1
|
Rühl S, Li Z, Srivastava S, Mari L, Guy CS, Yang M, Moldoveanu T, Green DR. Inhibition of BAK-mediated apoptosis by the BH3-only protein BNIP5. Cell Death Differ 2025; 32:320-336. [PMID: 39406920 PMCID: PMC11803101 DOI: 10.1038/s41418-024-01386-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 11/16/2024] Open
Abstract
BCL-2 family proteins regulate apoptosis by initiating mitochondrial outer membrane permeabilization (MOMP). Activation of the MOMP effectors BAX and BAK is controlled by the interplay of anti-apoptotic BCL-2 proteins (e.g., MCL-1) and pro-apoptotic BH3-only proteins (e.g., BIM). Using a genome-wide CRISPR-dCas9 transactivation screen we identified BNIP5 as an inhibitor of BAK-, but not BAX-induced apoptosis. BNIP5 blocked BAK activation in different cell types and in response to various cytotoxic therapies. The BH3 domain of BNIP5 was both necessary and sufficient to block BAK activation. Mechanistically, the BH3 domain of BNIP5 acts as a selective BAK activator, but a poor de-repressor of complexes between BAK and pro-survival BCL-2 family proteins. By promoting the binding of activated BAK to MCL-1 or BCL-xL, BNIP5 inhibits apoptosis when BAX is absent. Based on our observations, BNIP5 can act functionally as an anti-apoptotic BH3-only protein.
Collapse
Affiliation(s)
- Sebastian Rühl
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
- T3 Pharmaceuticals, Allschwil, Switzerland
| | - Zhenrui Li
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Shagun Srivastava
- Department of Biochemistry and Molecular Biology, UAMS College of Medicine, Little Rock, AR, 72205, USA
| | - Luigi Mari
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Clifford S Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mao Yang
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Tudor Moldoveanu
- Department of Biochemistry and Molecular Biology, UAMS College of Medicine, Little Rock, AR, 72205, USA
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
2
|
Suraweera CD, Espinoza B, Hinds MG, Kvansakul M. Mastering Death: The Roles of Viral Bcl-2 in dsDNA Viruses. Viruses 2024; 16:879. [PMID: 38932171 PMCID: PMC11209288 DOI: 10.3390/v16060879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Proteins of the Bcl-2 family regulate cellular fate via multiple mechanisms including apoptosis, autophagy, senescence, metabolism, inflammation, redox homeostasis, and calcium flux. There are several regulated cell death (RCD) pathways, including apoptosis and autophagy, that use distinct molecular mechanisms to elicit the death response. However, the same proteins/genes may be deployed in multiple biochemical pathways. In apoptosis, Bcl-2 proteins control the integrity of the mitochondrial outer membrane (MOM) by regulating the formation of pores in the MOM and apoptotic cell death. A number of prosurvival genes populate the genomes of viruses including those of the pro-survival Bcl-2 family. Viral Bcl-2 proteins are sequence and structural homologs of their cellular counterparts and interact with cellular proteins in apoptotic and autophagic pathways, potentially allowing them to modulate these pathways and determine cellular fate.
Collapse
Affiliation(s)
- Chathura D. Suraweera
- Genome Sciences and Cancer Division, The John Curtin School of Medical Research, Australian National University, Canberra 2601, Australia;
| | - Benjamin Espinoza
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, VIC 3086, Australia;
| | - Mark G. Hinds
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Marc Kvansakul
- Genome Sciences and Cancer Division, The John Curtin School of Medical Research, Australian National University, Canberra 2601, Australia;
| |
Collapse
|
3
|
Zhan J, Wang J, Liang Y, Wang L, Huang L, Liu S, Zeng X, Zeng E, Wang H. Apoptosis dysfunction: unravelling the interplay between ZBP1 activation and viral invasion in innate immune responses. Cell Commun Signal 2024; 22:149. [PMID: 38402193 PMCID: PMC10893743 DOI: 10.1186/s12964-024-01531-y] [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: 11/29/2023] [Accepted: 02/13/2024] [Indexed: 02/26/2024] Open
Abstract
Apoptosis plays a pivotal role in pathogen elimination and maintaining homeostasis. However, viruses have evolved strategies to evade apoptosis, enabling their persistence within the host. Z-DNA binding protein 1 (ZBP1) is a potent innate immune sensor that detects cytoplasmic nucleic acids and activates the innate immune response to clear pathogens. When apoptosis is inhibited by viral invasion, ZBP1 can be activated to compensate for the effect of apoptosis by triggering an innate immune response. This review examined the mechanisms of apoptosis inhibition and ZBP1 activation during viral invasion. The authors outlined the mechanisms of ZBP1-induced type I interferon, pyroptosis and necroptosis, as well as the crosstalk between ZBP1 and the cGAS-STING signalling pathway. Furthermore, ZBP1 can reverse the suppression of apoptotic signals induced by viruses. Intriguingly, a positive feedback loop exists in the ZBP1 signalling pathway, which intensifies the innate immune response while triggering a cytokine storm, leading to tissue and organ damage. The prudent use of ZBP1, which is a double-edged sword, has significant clinical implications for treating infections and inflammation.
Collapse
Affiliation(s)
- Jianhao Zhan
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- HuanKui Academy, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Jisheng Wang
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Yuqing Liang
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Lisha Wang
- HuanKui Academy, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Le Huang
- HuanKui Academy, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Shanshan Liu
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Xiaoping Zeng
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Medical College, Jinhua Polytechnic, Jinhua, Zhejiang Province, 321017, China
| | - Erming Zeng
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China.
| | - Hongmei Wang
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China.
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi Province, 330006, China.
- Medical College, Jinhua Polytechnic, Jinhua, Zhejiang Province, 321017, China.
| |
Collapse
|
4
|
Muthye V, Wasmuth JD. Proteome-wide comparison of tertiary protein structures reveals molecular mimicry in Plasmodium-human interactions. FRONTIERS IN PARASITOLOGY 2023; 2:1162697. [PMID: 39816809 PMCID: PMC11732093 DOI: 10.3389/fpara.2023.1162697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/05/2023] [Indexed: 01/18/2025]
Abstract
Introduction Molecular mimicry is a strategy used by parasites to evade the host's immune system and facilitate transmission to a new host. To date, high-throughput examples of molecular mimicry have been limited to comparing protein sequences. However, recent advances in the prediction of tertiary structural models, led by Deepmind's AlphaFold, enable the comparison of thousands of proteins from parasites and their hosts at the structural level, allowing for the identification of more mimics. Here, we present the first proteome-level search for tertiary structure similarity between proteins from Plasmodium falciparum, a malaria-causing parasite, and humans. Methods We assembled a database of experimentally-characterized protein tertiary structures (from the Protein Data Bank) and AlphaFold-generated protein tertiary structures from P. falciparum, human, and 15 negative control species, i.e., species not infected by P. falciparum. We aligned human and control structures to the parasite structures using Foldseek. Results We identified molecular mimicry in three proteins that have been previously proposed as mediators of Plasmodium-human interactions. By extending this approach to all P. falciparum proteins, we identified an additional 41 potential mimics that are supported by additional experimental data. Discussion Our findings demonstrate a valuable application of AlphaFold-derived tertiary structural models, and we discuss key considerations for its effective use in other host-parasite systems.
Collapse
Affiliation(s)
- Viraj Muthye
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions Research Training Network, University of Calgary, Calgary, AB, Canada
| | - James D. Wasmuth
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions Research Training Network, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
5
|
Nainu F, Ophinni Y, Shiratsuchi A, Nakanishi Y. Apoptosis and Phagocytosis as Antiviral Mechanisms. Subcell Biochem 2023; 106:77-112. [PMID: 38159224 DOI: 10.1007/978-3-031-40086-5_3] [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: 01/03/2024]
Abstract
Viruses are infectious entities that make use of the replication machinery of their hosts to produce more progenies, causing disease and sometimes death. To counter viral infection, metazoan hosts are equipped with various defense mechanisms, from the rapid-evoking innate immune responses to the most advanced adaptive immune responses. Previous research demonstrated that cells in fruit flies and mice infected with Drosophila C virus and influenza, respectively, undergo apoptosis, which triggers the engulfment of apoptotic virus-infected cells by phagocytes. This process involves the recognition of eat-me signals on the surface of virus-infected cells by receptors of specialized phagocytes, such as macrophages and neutrophils in mice and hemocytes in fruit flies, to facilitate the phagocytic elimination of virus-infected cells. Inhibition of phagocytosis led to severe pathologies and death in both species, indicating that apoptosis-dependent phagocytosis of virus-infected cells is a conserved antiviral mechanism in multicellular organisms. Indeed, our understanding of the mechanisms underlying apoptosis-dependent phagocytosis of virus-infected cells has shed a new perspective on how hosts defend themselves against viral infection. This chapter explores the mechanisms of this process and its potential for developing new treatments for viral diseases.
Collapse
Affiliation(s)
- Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia.
| | - Youdiil Ophinni
- Division of Clinical Virology, Center for Infectious Diseases, Kobe University Graduate School of Medicine, Kobe, Japan
- Laboratory of Host Defense, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Akiko Shiratsuchi
- Center for Medical Education, Sapporo Medical University, Sapporo, Japan
- Division of Biological Function and Regulation, Graduate School of Medicine, Sapporo Medical University, Sapporo, Japan
| | | |
Collapse
|
6
|
Suraweera CD, Hinds MG, Kvansakul M. Crystal Structures of Epstein-Barr Virus Bcl-2 Homolog BHRF1 Bound to Bid and Puma BH3 Motif Peptides. Viruses 2022; 14:v14102222. [PMID: 36298777 PMCID: PMC9609553 DOI: 10.3390/v14102222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/27/2022] [Accepted: 10/04/2022] [Indexed: 11/09/2022] Open
Abstract
Apoptosis is a powerful defense mechanism used by multicellular organisms to counteract viral infection. In response to premature host cell suicide, viruses have evolved numerous countermeasures to ensure cell viability to optimize their replication by encoding proteins homologous in structure and function to cellular pro-survival Bcl-2 proteins. Epstein-Barr virus (EBV), a member of the Gammaherpesviridae, encodes the Bcl-2 homolog BHRF1, a potent inhibitor of Bcl-2-mediated apoptosis. BHRF1 acts by directly targeting Bid and Puma, two proapoptotic proteins of the Bcl-2 family. Here, we determined the crystal structures of BHRF1 bound to peptides spanning the Bcl-2 binding motifs (Bcl-2 homology 3 motif, BH3) of Bid and Puma. BHRF1 engages BH3 peptides using the canonical ligand-binding groove of its Bcl-2 fold and maintains a salt bridge between an Arg residue with a conserved Asp residue in the BH3 motif mimicking the canonical ionic interaction seen in host Bcl-2:BH3 motif complexes. Furthermore, both Bid and Puma utilize a fifth binding pocket in the canonical ligand binding groove of BHRF1 to provide an additional hydrophobic interaction distinct from the interactions previously seen with Bak and Bim. These findings provide a structural basis for EBV-mediated suppression of host cell apoptosis and reveal the flexibility of virus encoded Bcl-2 proteins in mimicking key interactions from the endogenous host signaling pathways.
Collapse
Affiliation(s)
- Chathura D. Suraweera
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Mark G. Hinds
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
- Correspondence: (M.G.H.); (M.K.)
| | - Marc Kvansakul
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
- Correspondence: (M.G.H.); (M.K.)
| |
Collapse
|
7
|
Wolf P, Schoeniger A, Edlich F. Pro-apoptotic complexes of BAX and BAK on the outer mitochondrial membrane. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119317. [PMID: 35752202 DOI: 10.1016/j.bbamcr.2022.119317] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/02/2022] [Accepted: 06/15/2022] [Indexed: 06/15/2023]
Abstract
In multicellular organisms the regulated cell death apoptosis is critically important for both ontogeny and homeostasis. Mitochondria are indispensable for stress-induced apoptosis. The BCL-2 protein family controls mitochondrial apoptosis and initiates cell death through the pro-apoptotic activities of BAX and BAK at the outer mitochondrial membrane (OMM). Cellular survival is ensured by the retrotranslocation of mitochondrial BAX and BAK into the cytosol by anti-apoptotic BCL-2 proteins. BAX/BAK-dependent OMM permeabilization releases the mitochondrial cytochrome c (cyt c), which initiates activation of caspase-9. The caspase cascade leads to cell shrinkage, plasma membrane blebbing, chromatin condensation, and apoptotic body formation. Although it is clear that ultimately complexes of active BAX and BAK commit the cell to apoptosis, the nature of these complexes is still enigmatic. Excessive research has described a range of complexes, varying from a few molecules to several 10,000, in different systems. BAX/BAK complexes potentially form ring-like structures that could expose the inner mitochondrial membrane. It has been suggested that these pores allow the efflux of small proteins and even mitochondrial DNA. Here we summarize the current state of knowledge for mitochondrial BAX/BAK complexes and the interactions between these proteins and the membrane.
Collapse
Affiliation(s)
- Philipp Wolf
- Institute of Biochemistry, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Axel Schoeniger
- Institute of Biochemistry, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Frank Edlich
- Institute of Biochemistry, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany.
| |
Collapse
|
8
|
Verburg SG, Lelievre RM, Westerveld MJ, Inkol JM, Sun YL, Workenhe ST. Viral-mediated activation and inhibition of programmed cell death. PLoS Pathog 2022; 18:e1010718. [PMID: 35951530 PMCID: PMC9371342 DOI: 10.1371/journal.ppat.1010718] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Viruses are ubiquitous intracellular genetic parasites that heavily rely on the infected cell to complete their replication life cycle. This dependency on the host machinery forces viruses to modulate a variety of cellular processes including cell survival and cell death. Viruses are known to activate and block almost all types of programmed cell death (PCD) known so far. Modulating PCD in infected hosts has a variety of direct and indirect effects on viral pathogenesis and antiviral immunity. The mechanisms leading to apoptosis following virus infection is widely studied, but several modalities of PCD, including necroptosis, pyroptosis, ferroptosis, and paraptosis, are relatively understudied. In this review, we cover the mechanisms by which viruses activate and inhibit PCDs and suggest perspectives on how these affect viral pathogenesis and immunity.
Collapse
Affiliation(s)
- Shayla Grace Verburg
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Canada
| | | | | | - Jordon Marcus Inkol
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Canada
| | - Yi Lin Sun
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Canada
| | - Samuel Tekeste Workenhe
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Canada
| |
Collapse
|
9
|
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
|
10
|
Suraweera CD, Hinds MG, Kvansakul M. Structural Insight into KsBcl-2 Mediated Apoptosis Inhibition by Kaposi Sarcoma Associated Herpes Virus. Viruses 2022; 14:v14040738. [PMID: 35458468 PMCID: PMC9027176 DOI: 10.3390/v14040738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/23/2022] [Accepted: 03/29/2022] [Indexed: 02/05/2023] Open
Abstract
Numerous large DNA viruses have evolved sophisticated countermeasures to hijack the premature programmed cell death of host cells post-infection, including the expression of proteins homologous in sequence, structure, or function to cellular Bcl-2 proteins. Kaposi sarcoma herpes virus (KSHV), a member of the gammaherpesvirinae, has been shown to encode for KsBcl-2, a potent inhibitor of Bcl-2 mediated apoptosis. KsBcl-2 acts by directly engaging host pro-apoptotic Bcl-2 proteins including Bak, Bax and Bok, the BH3-only proteins; Bim, Bid, Bik, Hrk, Noxa and Puma. Here we determined the crystal structures of KsBcl-2 bound to the BH3 motif of pro-apoptotic proteins Bid and Puma. The structures reveal that KsBcl-2 engages pro-apoptotic BH3 motif peptides using the canonical ligand binding groove. Thus, the presence of the readily identifiable conserved BH1 motif sequence “NWGR” of KsBcl-2, as well as highly conserved Arg residue (R86) forms an ionic interaction with the conserved Asp in the BH3 motif in a manner that mimics the canonical ionic interaction seen in host Bcl-2:BH3 motif complexes. These findings provide a structural basis for KSHV mediated inhibition of host cell apoptosis and reveal the flexibility of virus encoded Bcl-2 proteins to mimic key interactions from endogenous host signalling pathways.
Collapse
Affiliation(s)
- Chathura D. Suraweera
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia;
| | - Mark G. Hinds
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3052, Australia
- Correspondence: (M.G.H.); (M.K.)
| | - Marc Kvansakul
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia;
- Correspondence: (M.G.H.); (M.K.)
| |
Collapse
|
11
|
Reddy C, Sankararamakrishnan R. Designing BH3-Mimetic Peptide Inhibitors for the Viral Bcl-2 Homologues A179L and BHRF1: Importance of Long-Range Electrostatic Interactions. ACS OMEGA 2021; 6:26976-26989. [PMID: 34693118 PMCID: PMC8529603 DOI: 10.1021/acsomega.1c03385] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/16/2021] [Indexed: 06/13/2023]
Abstract
Viruses have evolved strategies to prevent apoptosis of infected cells at early stages of infection. The viral proteins (vBcl-2s) from specific viral genes adopt a helical fold that is structurally similar to that of mammalian antiapoptotic Bcl-2 proteins and exhibit little sequence similarity. Hence, vBcl-2 homologues are attractive targets to prevent viral infection. However, very few studies have focused on developing inhibitors for vBcl-2 homologues. In this study, we have considered two vBcl-2 homologues, A179L from African swine fever virus and BHRF1 from Epstein-Barr virus. We generated two sets of 8000 randomized BH3-like sequences from eight wild-type proapoptotic BH3 peptides. During this process, the four conserved hydrophobic residues and an Asp residue were retained at their respective positions, and all other positions were substituted randomly without any bias. We constructed 8000 structures each for A179L and BHRF1 in complex with BH3-like sequences. Histograms of interaction energies calculated between the peptide and the protein resulted in negatively skewed distributions. The BH3-like peptides with high helical propensities selected from the negative tail of the respective interaction energy distributions exhibited more favorable interactions with A179L and BHRF1, and they are rich in basic residues. Molecular dynamics studies and electrostatic potential maps further revealed that both acidic and basic residues favorably interact with A179L, while only basic residues have the most favorable interactions with BHRF1. As in mammalian homologues, the role of long-range interactions and nonhotspot residues has to be taken into account while designing specific BH3-mimetic inhibitors for vBcl-2 homologues.
Collapse
Affiliation(s)
- Chinthakunta
Narendra Reddy
- Department
of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Ramasubbu Sankararamakrishnan
- Department
of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
- Mehta
Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, India
| |
Collapse
|
12
|
Structural Investigation of Orf Virus Bcl-2 Homolog ORFV125 Interactions with BH3-Motifs from BH3-Only Proteins Puma and Hrk. Viruses 2021; 13:v13071374. [PMID: 34372579 PMCID: PMC8310162 DOI: 10.3390/v13071374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 12/21/2022] Open
Abstract
Numerous viruses have evolved sophisticated countermeasures to hijack the early programmed cell death of host cells in response to infection, including the use of proteins homologous in sequence or structure to Bcl-2. Orf virus, a member of the parapoxviridae, encodes for the Bcl-2 homolog ORFV125, a potent inhibitor of Bcl-2-mediated apoptosis in the host. ORFV125 acts by directly engaging host proapoptotic Bcl-2 proteins including Bak and Bax as well as the BH3-only proteins Hrk and Puma. Here, we determined the crystal structures of ORFV125 bound to the BH3 motif of proapoptotic proteins Puma and Hrk. The structures reveal that ORFV125 engages proapoptotic BH3 motif peptides using the canonical ligand binding groove. An Arg located in the structurally equivalent BH1 region of ORFV125 forms an ionic interaction with the conserved Asp in the BH3 motif in a manner that mimics the canonical ionic interaction seen in host Bcl-2:BH3 motif complexes. These findings provide a structural basis for Orf virus-mediated inhibition of host cell apoptosis and reveal the flexibility of virus encoded Bcl-2 proteins to mimic key interactions from endogenous host signalling pathways.
Collapse
|
13
|
Fang Y, Peng K. Regulation of innate immune responses by cell death-associated caspases during virus infection. FEBS J 2021; 289:4098-4111. [PMID: 34089572 DOI: 10.1111/febs.16051] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/04/2021] [Accepted: 06/03/2021] [Indexed: 01/04/2023]
Abstract
Viruses are obligate intracellular pathogens that rely on cellular machinery for successful replication and dissemination. The host cells encode a number of different strategies to sense and restrict the invading viral pathogens. Caspase-mediated programmed cell death pathways that are triggered by virus infection, such as apoptosis and pyroptosis, provide a means for the infected cells to limit viral proliferation, leading to suicidal cell death (apoptosis) or lytic cell death and alerting uninfected cells to mount anti-viral responses (pyroptosis). However, some viruses can employ activated caspases to dampen the anti-viral responses and facilitate viral replication through cleavage of critical molecules of the innate immune pathways. The regulation of innate immune responses by caspase activation during virus infection has recently become an important topic. In this review, we briefly introduce the characteristics of different classes of caspases and the cell death pathways regulated by these caspases. We then describe how viruses trigger or dampen caspase activation during infection and how these activated caspases regulate three major innate immune response pathways of viral infections: the retinoic acid-inducible gene I-like receptor, toll-like receptor and cyclic GMP-AMP synthase-stimulator of interferon genes pathways.
Collapse
Affiliation(s)
- Yujie Fang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ke Peng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
14
|
Crystal structures of ORFV125 provide insight into orf virus-mediated inhibition of apoptosis. Biochem J 2021; 477:4527-4541. [PMID: 33175095 PMCID: PMC7719400 DOI: 10.1042/bcj20200776] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022]
Abstract
Premature apoptosis of cells is a strategy utilized by multicellular organisms to counter microbial threats. Orf virus (ORFV) is a large double-stranded DNA virus belonging to the poxviridae. ORFV encodes for an apoptosis inhibitory protein ORFV125 homologous to B-cell lymphoma 2 or Bcl-2 family proteins, which has been shown to inhibit host cell encoded pro-apoptotic Bcl-2 proteins. However, the structural basis of apoptosis inhibition by ORFV125 remains to be clarified. We show that ORFV125 is able to bind to a range of peptides spanning the BH3 motif of human pro-apoptotic Bcl-2 proteins including Bax, Bak, Puma and Hrk with modest to weak affinity. We then determined the crystal structures of ORFV125 alone as well as bound to the highest affinity ligand Bax BH3 motif. ORFV125 adopts a globular Bcl-2 fold comprising 7 α-helices, and utilizes the canonical Bcl-2 binding groove to engage pro-apoptotic host cell Bcl-2 proteins. In contrast with a previously predicted structure, ORFV125 adopts a domain-swapped dimeric topology, where the α1 helix from one protomer is swapped into a neighbouring unit. Furthermore, ORFV125 differs from the conserved architecture of the Bcl-2 binding groove and instead of α3 helix forming one of the binding groove walls, ORFV125 utilizes an extended α2 helix that comprises the equivalent region of helix α3. This results in a subtle variation of previously observed dimeric Bcl-2 architectures in other poxvirus and human encoded Bcl-2 proteins. Overall, our results provide a structural and mechanistic basis for orf virus-mediated inhibition of host cell apoptosis.
Collapse
|
15
|
Quarleri J, Cevallos C, Delpino MV. Apoptosis in infectious diseases as a mechanism of immune evasion and survival. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 125:1-24. [PMID: 33931136 DOI: 10.1016/bs.apcsb.2021.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In pluricellular organisms, apoptosis is indispensable for the development and homeostasis. During infection, apoptosis plays the main role in the elimination of infected cells. Infectious diseases control apoptosis, and this contributes to disease pathogenesis. Increased apoptosis may participate in two different ways. It can assist the dissemination of intracellular pathogens or induce immunosuppression to favor pathogen dissemination. In other conditions, apoptosis can benefit eradicate infectious agents from the host. Accordingly, bacteria, viruses, fungi, and parasites have developed strategies to inhibit host cell death by apoptosis to allow intracellular survival and persistence of the pathogen. The clarification of the intracellular signaling pathways, the receptors involved and the pathogen factors that interfere with apoptosis could disclose new therapeutic targets for blocking microbial actions on apoptotic pathways. In this review, we summarize the current knowledge on pathogen anti-apoptotic and apoptotic approaches and the mechanisms involving in disease.
Collapse
Affiliation(s)
- Jorge Quarleri
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Cintia Cevallos
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - María Victoria Delpino
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
16
|
Suraweera CD, Hinds MG, Kvansakul M. Poxviral Strategies to Overcome Host Cell Apoptosis. Pathogens 2020; 10:pathogens10010006. [PMID: 33374867 PMCID: PMC7823800 DOI: 10.3390/pathogens10010006] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 12/17/2022] Open
Abstract
Apoptosis is a form of cellular suicide initiated either via extracellular (extrinsic apoptosis) or intracellular (intrinsic apoptosis) cues. This form of programmed cell death plays a crucial role in development and tissue homeostasis in multicellular organisms and its dysregulation is an underlying cause for many diseases. Intrinsic apoptosis is regulated by members of the evolutionarily conserved B-cell lymphoma-2 (Bcl-2) family, a family that consists of pro- and anti-apoptotic members. Bcl-2 genes have also been assimilated by numerous viruses including pox viruses, in particular the sub-family of chordopoxviridae, a group of viruses known to infect almost all vertebrates. The viral Bcl-2 proteins are virulence factors and aid the evasion of host immune defenses by mimicking the activity of their cellular counterparts. Viral Bcl-2 genes have proved essential for the survival of virus infected cells and structural studies have shown that though they often share very little sequence identity with their cellular counterparts, they have near-identical 3D structures. However, their mechanisms of action are varied. In this review, we examine the structural biology, molecular interactions, and detailed mechanism of action of poxvirus encoded apoptosis inhibitors and how they impact on host–virus interactions to ultimately enable successful infection and propagation of viral infections.
Collapse
Affiliation(s)
- Chathura D. Suraweera
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia;
| | - Mark G. Hinds
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3052, Australia
- Correspondence: (M.G.H.); (M.K.)
| | - Marc Kvansakul
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia;
- Correspondence: (M.G.H.); (M.K.)
| |
Collapse
|
17
|
Cui C, Liang Q, Tang X, Xing J, Sheng X, Zhan W. Differential Apoptotic Responses of Hemocyte Subpopulations to White Spot Syndrome Virus Infection in Fenneropenaeus chinensis. Front Immunol 2020; 11:594390. [PMID: 33365030 PMCID: PMC7750459 DOI: 10.3389/fimmu.2020.594390] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/05/2020] [Indexed: 11/13/2022] Open
Abstract
The apoptosis of hemocytes plays an essential function in shrimp immune defense against pathogen invasions. In order to further elucidate the differential apoptotic responses of the granulocytes and the hyalinocytes in Fenneropenaeus chinensis post WSSV infection, the characteristics of apoptotic dynamics and viral proliferation in total hemocytes and hemocyte subpopulations were respectively investigated in the present work. The results showed that the apoptotic rate of hemocytes changed significantly, and the apoptosis-related genes also showed significantly differential expression responses during WSSV infection. Interestingly, we found that the apoptotic rate of virus-negative hemocytes was significantly higher than that of virus-positive hemocytes in the early stage of WSSV infection, while it was significantly lower than that of virus-positive cells in the middle and late infection stages. The difference of apoptosis between virus-positive and virus-negative hemocytes seems to be an important way for the WSSV to destroy the host’s immune system and facilitate the virus spread at different infection stages. It was further found that the apoptosis rate of granulocytes was always significantly higher than that of hyalinocytes during WSSV infection, indicating that granulocytes have a stronger apoptotic response to WSSV infection. Moreover, a higher viral load was detected in granulocytes, and the density of granulocytes decreased more rapidly post WSSV infection, indicating that the granulocytes are more susceptible and vulnerable to WSSV infection compared with the hyalinocytes. These results collectively demonstrated that the apoptotic response in shrimp hemocytes was significantly influenced by the WSSV infection, and the differential apoptotic response of granulocytes and hyalinocytes to WSSV indicated the differences of antiviral mechanisms between the two hemocyte subpopulations.
Collapse
Affiliation(s)
- Chuang Cui
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
| | - Qianrong Liang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
18
|
García-Murria MJ, Duart G, Grau B, Diaz-Beneitez E, Rodríguez D, Mingarro I, Martínez-Gil L. Viral Bcl2s' transmembrane domain interact with host Bcl2 proteins to control cellular apoptosis. Nat Commun 2020; 11:6056. [PMID: 33247105 PMCID: PMC7695858 DOI: 10.1038/s41467-020-19881-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 10/29/2020] [Indexed: 12/20/2022] Open
Abstract
Viral control of programmed cell death relies in part on the expression of viral analogs of the B-cell lymphoma 2 (Bcl2) protein known as viral Bcl2s (vBcl2s). vBcl2s control apoptosis by interacting with host pro- and anti-apoptotic members of the Bcl2 family. Here, we show that the carboxyl-terminal hydrophobic region of herpesviral and poxviral vBcl2s can operate as transmembrane domains (TMDs) and participate in their homo-oligomerization. Additionally, we show that the viral TMDs mediate interactions with cellular pro- and anti-apoptotic Bcl2 TMDs within the membrane. Furthermore, these intra-membrane interactions among viral and cellular proteins are necessary to control cell death upon an apoptotic stimulus. Therefore, their inhibition represents a new potential therapy against viral infections, which are characterized by short- and long-term deregulation of programmed cell death.
Collapse
Affiliation(s)
- Maria Jesús García-Murria
- Department of Biochemistry and Molecular Biology, Institut de Biotecnologia i Biomedicina, Universitat de València, 46100, Burjassot, Spain
| | - Gerard Duart
- Department of Biochemistry and Molecular Biology, Institut de Biotecnologia i Biomedicina, Universitat de València, 46100, Burjassot, Spain
| | - Brayan Grau
- Department of Biochemistry and Molecular Biology, Institut de Biotecnologia i Biomedicina, Universitat de València, 46100, Burjassot, Spain
| | - Elisabet Diaz-Beneitez
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Campus Universidad Autónoma, 28049, Madrid, Spain
| | - Dolores Rodríguez
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Campus Universidad Autónoma, 28049, Madrid, Spain
| | - Ismael Mingarro
- Department of Biochemistry and Molecular Biology, Institut de Biotecnologia i Biomedicina, Universitat de València, 46100, Burjassot, Spain
| | - Luis Martínez-Gil
- Department of Biochemistry and Molecular Biology, Institut de Biotecnologia i Biomedicina, Universitat de València, 46100, Burjassot, Spain.
| |
Collapse
|
19
|
Lasso G, Honig B, Shapira SD. A Sweep of Earth's Virome Reveals Host-Guided Viral Protein Structural Mimicry and Points to Determinants of Human Disease. Cell Syst 2020; 12:82-91.e3. [PMID: 33053371 PMCID: PMC7552982 DOI: 10.1016/j.cels.2020.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/03/2020] [Accepted: 09/18/2020] [Indexed: 12/17/2022]
Abstract
Viruses deploy genetically encoded strategies to coopt host machinery and support viral replicative cycles. Here, we use protein structure similarity to scan for molecular mimicry, manifested by structural similarity between viral and endogenous host proteins, across thousands of cataloged viruses and hosts spanning broad ecological niches and taxonomic range, including bacteria, plants and fungi, invertebrates, and vertebrates. This survey identified over 6,000,000 instances of structural mimicry; more than 70% of viral mimics cannot be discerned through protein sequence alone. We demonstrate that the manner and degree to which viruses exploit molecular mimicry varies by genome size and nucleic acid type and identify 158 human proteins that are mimicked by coronaviruses, providing clues about cellular processes driving pathogenesis. Our observations point to molecular mimicry as a pervasive strategy employed by viruses and indicate that the protein structure space used by a given virus is dictated by the host proteome. A record of this paper's transparent peer review process is included in the Supplemental Information.
Collapse
Affiliation(s)
- Gorka Lasso
- Department of Systems Biology, Columbia University Medical Center, New York, NY, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, USA; Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA
| | - Barry Honig
- Department of Systems Biology, Columbia University Medical Center, New York, NY, USA; Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, NY, USA; Zuckerman Mind Brain Behavior Institute, Columbia University Medical Center, New York, NY, USA; Department of Medicine, Columbia University, New York, NY, USA
| | - Sagi D Shapira
- Department of Systems Biology, Columbia University Medical Center, New York, NY, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
20
|
Diversity in the intrinsic apoptosis pathway of nematodes. Commun Biol 2020; 3:478. [PMID: 32859965 PMCID: PMC7456325 DOI: 10.1038/s42003-020-01208-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/03/2020] [Indexed: 11/08/2022] Open
Abstract
Early studies of the free-living nematode C. elegans informed us how BCL-2-regulated apoptosis in humans is regulated. However, subsequent studies showed C. elegans apoptosis has several unique features compared with human apoptosis. To date, there has been no detailed analysis of apoptosis regulators in nematodes other than C. elegans. Here, we discovered BCL-2 orthologues in 89 free-living and parasitic nematode taxa representing four evolutionary clades (I, III, IV and V). Unlike in C. elegans, 15 species possess multiple (two to five) BCL-2-like proteins, and some do not have any recognisable BCL-2 sequences. Functional studies provided no evidence that BAX/BAK proteins have evolved in nematodes, and structural studies of a BCL-2 protein from the basal clade I revealed it lacks a functionally important feature of the C. elegans orthologue. Clade I CED-4/APAF-1 proteins also possess WD40-repeat sequences associated with apoptosome assembly, not present in C. elegans, or other nematode taxa studied.
Collapse
|
21
|
Suraweera CD, Anasir MI, Chugh S, Javorsky A, Impey RE, Hasan Zadeh M, Soares da Costa TP, Hinds MG, Kvansakul M. Structural insight into tanapoxvirus-mediated inhibition of apoptosis. FEBS J 2020; 287:3733-3750. [PMID: 32412687 DOI: 10.1111/febs.15365] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/26/2020] [Accepted: 05/12/2020] [Indexed: 01/07/2023]
Abstract
Premature programmed cell death or apoptosis of cells is a strategy utilized by multicellular organisms to counter microbial threats. Tanapoxvirus (TANV) is a large double-stranded DNA virus belonging to the poxviridae that causes mild monkeypox-like infections in humans and primates. TANV encodes for a putative apoptosis inhibitory protein 16L. We show that TANV16L is able to bind to a range of peptides spanning the BH3 motif of human proapoptotic Bcl-2 proteins and is able to counter growth arrest of yeast induced by human Bak and Bax. We then determined the crystal structures of TANV16L bound to three identified interactors, Bax, Bim and Puma BH3. TANV16L adopts a globular Bcl-2 fold comprising 7 α-helices and utilizes the canonical Bcl-2 binding groove to engage proapoptotic host cell Bcl-2 proteins. Unexpectedly, TANV16L is able to adopt both a monomeric and a domain-swapped dimeric topology where the α1 helix from one protomer is swapped into a neighbouring unit. Despite adopting two different oligomeric forms, the canonical ligand binding groove in TANV16L remains unchanged from monomer to domain-swapped dimer. Our results provide a structural and mechanistic basis for tanapoxvirus-mediated inhibition of host cell apoptosis and reveal the capacity of Bcl-2 proteins to adopt differential oligomeric states whilst maintaining the canonical ligand binding groove in an unchanged state. DATABASE: Structural data are available in the Protein Data Bank (PDB) under the accession numbers 6TPQ, 6TQQ and 6TRR.
Collapse
Affiliation(s)
- Chathura D Suraweera
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Mohd Ishtiaq Anasir
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Srishti Chugh
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Airah Javorsky
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Rachael E Impey
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Mohammad Hasan Zadeh
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Tatiana P Soares da Costa
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Mark G Hinds
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - Marc Kvansakul
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| |
Collapse
|
22
|
Suraweera CD, Burton DR, Hinds MG, Kvansakul M. Crystal structures of the sheeppox virus encoded inhibitor of apoptosis SPPV14 bound to the proapoptotic BH3 peptides Hrk and Bax. FEBS Lett 2020; 594:2016-2026. [PMID: 32390192 DOI: 10.1002/1873-3468.13807] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022]
Abstract
Programmed death of infected cells is used by multicellular organisms to counter viral infections. Sheeppox virus encodes for SPPV14, a potent inhibitor of Bcl-2-mediated apoptosis. We reveal the structural basis of apoptosis inhibition by determining crystal structures of SPPV14 bound to BH3 motifs of proapoptotic Bax and Hrk. The structures show that SPPV14 engages BH3 peptides using the canonical ligand-binding groove. Unexpectedly, Arg84 from SPPV14 forms an ionic interaction with the conserved Asp in the BH3 motif in a manner that replaces the canonical ionic interaction seen in almost all host Bcl-2:BH3 motif complexes. These results reveal the flexibility of virus-encoded Bcl-2 proteins to mimic key interactions from endogenous host signalling pathways to retain BH3 binding and prosurvival functionality.
Collapse
Affiliation(s)
- Chathura D Suraweera
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Vic., Australia
| | - Denis R Burton
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Vic., Australia
| | - Mark G Hinds
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Vic., Australia
| | - Marc Kvansakul
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Vic., Australia
| |
Collapse
|
23
|
Neidel S, Torres AA, Ren H, Smith GL. Leaky scanning translation generates a second A49 protein that contributes to vaccinia virus virulence. J Gen Virol 2020; 101:533-541. [PMID: 32100702 PMCID: PMC7414448 DOI: 10.1099/jgv.0.001386] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 01/08/2020] [Indexed: 12/17/2022] Open
Abstract
Vaccinia virus (VACV) strain Western Reserve gene A49L encodes a small intracellular protein with a Bcl-2 fold that is expressed early during infection and has multiple functions. A49 co-precipitates with the E3 ubiquitin ligase β-TrCP and thereby prevents ubiquitylation and proteasomal degradation of IκBα, and consequently blocks activation of NF-κB. In a similar way, A49 stabilizes β-catenin, leading to activation of the wnt signalling pathway. However, a VACV strain expressing a mutant A49 that neither co-precipitates with β-TrCP nor inhibits NF-κB activation, is more virulent than a virus lacking A49, indicating that A49 has another function that also contributes to virulence. Here we demonstrate that gene A49L encodes a second, smaller polypeptide that is expressed via leaky scanning translation from methionine 20 and is unable to block NF-κB activation. Viruses engineered to express either only the large protein or only the small A49 protein both have lower virulence than wild-type virus and greater virulence than an A49L deletion mutant. This demonstrates that the small protein contributes to virulence by an unknown mechanism that is independent of NF-κB inhibition. Despite having a large genome with about 200 genes, this study illustrates how VACV makes efficient use of its coding potential and from gene A49L expresses a protein with multiple functions and multiple proteins with different functions.
Collapse
Affiliation(s)
- Sarah Neidel
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Alice A. Torres
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Hongwei Ren
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
- Present address: Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Geoffrey L. Smith
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| |
Collapse
|
24
|
The Bcl-2 Family: Ancient Origins, Conserved Structures, and Divergent Mechanisms. Biomolecules 2020; 10:biom10010128. [PMID: 31940915 PMCID: PMC7022251 DOI: 10.3390/biom10010128] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/18/2019] [Accepted: 01/09/2020] [Indexed: 12/13/2022] Open
Abstract
Intrinsic apoptosis, the response to intracellular cell death stimuli, is regulated by the interplay of the B-cell lymphoma 2 (Bcl-2) family and their membrane interactions. Bcl-2 proteins mediate a number of processes including development, homeostasis, autophagy, and innate and adaptive immune responses and their dysregulation underpins a host of diseases including cancer. The Bcl-2 family is characterized by the presence of conserved sequence motifs called Bcl-2 homology motifs, as well as a transmembrane region, which form the interaction sites and intracellular location mechanism, respectively. Bcl-2 proteins have been recognized in the earliest metazoans including Porifera (sponges), Placozoans, and Cnidarians (e.g., Hydra). A number of viruses have gained Bcl-2 homologs and subvert innate immunity and cellular apoptosis for their replication, but they frequently have very different sequences to their host Bcl-2 analogs. Though most mechanisms of apoptosis initiation converge on activation of caspases that destroy the cell from within, the numerous gene insertions, deletions, and duplications during evolution have led to a divergence in mechanisms of intrinsic apoptosis. Currently, the action of the Bcl-2 family is best understood in vertebrates and nematodes but new insights are emerging from evolutionarily earlier organisms. This review focuses on the mechanisms underpinning the activity of Bcl-2 proteins including their structures and interactions, and how they have changed over the course of evolution.
Collapse
|
25
|
Rahman MM, McFadden G. Oncolytic Virotherapy with Myxoma Virus. J Clin Med 2020; 9:jcm9010171. [PMID: 31936317 PMCID: PMC7020043 DOI: 10.3390/jcm9010171] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 12/25/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Oncolytic viruses are one of the most promising novel therapeutics for malignant cancers. They selectively infect and kill cancer cells while sparing the normal counterparts, expose cancer- specific antigens and activate the host immune system against both viral and tumor determinants. Oncolytic viruses can be used as monotherapy or combined with existing cancer therapies to become more potent. Among the many types of oncolytic viruses that have been developed thus far, members of poxviruses are the most promising candidates against diverse cancer types. This review summarizes recent advances that are made with oncolytic myxoma virus (MYXV), a member of the Leporipoxvirus genus. Unlike other oncolytic viruses, MYXV infects only rabbits in nature and causes no harm to humans or any other non-leporid animals. However, MYXV can selectively infect and kill cancer cells originating from human, mouse and other host species. This selective cancer tropism and safety profile have led to the testing of MYXV in various types of preclinical cancer models. The next stage will be successful GMP manufacturing and clinical trials that will bring MYXV from bench to bedside for the treatment of currently intractable malignancies.
Collapse
|
26
|
Biswas S, Noyce RS, Babiuk LA, Lung O, Bulach DM, Bowden TR, Boyle DB, Babiuk S, Evans DH. Extended sequencing of vaccine and wild-type capripoxvirus isolates provides insights into genes modulating virulence and host range. Transbound Emerg Dis 2019; 67:80-97. [PMID: 31379093 DOI: 10.1111/tbed.13322] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/22/2019] [Accepted: 07/30/2019] [Indexed: 11/29/2022]
Abstract
The genus Capripoxvirus in the subfamily Chordopoxvirinae, family Poxviridae, comprises sheeppox virus (SPPV), goatpox virus (GTPV) and lumpy skin disease virus (LSDV), which cause the eponymous diseases across parts of Africa, the Middle East and Asia. These diseases cause significant economic losses and can have a devastating impact on the livelihoods and food security of small farm holders. So far, only live classically attenuated SPPV, GTPV and LSDV vaccines are commercially available and the history, safety and efficacy of many have not been well established. Here, we report 13 new capripoxvirus genome sequences, including the hairpin telomeres, from both pathogenic field isolates and vaccine strains. We have also updated the genome annotations to incorporate recent advances in our understanding of poxvirus biology. These new genomes and genes grouped phenetically with other previously sequenced capripoxvirus strains, and these new alignments collectively identified several recurring alterations in genes thought to modulate virulence and host range. In particular, some of the many large capripoxvirus ankyrin and kelch-like proteins are commonly mutated in vaccine strains, while the variola virus B22R-like gene homolog has also been disrupted in many vaccine isolates. Among these vaccine isolates, frameshift mutations are especially common and clearly present a risk of reversion to wild type in vaccines bearing these mutations. A consistent pattern of gene inactivation from LSDV to GTPV and then SPPV is also observed, much like the pattern of gene loss in orthopoxviruses, but, rather surprisingly, the overall genome size of ~150 kbp remains relatively constant. These data provide new insights into the evolution of capripoxviruses and the determinants of pathogenicity and host range. They will find application in the development of new vaccines with better safety, efficacy and trade profiles.
Collapse
Affiliation(s)
- Siddhartha Biswas
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Ryan S Noyce
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Lorne A Babiuk
- Department of Agricultural, Food, and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada
| | - Oliver Lung
- National Centre for Foreign Animal Disease (NCFAD), Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | - Dieter M Bulach
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Vic., Australia
| | - Timothy R Bowden
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Vic., Australia
| | - David B Boyle
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Vic., Australia
| | - Shawn Babiuk
- National Centre for Foreign Animal Disease (NCFAD), Canadian Food Inspection Agency, Winnipeg, MB, Canada.,Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - David H Evans
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
27
|
Banjara S, Shimmon GL, Dixon LK, Netherton CL, Hinds MG, Kvansakul M. Crystal Structure of African Swine Fever Virus A179L with the Autophagy Regulator Beclin. Viruses 2019; 11:v11090789. [PMID: 31461953 PMCID: PMC6784060 DOI: 10.3390/v11090789] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/23/2019] [Accepted: 08/24/2019] [Indexed: 12/15/2022] Open
Abstract
Subversion of programmed cell death-based host defence systems is a prominent feature of infections by large DNA viruses. African swine fever virus (ASFV) is a large DNA virus and sole member of the Asfarviridae family that harbours the B-cell lymphoma 2 or Bcl-2 homolog A179L. A179L has been shown to bind to a range of cell death-inducing host proteins, including pro-apoptotic Bcl-2 proteins as well as the autophagy regulator Beclin. Here we report the crystal structure of A179L bound to the Beclin BH3 motif. A179L engages Beclin using the same canonical ligand-binding groove that is utilized to bind to pro-apoptotic Bcl-2 proteins. The mode of binding of Beclin to A179L mirrors that of Beclin binding to human Bcl-2 and Bcl-xL as well as murine γ-herpesvirus 68. The introduction of bulky hydrophobic residues into the A179L ligand-binding groove via site-directed mutagenesis ablates binding of Beclin to A179L, leading to a loss of the ability of A179L to modulate autophagosome formation in Vero cells during starvation. Our findings provide a mechanistic understanding for the potent autophagy inhibitory activity of A179L and serve as a platform for more detailed investigations into the role of autophagy during ASFV infection.
Collapse
Affiliation(s)
- Suresh Banjara
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | | | - Linda K Dixon
- Pirbright Institute, Ash Road, Pirbright, Surrey GU24 0NF, UK
| | | | - Mark G Hinds
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3050, Australia.
| | - Marc Kvansakul
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia.
| |
Collapse
|
28
|
Naderer T, Fulcher MC. Targeting apoptosis pathways in infections. J Leukoc Biol 2019; 103:275-285. [PMID: 29372933 DOI: 10.1189/jlb.4mr0717-286r] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/29/2017] [Accepted: 09/13/2017] [Indexed: 11/24/2022] Open
Abstract
The programmed cell death pathway of apoptosis is essential for mammalian development and immunity as it eliminates unwanted and dangerous cells. As part of the cellular immune response, apoptosis removes the replicative niche of intracellular pathogens and enables the resolution of infections. To subvert apoptosis, pathogens have evolved a diverse range of mechanisms. In some circumstances, however, pathogens express effector molecules that induce apoptotic cell death. In this review, we focus on selected host-pathogen interactions that affect apoptotic pathways. We discuss how pathogens control the fate of host cells and how this determines the outcome of infections. Finally, small molecule inhibitors that activate apoptosis in cancer cells can also induce apoptotic cell death of infected cells. This suggests that targeting host death factors to kill infected cells is a potential therapeutic option to treat infectious diseases.
Collapse
Affiliation(s)
- Thomas Naderer
- Biomedicine Discovery Institute and Department of Biochemistry & Molecular Biology, Monash University, Clayton, Australia
| | - Maria Cecilia Fulcher
- Biomedicine Discovery Institute and Department of Biochemistry & Molecular Biology, Monash University, Clayton, Australia
| |
Collapse
|
29
|
NF-κB activation is a turn on for vaccinia virus phosphoprotein A49 to turn off NF-κB activation. Proc Natl Acad Sci U S A 2019; 116:5699-5704. [PMID: 30819886 PMCID: PMC6431142 DOI: 10.1073/pnas.1813504116] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Vaccinia virus (VACV) encodes many proteins that inhibit innate immunity. For instance, protein A49 inhibits NF-κB activation by binding to β-TrCP. Here we show that A49 is phosphorylated on serine 7 and that this is necessary for binding β-TrCP and inhibition of NF-κB activation. Further, this phosphorylation occurs when the NF-κB pathway is stimulated and the kinase IKKβ is activated. Thus, A49 shows beautiful biological regulation, for activation of the pathway also activates the virus inhibitor of the pathway. The significance is seen in vivo, since VACVs expressing A49 S7A or S7E are less virulent than wild-type virus but more virulent than a virus lacking A49. Vaccinia virus protein A49 inhibits NF-κB activation by molecular mimicry and has a motif near the N terminus that is conserved in IκBα, β-catenin, HIV Vpu, and some other proteins. This motif contains two serines, and for IκBα and β-catenin, phosphorylation of these serines enables recognition by the E3 ubiquitin ligase β-TrCP. Binding of IκBα and β-catenin by β-TrCP causes their ubiquitylation and thereafter proteasome-mediated degradation. In contrast, HIV Vpu and VACV A49 are not degraded. This paper shows that A49 is phosphorylated at serine 7 but not serine 12 and that this is necessary and sufficient for binding β-TrCP and antagonism of NF-κB. Phosphorylation of A49 S7 occurs when NF-κB signaling is activated by addition of IL-1β or overexpression of TRAF6 or IKKβ, the kinase needed for IκBα phosphorylation. Thus, A49 shows beautiful biological regulation, for it becomes an NF-κB antagonist upon activation of NF-κB signaling. The virulence of viruses expressing mutant A49 proteins or lacking A49 (vΔA49) was tested. vΔA49 was attenuated compared with WT, but viruses expressing A49 that cannot bind β-TrCP or bind β-TrCP constitutively had intermediate virulence. So A49 promotes virulence by inhibiting NF-κB activation and by another mechanism independent of S7 phosphorylation and NF-κB antagonism. Last, a virus lacking A49 was more immunogenic than the WT virus.
Collapse
|
30
|
Banjara S, Mao J, Ryan TM, Caria S, Kvansakul M. Grouper iridovirus GIV66 is a Bcl-2 protein that inhibits apoptosis by exclusively sequestering Bim. J Biol Chem 2018; 293:5464-5477. [PMID: 29483196 DOI: 10.1074/jbc.ra117.000591] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 02/21/2018] [Indexed: 11/06/2022] Open
Abstract
Programmed cell death or apoptosis is a critical mechanism for the controlled removal of damaged or infected cells, and proteins of the Bcl-2 family are important arbiters of this process. Viruses have been shown to encode functional and structural homologs of Bcl-2 to counter premature host-cell apoptosis and ensure viral proliferation or survival. Grouper iridovirus (GIV) is a large DNA virus belonging to the Iridoviridae family and harbors GIV66, a putative Bcl-2-like protein and mitochondrially localized apoptosis inhibitor. However, the molecular and structural basis of GIV66-mediated apoptosis inhibition is currently not understood. To gain insight into GIV66's mechanism of action, we systematically evaluated its ability to bind peptides spanning the BH3 domain of pro-apoptotic Bcl-2 family members. Our results revealed that GIV66 harbors an unusually high level of specificity for pro-apoptotic Bcl-2 and displays affinity only for Bcl-2-like 11 (Bcl2L11 or Bim). Using crystal structures of both apo-GIV66 and GIV66 bound to the BH3 domain from Bim, we unexpectedly found that GIV66 forms dimers via an interface that results in occluded access to the canonical Bcl-2 ligand-binding groove, which breaks apart upon Bim binding. This observation suggests that GIV66 dimerization may affect GIV66's ability to bind host pro-death Bcl-2 proteins and enables highly targeted virus-directed suppression of host apoptosis signaling. Our findings provide a mechanistic understanding for the potent anti-apoptotic activity of GIV66 by identifying it as the first single-specificity, pro-survival Bcl-2 protein and identifying a pivotal role of Bim in GIV-mediated inhibition of apoptosis.
Collapse
Affiliation(s)
- Suresh Banjara
- From the Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia and
| | - Jiahao Mao
- From the Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia and
| | - Timothy M Ryan
- SAXS/WAXS, Australian Synchrotron, 800 Blackburn Road, Clayton, Victoria 3168, Australia
| | - Sofia Caria
- From the Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia and
| | - Marc Kvansakul
- From the Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia and
| |
Collapse
|
31
|
Structural and Functional Insight into Canarypox Virus CNP058 Mediated Regulation of Apoptosis. Viruses 2017; 9:v9100305. [PMID: 29053589 PMCID: PMC5691656 DOI: 10.3390/v9100305] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 10/15/2017] [Accepted: 10/18/2017] [Indexed: 12/21/2022] Open
Abstract
Programmed cell death or apoptosis is an important component of host defense systems against viral infection. The B-cell lymphoma 2 (Bcl-2) proteins family is the main arbiter of mitochondrially mediated apoptosis, and viruses have evolved sequence and structural mimics of Bcl-2 to subvert premature host cell apoptosis in response to viral infection. The sequencing of the canarypox virus genome identified a putative pro-survival Bcl-2 protein, CNP058. However, a role in apoptosis inhibition for CNP058 has not been identified to date. Here, we report that CNP058 is able to bind several host cell pro-death Bcl-2 proteins, including Bak and Bax, as well as several BH3 only-proteins including Bim, Bid, Bmf, Noxa, Puma, and Hrk with high to moderate affinities. We then defined the structural basis for CNP058 binding to pro-death Bcl-2 proteins by determining the crystal structure of CNP058 bound to Bim BH3. CNP058 adopts the conserved Bcl-2 like fold observed in cellular pro-survival Bcl-2 proteins, and utilizes the canonical ligand binding groove to bind Bim BH3. We then demonstrate that CNP058 is a potent inhibitor of ultraviolet (UV) induced apoptosis in a cell culture model. Our findings suggest that CNP058 is a potent inhibitor of apoptosis that is able to bind to BH3 domain peptides from a broad range of pro-death Bcl-2 proteins, and may play a key role in countering premature host apoptosis.
Collapse
|
32
|
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
|
33
|
Okamoto T, Suzuki T, Kusakabe S, Tokunaga M, Hirano J, Miyata Y, Matsuura Y. Regulation of Apoptosis during Flavivirus Infection. Viruses 2017; 9:v9090243. [PMID: 28846635 PMCID: PMC5618009 DOI: 10.3390/v9090243] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/19/2017] [Accepted: 08/25/2017] [Indexed: 02/06/2023] Open
Abstract
Apoptosis is a type of programmed cell death that regulates cellular homeostasis by removing damaged or unnecessary cells. Its importance in host defenses is highlighted by the observation that many viruses evade, obstruct, or subvert apoptosis, thereby blunting the host immune response. Infection with Flaviviruses such as Japanese encephalitis virus (JEV), Dengue virus (DENV) and West Nile virus (WNV) has been shown to activate several signaling pathways such as endoplasmic reticulum (ER)-stress and AKT/PI3K pathway, resulting in activation or suppression of apoptosis in virus-infected cells. On the other hands, expression of some viral proteins induces or protects apoptosis. There is a discrepancy between induction and suppression of apoptosis during flavivirus infection because the experimental situation may be different, and strong links between apoptosis and other types of cell death such as necrosis may make it more difficult. In this paper, we review the effects of apoptosis on viral propagation and pathogenesis during infection with flaviviruses.
Collapse
Affiliation(s)
- Toru Okamoto
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan.
| | - Tatsuya Suzuki
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan.
| | - Shinji Kusakabe
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan.
| | - Makoto Tokunaga
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan.
| | - Junki Hirano
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan.
| | - Yuka Miyata
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan.
| | - Yoshiharu Matsuura
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
34
|
Nichols DB, De Martini W, Cottrell J. Poxviruses Utilize Multiple Strategies to Inhibit Apoptosis. Viruses 2017; 9:v9080215. [PMID: 28786952 PMCID: PMC5580472 DOI: 10.3390/v9080215] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/31/2017] [Accepted: 08/02/2017] [Indexed: 12/11/2022] Open
Abstract
Cells have multiple means to induce apoptosis in response to viral infection. Poxviruses must prevent activation of cellular apoptosis to ensure successful replication. These viruses devote a substantial portion of their genome to immune evasion. Many of these immune evasion products expressed during infection antagonize cellular apoptotic pathways. Poxvirus products target multiple points in both the extrinsic and intrinsic apoptotic pathways, thereby mitigating apoptosis during infection. Interestingly, recent evidence indicates that poxviruses also hijack cellular means of eliminating apoptotic bodies as a means to spread cell to cell through a process called apoptotic mimicry. Poxviruses are the causative agent of many human and veterinary diseases. Further, there is substantial interest in developing these viruses as vectors for a variety of uses including vaccine delivery and as oncolytic viruses to treat certain human cancers. Therefore, an understanding of the molecular mechanisms through which poxviruses regulate the cellular apoptotic pathways remains a top research priority. In this review, we consider anti-apoptotic strategies of poxviruses focusing on three relevant poxvirus genera: Orthopoxvirus, Molluscipoxvirus, and Leporipoxvirus. All three genera express multiple products to inhibit both extrinsic and intrinsic apoptotic pathways with many of these products required for virulence.
Collapse
Affiliation(s)
- Daniel Brian Nichols
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07039, USA.
| | - William De Martini
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07039, USA.
| | - Jessica Cottrell
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07039, USA.
| |
Collapse
|
35
|
Anasir MI, Caria S, Skinner MA, Kvansakul M. Structural basis of apoptosis inhibition by the fowlpox virus protein FPV039. J Biol Chem 2017; 292:9010-9021. [PMID: 28411240 DOI: 10.1074/jbc.m116.768879] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 04/13/2017] [Indexed: 11/06/2022] Open
Abstract
Programmed cell death or apoptosis of infected host cells is an important defense mechanism in response to viral infections. This process is regulated by proapoptotic and prosurvival members of the B-cell lymphoma 2 (Bcl-2) protein family. To counter premature death of a virus-infected cell, poxviruses use a range of different molecular strategies including the mimicry of prosurvival Bcl-2 proteins. One such viral prosurvival protein is the fowlpox virus protein FPV039, which is a potent apoptosis inhibitor, but the precise molecular mechanism by which FPV039 inhibits apoptosis is unknown. To understand how fowlpox virus inhibits apoptosis, we examined FPV039 using isothermal titration calorimetry, small-angle X-ray scattering, and X-ray crystallography. Here, we report that the fowlpox virus prosurvival protein FPV039 promiscuously binds to cellular proapoptotic Bcl-2 and engages all major proapoptotic Bcl-2 proteins. Unlike other identified viral Bcl-2 proteins to date, FPV039 engaged with cellular proapoptotic Bcl-2 with affinities comparable with those of Bcl-2's endogenous cellular counterparts. Structural studies revealed that FPV039 adopts the conserved Bcl-2 fold observed in cellular prosurvival Bcl-2 proteins and closely mimics the structure of the prosurvival Bcl-2 family protein Mcl-1. Our findings suggest that FPV039 is a pan-Bcl-2 protein inhibitor that can engage all host BH3-only proteins, as well as Bcl-2-associated X, apoptosis regulator (Bax) and Bcl-2 antagonist/killer (Bak) proteins to inhibit premature apoptosis of an infected host cell. This work therefore provides a mechanistic platform to better understand FPV039-mediated apoptosis inhibition.
Collapse
Affiliation(s)
- Mohd Ishtiaq Anasir
- From the Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia and
| | - Sofia Caria
- From the Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia and
| | - Michael A Skinner
- Section of Virology, Faculty of Medicine, Imperial College London, London W2 1PZ, United Kingdom
| | - Marc Kvansakul
- From the Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia and
| |
Collapse
|
36
|
Banjara S, Caria S, Dixon LK, Hinds MG, Kvansakul M. Structural Insight into African Swine Fever Virus A179L-Mediated Inhibition of Apoptosis. J Virol 2017; 91:e02228-16. [PMID: 28053104 PMCID: PMC5331815 DOI: 10.1128/jvi.02228-16] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 12/21/2016] [Indexed: 11/20/2022] Open
Abstract
Programmed cell death is a tightly controlled process critical for the removal of damaged or infected cells. Pro- and antiapoptotic proteins of the Bcl-2 family are pivotal mediators of this process. African swine fever virus (ASFV) is a large DNA virus, the only member of the Asfarviridae family, and harbors A179L, a putative Bcl-2 like protein. A179L has been shown to bind to several proapoptotic Bcl-2 proteins; however, the hierarchy of binding and the structural basis for apoptosis inhibition are currently not understood. We systematically evaluated the ability of A179L to bind proapoptotic Bcl-2 family members and show that A179L is the first antiapoptotic Bcl-2 protein to bind to all major death-inducing mammalian Bcl-2 proteins. We then defined the structural basis for apoptosis inhibition of A179L by determining the crystal structures of A179L bound to both Bid and Bax BH3 motifs. Our findings provide a mechanistic understanding for the potent antiapoptotic activity of A179L by identifying it as the first panprodeath Bcl-2 binder and serve as a platform for more-detailed investigations into the role of A179L during ASFV infection.IMPORTANCE Numerous viruses have acquired strategies to subvert apoptosis by encoding proteins capable of sequestering proapoptotic host proteins. African swine fever virus (ASFV), a large DNA virus and the only member of the Asfarviridae family, encodes the protein A179L, which functions to prevent apoptosis. We show that A179L is unusual among antiapoptotic Bcl-2 proteins in being able to physically bind to all core death-inducing mammalian Bcl-2 proteins. Currently, little is known regarding the molecular interactions between A179L and the proapoptotic Bcl-2 members. Using the crystal structures of A179L bound to two of the identified proapoptotic Bcl-2 proteins, Bid and Bax, we now provide a three-dimensional (3D) view of how A179L sequesters host proapoptotic proteins, which is crucial for subverting premature host cell apoptosis.
Collapse
Affiliation(s)
- Suresh Banjara
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Sofia Caria
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | | | - Mark G Hinds
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Marc Kvansakul
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
37
|
Coutu J, Ryerson MR, Bugert J, Brian Nichols D. The Molluscum Contagiosum Virus protein MC163 localizes to the mitochondria and dampens mitochondrial mediated apoptotic responses. Virology 2017; 505:91-101. [PMID: 28235685 DOI: 10.1016/j.virol.2017.02.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 01/30/2017] [Accepted: 02/17/2017] [Indexed: 12/22/2022]
Abstract
Apoptosis is a powerful host cell defense to prevent viruses from completing replication. Poxviruses have evolved complex means to dampen cellular apoptotic responses. The poxvirus, Molluscum Contagiosum Virus (MCV), encodes numerous host interacting molecules predicted to antagonize immune responses. However, the function of the majority of these MCV products has not been characterized. Here, we show that the MCV MC163 protein localized to the mitochondria via an N-terminal mitochondrial localization sequence and transmembrane domain. Transient expression of the MC163 protein prevented mitochondrial membrane permeabilization (MMP), an event central to cellular apoptotic responses, induced by either Tumor Necrosis Factor alpha (TNF-α) or carbonyl cyanide 3-chlorophenylhydrazone (CCCP). MC163 expression prevented the release of a mitochondrial intermembrane space reporter protein when cells were challenged with TNF-α. Inhibition of MMP was also observed in cell lines stably expressing MC163. MC163 expression may contribute to the persistence of MCV lesions by dampening cellular apoptotic responses.
Collapse
Affiliation(s)
- Jesse Coutu
- Department of Biological Sciences, Seton Hall University, 400 S. Orange Ave, South Orange, NJ 07079, United States
| | - Melissa R Ryerson
- Department of Microbiology, University of Illinois, 601 S. Goodwin Ave., Champaign-Urbana, IL 61801, United States
| | - Joachim Bugert
- Institut für Mikrobiologie der Bundeswehr, Neuherbergstrasse, 1180937 München, Germany
| | - Daniel Brian Nichols
- Department of Biological Sciences, Seton Hall University, 400 S. Orange Ave, South Orange, NJ 07079, United States.
| |
Collapse
|
38
|
Caria S, Marshall B, Burton RL, Campbell S, Pantaki-Eimany D, Hawkins CJ, Barry M, Kvansakul M. The N Terminus of the Vaccinia Virus Protein F1L Is an Intrinsically Unstructured Region That Is Not Involved in Apoptosis Regulation. J Biol Chem 2016; 291:14600-8. [PMID: 27151220 DOI: 10.1074/jbc.m116.726851] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Indexed: 12/21/2022] Open
Abstract
Subversion of host cell apoptotic responses is a prominent feature of viral immune evasion strategies to prevent premature clearance of infected cells. Numerous poxviruses encode structural and functional homologs of the Bcl-2 family of proteins, and vaccinia virus harbors antiapoptotic F1L that potently inhibits the mitochondrial apoptotic checkpoint. Recently F1L has been assigned a caspase-9 inhibitory function attributed to an N-terminal α helical region of F1L spanning residues 1-15 (1) preceding the domain-swapped Bcl-2-like domains. Using a reconstituted caspase inhibition assay in yeast we found that unlike AcP35, a well characterized caspase-9 inhibitor from the insect virus Autographa californica multiple nucleopolyhedrovirus, F1L does not prevent caspase-9-mediated yeast cell death. Furthermore, we found that deletion of the F1L N-terminal region does not impede F1L antiapoptotic activity in the context of a viral infection. Solution analysis of the F1L N-terminal regions using small angle x-ray scattering indicates that the region of F1L spanning residues 1-50 located N-terminally from the Bcl-2 fold is an intrinsically unstructured region. We conclude that the N terminus of F1L is not involved in apoptosis inhibition and may act as a regulatory element in other signaling pathways in a manner reminiscent of other unstructured regulatory elements commonly found in mammalian prosurvival Bcl-2 members including Bcl-xL and Mcl-1.
Collapse
Affiliation(s)
- Sofia Caria
- From the La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, La Trobe University, Victoria 3086, Australia and
| | - Bevan Marshall
- From the La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, La Trobe University, Victoria 3086, Australia and
| | - Robyn-Lee Burton
- Li Ka Shing Institute for Virology, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Stephanie Campbell
- Li Ka Shing Institute for Virology, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Delara Pantaki-Eimany
- From the La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, La Trobe University, Victoria 3086, Australia and
| | - Christine J Hawkins
- From the La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, La Trobe University, Victoria 3086, Australia and
| | - Michele Barry
- Li Ka Shing Institute for Virology, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Marc Kvansakul
- From the La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, La Trobe University, Victoria 3086, Australia and
| |
Collapse
|
39
|
Pisklakova A, McKenzie B, Zemp F, Lun X, Kenchappa RS, Etame AB, Rahman MM, Reilly K, Pilon-Thomas S, McFadden G, Kurz E, Forsyth PA. M011L-deficient oncolytic myxoma virus induces apoptosis in brain tumor-initiating cells and enhances survival in a novel immunocompetent mouse model of glioblastoma. Neuro Oncol 2016; 18:1088-1098. [PMID: 26962017 DOI: 10.1093/neuonc/now006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Myxoma virus (MYXV) is a promising oncolytic agent and is highly effective against immortalized glioma cells but less effective against brain tumor initiating cells (BTICs), which are believed to mediate glioma development/recurrence. MYXV encodes various proteins to attenuate host cell apoptosis, including an antiapoptotic Bcl-2 homologue known as M011L. Such proteins may limit the ability of MYXV to kill BTICs, which have heightened resistance to apoptosis. We hypothesized that infecting BTICs with an M011L-deficient MYXV construct would overcome BTIC resistance to MYXV. METHODS We used patient-derived BTICs to evaluate the efficacy of M011L knockout virus (vMyx-M011L-KO) versus wild-type MYXV (vMyx-WT) and characterized the mechanism of virus-induced cell death in vitro. To extend our findings in a novel immunocompetent animal model, we derived, cultured, and characterized a C57Bl/6J murine BTIC (mBTIC0309) from a spontaneous murine glioma and evaluated vMyx-M011L-KO efficacy with and without temozolomide (TMZ) in mBTIC0309-bearing mice. RESULTS We demonstrated that vMyx-M011L-KO induces apoptosis in BTICs, dramatically increasing sensitivity to the virus. vMyx-WT failed to induce apoptosis as M011L protein prevented Bax activation and cytochrome c release. In vivo, intracranial implantation of mBTIC0309 generated tumors that closely recapitulated the pathological and molecular profile of human gliomas. Treatment of tumor-bearing mice with vMyx-M011L-KO significantly prolonged survival in immunocompetent-but not immunodeficient-mouse models, an effect that is significantly enhanced in combination with TMZ. CONCLUSIONS Our data suggest that vMyx-M011L-KO is an effective, well-tolerated, proapoptotic oncolytic virus and a strong candidate for clinical translation.
Collapse
Affiliation(s)
- Alexandra Pisklakova
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Brienne McKenzie
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Franz Zemp
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Xueqing Lun
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Rajappa S Kenchappa
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Arnold B Etame
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Masmudur M Rahman
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Karlyne Reilly
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Shari Pilon-Thomas
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Grant McFadden
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Ebba Kurz
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Peter A Forsyth
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| |
Collapse
|
40
|
Thébault S, Agez M, Chi X, Stojko J, Cura V, Telerman SB, Maillet L, Gautier F, Billas-Massobrio I, Birck C, Troffer-Charlier N, Karafin T, Honoré J, Senff-Ribeiro A, Montessuit S, Johnson CM, Juin P, Cianférani S, Martinou JC, Andrews DW, Amson R, Telerman A, Cavarelli J. TCTP contains a BH3-like domain, which instead of inhibiting, activates Bcl-xL. Sci Rep 2016; 6:19725. [PMID: 26813996 PMCID: PMC4728560 DOI: 10.1038/srep19725] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 12/17/2015] [Indexed: 01/28/2023] Open
Abstract
Translationally Controlled Tumor Protein (TCTP) is anti-apoptotic, key in development and cancer, however without the typical Bcl2 family members’ structure. Here we report that TCTP contains a BH3-like domain and forms heterocomplexes with Bcl-xL. The crystal structure of a Bcl-xL deletion variant-TCTP11–31 complex reveals that TCTP refolds in a helical conformation upon binding the BH3-groove of Bcl-xL, although lacking the h1-subregion interaction. Experiments using in vitro-vivo reconstituted systems and TCTP+/− mice indicate that TCTP activates the anti-apoptotic function of Bcl-xL, in contrast to all other BH3-proteins. Replacing the non-conserved h1 of TCTP by that of Bax drastically increases the affinity of this hybrid for Bcl-xL, modifying its biological properties. This work reveals a novel class of BH3-proteins potentiating the anti-apoptotic function of Bcl-xL.
Collapse
Affiliation(s)
- Stéphanie Thébault
- Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR 7104, INSERM U964, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France.,CNRS-UMR 8113, LBPA, École Normale Supérieure, 61 avenue du Président Wilson, 94235 Cachan, France
| | - Morgane Agez
- Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR 7104, INSERM U964, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France.,Institut Gustave Roussy, Unité Inserm U981, Bâtiment B2M, 114 rue Édouard-Vaillant, 94805 Villejuif, France
| | - Xiaoke Chi
- Sunnybrook Research Institute and Departments of Biochemistry and Medical Biophysics, University of Toronto, 2075 Bayview Ave., Toronto, Ontario, M4N 3M5, Canada.,Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St. W. Hamilton, Ontario, L8N 3Z5, Canada
| | - Johann Stojko
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), IPHC-DSA, Université de Strasbourg, CNRS, UMR7178, 25 rue Becquerel, 67087 Strasbourg, France
| | - Vincent Cura
- Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR 7104, INSERM U964, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Stéphanie B Telerman
- King's College London Centre for Stem Cells and Regenerative Medicine, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.,MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Laurent Maillet
- Center for Cancer Research Nantes-Angers, UMR 892 Inserm - 6299 CNRS/Université de Nantes, IRS-UN, 8 Quai Moncousu - BP 70721, 44007 Nantes Cedex 1
| | - Fabien Gautier
- Center for Cancer Research Nantes-Angers, UMR 892 Inserm - 6299 CNRS/Université de Nantes, IRS-UN, 8 Quai Moncousu - BP 70721, 44007 Nantes Cedex 1.,Institut de Cancérologie de l'Ouest, Centre René Gauducheau Bd Jacques Monod, 44805 Saint Herblain-Nantes cedex
| | - Isabelle Billas-Massobrio
- Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR 7104, INSERM U964, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Catherine Birck
- Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR 7104, INSERM U964, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Nathalie Troffer-Charlier
- Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR 7104, INSERM U964, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Teele Karafin
- CNRS-UMR 8113, LBPA, École Normale Supérieure, 61 avenue du Président Wilson, 94235 Cachan, France.,Institut Gustave Roussy, Unité Inserm U981, Bâtiment B2M, 114 rue Édouard-Vaillant, 94805 Villejuif, France
| | - Joane Honoré
- Institut Gustave Roussy, Unité Inserm U981, Bâtiment B2M, 114 rue Édouard-Vaillant, 94805 Villejuif, France
| | - Andrea Senff-Ribeiro
- CNRS-UMR 8113, LBPA, École Normale Supérieure, 61 avenue du Président Wilson, 94235 Cachan, France.,Institut Gustave Roussy, Unité Inserm U981, Bâtiment B2M, 114 rue Édouard-Vaillant, 94805 Villejuif, France
| | - Sylvie Montessuit
- Department of Cell Biology, University of Geneva, 30, quai Ansermet, 1211 Geneva 4, Switzerland
| | - Christopher M Johnson
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Philippe Juin
- Center for Cancer Research Nantes-Angers, UMR 892 Inserm - 6299 CNRS/Université de Nantes, IRS-UN, 8 Quai Moncousu - BP 70721, 44007 Nantes Cedex 1.,Institut de Cancérologie de l'Ouest, Centre René Gauducheau Bd Jacques Monod, 44805 Saint Herblain-Nantes cedex
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), IPHC-DSA, Université de Strasbourg, CNRS, UMR7178, 25 rue Becquerel, 67087 Strasbourg, France
| | - Jean-Claude Martinou
- Department of Cell Biology, University of Geneva, 30, quai Ansermet, 1211 Geneva 4, Switzerland
| | - David W Andrews
- Sunnybrook Research Institute and Departments of Biochemistry and Medical Biophysics, University of Toronto, 2075 Bayview Ave., Toronto, Ontario, M4N 3M5, Canada.,Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St. W. Hamilton, Ontario, L8N 3Z5, Canada
| | - Robert Amson
- CNRS-UMR 8113, LBPA, École Normale Supérieure, 61 avenue du Président Wilson, 94235 Cachan, France.,Institut Gustave Roussy, Unité Inserm U981, Bâtiment B2M, 114 rue Édouard-Vaillant, 94805 Villejuif, France
| | - Adam Telerman
- CNRS-UMR 8113, LBPA, École Normale Supérieure, 61 avenue du Président Wilson, 94235 Cachan, France.,Institut Gustave Roussy, Unité Inserm U981, Bâtiment B2M, 114 rue Édouard-Vaillant, 94805 Villejuif, France
| | - Jean Cavarelli
- Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR 7104, INSERM U964, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| |
Collapse
|
41
|
Abstract
High-resolution protein structures determined by X-ray crystallography or NMR have proven invaluable for deciphering the molecular mechanisms underlying the function of a vast range of proteins. Here, we describe methods to generate complexes of proteins belonging to the Bcl-2 family of proteins with either biological ligands or small molecule antagonists.
Collapse
Affiliation(s)
- Marc Kvansakul
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Physical Sciences 4, Room 205, Kingsbury Drive, Bundoora, Melbourne, VIC, 3086, Australia.,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Peter E Czabotar
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia. .,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, 3052, Australia.
| |
Collapse
|
42
|
The phospholipid code: a key component of dying cell recognition, tumor progression and host-microbe interactions. Cell Death Differ 2015; 22:1893-905. [PMID: 26450453 DOI: 10.1038/cdd.2015.122] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 08/05/2015] [Accepted: 08/06/2015] [Indexed: 02/06/2023] Open
Abstract
A significant effort is made by the cell to maintain certain phospholipids at specific sites. It is well described that proteins involved in intracellular signaling can be targeted to the plasma membrane and organelles through phospholipid-binding domains. Thus, the accumulation of a specific combination of phospholipids, denoted here as the 'phospholipid code', is key in initiating cellular processes. Interestingly, a variety of extracellular proteins and pathogen-derived proteins can also recognize or modify phospholipids to facilitate the recognition of dying cells, tumorigenesis and host-microbe interactions. In this article, we discuss the importance of the phospholipid code in a range of physiological and pathological processes.
Collapse
|
43
|
Kvansakul M, Hinds MG. The Bcl-2 family: structures, interactions and targets for drug discovery. Apoptosis 2015; 20:136-50. [PMID: 25398535 DOI: 10.1007/s10495-014-1051-7] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Two phylogenetically and structurally distinct groups of proteins regulate stress induced intrinsic apoptosis, the programmed disassembly of cells. Together they form the B cell lymphoma-2 (Bcl-2) family. Bcl-2 proteins appeared early in metazoan evolution and are identified by the presence of up to four short conserved sequence blocks known as Bcl-2 homology (BH) motifs, or domains. The simple BH3-only proteins bear only a BH3-motif and are intrinsically disordered proteins and antagonize or activate the other group, the multi-motif Bcl-2 proteins that have up to four BH motifs, BH1-BH4. Multi-motif Bcl-2 proteins are either pro-survival or pro-apoptotic in action and have remarkably similar α-helical bundle structures that provide a binding groove formed from the BH1, BH2, and BH3-motifs for their BH3-bearing antagonists. In mammals a network of interactions between Bcl-2 members regulates mitochondrial outer membrane permeability (MOMP) and efflux of cytochrome c and other death inducing factors from mitochondria to initiate the apoptotic caspase cascade, but the molecular events leading to MOMP are uncertain. Dysregulation of the Bcl-2 family occurs in many diseases and pathogenic viruses have assimilated pro-survival Bcl-2 proteins to evade immune responses. Their role in disease has made the Bcl-2 family the focus of drug design attempts and clinical trials are showing promise for 'BH3-mimics', drugs that mimic the ability of BH3-only proteins to neutralize selected pro-survival proteins to induce cell death in tumor cells. This review focuses on the structural biology of Bcl-2 family proteins, their interactions and attempts to harness them as targets for drug design.
Collapse
Affiliation(s)
- Marc Kvansakul
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, 3086, Australia,
| | | |
Collapse
|
44
|
Burton DR, Caria S, Marshall B, Barry M, Kvansakul M. Structural basis of Deerpox virus-mediated inhibition of apoptosis. ACTA ACUST UNITED AC 2015; 71:1593-603. [PMID: 26249341 DOI: 10.1107/s1399004715009402] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 05/18/2015] [Indexed: 01/21/2023]
Abstract
Apoptosis is a key innate defence mechanism to eliminate virally infected cells. To counteract premature host-cell apoptosis, poxviruses have evolved numerous molecular strategies, including the use of Bcl-2 proteins, to ensure their own survival. Here, it is reported that the Deerpox virus inhibitor of apoptosis, DPV022, only engages a highly restricted set of death-inducing Bcl-2 proteins, including Bim, Bax and Bak, with modest affinities. Structural analysis reveals that DPV022 adopts a Bcl-2 fold with a dimeric domain-swapped topology and binds pro-death Bcl-2 proteins via two conserved ligand-binding grooves found on opposite sides of the dimer. Structures of DPV022 bound to Bim, Bak and Bax BH3 domains reveal that a partial obstruction of the binding groove is likely to be responsible for the modest affinities of DPV022 for BH3 domains. These findings reveal that domain-swapped dimeric Bcl-2 folds are not unusual and may be found more widely in viruses. Furthermore, the modest affinities of DPV022 for pro-death Bcl-2 proteins suggest that two distinct classes of anti-apoptotic viral Bcl-2 proteins exist: those that are monomeric and tightly bind a range of death-inducing Bcl-2 proteins, and others such as DPV022 that are dimeric and only bind a very limited number of death-inducing Bcl-2 proteins with modest affinities.
Collapse
Affiliation(s)
- Denis R Burton
- Department of Biochemistry, La Trobe University, Melbourne, VIC 3058, Australia
| | - Sofia Caria
- Department of Biochemistry, La Trobe University, Melbourne, VIC 3058, Australia
| | - Bevan Marshall
- Department of Biochemistry, La Trobe University, Melbourne, VIC 3058, Australia
| | - Michele Barry
- Li Ka Shing Institute for Virology, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Marc Kvansakul
- Department of Biochemistry, La Trobe University, Melbourne, VIC 3058, Australia
| |
Collapse
|
45
|
Rajan S, Choi M, Baek K, Yoon HS. Bh3 induced conformational changes in Bcl-Xl revealed by crystal structure and comparative analysis. Proteins 2015; 83:1262-72. [PMID: 25907960 DOI: 10.1002/prot.24816] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 04/09/2015] [Accepted: 04/17/2015] [Indexed: 01/08/2023]
Abstract
Apoptosis or programmed cell death is a regulatory process in cells in response to stimuli perturbing physiological conditions. The Bcl-2 family of proteins plays an important role in regulating homeostasis during apoptosis. In the process, the molecular interactions among the three members of this family, the pro-apoptotic, anti-apoptotic and BH3-only proteins at the mitochondrial outer membrane define the fate of a cell. Here, we report the crystal structures of the human anti-apoptotic protein Bcl-XL in complex with BH3-only BID(BH3) and BIM(BH3) peptides determined at 2.0 Å and 1.5 Å resolution, respectively. The BH3 peptides bind to the canonical hydrophobic pocket in Bcl-XL and adopt an alpha helical conformation in the bound form. Despite a similar structural fold, a comparison with other BH3 complexes revealed structural differences due to their sequence variations. In the Bcl-XL-BID(BH3) complex we observed a large pocket, in comparison with other BH3 complexes, lined by residues from helices α1, α2, α3, and α5 located adjacent to the canonical hydrophobic pocket. These results suggest that there are differences in the mode of interactions by the BH3 peptides that may translate into functional differences in apoptotic regulation.
Collapse
Affiliation(s)
- Sreekanth Rajan
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Minjoo Choi
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Kwanghee Baek
- Department of Genetic Engineering, College of Life Sciences, Kyung Hee University, Yongin-Si, Gyeonggi-Do, 446-701, Republic of Korea
| | - Ho Sup Yoon
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore.,Department of Genetic Engineering, College of Life Sciences, Kyung Hee University, Yongin-Si, Gyeonggi-Do, 446-701, Republic of Korea
| |
Collapse
|
46
|
Fleming SB, Wise LM, Mercer AA. Molecular genetic analysis of orf virus: a poxvirus that has adapted to skin. Viruses 2015; 7:1505-39. [PMID: 25807056 PMCID: PMC4379583 DOI: 10.3390/v7031505] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 03/17/2015] [Accepted: 03/19/2015] [Indexed: 12/17/2022] Open
Abstract
Orf virus is the type species of the Parapoxvirus genus of the family Poxviridae. It induces acute pustular skin lesions in sheep and goats and is transmissible to humans. The genome is G+C rich, 138 kbp and encodes 132 genes. It shares many essential genes with vaccinia virus that are required for survival but encodes a number of unique factors that allow it to replicate in the highly specific immune environment of skin. Phylogenetic analysis suggests that both viral interleukin-10 and vascular endothelial growth factor genes have been "captured" from their host during the evolution of the parapoxviruses. Genes such as a chemokine binding protein and a protein that binds granulocyte-macrophage colony-stimulating factor and interleukin-2 appear to have evolved from a common poxvirus ancestral gene while three parapoxvirus nuclear factor (NF)-κB signalling pathway inhibitors have no homology to other known NF-κB inhibitors. A homologue of an anaphase-promoting complex subunit that is believed to manipulate the cell cycle and enhance viral DNA synthesis appears to be a specific adaptation for viral-replication in keratinocytes. The review focuses on the unique genes of orf virus, discusses their evolutionary origins and their role in allowing viral-replication in the skin epidermis.
Collapse
Affiliation(s)
- Stephen B Fleming
- Department of Microbiology and Immunology, 720 Cumberland St, University of Otago, Dunedin 9016, New Zealand.
| | - Lyn M Wise
- Department of Microbiology and Immunology, 720 Cumberland St, University of Otago, Dunedin 9016, New Zealand.
| | - Andrew A Mercer
- Department of Microbiology and Immunology, 720 Cumberland St, University of Otago, Dunedin 9016, New Zealand.
| |
Collapse
|
47
|
Variola virus F1L is a Bcl-2-like protein that unlike its vaccinia virus counterpart inhibits apoptosis independent of Bim. Cell Death Dis 2015; 6:e1680. [PMID: 25766319 PMCID: PMC4385930 DOI: 10.1038/cddis.2015.52] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 01/26/2015] [Accepted: 01/27/2015] [Indexed: 01/29/2023]
Abstract
Subversion of host cell apoptosis is an important survival strategy for viruses to ensure their own proliferation and survival. Certain viruses express proteins homologous in sequence, structure and function to mammalian pro-survival B-cell lymphoma 2 (Bcl-2) proteins, which prevent rapid clearance of infected host cells. In vaccinia virus (VV), the virulence factor F1L was shown to be a potent inhibitor of apoptosis that functions primarily be engaging pro-apoptotic Bim. Variola virus (VAR), the causative agent of smallpox, harbors a homolog of F1L of unknown function. We show that VAR F1L is a potent inhibitor of apoptosis, and unlike all other characterized anti-apoptotic Bcl-2 family members lacks affinity for the Bim Bcl-2 homology 3 (BH3) domain. Instead, VAR F1L engages Bid BH3 as well as Bak and Bax BH3 domains. Unlike its VV homolog, variola F1L only protects against Bax-mediated apoptosis in cellular assays. Crystal structures of variola F1L bound to Bid and Bak BH3 domains reveal that variola F1L forms a domain-swapped Bcl-2 fold, which accommodates Bid and Bak BH3 in the canonical Bcl-2-binding groove, in a manner similar to VV F1L. Despite the observed conservation of structure and sequence, variola F1L inhibits apoptosis using a startlingly different mechanism compared with its VV counterpart. Our results suggest that unlike during VV infection, Bim neutralization may not be required during VAR infection. As molecular determinants for the human-specific tropism of VAR remain essentially unknown, identification of a different mechanism of action and utilization of host factors used by a VAR virulence factor compared with its VV homolog suggest that studying VAR directly may be essential to understand its unique tropism.
Collapse
|
48
|
Neidel S, Maluquer de Motes C, Mansur DS, Strnadova P, Smith GL, Graham SC. Vaccinia virus protein A49 is an unexpected member of the B-cell Lymphoma (Bcl)-2 protein family. J Biol Chem 2015; 290:5991-6002. [PMID: 25605733 PMCID: PMC4358236 DOI: 10.1074/jbc.m114.624650] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 01/11/2015] [Indexed: 12/18/2022] Open
Abstract
Vaccinia virus (VACV) encodes several proteins that inhibit activation of the proinflammatory transcription factor nuclear factor κB (NF-κB). VACV protein A49 prevents translocation of NF-κB to the nucleus by sequestering cellular β-TrCP, a protein required for the degradation of the inhibitor of κB. A49 does not share overall sequence similarity with any protein of known structure or function. We solved the crystal structure of A49 from VACV Western Reserve to 1.8 Å resolution and showed, surprisingly, that A49 has the same three-dimensional fold as Bcl-2 family proteins despite lacking identifiable sequence similarity. Whereas Bcl-2 family members characteristically modulate cellular apoptosis, A49 lacks a surface groove suitable for binding BH3 peptides and does not bind proapoptotic Bcl-2 family proteins Bax or Bak. The N-terminal 17 residues of A49 do not adopt a single well ordered conformation, consistent with their proposed role in binding β-TrCP. Whereas pairs of A49 molecules interact symmetrically via a large hydrophobic surface in crystallo, A49 does not dimerize in solution or in cells, and we propose that this hydrophobic interaction surface may mediate binding to a yet undefined cellular partner. A49 represents the eleventh VACV Bcl-2 family protein and, despite these proteins sharing very low sequence identity, structure-based phylogenetic analysis shows that all poxvirus Bcl-2 proteins are structurally more similar to each other than they are to any cellular or herpesvirus Bcl-2 proteins. This is consistent with duplication and diversification of a single BCL2 family gene acquired by an ancestral poxvirus.
Collapse
Affiliation(s)
- Sarah Neidel
- From the Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom and
| | - Carlos Maluquer de Motes
- From the Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom and
| | - Daniel S Mansur
- the Department of Microbiology, Immunology, and Parasitology, Universidade Federal de Santa Catarina, Florianopolis, 88040-900 Brazil
| | - Pavla Strnadova
- From the Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom and
| | - Geoffrey L Smith
- From the Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom and
| | - Stephen C Graham
- From the Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom and
| |
Collapse
|
49
|
Zhou J, Grigoryan G. Rapid search for tertiary fragments reveals protein sequence-structure relationships. Protein Sci 2014; 24:508-24. [PMID: 25420575 DOI: 10.1002/pro.2610] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/21/2014] [Indexed: 12/31/2022]
Abstract
Finding backbone substructures from the Protein Data Bank that match an arbitrary query structural motif, composed of multiple disjoint segments, is a problem of growing relevance in structure prediction and protein design. Although numerous protein structure search approaches have been proposed, methods that address this specific task without additional restrictions and on practical time scales are generally lacking. Here, we propose a solution, dubbed MASTER, that is both rapid, enabling searches over the Protein Data Bank in a matter of seconds, and provably correct, finding all matches below a user-specified root-mean-square deviation cutoff. We show that despite the potentially exponential time complexity of the problem, running times in practice are modest even for queries with many segments. The ability to explore naturally plausible structural and sequence variations around a given motif has the potential to synthesize its design principles in an automated manner; so we go on to illustrate the utility of MASTER to protein structural biology. We demonstrate its capacity to rapidly establish structure-sequence relationships, uncover the native designability landscapes of tertiary structural motifs, identify structural signatures of binding, and automatically rewire protein topologies. Given the broad utility of protein tertiary fragment searches, we hope that providing MASTER in an open-source format will enable novel advances in understanding, predicting, and designing protein structure.
Collapse
Affiliation(s)
- Jianfu Zhou
- Department of Computer Science, Dartmouth College, Hanover, New Hampshire, 03755
| | | |
Collapse
|
50
|
Todt F, Cakir Z, Reichenbach F, Emschermann F, Lauterwasser J, Kaiser A, Ichim G, Tait SWG, Frank S, Langer HF, Edlich F. Differential retrotranslocation of mitochondrial Bax and Bak. EMBO J 2014; 34:67-80. [PMID: 25378477 DOI: 10.15252/embj.201488806] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The Bcl-2 proteins Bax and Bak can permeabilize the outer mitochondrial membrane and commit cells to apoptosis. Pro-survival Bcl-2 proteins control Bax by constant retrotranslocation into the cytosol of healthy cells. The stabilization of cytosolic Bax raises the question whether the functionally redundant but largely mitochondrial Bak shares this level of regulation. Here we report that Bak is retrotranslocated from the mitochondria by pro-survival Bcl-2 proteins. Bak is present in the cytosol of human cells and tissues, but low shuttling rates cause predominant mitochondrial Bak localization. Interchanging the membrane anchors of Bax and Bak reverses their subcellular localization compared to the wild-type proteins. Strikingly, the reduction of Bax shuttling to the level of Bak retrotranslocation results in full Bax toxicity even in absence of apoptosis induction. Thus, fast Bax retrotranslocation is required to protect cells from commitment to programmed death.
Collapse
Affiliation(s)
- Franziska Todt
- Institute for Biochemistry and Molecular Biology University of Freiburg, Freiburg, Germany Spemann Graduate School of Biology and Medicine SGBM, Freiburg, Germany Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Zeynep Cakir
- Institute for Biochemistry and Molecular Biology University of Freiburg, Freiburg, Germany Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Frank Reichenbach
- Institute for Biochemistry and Molecular Biology University of Freiburg, Freiburg, Germany Spemann Graduate School of Biology and Medicine SGBM, Freiburg, Germany Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Frederic Emschermann
- Section for Cardioimmunology, University of Tübingen, Tübingen, Germany Department of Cardiovascular Medicine, University Hospital University of Tübingen, Tübingen, Germany
| | - Joachim Lauterwasser
- Institute for Biochemistry and Molecular Biology University of Freiburg, Freiburg, Germany Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Andrea Kaiser
- Institute for Biochemistry and Molecular Biology University of Freiburg, Freiburg, Germany
| | - Gabriel Ichim
- Cancer Research UK Beatson Institute University of Glasgow, Glasgow, UK
| | - Stephen W G Tait
- Cancer Research UK Beatson Institute University of Glasgow, Glasgow, UK
| | - Stephan Frank
- Division of Neuropathology, Institute of Pathology Basel University Hospitals, Basel, Switzerland
| | - Harald F Langer
- Section for Cardioimmunology, University of Tübingen, Tübingen, Germany Department of Cardiovascular Medicine, University Hospital University of Tübingen, Tübingen, Germany
| | - Frank Edlich
- Institute for Biochemistry and Molecular Biology University of Freiburg, Freiburg, Germany Spemann Graduate School of Biology and Medicine SGBM, Freiburg, Germany BIOSS, Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|