer membrane, while trBak operates as a BH3-only sensitizer repressing antiapoptotic activities of trBcl-2L1 and trBcl-2L2. The crystal structure of a trBcl-2L2:trBak BH3 complex reveals that trBcl-2L2 uses the canonical Bcl-2 ligand binding groove to sequester trBak BH3, indicating that the structural basis for apoptosis control is conserved from T. adhaerens to mammals. Finally, we demonstrate that both trBax and trBak BH3 peptides bind selectively to human Bcl-2 homologs to sensitize cancer cells to chemotherapy treatment.
Collapse
Affiliation(s)
- Nikolay Popgeorgiev
- Université de Lyon, Centre de recherche en cancérologie de Lyon, U1052 INSERM, UMR CNRS 5286, Université Lyon I, Centre Léon Bérard, 28 rue Laennec, 69008 Lyon, France.
| | - Jaison D Sa
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Lea Jabbour
- Université de Lyon, Centre de recherche en cancérologie de Lyon, U1052 INSERM, UMR CNRS 5286, Université Lyon I, Centre Léon Bérard, 28 rue Laennec, 69008 Lyon, France
| | - Suresh Banjara
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Trang Thi Minh Nguyen
- Université de Lyon, Centre de recherche en cancérologie de Lyon, U1052 INSERM, UMR CNRS 5286, Université Lyon I, Centre Léon Bérard, 28 rue Laennec, 69008 Lyon, France
| | - Aida Akhavan-E-Sabet
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Rudy Gadet
- Université de Lyon, Centre de recherche en cancérologie de Lyon, U1052 INSERM, UMR CNRS 5286, Université Lyon I, Centre Léon Bérard, 28 rue Laennec, 69008 Lyon, France
| | - Nikola Ralchev
- Université de Lyon, Centre de recherche en cancérologie de Lyon, U1052 INSERM, UMR CNRS 5286, Université Lyon I, Centre Léon Bérard, 28 rue Laennec, 69008 Lyon, France
| | - Stéphen Manon
- Institut de Biochimie et de Génétique Cellulaires, UMR5095, CNRS et Université de Bordeaux, CS61390, 1 Rue Camille Saint-Saëns, 33000 Bordeaux, France
| | - Mark G Hinds
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne 3050, Australia
| | - Hans-Jürgen Osigus
- Institute of Animal Ecology, Division of Molecular Evolution, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Bernd Schierwater
- Institute of Animal Ecology, Division of Molecular Evolution, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
- Sackler Institute for Comparative Genomics, American Museum of Natural History, New York, NY 10024, USA
| | - Patrick O Humbert
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Ruth Rimokh
- Université de Lyon, Centre de recherche en cancérologie de Lyon, U1052 INSERM, UMR CNRS 5286, Université Lyon I, Centre Léon Bérard, 28 rue Laennec, 69008 Lyon, France
| | - Germain Gillet
- Université de Lyon, Centre de recherche en cancérologie de Lyon, U1052 INSERM, UMR CNRS 5286, Université Lyon I, Centre Léon Bérard, 28 rue Laennec, 69008 Lyon, France.
| | - Marc Kvansakul
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia.
| |
Collapse
|
25
|
Reconstituting the Mammalian Apoptotic Switch in Yeast. Genes (Basel) 2020; 11:genes11020145. [PMID: 32013249 PMCID: PMC7073680 DOI: 10.3390/genes11020145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 01/28/2020] [Accepted: 01/28/2020] [Indexed: 12/22/2022] Open
Abstract
Proteins of the Bcl-2 family regulate the permeabilization of the mitochondrial outer membrane that represents a crucial irreversible step in the process of induction of apoptosis in mammalian cells. The family consists of both proapoptotic proteins that facilitate the membrane permeabilization and antiapoptotic proteins that prevent it in the absence of an apoptotic signal. The molecular mechanisms, by which these proteins interact with each other and with the mitochondrial membranes, however, remain under dispute. Although yeast do not have apparent homologues of these apoptotic regulators, yeast cells expressing mammalian members of the Bcl-2 family have proved to be a valuable model system, in which action of these proteins can be effectively studied. This review focuses on modeling the activity of proapoptotic as well as antiapoptotic proteins of the Bcl-2 family in yeast.
Collapse
|
26
|
Flores-Romero H, Ros U, García-Sáez AJ. A lipid perspective on regulated cell death. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 351:197-236. [PMID: 32247580 DOI: 10.1016/bs.ircmb.2019.11.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lipids are fundamental to life as structural components of cellular membranes and for signaling. They are also key regulators of different cellular processes such as cell division, proliferation, and death. Regulated cell death (RCD) requires the engagement of lipids and lipid metabolism for the initiation and execution of its killing machinery. The permeabilization of lipid membranes is a hallmark of RCD that involves, for each kind of cell death, a unique lipid profile. While the permeabilization of the mitochondrial outer membrane allows the release of apoptotic factors to the cytosol during apoptosis, permeabilization of the plasma membrane facilitates the release of intracellular content in other nonapoptotic types of RCD like necroptosis and ferroptosis. Lipids and lipid membranes are important accessory molecules required for the activation of protein executors of cell death such as BAX in apoptosis and MLKL in necroptosis. Peroxidation of membrane phospholipids and the subsequent membrane destabilization is a prerequisite to ferroptosis. Here, we discuss how lipids are essential players in apoptosis, the most common form of RCD, and also their role in necroptosis and ferroptosis. Altogether, we aim to highlight the contribution of lipids and membrane dynamics in cell death regulation.
Collapse
Affiliation(s)
- Hector Flores-Romero
- Interfaculty Institute of Biochemistry, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Uris Ros
- Interfaculty Institute of Biochemistry, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Ana J García-Sáez
- Interfaculty Institute of Biochemistry, Eberhard-Karls-Universität Tübingen, Tübingen, Germany.
| |
Collapse
|
27
|
Kønig SM, Rissler V, Terkelsen T, Lambrughi M, Papaleo E. Alterations of the interactome of Bcl-2 proteins in breast cancer at the transcriptional, mutational and structural level. PLoS Comput Biol 2019; 15:e1007485. [PMID: 31825969 PMCID: PMC6927658 DOI: 10.1371/journal.pcbi.1007485] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 12/23/2019] [Accepted: 10/12/2019] [Indexed: 12/11/2022] Open
Abstract
Apoptosis is an essential defensive mechanism against tumorigenesis. Proteins of the B-cell lymphoma-2 (Bcl-2) family regulate programmed cell death by the mitochondrial apoptosis pathway. In response to intracellular stress, the apoptotic balance is governed by interactions of three distinct subgroups of proteins; the activator/sensitizer BH3 (Bcl-2 homology 3)-only proteins, the pro-survival, and the pro-apoptotic executioner proteins. Changes in expression levels, stability, and functional impairment of pro-survival proteins can lead to an imbalance in tissue homeostasis. Their overexpression or hyperactivation can result in oncogenic effects. Pro-survival Bcl-2 family members carry out their function by binding the BH3 short linear motif of pro-apoptotic proteins in a modular way, creating a complex network of protein-protein interactions. Their dysfunction enables cancer cells to evade cell death. The critical role of Bcl-2 proteins in homeostasis and tumorigenesis, coupled with mounting insight in their structural properties, make them therapeutic targets of interest. A better understanding of gene expression, mutational profile, and molecular mechanisms of pro-survival Bcl-2 proteins in different cancer types, could help to clarify their role in cancer development and may guide advancement in drug discovery. Here, we shed light on the pro-survival Bcl-2 proteins in breast cancer using different bioinformatic approaches, linking -omics with structural data. We analyzed the changes in the expression of the Bcl-2 proteins and their BH3-containing interactors in breast cancer samples. We then studied, at the structural level, a selection of interactions, accounting for effects induced by mutations found in the breast cancer samples. We find two complexes between the up-regulated Bcl2A1 and two down-regulated BH3-only candidates (i.e., Hrk and Nr4a1) as targets associated with reduced apoptosis in breast cancer samples for future experimental validation. Furthermore, we predict L99R, M75R as damaging mutations altering protein stability, and Y120C as a possible allosteric mutation from an exposed surface to the BH3-binding site.
Collapse
Affiliation(s)
- Simon Mathis Kønig
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Vendela Rissler
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Thilde Terkelsen
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Matteo Lambrughi
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Elena Papaleo
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
- Translational Disease Systems Biology, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Denis C, Sopková-de Oliveira Santos J, Bureau R, Voisin-Chiret AS. Hot-Spots of Mcl-1 Protein. J Med Chem 2019; 63:928-943. [DOI: 10.1021/acs.jmedchem.9b00983] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Camille Denis
- Normandie Univiversité, UNICAEN, CERMN, 14000 Caen, France
| | | | - Ronan Bureau
- Normandie Univiversité, UNICAEN, CERMN, 14000 Caen, France
| | | |
Collapse
|
29
|
Distelhorst CW, Bootman MD. Creating a New Cancer Therapeutic Agent by Targeting the Interaction between Bcl-2 and IP 3 Receptors. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a035196. [PMID: 31110129 DOI: 10.1101/cshperspect.a035196] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bcl-2 is a member of a family of proteins that regulate cell survival. Expression of Bcl-2 is aberrantly elevated in many types of cancer. Within cells of the immune system, Bcl-2 has a physiological role in regulating immune responses. However, in cancers arising from cells of the immune system Bcl-2 promotes cell survival and proliferation. This review summarizes discoveries over the past 30 years that have elucidated Bcl-2's role in the normal immune system, including its actions in regulating calcium (Ca2+) signals necessary for the immune response, and for Ca2+-mediated apoptosis at the end of an immune response. How Bcl-2 modulates the release of Ca2+ from intracellular stores via inositol 1,4,5-trisphosphate receptors (IP3R) is discussed, and in particular, the role of Bcl-2/IP3R interactions in promoting the survival of cancer cells by preventing Ca2+-mediated cell death. The development and usage of a peptide, referred to as TAT-Pep8, or more recently, BIRD-2, that induces death of cancer cells by inhibiting Bcl-2's control over IP3R-mediated Ca2+ elevation is discussed. Studies aimed at discovering a small molecule that mimics BIRD-2's anticancer mechanism of action are summarized, along with the prospect of such a compound becoming a novel therapeutic option for cancer.
Collapse
Affiliation(s)
- Clark W Distelhorst
- Departments of Medicine and Pharmacology, Case Western Reserve University School of Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio 44106, USA
| | - Martin D Bootman
- School of Life, Health, and Chemical Science, The Open University, Milton Keynes MK7 6AA, United Kingdom
| |
Collapse
|
30
|
Wang Z, He N, Guo Z, Niu C, Song T, Guo Y, Cao K, Wang A, Zhu J, Zhang X, Zhang Z. Proteolysis Targeting Chimeras for the Selective Degradation of Mcl-1/Bcl-2 Derived from Nonselective Target Binding Ligands. J Med Chem 2019; 62:8152-8163. [DOI: 10.1021/acs.jmedchem.9b00919] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
31
|
Aouacheria A, Navratil V, Combet C. Database and Bioinformatic Analysis of BCL-2 Family Proteins and BH3-Only Proteins. Methods Mol Biol 2019; 1877:23-43. [PMID: 30535996 DOI: 10.1007/978-1-4939-8861-7_2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BCL-2 proteins correspond to a structurally, functionally, and phylogenetically heterogeneous group of regulators that play crucial roles in the life and death of animal cells. Some of these regulators also represent therapeutic targets in human diseases including cancer. In the omics era, there is great need for easy data retrieval and fast analysis of the molecular players involved in cell death. In this chapter, we present generic and specific computational resources (such as the reference database BCL2DB) as well as bioinformatics tools that can be used to investigate BCL-2 homologs and BH3-only proteins.
Collapse
Affiliation(s)
- Abdel Aouacheria
- ISEM, Institut des Sciences de l'Evolution de Montpellier, Université de Montpellier, UMR 5554, CNRS, IRD, EPHE, Montpellier, France.
| | - Vincent Navratil
- PRABI, Rhône Alpes Bioinformatics Center, UCBL, Lyon1, Université de Lyon, Lyon, France
| | - Christophe Combet
- Centre de Recherche en Cancérologie de Lyon, UMR Inserm U1052, CNRS 5286, Université Claude Bernard Lyon 1, Centre Léon Bérard, Lyon, France
| |
Collapse
|
32
|
Flores-Romero H, Landeta O, Ugarte-Uribe B, Cosentino K, García-Porras M, García-Sáez AJ, Basañez G. BFL1 modulates apoptosis at the membrane level through a bifunctional and multimodal mechanism showing key differences with BCLXL. Cell Death Differ 2018; 26:1880-1894. [PMID: 30560933 PMCID: PMC6748131 DOI: 10.1038/s41418-018-0258-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/13/2018] [Accepted: 11/27/2018] [Indexed: 12/17/2022] Open
Abstract
BFL1 is a relatively understudied member of the BCL2 protein family which has been implicated in the pathogenesis and chemoresistance of a variety of human cancers, including hematological malignancies and solid tumours. BFL1 is generally considered to have an antiapoptotic function, although its precise mode of action remains unclear. By quantitatively analyzing BFL1 action in synthetic membrane models and in cells, we found that BFL1 inhibits apoptosis through three distinct mechanisms which are similar but not identical to those of BCLXL, the paradigmatic antiapoptotic BCL2 family protein. Strikingly, alterations in lipid composition during apoptosis activate a prodeath function of BFL1 that is based on noncanonical oligomerization of the protein and breaching of the permeability barrier of the outer mitochondrial membrane (OMM). This lipid-triggered prodeath function of BFL1 is absent in BCLXL and also differs from that of the apoptotic effector BAX, which sets it apart from other BCL2 family members. Our findings support a new model in which BFL1 modulates apoptosis through a bifunctional and multimodal mode of action that is distinctly regulated by OMM lipids compared to BCLXL.
Collapse
Affiliation(s)
- Hector Flores-Romero
- Instituto Biofisika (CSIC, UPV/EHU), Parque Científico de la UPV/EHU, Barrio Sarriena s/n, Leioa, 48940, Bizkaia, Spain. .,Interfaculty Institute of Biochemistry, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 4, Tübingen, 72076, Germany.
| | - Olatz Landeta
- Instituto Biofisika (CSIC, UPV/EHU), Parque Científico de la UPV/EHU, Barrio Sarriena s/n, Leioa, 48940, Bizkaia, Spain.,Departmento de Bioquímica y Biología Molecular, Universidad del País Vasco (UPV/EHU), Barrio Sarriena s/n, Leioa, 48940, Bizkaia, Spain
| | - Begoña Ugarte-Uribe
- Interfaculty Institute of Biochemistry, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 4, Tübingen, 72076, Germany.,Departmento de Bioquímica y Biología Molecular, Universidad del País Vasco (UPV/EHU), Barrio Sarriena s/n, Leioa, 48940, Bizkaia, Spain
| | - Katia Cosentino
- Interfaculty Institute of Biochemistry, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 4, Tübingen, 72076, Germany
| | - Miguel García-Porras
- Instituto Biofisika (CSIC, UPV/EHU), Parque Científico de la UPV/EHU, Barrio Sarriena s/n, Leioa, 48940, Bizkaia, Spain
| | - Ana J García-Sáez
- Interfaculty Institute of Biochemistry, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 4, Tübingen, 72076, Germany
| | - Gorka Basañez
- Instituto Biofisika (CSIC, UPV/EHU), Parque Científico de la UPV/EHU, Barrio Sarriena s/n, Leioa, 48940, Bizkaia, Spain.
| |
Collapse
|
33
|
Teng X, Yau E, Sing C, Hardwick JM. Whi2 signals low leucine availability to halt yeast growth and cell death. FEMS Yeast Res 2018; 18:5083179. [PMID: 30165592 PMCID: PMC6149368 DOI: 10.1093/femsyr/foy095] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 08/26/2018] [Indexed: 12/16/2022] Open
Abstract
Cells are exquisitely tuned to environmental ques. Amino acid availability is rapidly sensed, allowing cells to adjust molecular processes and implement short or long-term metabolic shifts accordingly. How levels of most individual amino acids may be sensed and subsequently signaled to inform cells of their nutrient status is largely unknown. We made the unexpected observation that small changes in the levels of specific amino acids can have a profound effect on yeast cell growth, leading to the identification of yeast Whi2 as a negative regulator of cell growth in low amino acids. Although Whi2 was originally thought to be fungi-specific, Whi2 appears to share a conserved structural domain found in a family of 25 largely uncharacterized human genes encoding the KCTD (potassium channel tetramerization domain) protein family. Insights gained from yeast Whi2 are likely to be revealing about human KCTDs, many of which have been implicated or demonstrated to cause disease when mutated. Here we report new evidence that Whi2 responds to specific amino acids in the medium, particularly low leucine levels. We also discuss the known pathways of amino acid signaling and potential points of regulation by Whi2 in nutrient signaling in yeast and mammals.
Collapse
Affiliation(s)
- Xinchen Teng
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205-2103, USA
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, 215123 Suzhou, Jiangsu Province, People's Republic of China
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205-2103, USA
| | - Eric Yau
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205-2103, USA
| | - Cierra Sing
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205-2103, USA
| | - J Marie Hardwick
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205-2103, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205-2103, USA
| |
Collapse
|
34
|
Oudenaarden CRL, van de Ven RAH, Derksen PWB. Re-inforcing the cell death army in the fight against breast cancer. J Cell Sci 2018; 131:131/16/jcs212563. [DOI: 10.1242/jcs.212563] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
ABSTRACT
Metastatic breast cancer is responsible for most breast cancer-related deaths. Disseminated cancer cells have developed an intrinsic ability to resist anchorage-dependent apoptosis (anoikis). Anoikis is caused by the absence of cellular adhesion, a process that underpins lumen formation and maintenance during mammary gland development and homeostasis. In healthy cells, anoikis is mostly governed by B-cell lymphoma-2 (BCL2) protein family members. Metastatic cancer cells, however, have often developed autocrine BCL2-dependent resistance mechanisms to counteract anoikis. In this Review, we discuss how a pro-apoptotic subgroup of the BCL2 protein family, known as the BH3-only proteins, controls apoptosis and anoikis during mammary gland homeostasis and to what extent their inhibition confers tumor suppressive functions in metastatic breast cancer. Specifically, the role of the two pro-apoptotic BH3-only proteins BCL2-modifying factor (BMF) and BCL2-interacting mediator of cell death (BIM) will be discussed here. We assess current developments in treatment that focus on mimicking the function of the BH3-only proteins to induce apoptosis, and consider their applicability to restore normal apoptotic responses in anchorage-independent disseminating tumor cells.
Collapse
Affiliation(s)
- Clara R. L. Oudenaarden
- UMC Utrecht, Department of Pathology, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
- Lund University, Department of Experimental Oncology, Scheelevägen 2, 22363 Lund, Sweden
| | - Robert A. H. van de Ven
- UMC Utrecht, Department of Pathology, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
- Harvard Medical School, Department of Cell Biology, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Patrick W. B. Derksen
- UMC Utrecht, Department of Pathology, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| |
Collapse
|
35
|
Jin Y, You L, Kim HJ, Lee HW. Telomerase Reverse Transcriptase Contains a BH3-Like Motif and Interacts with BCL-2 Family Members. Mol Cells 2018; 41:684-694. [PMID: 29937479 PMCID: PMC6078858 DOI: 10.14348/molcells.2018.0206] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 05/21/2018] [Indexed: 12/12/2022] Open
Abstract
Upregulation of human telomerase reverse transcriptase (hTERT) expression is an important factor in the cellular survival and cancer. Although growing evidence suggests that hTERT inhibits cellular apoptosis by telomere-independent functions, the mechanisms involved are not fully understood. Here, we show that hTERT contains a BH3-like motif, a short peptide sequence found in BCL-2 family proteins, and interacts with anti-apoptotic BCL-2 family proteins MCL-1 and BCL-xL, suggesting a functional link between hTERT and the mitochondrial pathway of apoptosis. Additionally, we propose that hTERT can be categorized into the atypical BH3-only proteins that promote cellular survival, possibly due to the non-canonical interaction between hTERT and antiapoptotic proteins. Although the detailed mechanisms underlying the hTERT BH3-like motif functions and interactions between hTERT and BCL-2 family proteins have not been elucidated, this work proposes a possible connection between hTERT and BCL-2 family members and reconsiders the role of the BH3-like motif as an interaction motif.
Collapse
Affiliation(s)
- Young Jin
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722,
Korea
| | - Long You
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722,
Korea
| | - Hye Jeong Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722,
Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722,
Korea
| |
Collapse
|
36
|
Abstract
This question of whether fungi undergo apoptosis-like programmed cell death can be separated into two questions. One question is about applying the term "apoptosis" to fungi, and the other is a more challenging question of whether fungi have evolved mechanisms that inflict self-injury. The answers to both questions depend on the definitions applied to "apoptosis" and "programmed cell death." Considering how these and other cell death terms originated and are currently defined for animals, some confusion arises when the terms are applied to fungi. While it is difficult to defend the concept of fungal apoptosis, the more interesting issue is whether fungi will eventually be found to encode programmed or extemporaneous self-destructive processes, as suggested by intriguing new findings.
Collapse
Affiliation(s)
- J Marie Hardwick
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
37
|
Distelhorst CW. Targeting Bcl-2-IP 3 receptor interaction to treat cancer: A novel approach inspired by nearly a century treating cancer with adrenal corticosteroid hormones. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1795-1804. [PMID: 30053503 DOI: 10.1016/j.bbamcr.2018.07.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/18/2018] [Accepted: 07/19/2018] [Indexed: 12/12/2022]
Abstract
Bcl-2 inhibits cell death by at least two different mechanisms. On the one hand, its BH3 domain binds to pro-apoptotic proteins such as Bim and prevents apoptosis induction. On the other hand, the BH4 domain of Bcl-2 binds to the inositol 1,4,5-trisphosphate receptor (IP3R), preventing Ca2+ signals that mediate cell death. In normal T-cells, Bcl-2 levels increase during the immune response, protecting against cell death, and then decline as apoptosis ensues and the immune response dissipates. But in many cancers Bcl-2 is aberrantly expressed and exploited to prevent cell death by inhibiting IP3R-mediated Ca2+ elevation. This review summarizes what is known about the mechanism of Bcl-2's control over IP3R-mediated Ca2+ release and cell death induction. Early insights into the role of Ca2+ elevation in corticosteroid-mediated cell death serves as a model for how targeting IP3R-mediated Ca2+ elevation can be a highly effective therapeutic approach for different types of cancer. Moreover, the successful development of ABT-199 (Venetoclax), a small molecule targeting the BH3 domain of Bcl-2 but without effects on Ca2+, serves as proof of principle that targeting Bcl-2 can be an effective therapeutic approach. BIRD-2, a synthetic peptide that inhibits Bcl-2-IP3R interaction, induces cell death induction in ABT-199 (Venetoclax)-resistant cancer models, attesting to the value of developing therapeutic agents that selectively target Bcl-2-IP3R interaction, inducing Ca2+-mediated cell death.
Collapse
Affiliation(s)
- Clark W Distelhorst
- Case Western University School of Medicine, Case Comprehensive Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, United States of America.
| |
Collapse
|
38
|
TRIM17 and TRIM28 antagonistically regulate the ubiquitination and anti-apoptotic activity of BCL2A1. Cell Death Differ 2018; 26:902-917. [PMID: 30042493 DOI: 10.1038/s41418-018-0169-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 06/28/2018] [Accepted: 07/06/2018] [Indexed: 01/20/2023] Open
Abstract
BCL2A1 is an anti-apoptotic member of the BCL-2 family that contributes to chemoresistance in a subset of tumors. BCL2A1 has a short half-life due to its constitutive processing by the ubiquitin-proteasome system. This constitutes a major tumor-suppressor mechanism regulating BCL2A1 function. However, the enzymes involved in the regulation of BCL2A1 protein stability are currently unknown. Here, we provide the first insight into the regulation of BCL2A1 ubiquitination. We present evidence that TRIM28 is an E3 ubiquitin-ligase for BCL2A1. Indeed, endogenous TRIM28 and BCL2A1 bind to each other at the mitochondria and TRIM28 knock-down decreases BCL2A1 ubiquitination. We also show that TRIM17 stabilizes BCL2A1 by blocking TRIM28 from binding and ubiquitinating BCL2A1, and that GSK3 is involved in the phosphorylation-mediated inhibition of BCL2A1 degradation. BCL2A1 and its close relative MCL1 are thus regulated by common factors but with opposite outcome. Finally, overexpression of TRIM28 or knock-out of TRIM17 reduced BCLA1 protein levels and restored sensitivity of melanoma cells to BRAF-targeted therapy. Therefore, our data describe a molecular rheostat in which two proteins of the TRIM family antagonistically regulate BCL2A1 stability and modulate cell death.
Collapse
|
39
|
Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, Annicchiarico-Petruzzelli M, Antonov AV, Arama E, Baehrecke EH, Barlev NA, Bazan NG, Bernassola F, Bertrand MJM, Bianchi K, Blagosklonny MV, Blomgren K, Borner C, Boya P, Brenner C, Campanella M, Candi E, Carmona-Gutierrez D, Cecconi F, Chan FKM, Chandel NS, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Cohen GM, Conrad M, Cubillos-Ruiz JR, Czabotar PE, D'Angiolella V, Dawson TM, Dawson VL, De Laurenzi V, De Maria R, Debatin KM, DeBerardinis RJ, Deshmukh M, Di Daniele N, Di Virgilio F, Dixit VM, Dixon SJ, Duckett CS, Dynlacht BD, El-Deiry WS, Elrod JW, Fimia GM, Fulda S, García-Sáez AJ, Garg AD, Garrido C, Gavathiotis E, Golstein P, Gottlieb E, Green DR, Greene LA, Gronemeyer H, Gross A, Hajnoczky G, Hardwick JM, Harris IS, Hengartner MO, Hetz C, Ichijo H, Jäättelä M, Joseph B, Jost PJ, Juin PP, Kaiser WJ, Karin M, Kaufmann T, Kepp O, Kimchi A, Kitsis RN, Klionsky DJ, Knight RA, Kumar S, Lee SW, Lemasters JJ, Levine B, Linkermann A, Lipton SA, Lockshin RA, López-Otín C, Lowe SW, Luedde T, Lugli E, MacFarlane M, Madeo F, Malewicz M, Malorni W, Manic G, et alGalluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, Annicchiarico-Petruzzelli M, Antonov AV, Arama E, Baehrecke EH, Barlev NA, Bazan NG, Bernassola F, Bertrand MJM, Bianchi K, Blagosklonny MV, Blomgren K, Borner C, Boya P, Brenner C, Campanella M, Candi E, Carmona-Gutierrez D, Cecconi F, Chan FKM, Chandel NS, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Cohen GM, Conrad M, Cubillos-Ruiz JR, Czabotar PE, D'Angiolella V, Dawson TM, Dawson VL, De Laurenzi V, De Maria R, Debatin KM, DeBerardinis RJ, Deshmukh M, Di Daniele N, Di Virgilio F, Dixit VM, Dixon SJ, Duckett CS, Dynlacht BD, El-Deiry WS, Elrod JW, Fimia GM, Fulda S, García-Sáez AJ, Garg AD, Garrido C, Gavathiotis E, Golstein P, Gottlieb E, Green DR, Greene LA, Gronemeyer H, Gross A, Hajnoczky G, Hardwick JM, Harris IS, Hengartner MO, Hetz C, Ichijo H, Jäättelä M, Joseph B, Jost PJ, Juin PP, Kaiser WJ, Karin M, Kaufmann T, Kepp O, Kimchi A, Kitsis RN, Klionsky DJ, Knight RA, Kumar S, Lee SW, Lemasters JJ, Levine B, Linkermann A, Lipton SA, Lockshin RA, López-Otín C, Lowe SW, Luedde T, Lugli E, MacFarlane M, Madeo F, Malewicz M, Malorni W, Manic G, Marine JC, Martin SJ, Martinou JC, Medema JP, Mehlen P, Meier P, Melino S, Miao EA, Molkentin JD, Moll UM, Muñoz-Pinedo C, Nagata S, Nuñez G, Oberst A, Oren M, Overholtzer M, Pagano M, Panaretakis T, Pasparakis M, Penninger JM, Pereira DM, Pervaiz S, Peter ME, Piacentini M, Pinton P, Prehn JHM, Puthalakath H, Rabinovich GA, Rehm M, Rizzuto R, Rodrigues CMP, Rubinsztein DC, Rudel T, Ryan KM, Sayan E, Scorrano L, Shao F, Shi Y, Silke J, Simon HU, Sistigu A, Stockwell BR, Strasser A, Szabadkai G, Tait SWG, Tang D, Tavernarakis N, Thorburn A, Tsujimoto Y, Turk B, Vanden Berghe T, Vandenabeele P, Vander Heiden MG, Villunger A, Virgin HW, Vousden KH, Vucic D, Wagner EF, Walczak H, Wallach D, Wang Y, Wells JA, Wood W, Yuan J, Zakeri Z, Zhivotovsky B, Zitvogel L, Melino G, Kroemer G. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ 2018; 25:486-541. [PMID: 29362479 PMCID: PMC5864239 DOI: 10.1038/s41418-017-0012-4] [Show More Authors] [Citation(s) in RCA: 4383] [Impact Index Per Article: 626.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 02/06/2023] Open
Abstract
Over the past decade, the Nomenclature Committee on Cell Death (NCCD) has formulated guidelines for the definition and interpretation of cell death from morphological, biochemical, and functional perspectives. Since the field continues to expand and novel mechanisms that orchestrate multiple cell death pathways are unveiled, we propose an updated classification of cell death subroutines focusing on mechanistic and essential (as opposed to correlative and dispensable) aspects of the process. As we provide molecularly oriented definitions of terms including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, parthanatos, entotic cell death, NETotic cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence, and mitotic catastrophe, we discuss the utility of neologisms that refer to highly specialized instances of these processes. The mission of the NCCD is to provide a widely accepted nomenclature on cell death in support of the continued development of the field.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Paris Descartes/Paris V University, Paris, France.
| | - Ilio Vitale
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Stuart A Aaronson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John M Abrams
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dieter Adam
- Institute of Immunology, Kiel University, Kiel, Germany
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Emad S Alnemri
- Department of Biochemistry and Molecular Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lucia Altucci
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Ivano Amelio
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
| | - David W Andrews
- Biological Sciences, Sunnybrook Research Institute, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | | | - Alexey V Antonov
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
| | - Eli Arama
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Nickolai A Barlev
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, Louisiana State University School of Medicine, New Orleans, LA, USA
| | - Francesca Bernassola
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Mathieu J M Bertrand
- VIB Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Katiuscia Bianchi
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, Albert Ludwigs University, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), Faculty of Medicine, Albert Ludwigs University, Freiburg, Germany
| | - Patricia Boya
- Department of Cellular and Molecular Biology, Center for Biological Investigation (CIB), Spanish National Research Council (CSIC), Madrid, Spain
| | - Catherine Brenner
- INSERM U1180, Châtenay Malabry, France
- University of Paris Sud/Paris Saclay, Orsay, France
| | - Michelangelo Campanella
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, London, UK
- University College London Consortium for Mitochondrial Research, London, UK
| | - Eleonora Candi
- Biochemistry Laboratory, Dermopatic Institute of Immaculate (IDI) IRCCS, Rome, Italy
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | | | - Francesco Cecconi
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Unit of Cell Stress and Survival, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Francis K-M Chan
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Navdeep S Chandel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emily H Cheng
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jerry E Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John A Cidlowski
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Aaron Ciechanover
- Technion Integrated Cancer Center (TICC), The Ruth and Bruce Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Gerald M Cohen
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Marcus Conrad
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health (GmbH), Munich, Germany
| | - Juan R Cubillos-Ruiz
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Peter E Czabotar
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Vincenzo D'Angiolella
- Cancer Research UK and Medical Research Council Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vincenzo De Laurenzi
- Department of Medical, Oral and Biotechnological Sciences, CeSI-MetUniversity of Chieti-Pescara "G. d'Annunzio", Chieti, Italy
| | - Ruggero De Maria
- Institute of General Pathology, Catholic University "Sacro Cuore", Rome, Italy
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mohanish Deshmukh
- Department of Cell Biology and Physiology, Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
| | - Nicola Di Daniele
- Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Vishva M Dixit
- Department of Physiological Chemistry, Genentech, South San Francisco, CA, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Colin S Duckett
- Baylor Scott & White Research Institute, Baylor College of Medicine, Dallas, TX, USA
| | - Brian D Dynlacht
- Department of Pathology, New York University School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - John W Elrod
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine at Temple University School of Medicine, Philadelphia, PA, USA
| | - Gian Maria Fimia
- National Institute for Infectious Diseases IRCCS "Lazzaro Spallanzani", Rome, Italy
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site, Frankfurt, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ana J García-Sáez
- Interfaculty Institute of Biochemistry, Tübingen University, Tübingen, Germany
| | - Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Carmen Garrido
- INSERM U1231 "Lipides Nutrition Cancer", Dijon, France
- Faculty of Medicine, University of Burgundy France Comté, Dijon, France
- Cancer Centre Georges François Leclerc, Dijon, France
| | - Evripidis Gavathiotis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Pierre Golstein
- Immunology Center of Marseille-Luminy, Aix Marseille University, Marseille, France
| | - Eyal Gottlieb
- Technion Integrated Cancer Center (TICC), The Ruth and Bruce Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Douglas R Green
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Lloyd A Greene
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Hinrich Gronemeyer
- Team labeled "Ligue Contre le Cancer", Department of Functional Genomics and Cancer, Institute of Genetics and Molecular and Cellular Biology (IGBMC), Illkirch, France
- CNRS UMR 7104, Illkirch, France
- INSERM U964, Illkirch, France
- University of Strasbourg, Illkirch, France
| | - Atan Gross
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Gyorgy Hajnoczky
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - J Marie Hardwick
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Isaac S Harris
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Cellular and Molecular Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Marja Jäättelä
- Cell Death and Metabolism Unit, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Bertrand Joseph
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Philipp J Jost
- III Medical Department for Hematology and Oncology, Technical University Munich, Munich, Germany
| | - Philippe P Juin
- Team 8 "Stress adaptation and tumor escape", CRCINA-INSERM U1232, Nantes, France
- University of Nantes, Nantes, France
- University of Angers, Angers, France
- Institute of Cancer Research in Western France, Saint-Herblain, France
| | - William J Kaiser
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, TX, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, University of California San Diego, La Jolla, CA, USA
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Oliver Kepp
- Paris Descartes/Paris V University, Paris, France
- Faculty of Medicine, Paris Sud/Paris XI University, Kremlin-Bicêtre, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Campus, Villejuif, France
- Team 11 labeled "Ligue Nationale contre le Cancer", Cordeliers Research Center, Paris, France
- INSERM U1138, Paris, France
- Pierre et Marie Curie/Paris VI University, Paris, France
| | - Adi Kimchi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Richard N Kitsis
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Daniel J Klionsky
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Richard A Knight
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Sam W Lee
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - John J Lemasters
- Center for Cell Death, Injury and Regeneration, Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
- Center for Cell Death, Injury and Regeneration, Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Beth Levine
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andreas Linkermann
- Division of Nephrology, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Stuart A Lipton
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Richard A Lockshin
- Department of Biology, St. John's University, Queens, NY, USA
- Queens College of the City University of New York, Queens, NY, USA
| | - Carlos López-Otín
- Departament of Biochemistry and Molecular Biology, Faculty of Medicine, University Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - Scott W Lowe
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tom Luedde
- Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Enrico Lugli
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Humanitas Flow Cytometry Core, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Marion MacFarlane
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
| | - Frank Madeo
- Department Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Michal Malewicz
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
| | - Walter Malorni
- National Centre for Gender Medicine, Italian National Institute of Health (ISS), Rome, Italy
| | - Gwenola Manic
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Seamus J Martin
- Departments of Genetics, Trinity College, University of Dublin, Dublin 2, Ireland
| | - Jean-Claude Martinou
- Department of Cell Biology, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM), Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
- Cancer Genomics Center, Amsterdam, The Netherlands
| | - Patrick Mehlen
- Apoptosis, Cancer and Development laboratory, CRCL, Lyon, France
- Team labeled "La Ligue contre le Cancer", Lyon, France
- LabEx DEVweCAN, Lyon, France
- INSERM U1052, Lyon, France
- CNRS UMR5286, Lyon, France
- Department of Translational Research and Innovation, Léon Bérard Cancer Center, Lyon, France
| | - Pascal Meier
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Mary-Jean Mitchell Green Building, Chester Beatty Laboratories, London, UK
| | - Sonia Melino
- Department of Chemical Sciences and Technologies, University of Rome, Tor Vergata, Rome, Italy
| | - Edward A Miao
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, NC, USA
| | - Jeffery D Molkentin
- Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ute M Moll
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Cristina Muñoz-Pinedo
- Cell Death Regulation Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Shigekazu Nagata
- Laboratory of Biochemistry and Immunology, World Premier International (WPI) Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Gabriel Nuñez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Andrew Oberst
- Department of Immunology, University of Washington, Seattle, WA, USA
- Center for Innate Immunity and Immune Disease, Seattle, WA, USA
| | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute, Rehovot, Israel
| | - Michael Overholtzer
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michele Pagano
- Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York University School of Medicine, New York, NY, USA
| | - Theocharis Panaretakis
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Manolis Pasparakis
- Institute for Genetics, Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Campus Vienna BioCentre, Vienna, Austria
| | - David M Pereira
- REQUIMTE/LAQV, Laboratory of Pharmacognosy, Department of Chemistry, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
- National University Cancer Institute, National University Health System (NUHS), Singapore, Singapore
| | - Marcus E Peter
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mauro Piacentini
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- National Institute for Infectious Diseases IRCCS "Lazzaro Spallanzani", Rome, Italy
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
- LTTA center, University of Ferrara, Ferrara, Italy
- Maria Cecilia Hospital, GVM Care & Research, Health Science Foundation, Cotignola, Italy
| | - Jochen H M Prehn
- Department of Physiology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Hamsa Puthalakath
- Department of Biochemistry, La Trobe University, Victoria, Australia
| | - Gabriel A Rabinovich
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine (IBYME), National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
- Department of Biological Chemistry, Faculty of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Markus Rehm
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, Stuttgart, Germany
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Cecilia M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research (CIMR), University of Cambridge, Cambridge, UK
| | - Thomas Rudel
- Department of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Kevin M Ryan
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Emre Sayan
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Luca Scorrano
- Department of Biology, University of Padua, Padua, Italy
- Venetian Institute of Molecular Medicine, Padua, Italy
| | - Feng Shao
- National Institute of Biological Sciences, Beijing, China
| | - Yufang Shi
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Chinese Academy of Sciences, Shanghai, China
- Jiangsu Key Laboratory of Stem Cells and Medicinal Biomaterials, Institutes for Translational Medicine, Soochow University, Suzhou, China
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - John Silke
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
- Division of Inflammation, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Antonella Sistigu
- Institute of General Pathology, Catholic University "Sacro Cuore", Rome, Italy
- Unit of Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Chemistry, Columbia University, New York, NY, USA
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Gyorgy Szabadkai
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Department of Cell and Developmental Biology, University College London Consortium for Mitochondrial Research, London, UK
- Francis Crick Institute, London, UK
| | | | - Daolin Tang
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- Center for DAMP Biology, Guangzhou Medical University, Guangzhou, Guangdong, China
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou Medical University, Guangzhou, Guangdong, China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou Medical University, Guangzhou, Guangdong, China
- Key Laboratory for Protein Modification and Degradation of Guangdong Province, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas Medical School, University of Crete, Heraklion, Greece
| | - Andrew Thorburn
- Department of Pharmacology, University of Colorado, Aurora, CO, USA
| | | | - Boris Turk
- Department Biochemistry and Molecular Biology, "Jozef Stefan" Institute, Ljubljana, Slovenia
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Tom Vanden Berghe
- VIB Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter Vandenabeele
- VIB Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Andreas Villunger
- Division of Developmental Immunology, Innsbruck Medical University, Innsbruck, Austria
| | - Herbert W Virgin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Domagoj Vucic
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, CA, USA
| | - Erwin F Wagner
- Genes, Development and Disease Group, Cancer Cell Biology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Henning Walczak
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, London, UK
| | - David Wallach
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Ying Wang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Will Wood
- School of Cellular and Molecular Medicine, Faculty of Biomedical Sciences, University of Bristol, Bristol, UK
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zahra Zakeri
- Department of Biology, Queens College of the City University of New York, Queens, NY, USA
| | - Boris Zhivotovsky
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Laurence Zitvogel
- Faculty of Medicine, Paris Sud/Paris XI University, Kremlin-Bicêtre, France
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Gerry Melino
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Guido Kroemer
- Paris Descartes/Paris V University, Paris, France.
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Campus, Villejuif, France.
- Team 11 labeled "Ligue Nationale contre le Cancer", Cordeliers Research Center, Paris, France.
- INSERM U1138, Paris, France.
- Pierre et Marie Curie/Paris VI University, Paris, France.
- Biology Pole, European Hospital George Pompidou, AP-HP, Paris, France.
| |
Collapse
|
40
|
Ivanova H, Ritaine A, Wagner L, Luyten T, Shapovalov G, Welkenhuyzen K, Seitaj B, Monaco G, De Smedt H, Prevarskaya N, Yule DI, Parys JB, Bultynck G. The trans-membrane domain of Bcl-2α, but not its hydrophobic cleft, is a critical determinant for efficient IP3 receptor inhibition. Oncotarget 2018; 7:55704-55720. [PMID: 27494888 PMCID: PMC5342447 DOI: 10.18632/oncotarget.11005] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 07/18/2016] [Indexed: 12/29/2022] Open
Abstract
The anti-apoptotic Bcl-2 protein is emerging as an efficient inhibitor of IP3R function, contributing to its oncogenic properties. Yet, the underlying molecular mechanisms remain not fully understood. Using mutations or pharmacological inhibition to antagonize Bcl-2's hydrophobic cleft, we excluded this functional domain as responsible for Bcl-2-mediated IP3Rs inhibition. In contrast, the deletion of the C-terminus, containing the trans-membrane domain, which is only present in Bcl-2α, but not in Bcl-2β, led to impaired inhibition of IP3R-mediated Ca2+ release and staurosporine-induced apoptosis. Strikingly, the trans-membrane domain was sufficient for IP3R binding and inhibition. We therefore propose a novel model, in which the Bcl-2's C-terminus serves as a functional anchor, which beyond mere ER-membrane targeting, underlies efficient IP3R inhibition by (i) positioning the BH4 domain in the close proximity of its binding site on IP3R, thus facilitating their interaction; (ii) inhibiting IP3R-channel openings through a direct interaction with the C-terminal region of the channel downstream of the channel-pore. Finally, since the hydrophobic cleft of Bcl-2 was not involved in IP3R suppression, our findings indicate that ABT-199 does not interfere with IP3R regulation by Bcl-2 and its mechanism of action as a cell-death therapeutic in cancer cells likely does not involve Ca2+ signaling.
Collapse
Affiliation(s)
- Hristina Ivanova
- Katholieke Universiteit Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Cancer Institute (LKI), BE-3000 Leuven, Belgium
| | - Abigael Ritaine
- Inserm U-1003, Equipe Labellisée par la Ligue Nationale Contre le Cancer et LABEX (Laboratoire d'excellence), Université Lille1, 59655 Villeneuve d'Ascq, France
| | - Larry Wagner
- University of Rochester Medical Center School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Tomas Luyten
- Katholieke Universiteit Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Cancer Institute (LKI), BE-3000 Leuven, Belgium
| | - George Shapovalov
- Inserm U-1003, Equipe Labellisée par la Ligue Nationale Contre le Cancer et LABEX (Laboratoire d'excellence), Université Lille1, 59655 Villeneuve d'Ascq, France
| | - Kirsten Welkenhuyzen
- Katholieke Universiteit Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Cancer Institute (LKI), BE-3000 Leuven, Belgium
| | - Bruno Seitaj
- Katholieke Universiteit Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Cancer Institute (LKI), BE-3000 Leuven, Belgium
| | - Giovanni Monaco
- Katholieke Universiteit Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Cancer Institute (LKI), BE-3000 Leuven, Belgium
| | - Humbert De Smedt
- Katholieke Universiteit Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Cancer Institute (LKI), BE-3000 Leuven, Belgium
| | - Natalia Prevarskaya
- Inserm U-1003, Equipe Labellisée par la Ligue Nationale Contre le Cancer et LABEX (Laboratoire d'excellence), Université Lille1, 59655 Villeneuve d'Ascq, France
| | - David I Yule
- University of Rochester Medical Center School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Jan B Parys
- Katholieke Universiteit Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Cancer Institute (LKI), BE-3000 Leuven, Belgium
| | - Geert Bultynck
- Katholieke Universiteit Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Cancer Institute (LKI), BE-3000 Leuven, Belgium
| |
Collapse
|
41
|
Intra- and Intercellular Quality Control Mechanisms of Mitochondria. Cells 2017; 7:cells7010001. [PMID: 29278362 PMCID: PMC5789274 DOI: 10.3390/cells7010001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/22/2017] [Accepted: 12/23/2017] [Indexed: 12/29/2022] Open
Abstract
Mitochondria function to generate ATP and also play important roles in cellular homeostasis, signaling, apoptosis, autophagy, and metabolism. The loss of mitochondrial function results in cell death and various types of diseases. Therefore, quality control of mitochondria via intra- and intercellular pathways is crucial. Intracellular quality control consists of biogenesis, fusion and fission, and degradation of mitochondria in the cell, whereas intercellular quality control involves tunneling nanotubes and extracellular vesicles. In this review, we outline the current knowledge on the intra- and intercellular quality control mechanisms of mitochondria.
Collapse
|
42
|
The Bcl-2 Family in Host-Virus Interactions. Viruses 2017; 9:v9100290. [PMID: 28984827 PMCID: PMC5691641 DOI: 10.3390/v9100290] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 10/01/2017] [Accepted: 10/03/2017] [Indexed: 12/13/2022] Open
Abstract
Members of the B cell lymphoma-2 (Bcl-2) family are pivotal arbiters of mitochondrially mediated apoptosis, a process of fundamental importance during tissue development, homeostasis, and disease. At the structural and mechanistic level, the mammalian members of the Bcl-2 family are increasingly well understood, with their interplay ultimately deciding the fate of a cell. Dysregulation of Bcl-2-mediated apoptosis underlies a plethora of diseases, and numerous viruses have acquired homologs of Bcl-2 to subvert host cell apoptosis and autophagy to prevent premature death of an infected cell. Here we review the structural biology, interactions, and mechanisms of action of virus-encoded Bcl-2 proteins, and how they impact on host-virus interactions to ultimately enable successful establishment and propagation of viral infections.
Collapse
|
43
|
Aouacheria A, Baghdiguian S, Lamb HM, Huska JD, Pineda FJ, Hardwick JM. Connecting mitochondrial dynamics and life-or-death events via Bcl-2 family proteins. Neurochem Int 2017; 109:141-161. [PMID: 28461171 DOI: 10.1016/j.neuint.2017.04.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 04/17/2017] [Indexed: 12/12/2022]
Abstract
The morphology of a population of mitochondria is the result of several interacting dynamical phenomena, including fission, fusion, movement, elimination and biogenesis. Each of these phenomena is controlled by underlying molecular machinery, and when defective can cause disease. New understanding of the relationships between form and function of mitochondria in health and disease is beginning to be unraveled on several fronts. Studies in mammals and model organisms have revealed that mitochondrial morphology, dynamics and function appear to be subject to regulation by the same proteins that regulate apoptotic cell death. One protein family that influences mitochondrial dynamics in both healthy and dying cells is the Bcl-2 protein family. Connecting mitochondrial dynamics with life-death pathway forks may arise from the intersection of Bcl-2 family proteins with the proteins and lipids that determine mitochondrial shape and function. Bcl-2 family proteins also have multifaceted influences on cells and mitochondria, including calcium handling, autophagy and energetics, as well as the subcellular localization of mitochondrial organelles to neuronal synapses. The remarkable range of physical or functional interactions by Bcl-2 family proteins is challenging to assimilate into a cohesive understanding. Most of their effects may be distinct from their direct roles in apoptotic cell death and are particularly apparent in the nervous system. Dual roles in mitochondrial dynamics and cell death extend beyond BCL-2 family proteins. In this review, we discuss many processes that govern mitochondrial structure and function in health and disease, and how Bcl-2 family proteins integrate into some of these processes.
Collapse
Affiliation(s)
- Abdel Aouacheria
- Institute of Evolutionary Sciences of Montpellier (ISEM), CNRS UMR 5554, University of Montpellier, Place Eugène Bataillon, 34095 Montpellier, France
| | - Stephen Baghdiguian
- Institute of Evolutionary Sciences of Montpellier (ISEM), CNRS UMR 5554, University of Montpellier, Place Eugène Bataillon, 34095 Montpellier, France
| | - Heather M Lamb
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe St., Baltimore, MD 21205, USA
| | - Jason D Huska
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe St., Baltimore, MD 21205, USA
| | - Fernando J Pineda
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe St., Baltimore, MD 21205, USA; Department of Biostatistics, Johns Hopkins University, Bloomberg School of Public Health, 615 North Wolfe St., Baltimore, MD 21205, USA
| | - J Marie Hardwick
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe St., Baltimore, MD 21205, USA.
| |
Collapse
|
44
|
Birkinshaw RW, Czabotar PE. The BCL-2 family of proteins and mitochondrial outer membrane permeabilisation. Semin Cell Dev Biol 2017; 72:152-162. [PMID: 28396106 DOI: 10.1016/j.semcdb.2017.04.001] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 03/03/2017] [Accepted: 04/06/2017] [Indexed: 01/24/2023]
Abstract
Apoptosis is a form of programmed cell death critical for the development and homeostasis of multicellular organisms. A key event within the mitochondrial pathway to apoptosis is the permeabilisation of the mitochondrial outer membrane (MOM), a point of no return in apoptotic progression. This event is governed by a complex interplay of interactions between BCL-2 family members. Here we discuss the roles of opposing factions within the family. We focus on the structural details of these interactions, how they promote or prevent apoptosis and recent developments towards understanding the conformational changes of BAK and BAX that lead to MOM permeabilisation. These interactions and structural insights are of particular interest for drug discovery, as highlighted by the development of therapeutics that target pro-survival family members and restore apoptosis in cancer cells.
Collapse
Affiliation(s)
- Richard W Birkinshaw
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Peter E Czabotar
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
45
|
Ivanova H, Luyten T, Decrock E, Vervliet T, Leybaert L, Parys JB, Bultynck G. The BH4 domain of Bcl-2 orthologues from different classes of vertebrates can act as an evolutionary conserved inhibitor of IP 3 receptor channels. Cell Calcium 2017; 62:41-46. [PMID: 28179071 DOI: 10.1016/j.ceca.2017.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/19/2017] [Accepted: 01/19/2017] [Indexed: 02/05/2023]
Abstract
Ca2+ signalling plays an important role in various physiological processes in vertebrates. In mammals, the highly conserved anti-apoptotic B-cell lymphoma-2 (Bcl-2) protein is an important modulator of the inositol 1,4,5-trisphosphate receptor (IP3R), i.e. the main intracellular Ca2+ - release channel located at the endoplasmic reticulum (ER). The Bcl-2 Homology (BH) 4 domain of Bcl-2 (BH4-Bcl-2) is a critical determinant for inhibiting IP3Rs, by directly targeting a region in the modulatory domain of the receptor (domain 3). In this paper, we aimed to track the evolutionary history of IP3R regulation by the BH4 domain of Bcl-2 orthologues from different classes of vertebrates, including Osteichthyes, Amphibia, Reptilia, Aves and Mammalia. The high degree of conservation of the BH4 sequences correlated with the ability of all tested peptides to bind to the domain 3 of mouse IP3R1 in GST-pull downs and their overall ability to inhibit IP3-induced Ca2+ release (IICR) in permeabilized cells. Nevertheless, the BH4 domains differed in their potency to suppress IICR. The peptide derived from X. laevis was the least potent inhibitor. We identified a critical residue in BH4-Bcl-2 from H. sapiens, Thr7, which is replaced by Gly7 in X. laevis. Compared to the wild type X. laevis BH4-Bcl-2, a "humanized" version of the peptide (BH4-Bcl-2 Gly7Thr), displayed increased IP3R-inhibitory properties. Despite the differences in the inhibitory efficiency, our data indicate that the BH4 domain of Bcl-2 orthologues from different classes of vertebrates can act as a binding partner and inhibitor of IP3R channels.
Collapse
Affiliation(s)
- Hristina Ivanova
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and, Herestraat 49, BE-3000, Leuven, Belgium
| | - Tomas Luyten
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and, Herestraat 49, BE-3000, Leuven, Belgium
| | - Elke Decrock
- Ghent University, Physiology Group, Department of Basic Medical Sciences, BE-9000, Ghent, Belgium
| | - Tim Vervliet
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and, Herestraat 49, BE-3000, Leuven, Belgium
| | - Luc Leybaert
- Ghent University, Physiology Group, Department of Basic Medical Sciences, BE-9000, Ghent, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and, Herestraat 49, BE-3000, Leuven, Belgium; KU Leuven, Leuven Cancer Institute (LKI), BE-3000, Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and, Herestraat 49, BE-3000, Leuven, Belgium; KU Leuven, Leuven Cancer Institute (LKI), BE-3000, Leuven, Belgium.
| |
Collapse
|
46
|
Caria S, Hinds MG, Kvansakul M. Structural insight into an evolutionarily ancient programmed cell death regulator - the crystal structure of marine sponge BHP2 bound to LB-Bak-2. Cell Death Dis 2017; 8:e2543. [PMID: 28079890 PMCID: PMC5386376 DOI: 10.1038/cddis.2016.469] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 11/14/2016] [Accepted: 11/17/2016] [Indexed: 12/25/2022]
Abstract
Sponges of the porifera family harbor some of the evolutionary most ancient orthologs of the B-cell lymphoma-2 (Bcl-2) family, a protein family critical to regulation of apoptosis. The genome of the sponge Geodia cydonium contains the putative pro-survival Bcl-2 homolog BHP2, which protects sponge tissue as well as mammalian Hek-293 and NIH-3T3 cells against diverse apoptotic stimuli. The Lake Baikal demosponge Lubomirskia baicalensis has been shown to encode both putative pro-survival Bcl-2 (LB-Bcl-2) and pro-apoptotic Bcl-2 members (LB-Bak-2), which have been implied in axis formation (branches) in L. baicalensis. However, the molecular mechanism of action of sponge-encoded orthologs of Bcl-2 remains to be clarified. Here, we report that the pro-survival Bcl-2 ortholog BHP2 from G. cydonium is able to bind the BH3 motif of a pro-apoptotic Bcl-2 protein, LB-Bak-2 of the sponge L. baicalensis. Furthermore, we determined the crystal structure of BHP2 bound to LB-Bak-2, which revealed that using a binding groove conserved across all pro-survival Bcl-2 proteins, BHP2 binds multi-motif Bax-like proteins through their BH3-binding regions. However, BHP2 discriminates against BH3-only bearing proteins by blocking access to a hydrophobic pocket that is critical for BH3 motif binding in pro-survival Bcl-2 proteins from higher organisms. This differential binding mode is reflected in a structure-based phylogenetic comparison of BHP2 with other Bcl-2 family members, which revealed that BHP2 does not cluster with either Bcl-2 members of higher organisms or pathogen-encoded homologs, and assumes a discrete position. Our findings suggest that the molecular machinery and mechanisms for executing Bcl-2-mediated apoptosis as observed in mammals are evolutionary ancient, with early regulation of apoptotic machineries closely resembling their modern counterparts in mammals rather than Caenorhabditis elegans or drosophila.
Collapse
Affiliation(s)
- Sofia Caria
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne,Victoria 3086, Australia
| | - Mark G Hinds
- Department of Chemistry & Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Marc Kvansakul
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne,Victoria 3086, Australia
| |
Collapse
|
47
|
Abstract
Apoptosis is a form of programmed cell death that is critical for basic human development and physiology. One of the more important surprises in cell biology in the last two decades is the extent to which mitochondria represent a physical point of convergence for many apoptosis-inducing signals in mammalian cells. Mitochondria not only adjudicate the decision of whether or not to commit to cell death, but also release toxic proteins culminating in widespread proteolysis, nucleolysis, and cell engulfment. Interactions among BCL-2 family proteins at the mitochondrial outer membrane control the release of these toxic proteins and, by extension, control cellular commitment to apoptosis. This pathway is particularly relevant to cancer treatment, as most cancer chemotherapies trigger mitochondrial-mediated apoptosis. In this Review, we discuss recent advances in the BCL-2 family interactions, their control by upstream factors, and how the mitochondria itself alters these interactions. We also highlight recent clinical insights into mitochondrial-mediated apoptosis and novel cancer therapies that exploit this pathway.
Collapse
Affiliation(s)
- Patrick D Bhola
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Anthony Letai
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| |
Collapse
|
48
|
Genes CM, de Lucio H, González VM, Sánchez-Murcia PA, Rico E, Gago F, Fasel N, Jiménez-Ruiz A. A functional BH3 domain in an aquaporin from Leishmania infantum. Cell Death Discov 2016; 2:16043. [PMID: 27551533 PMCID: PMC4979448 DOI: 10.1038/cddiscovery.2016.43] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 05/15/2016] [Indexed: 02/08/2023] Open
Abstract
Despite the absence of sequences showing significant similarity to any of the members of the Bcl-2 family of proteins in protozoa, experiments carried out in yeast or trypanosomatids have demonstrated that ectopic expression of some of these members alters their response to different death stimuli. Because the BH3 domain is the smallest common signature in all the proteins of this family of apoptosis regulators and also because they are essential for molecular interactions between antagonistic members, we looked for sequences with significant similarity to the BH3 motif in the Leishmania infantum genome. Among the top scoring ones, we found the MYLALQNLGDEV amino-acid stretch at the C terminus of a previously described aquaporin, now renamed as Li-BH3AQP. This motif is highly conserved in homologous proteins from other species of the Leishmania genus. The association of Li-BH3AQP with human Bcl-XL was demonstrated by both co-immunoprecipitation and yeast two-hybrid experiments. Ectopic expression of Li-BH3AQP reduced viability of HeLa cells and this deleterious effect was abrogated by the simultaneous overexpression of Bcl-XL. Although we were not able to demonstrate a reduction in parasite viability when the protein was overexpressed in Leishmania promastigotes, a prodeath effect could be observed when the parasites overexpressing Li-BH3AQP were treated with staurosporine or antimycin A. Surprisingly, these parasites were more resistant, compared with wild-type parasites, to hypotonic stress or nutrient deprivation. The prodeath activity was abolished upon replacement of two highly conserved amino acids in this BH3 domain. Taken together, these results point to Li-BH3AQP as the first non-enzymatic protein ever described in trypanosomatids that is involved in cell death.
Collapse
Affiliation(s)
- C M Genes
- Departamento de Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares 28805, Spain
| | - H de Lucio
- Departamento de Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares 28805, Spain
| | - V M González
- Laboratory of aptamers, Departamento de Bioquímica-Investigación, IRYCIS-Hospital Ramón y Cajal, Madrid, Spain
| | - P A Sánchez-Murcia
- Departamento de Ciencias Biomédicas, Universidad de Alcalá, Facultad de Medicina, Alcalá de Henares 28805, Spain
| | - E Rico
- Departamento de Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares 28805, Spain
| | - F Gago
- Departamento de Ciencias Biomédicas, Universidad de Alcalá, Facultad de Medicina, Alcalá de Henares 28805, Spain
| | - N Fasel
- Department of Biochemistry, University of Lausanne, 155 Chemin des Boveresses, Epalinges 1066, Switzerland
| | - A Jiménez-Ruiz
- Departamento de Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares 28805, Spain
| |
Collapse
|
49
|
Masoudi-Sobhanzadeh Y, Motieghader H. World Competitive Contests (WCC) algorithm: A novel intelligent optimization algorithm for biological and non-biological problems. INFORMATICS IN MEDICINE UNLOCKED 2016; 3:15-28. [PMID: 32363231 PMCID: PMC7185599 DOI: 10.1016/j.imu.2016.06.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/29/2016] [Accepted: 06/27/2016] [Indexed: 02/02/2023] Open
Abstract
Since different sciences face lots of problems which cannot be solved in reasonable time order, we need new methods and algorithms for getting acceptable answers in proper time order. In the present study, a novel intelligent optimization algorithm, known as WCC (World Competitive Contests), has been proposed and applied to find the transcriptional factor binding sites (TFBS) and eight benchmark functions discovery processes. We recognize the need to introduce an intelligent optimization algorithm because the TFBS discovery is a biological and an NP-Hard problem. Although there are some intelligent algorithms for the purpose of solving the above-mentioned problems, an optimization algorithm with good and acceptable performance, which is based on the real parameters, is essential. Like the other optimization algorithms, the proposed algorithm starts with the first population of teams. After teams are put into different groups, they will begin competing against their rival teams. The highly qualified teams will ascend to the elimination stage and will play each other in the next rounds. The other teams will wait for a new season to start. In this paper, we’re going to implement our proposed algorithm and compare it with five famous optimization algorithms from the perspective of the following: the obtained results, stability, convergence, standard deviation and elapsed time, which are applied to the real and randomly created datasets with different motif sizes. According to our obtained results, in many cases, the WCC׳s performance is better than the other algorithms’. A new optimization algorithm which is named as WCC (World Competitive Contests) and is inspired by human sport rules is introduced. WCC is applied to TFBS (transcriptional factor binding sites) problem and eight benchmark functions. WCC is compared with the five known optimization algorithms. Experimental results show WCC’s remarkable performance relative to other optimization algorithms on eight benchmark functions and TFBS problem. WCC is implementable on the problems which can’t be solved in reasonable time order (NP problems).
Collapse
Affiliation(s)
- Yosef Masoudi-Sobhanzadeh
- Department of Computer Engineering, Payam Noor University, Tabriz, Iran.,Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Habib Motieghader
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| |
Collapse
|
50
|
Renault TT, Dejean LM, Manon S. A brewing understanding of the regulation of Bax function by Bcl-xL and Bcl-2. Mech Ageing Dev 2016; 161:201-210. [PMID: 27112371 DOI: 10.1016/j.mad.2016.04.007] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/19/2016] [Accepted: 04/22/2016] [Indexed: 12/17/2022]
Abstract
Bcl-2 family members form a network of protein-protein interactions that regulate apoptosis through permeabilization of the mitochondrial outer membrane. Deciphering this intricate network requires streamlined experimental models, including the heterologous expression in yeast. This approach had previously enabled researchers to identify domains and residues that underlie the conformational changes driving the translocation, the insertion and the oligomerization of the pro-apoptotic protein Bax at the level of the mitochondrial outer membrane. Recent studies that combine experiments in yeast and in mammalian cells have shown the unexpected effect of the anti-apoptotic protein Bcl-xL on the priming of Bax. As demonstrated with the BH3-mimetic molecule ABT-737, this property of Bcl-xL, and of Bcl-2, is crucial to elaborate about how apoptosis could be reactivated in tumoral cells.
Collapse
Affiliation(s)
- Thibaud T Renault
- Helmholtz Center for Infection Research, Junior Research Group Infection Biology of Salmonella, Inhoffenstraße 7, 38124 Braunschweig, Germany; Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Laurent M Dejean
- California State University of Fresno, Department of Chemistry, 2555 E. San Ramon Ave M/S SB70, Fresno, CA 93740-8034, USA
| | - Stéphen Manon
- CNRS, UMR5095, 1 Rue Camille Saint-Saëns, 33077 Bordeaux, France; Université de Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, France.
| |
Collapse
|