1
|
Morano AA, Xu W, Navarro FM, Shadija N, Dvorin JD, Ke H. The dynamin-related protein PfDyn2 is essential for both apicoplast and mitochondrial fission in Plasmodium falciparum. mBio 2025; 16:e0303624. [PMID: 39611847 PMCID: PMC11708027 DOI: 10.1128/mbio.03036-24] [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: 10/02/2024] [Accepted: 11/08/2024] [Indexed: 11/30/2024] Open
Abstract
Dynamins, or dynamin-related proteins (DRPs), are large mechano-sensitive GTPases that mediate membrane dynamics or organellar fission/fusion events. Plasmodium falciparum encodes three dynamin-like proteins whose functions are poorly understood. Here, we demonstrate that one of these dynamin-related proteins, PfDyn2, is required to divide both the apicoplast and the mitochondrion, a striking divergence from the biology of related parasites. Using super-resolution and ultrastructure expansion microscopy (U-ExM), we show that PfDyn2 is expressed in dividing schizonts, and that it localizes to both the apicoplast and the mitochondrion. Our use of long-term, live-cell microscopy allows for the visualization of apicoplast and mitochondrial division in live parasites at super resolution for the first time, and demonstrates that in PfDyn2-deficient parasites, while the apicoplast and mitochondrion increase in size and complexity, they do not undergo fission. We also show that these organellar fission defects prevent successful individualization of the schizont mass and the formation of new daughter cells, or merozoites because the basal complex, the cytokinetic ring of Plasmodium, cannot fully contract in PfDyn2-deficient parasites, a phenotype secondary to physical blockage by undivided organelles occluding the ring. PfDyn2's singular role in mediating both apicoplast and mitochondrial fission has not been observed in other organisms possessing two endosymbiotic organelles, including other Apicomplexans, thus reflecting a unique, potentially exploitable method of organellar division in P. falciparum.IMPORTANCEPlasmodium falciparum remains a significant global pathogen, causing over 200 million infections and over 600,000 deaths per year. One significant obstacle to the control of malaria is increasing resistance to first-line artemisinin-based antimalarials. Another is a lack of basic knowledge about the cell biology of the parasite. Along with the mitochondrion, Plasmodium contains a second organelle descended from an endosymbiotic event, the apicoplast. Both organelles are common targets for antimalarials, but because many proteins involved in organellar fission are not conserved in Plasmodium, until now, the mechanisms underlying apicoplast and mitochondrial division have been unknown. In this study, we demonstrate that PfDyn2, a dynamin-related protein (DRP), is required for the division of both organelles. We also show that defects in organellar division hinder segmentation of the schizont and formation of invasive merozoites by preventing full contraction of the basal complex. By demonstrating its necessity for the proper division of both the apicoplast and the mitochondria, this study highlights PfDyn2 as a potential target for new antimalarials.
Collapse
Affiliation(s)
- Alexander A. Morano
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | - Wei Xu
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Francesca M. Navarro
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | - Neeta Shadija
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Jeffrey D. Dvorin
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Hangjun Ke
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2
|
Morano AA, Xu W, Shadija N, Dvorin JD, Ke H. The dynamin-related protein Dyn2 is essential for both apicoplast and mitochondrial fission in Plasmodium falciparum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585229. [PMID: 38559241 PMCID: PMC10980034 DOI: 10.1101/2024.03.15.585229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Dynamins, or dynamin-related proteins (DRPs), are large mechano-sensitive GTPases mediating membrane dynamics or organellar fission/fusion events. Plasmodium falciparum encodes three dynamin-like proteins whose functions are poorly understood. Here, we demonstrate that PfDyn2 mediates both apicoplast and mitochondrial fission. Using super-resolution and ultrastructure expansion microscopy, we show that PfDyn2 is expressed in the schizont stage and localizes to both the apicoplast and mitochondria. Super-resolution long-term live cell microscopy shows that PfDyn2-deficient parasites cannot complete cytokinesis because the apicoplast and mitochondria do not undergo fission. Further, the basal complex or cytokinetic ring in Plasmodium cannot fully contract upon PfDyn2 depletion, a phenotype secondary to physical blockage of undivided organelles in the middle of the ring. Our data suggest that organellar fission defects result in aberrant schizogony, generating unsuccessful merozoites. The unique biology of PfDyn2, mediating both apicoplast and mitochondrial fission, has not been observed in other organisms possessing two endosymbiotic organelles. Highlights PfDyn2 is essential for schizont-stage development.PfDyn2 mediates both apicoplast and mitochondrial fission.Deficiency of PfDyn2 leads to organellar fission failures and blockage of basal complex contraction.Addition of apicoplast-derived metabolite IPP does not rescue the growth defects.
Collapse
|
3
|
Rivera-Cuevas Y, Carruthers VB. The multifaceted interactions between pathogens and host ESCRT machinery. PLoS Pathog 2023; 19:e1011344. [PMID: 37141275 PMCID: PMC10159163 DOI: 10.1371/journal.ppat.1011344] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
Abstract
The Endosomal Sorting Complex Required for Transport (ESCRT) machinery consists of multiple protein complexes that coordinate vesicle budding away from the host cytosol. ESCRTs function in many fundamental cellular processes including the biogenesis of multivesicular bodies and exosomes, membrane repair and restoration, and cell abscission during cytokinesis. Work over the past 2 decades has shown that a diverse cohort of viruses critically rely upon host ESCRT machinery for virus replication and envelopment. More recent studies reported that intracellular bacteria and the intracellular parasite Toxoplasma gondii benefit from, antagonize, or exploit host ESCRT machinery to preserve their intracellular niche, gain resources, or egress from infected cells. Here, we review how intracellular pathogens interact with the ESCRT machinery of their hosts, highlighting the variety of strategies they use to bind ESCRT complexes using short linear amino acid motifs like those used by ESCRTs to sequentially assemble on target membranes. Future work exposing new mechanisms of this molecular mimicry will yield novel insight of how pathogens exploit host ESCRT machinery and how ESCRTs facilitate key cellular processes.
Collapse
Affiliation(s)
- Yolanda Rivera-Cuevas
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Vern B. Carruthers
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| |
Collapse
|
4
|
Gurunathan S, Kang MH, Qasim M, Khan K, Kim JH. Biogenesis, Membrane Trafficking, Functions, and Next Generation Nanotherapeutics Medicine of Extracellular Vesicles. Int J Nanomedicine 2021; 16:3357-3383. [PMID: 34040369 PMCID: PMC8140893 DOI: 10.2147/ijn.s310357] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/25/2021] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of membrane-limited vesicles and multi-signal messengers loaded with biomolecules. Exosomes and ectosomes are two different types of EVs generated by all cell types. Their formation depends on local microdomains assembled in endocytic membranes for exosomes and in the plasma membrane for ectosomes. Further, EV release is a fundamental process required for intercellular communication in both normal physiology and pathological conditions to transmit/exchange bioactive molecules to recipient cells and the extracellular environment. The unique structure and composition of EVs enable them to serve as natural nanocarriers, and their physicochemical properties and biological functions can be used to develop next-generation nano and precision medicine. Knowledge of the cellular processes that govern EVs biology and membrane trafficking is essential for their clinical applications. However, in this rapidly expanding field, much remains unknown regarding EV origin, biogenesis, cargo sorting, and secretion, as well as EV-based theranostic platform generation. Hence, we present a comprehensive overview of the recent advances in biogenesis, membrane trafficking, and functions of EVs, highlighting the impact of nanoparticles and oxidative stress on EVs biogenesis and release and finally emphasizing the role of EVs as nanotherapeutic agents.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Min-Hee Kang
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Muhammad Qasim
- Center of Bioengineering and Nanomedicine, Department of Food Science, University of Otago, Dunedin, 9054, New Zealand
| | - Khalid Khan
- Science and Technology KPK, Peshawar, Pakistan
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| |
Collapse
|
5
|
Koohsarian P, Talebi A, Rahnama MA, Zomorrod MS, Kaviani S, Jalili A. Reviewing the role of cardiac exosomes in myocardial repair at a glance. Cell Biol Int 2021; 45:1352-1363. [PMID: 33289229 DOI: 10.1002/cbin.11515] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 11/14/2020] [Accepted: 11/28/2020] [Indexed: 12/11/2022]
Abstract
Exosome-based therapy is an emerging novel approach for myocardial infarction (MI) treatment. Exosomes are identified as extracellular vesicles that are produced within multivesicular bodies in the cells' cytosols and then are secreted from the cells. Exosomes are 30-100 nm in diameter that are released from viable cells and are different from other secreted vesicles such as apoptotic bodies and microvesicles in their origin and contents such as RNAs, proteins, and nucleic acid. The recent advances in exosome research have demonstrated the role of these bionanovesicles in the physiological, pathological, and molecular aspects of the heart. The results of in vitro and preclinical models have shown that exosomes from different cardiac cells can improve cardiac function following MI. For example, mesenchymal stem cells (MSCs) and cardiac progenitor cells (CPCs) containing exosomes can affect the proliferation, survival, and differentiation of cardiac fibroblasts and cardiomyocytes. Moreover, MSCs- and CPCs-derived exosomes can enhance the migration of endothelial cells. Exosome-based therapy approaches augment the cardiac function by multiple means, such as reducing fibrosis, stimulation of vascular angiogenesis, and proliferation of cardiomyocytes that result in replacing damaged heart tissue with newly generated functional myocytes. This review article aims to briefly discuss the recent advancements in the role of secreted exosomes in myocardial repair by focusing on cardiac cells-derived exosomes.
Collapse
Affiliation(s)
- Parisa Koohsarian
- Department of Hematology and Cell Therapy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Athar Talebi
- Department of Nervous System, Stem Cell Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Mahshid A Rahnama
- Department of Hematology and Cell Therapy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mina S Zomorrod
- Department of Hematology and Cell Therapy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeid Kaviani
- Department of Hematology and Cell Therapy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Arsalan Jalili
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Hematopoetic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Rathbun LI, Colicino EG, Manikas J, O'Connell J, Krishnan N, Reilly NS, Coyne S, Erdemci-Tandogan G, Garrastegui A, Freshour J, Santra P, Manning ML, Amack JD, Hehnly H. Cytokinetic bridge triggers de novo lumen formation in vivo. Nat Commun 2020; 11:1269. [PMID: 32152267 PMCID: PMC7062744 DOI: 10.1038/s41467-020-15002-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 02/14/2020] [Indexed: 02/03/2023] Open
Abstract
Multicellular rosettes are transient epithelial structures that serve as intermediates during diverse organ formation. We have identified a unique contributor to rosette formation in zebrafish Kupffer's vesicle (KV) that requires cell division, specifically the final stage of mitosis termed abscission. KV utilizes a rosette as a prerequisite before forming a lumen surrounded by ciliated epithelial cells. Our studies identify that KV-destined cells remain interconnected by cytokinetic bridges that position at the rosette's center. These bridges act as a landmark for directed Rab11 vesicle motility to deliver an essential cargo for lumen formation, CFTR (cystic fibrosis transmembrane conductance regulator). Here we report that premature bridge cleavage through laser ablation or inhibiting abscission using optogenetic clustering of Rab11 result in disrupted lumen formation. We present a model in which KV mitotic cells strategically place their cytokinetic bridges at the rosette center, where Rab11-associated vesicles transport CFTR to aid in lumen establishment.
Collapse
Affiliation(s)
- L I Rathbun
- Biology Department, Syracuse University, Syracuse, New York, USA
| | - E G Colicino
- Biology Department, Syracuse University, Syracuse, New York, USA
- Department of Cell and Developmental Biology, SUNY Upstate Medical School, Syracuse, New York, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - J Manikas
- Biology Department, Syracuse University, Syracuse, New York, USA
| | - J O'Connell
- Biology Department, Syracuse University, Syracuse, New York, USA
| | - N Krishnan
- Biology Department, Syracuse University, Syracuse, New York, USA
| | - N S Reilly
- Department of Physics and Astronomy, University of Rochester, Rochester, New York, USA
| | - S Coyne
- Department of Cell and Developmental Biology, SUNY Upstate Medical School, Syracuse, New York, USA
- Department of Biology, SUNY Geneseo, Geneseo, New York, USA
| | | | - A Garrastegui
- Biology Department, Syracuse University, Syracuse, New York, USA
| | - J Freshour
- Biology Department, Syracuse University, Syracuse, New York, USA
| | - P Santra
- Department of Cell and Developmental Biology, SUNY Upstate Medical School, Syracuse, New York, USA
| | - M L Manning
- Department of Physics, Syracuse University, Syracuse, New York, USA
| | - J D Amack
- Department of Cell and Developmental Biology, SUNY Upstate Medical School, Syracuse, New York, USA
| | - H Hehnly
- Biology Department, Syracuse University, Syracuse, New York, USA.
| |
Collapse
|
7
|
Abstract
The WW domain is a modular protein structure that recognizes the proline-rich Pro-Pro-x-Tyr (PPxY) motif contained in specific target proteins. The compact modular nature of the WW domain makes it ideal for mediating interactions between proteins in complex networks and signaling pathways of the cell (e.g. the Hippo pathway). As a result, WW domains play key roles in a plethora of both normal and disease processes. Intriguingly, RNA and DNA viruses have evolved strategies to hijack cellular WW domain-containing proteins and thereby exploit the modular functions of these host proteins for various steps of the virus life cycle, including entry, replication, and egress. In this review, we summarize key findings in this rapidly expanding field, in which new virus-host interactions continue to be identified. Further unraveling of the molecular aspects of these crucial virus-host interactions will continue to enhance our fundamental understanding of the biology and pathogenesis of these viruses. We anticipate that additional insights into these interactions will help support strategies to develop a new class of small-molecule inhibitors of viral PPxY-host WW-domain interactions that could be used as antiviral therapeutics.
Collapse
Affiliation(s)
- Ariel Shepley-McTaggart
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Hao Fan
- Bioinformatics Institute, Agency for Science, Technology, and Research (A*STAR), 30 Biopolis Street, Matrix #07-01, Singapore 138671.,Department of Biological Sciences (DBS), National University of Singapore, Singapore 119077.,Center for Computational Biology, DUKE-NUS Medical School, Singapore 169857
| | - Marius Sudol
- Department of Physiology, National University of Singapore, Singapore 119077.,Laboratory of Cancer Signaling and Domainopathies, Yong Loo Li School of Medicine, Block MD9, 2 Medical Drive #04-01, Singapore 117597.,Mechanobiology Institute, T-Lab, 5A Engineering Drive 1, Singapore 117411.,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Ronald N Harty
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
8
|
A Quantitative Genetic Interaction Map of HIV Infection. Mol Cell 2020; 78:197-209.e7. [PMID: 32084337 DOI: 10.1016/j.molcel.2020.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 01/10/2020] [Accepted: 02/02/2020] [Indexed: 12/16/2022]
Abstract
We have developed a platform for quantitative genetic interaction mapping using viral infectivity as a functional readout and constructed a viral host-dependency epistasis map (vE-MAP) of 356 human genes linked to HIV function, comprising >63,000 pairwise genetic perturbations. The vE-MAP provides an expansive view of the genetic dependencies underlying HIV infection and can be used to identify drug targets and study viral mutations. We found that the RNA deadenylase complex, CNOT, is a central player in the vE-MAP and show that knockout of CNOT1, 10, and 11 suppressed HIV infection in primary T cells by upregulating innate immunity pathways. This phenotype was rescued by deletion of IRF7, a transcription factor regulating interferon-stimulated genes, revealing a previously unrecognized host signaling pathway involved in HIV infection. The vE-MAP represents a generic platform that can be used to study the global effects of how different pathogens hijack and rewire the host during infection.
Collapse
|
9
|
Skinkle AD, Levental KR, Levental I. Cell-Derived Plasma Membrane Vesicles Are Permeable to Hydrophilic Macromolecules. Biophys J 2020; 118:1292-1300. [PMID: 32053777 DOI: 10.1016/j.bpj.2019.12.040] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 12/09/2019] [Accepted: 12/23/2019] [Indexed: 12/22/2022] Open
Abstract
Giant plasma membrane vesicles (GPMVs) are a widely used experimental platform for biochemical and biophysical analysis of isolated mammalian plasma membranes (PMs). A core advantage of these vesicles is that they maintain the native lipid and protein diversity of the PM while affording the experimental flexibility of synthetic giant vesicles. In addition to fundamental investigations of PM structure and composition, GPMVs have been used to evaluate the binding of proteins and small molecules to cell-derived membranes and the permeation of drug-like molecules through them. An important assumption of such experiments is that GPMVs are sealed, i.e., that permeation occurs by diffusion through the hydrophobic core rather than through hydrophilic pores. Here, we demonstrate that this assumption is often incorrect. We find that most GPMVs isolated using standard preparations are passively permeable to various hydrophilic solutes as large as 40 kDa, in contrast to synthetic giant unilamellar vesicles. We attribute this leakiness to stable, relatively large, and heterogeneous pores formed by rupture of vesicles from cells. Finally, we identify preparation conditions that minimize poration and allow evaluation of sealed GPMVs. These unexpected observations of GPMV poration are important for interpreting experiments utilizing GPMVs as PM models, particularly for drug permeation and membrane asymmetry.
Collapse
Affiliation(s)
- Allison D Skinkle
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas; Biological and Biomedical Sciences Program, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kandice R Levental
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Ilya Levental
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas.
| |
Collapse
|
10
|
Martín-Moreno A, Muñoz-Fernández MA. Dendritic Cells, the Double Agent in the War Against HIV-1. Front Immunol 2019; 10:2485. [PMID: 31708924 PMCID: PMC6820366 DOI: 10.3389/fimmu.2019.02485] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/04/2019] [Indexed: 12/19/2022] Open
Abstract
Human Immunodeficiency Virus (HIV) infects cells from the immune system and has thus developed tools to circumvent the host immunity and use it in its advance. Dendritic cells (DCs) are the first immune cells to encounter the HIV, and being the main antigen (Ag) presenting cells, they link the innate and the adaptive immune responses. While DCs work to promote an efficient immune response and halt the infection, HIV-1 has ways to take advantage of their role and uses DCs to gain faster and more efficient access to CD4+ T cells. Due to their ability to activate a specific immune response, DCs are promising candidates to achieve the functional cure of HIV-1 infection, but knowing the molecular partakers that determine the relationship between virus and cell is the key for the rational and successful design of a DC-based therapy. In this review, we summarize the current state of knowledge on how both DC subsets (myeloid and plasmacytoid DCs) act in presence of HIV-1, and focus on different pathways that the virus can take after binding to DC. First, we explore the consequences of HIV-1 recognition by each receptor on DCs, including CD4 and DC-SIGN. Second, we look at cellular mechanisms that prevent productive infection and weapons that turn cellular defense into a Trojan horse that hides the virus all the way to T cell. Finally, we discuss the possible outcomes of DC-T cell contact.
Collapse
Affiliation(s)
- Alba Martín-Moreno
- Sección de Inmunología, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain.,Instituto Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Mª Angeles Muñoz-Fernández
- Sección de Inmunología, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain.,Instituto Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Spanish HIV-HGM BioBank, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER BBN), Madrid, Spain
| |
Collapse
|
11
|
Cao J, Zhong MB, Toro CA, Zhang L, Cai D. Endo-lysosomal pathway and ubiquitin-proteasome system dysfunction in Alzheimer's disease pathogenesis. Neurosci Lett 2019; 703:68-78. [PMID: 30890471 PMCID: PMC6760990 DOI: 10.1016/j.neulet.2019.03.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/19/2019] [Accepted: 03/11/2019] [Indexed: 01/04/2023]
Abstract
Several lines of evidence have shown that defects in the endo-lysosomal autophagy degradation pathway and the ubiquitin-proteasome system play a role in Alzheimer's Disease (AD) pathogenesis and pathophysiology. Early pathological changes, such as marked enlargement of endosomal compartments, gradual accumulation of autophagic vacuoles (AVs) and lysosome dyshomeostasis, are well-recognized in AD. In addition to these pathological indicators, many genetic variants of key regulators in the endo-lysosomal autophagy networks and the ubiquitin-proteasome system have been found to be associated with AD. Furthermore, altered expression levels of key proteins in these pathways have been found in AD human brain tissues, primary cells and AD mouse models. In this review, we discuss potential disease mechanisms underlying the dysregulation of protein homeostasis governing systems. While the importance of two major protein degradation pathways in AD pathogenesis has been highlighted, targeted therapy at key components of these pathways has great potential in developing novel therapeutic interventions for AD. Future investigations are needed to define molecular mechanisms by which these complex regulatory systems become malfunctional at specific stages of AD development and progression, which will facilitate future development of novel therapeutic interventions. It is also critical to investigate all key components of the protein degradation pathways, both upstream and downstream, to improve our abilities to manipulate transport pathways with higher efficacy and less side effects.
Collapse
Affiliation(s)
- Jiqing Cao
- Research and Development, James J Peters VA Medical Center, Bronx, NY 10468, United States; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; The Central Hospital of The Hua Zhong University of Science and Technology, Wuhan, China.
| | - Margaret B Zhong
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Barnard College of Columbia University, New York, NY 10027, United States.
| | - Carlos A Toro
- Research and Development, James J Peters VA Medical Center, Bronx, NY 10468, United States; National Center for the Medical Consequences of Spinal Cord Injury, James J Peters VA Medical Center, Bronx, NY 10468, United States; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| | - Larry Zhang
- Research and Development, James J Peters VA Medical Center, Bronx, NY 10468, United States; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| | - Dongming Cai
- Research and Development, James J Peters VA Medical Center, Bronx, NY 10468, United States; Neurology Section, James J Peters VA Medical Center, Bronx, NY 10468, United States; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; The Central Hospital of The Hua Zhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
12
|
Johnson TB, Mach C, Grove R, Kelly R, Van Cott K, Blum P. Secretion and fusion of biogeochemically active archaeal membrane vesicles. GEOBIOLOGY 2018; 16:659-673. [PMID: 30019522 DOI: 10.1111/gbi.12306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/06/2018] [Accepted: 06/11/2018] [Indexed: 06/08/2023]
Abstract
Microbes belonging to the genus Metallosphaera oxidize sulfidic minerals. These organisms thrive at temperature extremes and are members of the archaeal phylum Crenarchaeota. Because they can employ a lithoautotrophic metabolism, energy availability likely limits their activity raising questions about how they conduct biogeochemical activity. Vesicles are membrane encapsulated structures produced by all biological lineages but using very different mechanisms. Across the Crenarchaeota, it has been proposed that a eukaryotic-like Endosomal Sorting Complex Required for Transport system promotes formation of these structures but in response to unknown signals and for undefined purposes. To address such questions, Metallosphaera sedula vesicle formation and function were studied under lithoautotrophic conditions. Energy deprivation was evaluated and found to stimulate vesicle synthesis while energy excess repressed vesicle formation. Purified vesicles adhered rapidly to the primary copper ore, chalcopyrite, and formed compact monolayers. These vesicle monolayers catalyzed iron oxidation and solubilization of mineralized copper in a time-dependent process. As these activities were membrane associated, their potential transfer by vesicle fusion to M. sedula cells was examined. Fluorophore-loaded vesicles rapidly transferred fluorescence under environmentally relevant conditions. Vesicles from a related archaeal species were also capable of fusion; however, this process was species-specific as vesicles from different species were incapable of fusion. In addition, vesicles produced by a copper-resistant M. sedula cell line transferred copper extrusion capacity along with improved viability over mutant M. sedula cells lacking copper transport proteins. Membrane vesicles may therefore play a role in modulating energy-related traits in geochemical environments by fusion-mediated protein delivery.
Collapse
Affiliation(s)
- Tyler B Johnson
- Center for Genetics, School of Biological Sciences, University of Nebraska, Lincoln, Nebraska
| | - Collin Mach
- Center for Genetics, School of Biological Sciences, University of Nebraska, Lincoln, Nebraska
| | - Ryan Grove
- Department of Biochemistry and the Redox Biology Center, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Robert Kelly
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina
| | - Kevin Van Cott
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska
| | - Paul Blum
- Center for Genetics, School of Biological Sciences, University of Nebraska, Lincoln, Nebraska
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, California
| |
Collapse
|
13
|
Gireud-Goss M, Reyes S, Wilson M, Farley M, Memarzadeh K, Srinivasan S, Sirisaengtaksin N, Yamashita S, Tsunoda S, Lang FF, Waxham MN, Bean AJ. Distinct mechanisms enable inward or outward budding from late endosomes/multivesicular bodies. Exp Cell Res 2018; 372:1-15. [PMID: 30144444 DOI: 10.1016/j.yexcr.2018.08.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 12/25/2022]
Abstract
Regulating the residence time of membrane proteins on the cell surface can modify their response to extracellular cues and allow for cellular adaptation in response to changing environmental conditions. The fate of membrane proteins that are internalized from the plasma membrane and arrive at the limiting membrane of the late endosome/multivesicular body (MVB) is dictated by whether they remain on the limiting membrane, bud into internal MVB vesicles, or bud outwardly from the membrane. The molecular details underlying the disposition of membrane proteins that transit this pathway and the mechanisms regulating these trafficking events are unclear. We established a cell-free system that reconstitutes budding of membrane protein cargo into internal MVB vesicles and onto vesicles that bud outwardly from the MVB membrane. Both budding reactions are cytosol-dependent and supported by Saccharomyces cerevisiae (yeast) cytosol. We observed that inward and outward budding from the MVB membrane are mechanistically distinct but may be linked, such that inhibition of inward budding triggers a re-routing of cargo from inward to outward budding vesicles, without affecting the number of vesicles that bud outwardly from MVBs.
Collapse
Affiliation(s)
- Monica Gireud-Goss
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA.
| | - Sahily Reyes
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA.
| | - Marenda Wilson
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA.
| | - Madeline Farley
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA.
| | - Kimiya Memarzadeh
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA.
| | | | - Natalie Sirisaengtaksin
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA.
| | - Shinji Yamashita
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Susan Tsunoda
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - M Neal Waxham
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | - Andrew J Bean
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Biochemistry and Cell Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA; Department of Pediatrics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
14
|
Han Z, Schwoerer MP, Hicks P, Liang J, Ruthel G, Berry CT, Freedman BD, Sagum CA, Bedford MT, Sidhu SS, Sudol M, Harty RN. Host Protein BAG3 is a Negative Regulator of Lassa VLP Egress. Diseases 2018; 6:diseases6030064. [PMID: 30011814 PMCID: PMC6163595 DOI: 10.3390/diseases6030064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 12/20/2022] Open
Abstract
Lassa fever virus (LFV) belongs to the Arenaviridae family and can cause acute hemorrhagic fever in humans. The LFV Z protein plays a central role in virion assembly and egress, such that independent expression of LFV Z leads to the production of virus-like particles (VLPs) that mimic egress of infectious virus. LFV Z contains both PTAP and PPPY L-domain motifs that are known to recruit host proteins that are important for mediating efficient virus egress and spread. The viral PPPY motif is known to interact with specific host WW-domain bearing proteins. Here we identified host WW-domain bearing protein BCL2 Associated Athanogene 3 (BAG3) as a LFV Z PPPY interactor using our proline-rich reading array of WW-domain containing mammalian proteins. BAG3 is a stress-induced molecular co-chaperone that functions to regulate cellular protein homeostasis and cell survival via Chaperone-Assisted Selective Autophagy (CASA). Similar to our previously published findings for the VP40 proteins of Ebola and Marburg viruses, our results using VLP budding assays, BAG3 knockout cells, and confocal microscopy indicate that BAG3 is a WW-domain interactor that negatively regulates egress of LFV Z VLPs, rather than promoting VLP release. Our results suggest that CASA and specifically BAG3 may represent a novel host defense mechanism, whereby BAG3 may dampen egress of several hemorrhagic fever viruses by interacting and interfering with the budding function of viral PPxY-containing matrix proteins.
Collapse
Affiliation(s)
- Ziying Han
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Michael P Schwoerer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Philip Hicks
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Jingjing Liang
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Gordon Ruthel
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Corbett T Berry
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Bruce D Freedman
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Cari A Sagum
- Department of Epigenetics & Molecular Carcinogenesis, M.D. Anderson Cancer Center, University of Texas, Smithville, TX 78957, USA.
| | - Mark T Bedford
- Department of Epigenetics & Molecular Carcinogenesis, M.D. Anderson Cancer Center, University of Texas, Smithville, TX 78957, USA.
| | - Sachdev S Sidhu
- Department of Molecular Genetics, University of Toronto, Toronto, ON M1C 1A4, Canada.
| | - Marius Sudol
- Department of Physiology, Institute for Molecular and Cell Biology (IMCB, AStar), National University of Singapore, Singapore 119077, Singapore.
| | - Ronald N Harty
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
15
|
Park M, Upton D, Blackmon M, Dixon V, Craver S, Neal D, Perkins D. Anacardic acid inhibits pancreatic cancer cell growth, and potentiates chemotherapeutic effect by Chmp1A - ATM - p53 signaling pathway. Altern Ther Health Med 2018; 18:71. [PMID: 29463243 PMCID: PMC5819688 DOI: 10.1186/s12906-018-2139-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 02/14/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Pancreatic cancer is one of the leading causes of cancer related death and its incidence has risen steadily. Although anticancer drugs have been developed based on the new molecular findings, the drugs have produced unsatisfactory results due to toxicity and resistance. Thus, a complementary therapeutic intervention is urgently needed for pancreatic cancer patients. METHODS The aim of this study was to assess the potential therapeutic effect of Anacardic acid on pancreatic cancer in vitro and elucidate its underlying mechanisms. Human pancreatic cancer cells were treated with Anacardic acid and assessed for the cytotoxic effect using MTT and spheroid formation assays. Using the same methods, the synergy between Anacardic acid and 5-Fluorouracil or Gemcitabine was determined. To elucidate the underlying molecular mechanisms, Western blot analysis and immunocytochemistry were performed on cancer cells treated with Anacardic acid alone or in combination with 5-Fluorouracil or Gemcitabine. Chromatin Modifying Protein 1A (Chmp1A), Ataxia Telangiectasia Mutated (ATM), and p53 were the primary signaling molecules examined. In addition, Chmp1A was silenced with shRNA to examine the necessity of Chmp1A for the anticancer effect of Anacardic acid, 5-Fluorouracil, or Gemcitabine. RESULTS Anacardic acid induced an anticancer effect in pancreatic cancer cell lines in a dose dependent manner, and increased the cytotoxicity of 5-Fluorouracil or Gemcitabine in MTT cell viability assays. In spheroid formation assays, spheroids formed were smaller in size and in number upon Anacardic acid treatment compared to control. Mechanistically, Anacardic acid exerted its anticancer activity via the activation of Chmp1A, ATM, and p53. Interestingly, 5-Fluorouracil and Gemcitabine also induced an increase in Chmp1A protein level, suggesting that Chmp1A might mediate the cytotoxic action of chemotherapeutics. Silencing experiments indicate that Chmp1A is required for the action of Anacardic acid, but not for 5-Fluorouracil or Gemcitabine. CONCLUSIONS Our data suggests that Anacardic Acid might be a promising complementary supplement to slow the initiation or progression of pancreatic cancer.
Collapse
|
16
|
Filovirus proteins for antiviral drug discovery: Structure/function of proteins involved in assembly and budding. Antiviral Res 2018; 150:183-192. [DOI: 10.1016/j.antiviral.2017.12.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/20/2017] [Accepted: 12/28/2017] [Indexed: 01/30/2023]
|
17
|
The ins and outs of eukaryotic viruses: Knowledge base and ontology of a viral infection. PLoS One 2017; 12:e0171746. [PMID: 28207819 PMCID: PMC5313201 DOI: 10.1371/journal.pone.0171746] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 01/25/2017] [Indexed: 12/19/2022] Open
Abstract
Viruses are genetically diverse, infect a wide range of tissues and host cells and follow unique processes for replicating themselves. All these processes were investigated and indexed in ViralZone knowledge base. To facilitate standardizing data, a simple ontology of viral life-cycle terms was developed to provide a common vocabulary for annotating data sets. New terminology was developed to address unique viral replication cycle processes, and existing terminology was modified and adapted. The virus life-cycle is classically described by schematic pictures. Using this ontology, it can be represented by a combination of successive terms: “entry”, “latency”, “transcription”, “replication” and “exit”. Each of these parts is broken down into discrete steps. For example Zika virus “entry” is broken down in successive steps: “Attachment”, “Apoptotic mimicry”, “Viral endocytosis/ macropinocytosis”, “Fusion with host endosomal membrane”, “Viral factory”. To demonstrate the utility of a standard ontology for virus biology, this work was completed by annotating virus data in the ViralZone, UniProtKB and Gene Ontology databases.
Collapse
|
18
|
Chutiwitoonchai N, Siarot L, Takeda E, Shioda T, Ueda M, Aida Y. HIV-1 Vpr Abrogates the Effect of TSG101 Overexpression to Support Virus Release. PLoS One 2016; 11:e0163100. [PMID: 27648839 PMCID: PMC5029901 DOI: 10.1371/journal.pone.0163100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 09/02/2016] [Indexed: 11/18/2022] Open
Abstract
HIV-1 budding requires interaction between Gag and cellular TSG101 to initiate viral particle assembly and release via the endosomal sorting complexes required for transport (ESCRT) pathway. However, some reports show that overexpression of TSG101 inhibits virus release by disruption of Gag targeting process. Since a HIV-1 accessory protein, Vpr binds to Gag p6 domain at the position close to the binding site for TSG101, whether Vpr implicates TSG101 overexpression effect has not been investigated. Here, we found that Vpr abrogates TSG101 overexpression effect to rescue viral production. Co-transfection of TSG101 and Gag with Vpr prevented TSG101-induced Gag accumulation in endosomes and lysosomes. In addition, Vpr rescued virus-like particle (VLP) production in a similar manner as a lysosomal inhibitor, Bafilomycin A1 indicating that Vpr inhibits TSG101-induced Gag downregulation via lysosomal pathway. Vpr and Gag interaction is required to counteract TSG101 overexpression effect since Vpr A30F mutant which is unable to interact with Gag and incorporate into virions, reduced ability to prevent Gag accumulation and to rescue VLP production. In addition, GST pull-down assays and Biacore analysis revealed that Vpr competed with TSG101 for Gag binding. These results indicate that Vpr overcomes the effects of TSG101 overexpression to support viral production by competing with TSG101 to bind Gag.
Collapse
Affiliation(s)
- Nopporn Chutiwitoonchai
- Viral Infectious Diseases Unit, RIKEN, Wako, Saitama, Japan
- Japan Foundation for AIDS Prevention, Chiyoda-ku, Tokyo, Japan
| | - Lowela Siarot
- Viral Infectious Diseases Unit, RIKEN, Wako, Saitama, Japan
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Eri Takeda
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Tatsuo Shioda
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Motoki Ueda
- Nano Medical Engineering Laboratory, RIKEN, Wako, Saitama, Japan
| | - Yoko Aida
- Viral Infectious Diseases Unit, RIKEN, Wako, Saitama, Japan
- * E-mail:
| |
Collapse
|
19
|
Kumar N, Barua S, Thachamvally R, Tripathi BN. Systems Perspective of Morbillivirus Replication. J Mol Microbiol Biotechnol 2016; 26:389-400. [DOI: 10.1159/000448842] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 08/02/2016] [Indexed: 11/19/2022] Open
Abstract
Systems biology refers to system-wide changes in biological components such as RNA/DNA (genomics), protein (proteomics) and lipids (lipidomics). In this review, we provide comprehensive information about morbillivirus replication. Besides discussing the role of individual viral/host proteins in virus replication, we also discuss how systems-level analyses could improve our understanding of morbillivirus replication, host-pathogen interaction, immune response and disease resistance. Finally, we discuss how viroinformatics is likely to provide important insights for understanding genome-genome, genome-protein and protein-protein interactions.
Collapse
|
20
|
Christ L, Wenzel EM, Liestøl K, Raiborg C, Campsteijn C, Stenmark H. ALIX and ESCRT-I/II function as parallel ESCRT-III recruiters in cytokinetic abscission. J Cell Biol 2016; 212:499-513. [PMID: 26929449 PMCID: PMC4772496 DOI: 10.1083/jcb.201507009] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cytokinetic abscission, the final stage of cell division, is mediated by the ESCRT machinery. Here, Christ et al. dissect the regulation of ESCRT-III recruitment and abscission timing and identify an intersection with abscission checkpoint signaling in cells with chromatin bridges. Cytokinetic abscission, the final stage of cell division where the two daughter cells are separated, is mediated by the endosomal sorting complex required for transport (ESCRT) machinery. The ESCRT-III subunit CHMP4B is a key effector in abscission, whereas its paralogue, CHMP4C, is a component in the abscission checkpoint that delays abscission until chromatin is cleared from the intercellular bridge. How recruitment of these components is mediated during cytokinesis remains poorly understood, although the ESCRT-binding protein ALIX has been implicated. Here, we show that ESCRT-II and the ESCRT-II–binding ESCRT-III subunit CHMP6 cooperate with ESCRT-I to recruit CHMP4B, with ALIX providing a parallel recruitment arm. In contrast to CHMP4B, we find that recruitment of CHMP4C relies predominantly on ALIX. Accordingly, ALIX depletion leads to furrow regression in cells with chromosome bridges, a phenotype associated with abscission checkpoint signaling failure. Collectively, our work reveals a two-pronged recruitment of ESCRT-III to the cytokinetic bridge and implicates ALIX in abscission checkpoint signaling.
Collapse
Affiliation(s)
- Liliane Christ
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, N-0379 Oslo, Norway Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, N-0379 Oslo, Norway
| | - Eva M Wenzel
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, N-0379 Oslo, Norway Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, N-0379 Oslo, Norway
| | - Knut Liestøl
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, N-0379 Oslo, Norway Department of Informatics, University of Oslo, N-0373 Oslo, Norway
| | - Camilla Raiborg
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, N-0379 Oslo, Norway Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, N-0379 Oslo, Norway
| | - Coen Campsteijn
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, N-0379 Oslo, Norway Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, N-0379 Oslo, Norway
| | - Harald Stenmark
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, N-0379 Oslo, Norway Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, N-0379 Oslo, Norway
| |
Collapse
|
21
|
Abstract
Exosomes are extracellular vesicles first described as such 30 years ago and since implicated in cell-cell communication and the transmission of disease states, and explored as a means of drug discovery. Yet fundamental questions about their biology remain unanswered. Here I explore what exosomes are, highlight the difficulties in studying them and explain the current definition and some of the outstanding issues in exosome biology.
Collapse
Affiliation(s)
- James R Edgar
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK.
| |
Collapse
|
22
|
Park A, Yun T, Vigant F, Pernet O, Won ST, Dawes BE, Bartkowski W, Freiberg AN, Lee B. Nipah Virus C Protein Recruits Tsg101 to Promote the Efficient Release of Virus in an ESCRT-Dependent Pathway. PLoS Pathog 2016; 12:e1005659. [PMID: 27203423 PMCID: PMC4874542 DOI: 10.1371/journal.ppat.1005659] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 05/04/2016] [Indexed: 12/21/2022] Open
Abstract
The budding of Nipah virus, a deadly member of the Henipavirus genus within the Paramyxoviridae, has been thought to be independent of the host ESCRT pathway, which is critical for the budding of many enveloped viruses. This conclusion was based on the budding properties of the virus matrix protein in the absence of other virus components. Here, we find that the virus C protein, which was previously investigated for its role in antagonism of innate immunity, recruits the ESCRT pathway to promote efficient virus release. Inhibition of ESCRT or depletion of the ESCRT factor Tsg101 abrogates the C enhancement of matrix budding and impairs live Nipah virus release. Further, despite the low sequence homology of the C proteins of known henipaviruses, they all enhance the budding of their cognate matrix proteins, suggesting a conserved and previously unknown function for the henipavirus C proteins. Nipah virus is a deadly pathogen (40–100% mortality) that has yearly outbreaks in Southeast Asia, resulting from spillover from its natural fruit bat reservoir. The viral C protein is one of only nine virus proteins, but its role in promoting virus replication is not fully understood. Here, we found that the C protein promotes the efficient release of budding Nipah virus from infected cells. It does so by recruiting an essential factor in the host ESCRT complex, Tsg101. The ESCRT complex has well-characterized functions in mediating membrane pinching off events that resemble virus budding. Further, we found that the C proteins of related viruses within the same genus (Henipavirus) also promote virus budding, suggesting that this previously unknown function of the henipavirus C proteins is conserved. This work illuminates the basic biology of henipaviruses with significant outbreak and public health concern, and opens the door to further lines of inquiry.
Collapse
Affiliation(s)
- Arnold Park
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California, United States of America
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Tatyana Yun
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Frederic Vigant
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Olivier Pernet
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California, United States of America
| | - Sohui T. Won
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Brian E. Dawes
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Wojciech Bartkowski
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California, United States of America
| | - Alexander N. Freiberg
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Benhur Lee
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California, United States of America
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
23
|
Abstract
Why some viruses are enveloped while others lack an outer lipid bilayer is a major question in viral evolution but one that has received relatively little attention. The viral envelope serves several functions, including protecting the RNA or DNA molecule(s), evading recognition by the immune system, and facilitating virus entry. Despite these commonalities, viral envelopes come in a wide variety of shapes and configurations. The evolution of the viral envelope is made more puzzling by the fact that nonenveloped viruses are able to infect a diverse range of hosts across the tree of life. We reviewed the entry, transmission, and exit pathways of all (101) viral families on the 2013 International Committee on Taxonomy of Viruses (ICTV) list. By doing this, we revealed a strong association between the lack of a viral envelope and the presence of a cell wall in the hosts these viruses infect. We were able to propose a new hypothesis for the existence of enveloped and nonenveloped viruses, in which the latter represent an adaptation to cells surrounded by a cell wall, while the former are an adaptation to animal cells where cell walls are absent. In particular, cell walls inhibit viral entry and exit, as well as viral transport within an organism, all of which are critical waypoints for successful infection and spread. Finally, we discuss how this new model for the origin of the viral envelope impacts our overall understanding of virus evolution.
Collapse
|
24
|
Abstract
Eukaryotic cells have been confronted throughout their evolution with potentially lethal plasma membrane injuries, including those caused by osmotic stress, by infection from bacterial toxins and parasites, and by mechanical and ischemic stress. The wounded cell can survive if a rapid repair response is mounted that restores boundary integrity. Calcium has been identified as the key trigger to activate an effective membrane repair response that utilizes exocytosis and endocytosis to repair a membrane tear, or remove a membrane pore. We here review what is known about the cellular and molecular mechanisms of membrane repair, with particular emphasis on the relevance of repair as it relates to disease pathologies. Collective evidence reveals membrane repair employs primitive yet robust molecular machinery, such as vesicle fusion and contractile rings, processes evolutionarily honed for simplicity and success. Yet to be fully understood is whether core membrane repair machinery exists in all cells, or whether evolutionary adaptation has resulted in multiple compensatory repair pathways that specialize in different tissues and cells within our body.
Collapse
Affiliation(s)
- Sandra T Cooper
- Institute for Neuroscience and Muscle Research, Kids Research Institute, The Children's Hospital at Westmead, Sydney, New South Wales, Australia; Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia; and Department of Cellular Biology and Anatomy, Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, Georgia
| | - Paul L McNeil
- Institute for Neuroscience and Muscle Research, Kids Research Institute, The Children's Hospital at Westmead, Sydney, New South Wales, Australia; Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia; and Department of Cellular Biology and Anatomy, Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, Georgia
| |
Collapse
|
25
|
Peñalva MA, Lucena-Agell D, Arst HN. Liaison alcaline: Pals entice non-endosomal ESCRTs to the plasma membrane for pH signaling. Curr Opin Microbiol 2015; 22:49-59. [PMID: 25460796 DOI: 10.1016/j.mib.2014.09.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/02/2014] [Accepted: 09/12/2014] [Indexed: 12/21/2022]
Abstract
The alkaline pH-responsive Pal/Rim signal transduction pathway mediating regulation of gene expression by ambient pH has been extensively studied in Aspergillus nidulans and Saccharomyces cerevisiae. In A. nidulans, PalH, PalI, PalF, PalC, PalA and PalB are required for the proteolytic activation of the executing transcription factor PacC. Although necessary, Pal proteins are insufficient to transmit the signal, which additionally requires ESCRT-I, II and Vps20 with Snf7 in ESCRT-III. Although this initially suggested cooperation between a plasma membrane sensor and an ESCRT-containing Pal complex on endosomes, recent evidence convincingly indicates that pH signaling actually takes place in plasma membrane-associated foci in which Pal proteins and an ESCRT-III polymer scaffold cooperate for pH signaling purposes, representing another non-endosomal role of ESCRT components.
Collapse
Affiliation(s)
- Miguel A Peñalva
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas CSIC, Ramiro de Maeztu 9, Madrid 28040, Spain.
| | | | | |
Collapse
|
26
|
Quantitative Proteomic Analysis of BHK-21 Cells Infected with Foot-and-Mouth Disease Virus Serotype Asia 1. PLoS One 2015; 10:e0132384. [PMID: 26161868 PMCID: PMC4498813 DOI: 10.1371/journal.pone.0132384] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 06/12/2015] [Indexed: 01/03/2023] Open
Abstract
Stable isotope labeling with amino acids in cell culture (SILAC) was used to quantitatively study the host cell gene expression profile, in order to achieve an unbiased overview of the protein expression changes in BHK-21 cells infected with FMDV serotype Asia 1. The SILAC-based approach identified overall 2,141 proteins, 153 of which showed significant alteration in the expression level 6 h post FMDV infection (57 up-regulated and 96 down-regulated). Among these proteins, six cellular proteins, including three down-regulated (VPS28, PKR, EVI5) and three up-regulated (LYPLA1, SEC62 and DARs), were selected according to the significance of the changes and/or the relationship with PKR. The expression level and pattern of the selected proteins were validated by immunoblotting and confocal microscopy. Furthermore, the functions of these cellular proteins were assessed by small interfering RNA-mediated depletion, and their functional importance in the replication of FMDV was demonstrated by western blot, reverse transcript PCR (RT-PCR) and 50% Tissue Culture Infective Dose (TCID50). The results suggest that FMDV infection may have effects on the expression of specific cellular proteins to create more favorable conditions for FMDV infection. This study provides novel data that can be utilized to understand the interactions between FMDV and the host cell.
Collapse
|
27
|
Dutta S, Saha N, Ray A, Sarkar S. Significantly Diverged Did2/Vps46 Orthologues from the Protozoan Parasite Giardia lamblia. Curr Microbiol 2015; 71:333-40. [PMID: 26068593 DOI: 10.1007/s00284-015-0844-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 04/13/2015] [Indexed: 10/23/2022]
Abstract
The endosomal compartment performs extensive sorting functions in most eukaryotes, some of which are accomplished with the help of the multivesicular body (MVB) sorting pathway. This pathway depends on the sequential action of complexes, termed the endosomal sorting complex required for transport (ESCRT). After successful sorting, the crucial step of recycling of the ESCRT complex components requires the activation of the AAA ATPase Vps4, and Did2/Vps46 plays an important role in this activation event. The endolysosomal system of the protozoan parasite Giardia lamblia appears to lack complexity, for instead of having distinct early endosomes, late endosomes and lysosomes, there are only peripheral vesicles (PVs) that are located close to the cell periphery. Additionally, comparative genomics studies predict the presence of only a subset of the ESCRT components in G. lamblia. Thus, it is possible that the MVB pathway is not functional in G. lamblia. To address this issue, the present study focused on the two putative orthologues of Did2/Vps46 of G. lamblia as their function is likely to be pivotal for a functional MVB sorting pathway. In spite of considerable sequence divergence, compared to other eukaryotic orthologues, the proteins encoded by both these genes have the ability to function as Did2/Vps46 in the context of the yeast ESCRT pathway. Furthermore, they also localized to the cellular periphery, where PVs are also located. Thus, this report is the first to provide experimental evidence indicating the presence of a functional ESCRT component in G. lamblia by characterizing the putative Did2/Vps46 orthologues.
Collapse
Affiliation(s)
- Somnath Dutta
- Department of Biochemistry (Room 226), Bose Institute, Centenary Campus, P 1/12 C.I.T. Scheme VII M, Kolkata, 700054, West Bengal, India
| | | | | | | |
Collapse
|
28
|
Madara JJ, Han Z, Ruthel G, Freedman BD, Harty RN. The multifunctional Ebola virus VP40 matrix protein is a promising therapeutic target. Future Virol 2015; 10:537-546. [PMID: 26120351 DOI: 10.2217/fvl.15.6] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The highly virulent nature of Ebola virus, evident from the 2014 West African pandemic, highlights the need to develop vaccines or therapeutic agents that limit the pathogenesis and spread of this virus. While vaccines represent an obvious approach, targeting virus interactions with host proteins that critically regulate the virus lifecycle also represent important therapeutic strategies. Among Ebola virus proteins at this critical interface is its matrix protein, VP40, which is abundantly expressed during infection and plays a number of critical roles in the viral lifecycle. In addition to regulating viral transcription, VP40 coordinates virion assembly and budding from infected cells. Details of the molecular mechanisms underpinning these essential functions are currently being elucidated, with a particular emphasis on its interactions with host proteins that control virion assembly and egress. This review focuses on the strategies geared toward developing novel therapeutic agents that target VP40-specific control of host functions critical to virion transcription, assembly and egress.
Collapse
Affiliation(s)
- Jonathan J Madara
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | - Ziying Han
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | - Gordon Ruthel
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | - Bruce D Freedman
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | - Ronald N Harty
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104, USA
| |
Collapse
|
29
|
Ihms EC, Foster MP. MESMER: minimal ensemble solutions to multiple experimental restraints. Bioinformatics 2015; 31:1951-8. [PMID: 25673340 DOI: 10.1093/bioinformatics/btv079] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 02/03/2015] [Indexed: 11/12/2022] Open
Abstract
MOTIVATION Macromolecular structures and interactions are intrinsically heterogeneous, temporally adopting a range of configurations that can confound the analysis of data from bulk experiments. To obtain quantitative insights into heterogeneous systems, an ensemble-based approach can be employed, in which predicted data computed from a collection of models is compared to the observed experimental results. By simultaneously fitting orthogonal structural data (e.g. small-angle X-ray scattering, nuclear magnetic resonance residual dipolar couplings, dipolar electron-electron resonance spectra), the range and population of accessible macromolecule structures can be probed. RESULTS We have developed MESMER, software that enables the user to identify ensembles that can recapitulate experimental data by refining thousands of component collections selected from an input pool of potential structures. The MESMER suite includes a powerful graphical user interface (GUI) to streamline usage of the command-line tools, calculate data from structure libraries and perform analyses of conformational and structural heterogeneity. To allow for incorporation of other data types, modular Python plugins enable users to compute and fit data from nearly any type of quantitative experimental data. RESULTS Conformational heterogeneity in three macromolecular systems was analyzed with MESMER, demonstrating the utility of the streamlined, user-friendly software. AVAILABILITY AND IMPLEMENTATION https://code.google.com/p/mesmer/
Collapse
Affiliation(s)
- Elihu C Ihms
- Ohio State University Biophysics Program, Department of Chemistry and Biochemistry, and Center for RNA Biology, The Ohio State University, Columbus, OH, USA Ohio State University Biophysics Program, Department of Chemistry and Biochemistry, and Center for RNA Biology, The Ohio State University, Columbus, OH, USA
| | - Mark P Foster
- Ohio State University Biophysics Program, Department of Chemistry and Biochemistry, and Center for RNA Biology, The Ohio State University, Columbus, OH, USA Ohio State University Biophysics Program, Department of Chemistry and Biochemistry, and Center for RNA Biology, The Ohio State University, Columbus, OH, USA Ohio State University Biophysics Program, Department of Chemistry and Biochemistry, and Center for RNA Biology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
30
|
Martinez JP, Sasse F, Brönstrup M, Diez J, Meyerhans A. Antiviral drug discovery: broad-spectrum drugs from nature. Nat Prod Rep 2015; 32:29-48. [PMID: 25315648 DOI: 10.1039/c4np00085d] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Covering: up to April 2014. The development of drugs with broad-spectrum antiviral activities is a long pursued goal in drug discovery. It has been shown that blocking co-opted host-factors abrogates the replication of many viruses, yet the development of such host-targeting drugs has been met with scepticism mainly due to toxicity issues and poor translation to in vivo models. With the advent of new and more powerful screening assays and prediction tools, the idea of a drug that can efficiently treat a wide range of viral infections by blocking specific host functions has re-bloomed. Here we critically review the state-of-the-art in broad-spectrum antiviral drug discovery. We discuss putative targets and treatment strategies, with particular focus on natural products as promising starting points for antiviral lead development.
Collapse
Affiliation(s)
- J P Martinez
- Infection Biology Group, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
| | | | | | | | | |
Collapse
|
31
|
Bhutta MS, McInerny CJ, Gould GW. ESCRT function in cytokinesis: location, dynamics and regulation by mitotic kinases. Int J Mol Sci 2014; 15:21723-39. [PMID: 25429432 PMCID: PMC4284674 DOI: 10.3390/ijms151221723] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 11/17/2014] [Accepted: 11/17/2014] [Indexed: 01/22/2023] Open
Abstract
Mammalian cytokinesis proceeds by constriction of an actomyosin ring and furrow ingression, resulting in the formation of the midbody bridge connecting two daughter cells. At the centre of the midbody resides the Flemming body, a dense proteinaceous ring surrounding the interlocking ends of anti-parallel microtubule arrays. Abscission, the terminal step of cytokinesis, occurs near the Flemming body. A series of broad processes govern abscission: the initiation and stabilisation of the abscission zone, followed by microtubule severing and membrane scission-The latter mediated by the endosomal sorting complex required for transport (ESCRT) proteins. A key goal of cell and developmental biologists is to develop a clear understanding of the mechanisms that underpin abscission, and how the spatiotemporal coordination of these events with previous stages in cell division is accomplished. This article will focus on the function and dynamics of the ESCRT proteins in abscission and will review recent work, which has begun to explore how these complex protein assemblies are regulated by the cell cycle machinery.
Collapse
Affiliation(s)
- Musab S Bhutta
- Henry Wellcome Laboratory of Cell Biology, Davidson Building, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| | - Christopher J McInerny
- Henry Wellcome Laboratory of Cell Biology, Davidson Building, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| | - Gwyn W Gould
- Henry Wellcome Laboratory of Cell Biology, Davidson Building, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
32
|
Bhutta MS, Roy B, Gould GW, McInerny CJ. A complex network of interactions between mitotic kinases, phosphatases and ESCRT proteins regulates septation and membrane trafficking in S. pombe. PLoS One 2014; 9:e111789. [PMID: 25356547 PMCID: PMC4214795 DOI: 10.1371/journal.pone.0111789] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 08/27/2014] [Indexed: 11/18/2022] Open
Abstract
Cytokinesis and cell separation are critical events in the cell cycle. We show that Endosomal Sorting Complex Required for Transport (ESCRT) genes are required for cell separation in Schizosaccharomyces pombe. We identify genetic interactions between ESCRT proteins and polo and aurora kinases and Cdc14 phosphatase that manifest as impaired growth and exacerbated defects in septation, suggesting that the encoded proteins function together to control these processes. Furthermore, we observed defective endosomal sorting in mutants of plo1, ark1 and clp1, as has been reported for ESCRT mutants, consistent with a role for these kinases in the control of ESCRT function in membrane traffic. Multiple observations indicate functional interplay between polo and ESCRT components: firstly, two-hybrid in vivo interactions are reported between Plo1p and Sst4p, Vps28p, Vps25p, Vps20p and Vps32p; secondly, co-immunoprecipitation of human homologues of Vps20p, Vps32p, Vps24p and Vps2p by human Plk1; and thirdly, in vitro phosphorylation of budding yeast Vps32p and Vps20p by polo kinase. Two-hybrid analyses also identified interactions between Ark1p and Vps20p and Vps32p, and Clp1p and Vps28p. These experiments indicate a network of interactions between ESCRT proteins, plo1, ark1 and clp1 that coordinate membrane trafficking and cell separation in fission yeast.
Collapse
Affiliation(s)
- Musab S. Bhutta
- Henry Wellcome Laboratory of Cell Biology, Davidson Building, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Brinta Roy
- Henry Wellcome Laboratory of Cell Biology, Davidson Building, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Gwyn W. Gould
- Henry Wellcome Laboratory of Cell Biology, Davidson Building, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Christopher J. McInerny
- Henry Wellcome Laboratory of Cell Biology, Davidson Building, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
33
|
Brodsky FM, Sosa RT, Ybe JA, O'Halloran TJ. Unconventional functions for clathrin, ESCRTs, and other endocytic regulators in the cytoskeleton, cell cycle, nucleus, and beyond: links to human disease. Cold Spring Harb Perspect Biol 2014; 6:a017004. [PMID: 25183831 DOI: 10.1101/cshperspect.a017004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The roles of clathrin, its regulators, and the ESCRT (endosomal sorting complex required for transport) proteins are well defined in endocytosis. These proteins can also participate in intracellular pathways that are independent of endocytosis and even independent of the membrane trafficking function of these proteins. These nonendocytic functions involve unconventional biochemical interactions for some endocytic regulators, but can also exploit known interactions for nonendocytic functions. The molecular basis for the involvement of endocytic regulators in unconventional functions that influence the cytoskeleton, cell cycle, signaling, and gene regulation are described here. Through these additional functions, endocytic regulators participate in pathways that affect infection, glucose metabolism, development, and cellular transformation, expanding their significance in human health and disease.
Collapse
Affiliation(s)
- Frances M Brodsky
- Department of Bioengineering and Therapeutic Sciences, Departments of Pharmaceutical Chemistry and Microbiology and Immunology, The G.W. Hooper Foundation, University of California, San Francisco, San Francisco, California 94143-0552
| | - R Thomas Sosa
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, University of Texas, Austin, Texas 78712-1095
| | - Joel A Ybe
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, Indiana 47405
| | - Theresa J O'Halloran
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, University of Texas, Austin, Texas 78712-1095
| |
Collapse
|
34
|
Diaz-Rohrer B, Levental KR, Levental I. Rafting through traffic: Membrane domains in cellular logistics. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:3003-3013. [PMID: 25130318 DOI: 10.1016/j.bbamem.2014.07.029] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/28/2014] [Accepted: 07/31/2014] [Indexed: 01/03/2023]
Abstract
The intricate and tightly regulated organization of eukaryotic cells into spatially and functionally distinct membrane-bound compartments is a defining feature of complex organisms. These compartments are defined by their lipid and protein compositions, with their limiting membrane as the functional interface to the rest of the cell. Thus, proper segregation of membrane proteins and lipids is necessary for the maintenance of organelle identity, and this segregation must be maintained despite extensive, rapid membrane exchange between compartments. Sorting processes of high efficiency and fidelity are required to avoid potentially deleterious mis-targeting and maintain cellular function. Although much molecular machinery associated with membrane traffic (i.e. membrane budding/fusion/fission) has been characterized both structurally and biochemically, the mechanistic details underlying the tightly regulated distribution of membranes between subcellular locations remain to be elucidated. This review presents evidence for the role of ordered lateral membrane domains known as lipid rafts in both biosynthetic sorting in the late secretory pathway, as well as endocytosis and recycling to/from the plasma membrane. Although such evidence is extensive and the involvement of membrane domains in sorting is definitive, specific mechanistic details for raft-dependent sorting processes remain elusive.
Collapse
Affiliation(s)
- Blanca Diaz-Rohrer
- University of Texas Health Science Center at Houston, 6431 Fannin St, Houston, TX 77030, USA
| | - Kandice R Levental
- University of Texas Health Science Center at Houston, 6431 Fannin St, Houston, TX 77030, USA
| | - Ilya Levental
- University of Texas Health Science Center at Houston, 6431 Fannin St, Houston, TX 77030, USA; Cancer Prevention and Research Institute of Texas, USA.
| |
Collapse
|
35
|
Paramyxovirus glycoprotein incorporation, assembly and budding: a three way dance for infectious particle production. Viruses 2014; 6:3019-54. [PMID: 25105277 PMCID: PMC4147685 DOI: 10.3390/v6083019] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 07/24/2014] [Accepted: 07/25/2014] [Indexed: 12/21/2022] Open
Abstract
Paramyxoviruses are a family of negative sense RNA viruses whose members cause serious diseases in humans, such as measles virus, mumps virus and respiratory syncytial virus; and in animals, such as Newcastle disease virus and rinderpest virus. Paramyxovirus particles form by assembly of the viral matrix protein, the ribonucleoprotein complex and the surface glycoproteins at the plasma membrane of infected cells and subsequent viral budding. Two major glycoproteins expressed on the viral envelope, the attachment protein and the fusion protein, promote attachment of the virus to host cells and subsequent virus-cell membrane fusion. Incorporation of the surface glycoproteins into infectious progeny particles requires coordinated interplay between the three viral structural components, driven primarily by the matrix protein. In this review, we discuss recent progress in understanding the contributions of the matrix protein and glycoproteins in driving paramyxovirus assembly and budding while focusing on the viral protein interactions underlying this process and the intracellular trafficking pathways for targeting viral components to assembly sites. Differences in the mechanisms of particle production among the different family members will be highlighted throughout.
Collapse
|
36
|
Elia N, Ott C, Lippincott-Schwartz J. Incisive imaging and computation for cellular mysteries: lessons from abscission. Cell 2014; 155:1220-31. [PMID: 24315094 DOI: 10.1016/j.cell.2013.11.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Indexed: 02/06/2023]
Abstract
The final cleavage event that terminates cell division, abscission of the small, dense intercellular bridge, has been particularly challenging to resolve. Here, we describe imaging innovations that helped answer long-standing questions about the mechanism of abscission. We further explain how computational modeling of high-resolution data was employed to test hypotheses and generate additional insights. We present the model that emerges from application of these complimentary approaches. Similar experimental strategies will undoubtedly reveal exciting details about other underresolved cellular structures.
Collapse
Affiliation(s)
- Natalie Elia
- Department of Life Sciences and the NIBN, Ben Gurion University of the Negev, Beer Sheva 84105, Israel.
| | | | | |
Collapse
|
37
|
Abstract
Phosphoinositides (PIs) make up only a small fraction of cellular phospholipids, yet they control almost all aspects of a cell's life and death. These lipids gained tremendous research interest as plasma membrane signaling molecules when discovered in the 1970s and 1980s. Research in the last 15 years has added a wide range of biological processes regulated by PIs, turning these lipids into one of the most universal signaling entities in eukaryotic cells. PIs control organelle biology by regulating vesicular trafficking, but they also modulate lipid distribution and metabolism via their close relationship with lipid transfer proteins. PIs regulate ion channels, pumps, and transporters and control both endocytic and exocytic processes. The nuclear phosphoinositides have grown from being an epiphenomenon to a research area of its own. As expected from such pleiotropic regulators, derangements of phosphoinositide metabolism are responsible for a number of human diseases ranging from rare genetic disorders to the most common ones such as cancer, obesity, and diabetes. Moreover, it is increasingly evident that a number of infectious agents hijack the PI regulatory systems of host cells for their intracellular movements, replication, and assembly. As a result, PI converting enzymes began to be noticed by pharmaceutical companies as potential therapeutic targets. This review is an attempt to give an overview of this enormous research field focusing on major developments in diverse areas of basic science linked to cellular physiology and disease.
Collapse
Affiliation(s)
- Tamas Balla
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|
38
|
Agromayor M, Martin-Serrano J. Knowing when to cut and run: mechanisms that control cytokinetic abscission. Trends Cell Biol 2013; 23:433-41. [PMID: 23706391 DOI: 10.1016/j.tcb.2013.04.006] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 04/07/2013] [Accepted: 04/15/2013] [Indexed: 10/26/2022]
Abstract
Abscission, the final step of cytokinesis, mediates the severing of the membrane tether, or midbody, that connects two daughter cells. It is now recognized that abscission is a complex process requiring tight spatiotemporal regulation of its machinery to ensure equal chromosome segregation and cytoplasm content distribution between daughter cells. Failure to coordinate these events results in genetic damage. Here, we review recent evidence suggesting that proper abscission timing is coordinated by cytoskeletal rearrangements and recruitment of regulators of the Endosomal Sorting Complex Required for Transport (ESCRT) machinery such as CEP55 and MIT-domain-containing protein 1 (MITD1) to the abscission site. Additionally, we discuss the surveillance mechanism known as the Aurora B-mediated abscission checkpoint (NoCut), which prevents genetic damage by ensuring proper abscission delay when chromatin is trapped at the midbody.
Collapse
Affiliation(s)
- Monica Agromayor
- Department of Infectious Diseases, King's College London School of Medicine, London SE1 9RT, UK
| | | |
Collapse
|
39
|
Abstract
Ubiquitylation is a reversible post-translational modification that has emerged as a key regulator of most complex cellular processes. It may rival phosphorylation in scope and exceed it in complexity. The dynamic nature of ubiquitylation events is important for governing protein stability, maintaining ubiquitin homeostasis and controlling ubiquitin-dependent signalling pathways. The human genome encodes ~80 active deubiquitylating enzymes (DUBs, also referred to as deubiquitinases), which exhibit distinct specificity profiles towards the various ubiquitin chain topologies. As a result of their ability to reverse ubiquitylation, these enzymes control a broad range of key cellular processes. In this Commentary we discuss the cellular functions of DUBs, such as their role in governing membrane traffic and protein quality control. We highlight two key signalling pathways--the Wnt and transforming growth factor β (TGF-β) pathways, for which dynamic ubiquitylation has emerged as a key regulator. We also discuss the roles of DUBs in the nucleus, where they govern transcriptional activity and DNA repair pathways.
Collapse
Affiliation(s)
- Michael J Clague
- Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Crown Street, L69 3BX Liverpool, UK.
| | | | | |
Collapse
|
40
|
Chen CT, Ettinger AW, Huttner WB, Doxsey SJ. Resurrecting remnants: the lives of post-mitotic midbodies. Trends Cell Biol 2012; 23:118-28. [PMID: 23245592 DOI: 10.1016/j.tcb.2012.10.012] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 10/23/2012] [Accepted: 10/25/2012] [Indexed: 02/01/2023]
Abstract
Around a century ago, the midbody (MB) was described as a structural assembly within the intercellular bridge during cytokinesis that served to connect the two future daughter cells. The MB has become the focus of intense investigation through the identification of a growing number of diverse cellular and molecular pathways that localize to the MB and contribute to its cytokinetic functions, ranging from selective vesicle trafficking and regulated microtubule (MT), actin, and endosomal sorting complex required for transport (ESCRT) filament assembly and disassembly to post-translational modification, such as ubiquitination. More recent studies have revealed new and unexpected functions of MBs in post-mitotic cells. In this review, we provide a historical perspective, discuss exciting new roles for MBs beyond their cytokinetic function, and speculate on their potential contributions to pluripotency.
Collapse
Affiliation(s)
- Chun-Ting Chen
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | |
Collapse
|
41
|
Carrasquillo R, Tian D, Krishna S, Pollak MR, Greka A, Schlöndorff J. SNF8, a member of the ESCRT-II complex, interacts with TRPC6 and enhances its channel activity. BMC Cell Biol 2012; 13:33. [PMID: 23171048 PMCID: PMC3520717 DOI: 10.1186/1471-2121-13-33] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 10/23/2012] [Indexed: 11/10/2022] Open
Abstract
Background Transient receptor potential canonical (TRPC) channels are non-selective cation channels involved in receptor-mediated calcium signaling in diverse cells and tissues. The canonical transient receptor potential 6 (TRPC6) has been implicated in several pathological processes, including focal segmental glomerulosclerosis (FSGS), cardiac hypertrophy, and pulmonary hypertension. The two large cytoplasmic segments of the cation channel play a critical role in the proper regulation of channel activity, and are involved in several protein-protein interactions. Results Here we report that SNF8, a component of the endosomal sorting complex for transport-II (ESCRT-II) complex, interacts with TRPC6. The interaction was initially observed in a yeast two-hybrid screen using the amino-terminal cytoplasmic domain of TRPC6 as bait, and confirmed by co-immunoprecipitation from eukaryotic cell extracts. The amino-terminal 107 amino acids are necessary and sufficient for the interaction. Overexpression of SNF8 enhances both wild-type and gain-of-function mutant TRPC6-mediated whole-cell currents in HEK293T cells. Furthermore, activation of NFAT-mediated transcription by gain-of-function mutants is enhanced by overexpression of SNF8, and partially inhibited by RNAi mediated knockdown of SNF8. Although the ESCRT-II complex functions in the endocytosis and lysosomal degradation of transmembrane proteins, SNF8 overexpression does not alter the amount of TRPC6 present on the cell surface. Conclusion SNF8 is novel binding partner of TRPC6, binding to the amino-terminal cytoplasmic domain of the channel. Modulating SNF8 expression levels alters the TRPC6 channel current and can modulate activation of NFAT-mediated transcription downstream of gain-of-function mutant TRPC6. Taken together, these results identify SNF8 as a novel regulator of TRPC6.
Collapse
Affiliation(s)
- Robert Carrasquillo
- Division of Nephrology, Beth Israel Deaconess Medical Center, Research North 304B, 99 Brookline Ave, Boston, MA 02215, USA
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
As intracellular pathogens, enveloped viruses must usurp the host cell machinery for many stages of the viral life cycle in order to produce a new generation of infectious virions. In one of the less understood steps of viral assembly, viral components including the transmembrane glycoproteins, structural proteins and the viral genome must be targeted to the site of viral budding, where they assemble and are incorporated into a newly formed virion that gains a lipid envelope from a cellular membrane. Recent work has revealed that the cellular recycling endosome pathway, in particular Rab11, plays an important role in the assembly of negative-strand RNA viruses such as respiratory syncytial virus, influenza A virus, Andes virus and Sendai virus. The present mini-review discusses this emerging field and explores the potential roles of the Rab11 pathway in the trafficking, assembly and budding steps of these viruses.
Collapse
|
43
|
Effantin G, Dordor A, Sandrin V, Martinelli N, Sundquist WI, Schoehn G, Weissenhorn W. ESCRT-III CHMP2A and CHMP3 form variable helical polymers in vitro and act synergistically during HIV-1 budding. Cell Microbiol 2012; 15:213-26. [PMID: 23051622 DOI: 10.1111/cmi.12041] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 10/03/2012] [Accepted: 10/04/2012] [Indexed: 12/23/2022]
Abstract
The endosomal sorting complex required for transport-III (ESCRT-III) proteins are essential for budding of some enveloped viruses, for the formation of intraluminal vesicles at the endosome and for the abscission step of cytokinesis. ESCRT-III proteins form polymers that constrict membrane tubes, leading to fission. We have used electron cryomicroscopy to determine the molecular organization of pleiomorphic ESCRT-III CHMP2A-CHMP3 polymers. The three-dimensional reconstruction at 22 Å resolution reveals a helical organization of filaments of CHMP molecules organized in a head-to-tail fashion. Protease susceptibility experiments indicate that polymerization is achieved via conformational changes that increase the protomer stability. Combinatorial siRNA knockdown experiments indicate that CHMP3 contributes synergistically to HIV-1 budding, and the CHMP3 contribution is ~ 10-fold more pronounced in concert with CHMP2A than with CHMP2B. This is consistent with surface plasmon resonance affinity measurements that suggest sequential CHMP4B-CHMP3-CHMP2A recruitment while showing that both CHMP2A and CHMP2B interact with CHMP4B, in agreement with their redundant functions in HIV-1 budding. Our data thus indicate that the CHMP2A-CHMP3 polymer observed in vitro contributes to HIV-1 budding by assembling on CHMP4B polymers.
Collapse
Affiliation(s)
- Grégory Effantin
- Unit of Virus Host Cell Interactions, UMI 3265, Université Joseph Fourier-EMBL-CNRS, 6 rue Jules Horowitz, 38042 Grenoble Cedex 9, France
| | | | | | | | | | | | | |
Collapse
|
44
|
Borio CS, Bilen MF, Argüelles MH, Goñi SE, Iserte JA, Glikmann G, Lozano ME. Antigen vehiculization particles based on the Z protein of Junin virus. BMC Biotechnol 2012; 12:80. [PMID: 23121996 PMCID: PMC3534497 DOI: 10.1186/1472-6750-12-80] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 10/25/2012] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Arenavirus matrix protein Z plays an important role in virus budding and is able to generate enveloped virus-like-particles (VLPs) in absence of any other viral proteins. In these VLPs, Z protein is associated to the plasma membrane inner surface by its myristoyl residue. Budding induction and vesicle formation properties can be exploited to generate enveloped VLPs platform. These structures can be designed to carry specific antigen in the inner side or on the surface of VLPs.Vaccines based on VLPs are a highly effective type of subunit vaccines that mimic the overall structure of virus particles in absence of viral nucleic acid, being noninfectious.In this work we assayed the capacity of Junin Z protein to produce VLPs carrying the green fluorescent protein (eGFP), as a model antigen. RESULTS In this report the Junin Z protein ability to produce VLPs from 293T cells and its capacity to deliver a specific antigen (eGFP) fused to Z was evaluated. Confocal microscopy showed a particular membrane bending in cells expressing Z and a spot welded distribution in the cytoplasm. VLPs were detected by TEM (transmission electron microscopy) and were purified from cell supernatant. The proteinase protection assay demonstrated the VLPs integrity and the absence of degradation of the fused antigen, thus indicating its internal localization. Finally, immunization of mice with purified VLPs produced high titres of anti-eGFP antibodies compared to the controls. CONCLUSIONS It was proved that VLPs can be generated from cells transfected with a fusion Junin virus Z-eGFP protein in absence of any other viral protein, and the capacity of Z protein to support fusions at the C-terminal, without impairing its budding activity, allowing vehiculization of specific antigens into VLPs.
Collapse
Affiliation(s)
- Cristina S Borio
- LIGBCM-AVEZ, Department of Science and Technology, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina.
| | | | | | | | | | | | | |
Collapse
|
45
|
Frankenberg N, Lischka P, Pepperl-Klindworth S, Stamminger T, Plachter B. Nucleocytoplasmic shuttling and CRM1-dependent MHC class I peptide presentation of human cytomegalovirus pp65. Med Microbiol Immunol 2012; 201:567-79. [PMID: 22965172 DOI: 10.1007/s00430-012-0269-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 08/25/2012] [Indexed: 10/27/2022]
Abstract
The phosphoprotein 65 (pp65) of human cytomegalovirus is a prominent target of the antiviral CD8 T lymphocyte response. This study focused on investigating the properties of pp65 that render it a privileged antigen. It was found that pp65 was metabolically stable. The tegument protein was introduced into MHC class I presentation following its delivery via non-replicating dense bodies. No ubiquitination was found on particle-associated pp65. Proof was obtained that pp65 was a nucleocytoplasmic shuttle protein, using heterokaryon analyses. Based on this finding, inhibition experiments showed that presentation of particle-derived pp65 by HLA-A2 was sensitive to the impairment of the CRM1-mediated nuclear export pathway. The data support the idea that particle-derived pp65 can serve as a nuclear reservoir for proteasomal processing and MHC class I presentation, following its CRM1-dependent nuclear export. The presentation of pp65-derived peptides was also impaired by CRM1-inhibition following de novo synthesis of the tegument protein. However, pp65 protein levels were also reduced when blocking CRM1-mediated export after transient expression. This indicated that pp65 expression rather than direct interference with its own nuclear export was responsible for its reduced presentation in this case. The functionality of CRM1-mediated nuclear export is thus important for the presentation of pp65-derived peptides in the context of MHC class I on organ cells, both after exogenous uptake and after de novo synthesis of the tegument protein, but different mechanisms may account for either case.
Collapse
Affiliation(s)
- Nadine Frankenberg
- Institute for Virology, University Medical Center Mainz, Obere Zahlbacher Str. 67, 55101 Mainz, Germany
| | | | | | | | | |
Collapse
|
46
|
In vitro reconstitution of the ordered assembly of the endosomal sorting complex required for transport at membrane-bound HIV-1 Gag clusters. Proc Natl Acad Sci U S A 2012; 109:16928-33. [PMID: 23027949 DOI: 10.1073/pnas.1211759109] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Most membrane-enveloped viruses depend on host proteins of the endosomal sorting complex required for transport (ESCRT) machinery for their release. HIV-1 is the prototypic ESCRT-dependent virus. The direct interactions between HIV-1 and the early ESCRT factors TSG101 and ALIX have been mapped in detail. However, the full pathway of ESCRT recruitment to HIV-1 budding sites, which culminates with the assembly of the late-acting CHMP4, CHMP3, CHMP2, and CHMP1 subunits, is less completely understood. Here, we report the biochemical reconstitution of ESCRT recruitment to viral assembly sites, using purified proteins and giant unilamellar vesicles. The myristylated full-length Gag protein of HIV-1 was purified to monodispersity. Myr-Gag forms clusters on giant unilamellar vesicle membranes containing the plasma membrane lipid PI(4,5)P(2). These Gag clusters package a fluorescent oligonucleotide, and recruit early ESCRT complexes ESCRT-I or ALIX with the appropriate dependence on the Gag PTAP and LYP(X)(n)L motifs. ALIX directly recruits the key ESCRT-III subunit CHMP4. ESCRT-I can only recruit CHMP4 when ESCRT-II and CHMP6 are present as intermediary factors. Downstream of CHMP4, CHMP3 and CHMP2 assemble synergistically, with the presence of both subunits required for efficient recruitment. The very late-acting factor CHMP1 is not recruited unless the pathway is completed through CHMP3 and CHMP2. These findings define the minimal sets of components needed to complete ESCRT assembly at HIV-1 budding sites, and provide a starting point for in vitro structural and biophysical dissection of the system.
Collapse
|
47
|
Troost T, Jaeckel S, Ohlenhard N, Klein T. The tumour suppressor Lethal (2) giant discs is required for the function of the ESCRT-III component Shrub/CHMP4. J Cell Sci 2012; 125:763-76. [PMID: 22389409 DOI: 10.1242/jcs.097261] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Recent work indicates that defects in late phases of the endosomal pathway caused by loss of function of the tumour suppressor gene lethal (2) giant discs (lgd) or the function of the ESCRT complexes I-III result in the ligand-independent activation of the Notch pathway in all imaginal disc cells in Drosophila melanogaster. lgd encodes a member of an uncharacterised protein family, whose members contain one C2 domain and four repeats of the DM14 domain. The function of the DM14 domain is unknown. We here report a detailed structure-function analysis of Lgd protein, which reveals that the DM14 domains are essential for the function of Lgd and act in a redundant manner. Moreover, our analysis indicates that the DM14 domain provides the specific function, whereas the C2 domain is required for the subcellular location of Lgd. We found that Lgd interacts directly with the ESCRT-III subunit Shrub through the DM14 domains. The interaction is required for the function of Shrub, indicating that Lgd contributes to the function of the ESCRT-III complex. Furthermore, our genetic studies indicate that the activation of Notch in ESCRT and lgd mutant cells occurs in a different manner and that the activity of Shrub and other ESCRT components are required for the activation of Notch in lgd mutant cells.
Collapse
Affiliation(s)
- Tobias Troost
- Institut für Genetik, Heinrich-Heine-Universität Düsseldorf, Universitätsstrasse1, 40225, Düsseldorf, Germany
| | | | | | | |
Collapse
|
48
|
Boura E, Ivanov V, Carlson LA, Mizuuchi K, Hurley JH. Endosomal sorting complex required for transport (ESCRT) complexes induce phase-separated microdomains in supported lipid bilayers. J Biol Chem 2012; 287:28144-51. [PMID: 22718754 DOI: 10.1074/jbc.m112.378646] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The endosomal sorting complex required for transport (ESCRT) system traffics ubiquitinated cargo to lysosomes via an unusual membrane budding reaction that is directed away from the cytosol. Here, we show that human ESCRT-II self-assembles into clusters of 10-100 molecules on supported lipid bilayers. The ESCRT-II clusters are functional in that they bind to ubiquitin and the ESCRT-III subunit VPS20 at nanomolar concentrations on membranes with the same stoichiometries observed in solution and in crystals. The clusters only form when cholesterol is included in the lipid mixture at >10 mol %. The clusters induce the formation of ordered membrane domains that exclude the dye 1,1'-dioctadecyl-3,3,3',3'-tetramethylindodicarbo-cyanine perchlorate. These results show that ESCRT complexes are capable of inducing lateral lipid phase separation under conditions where the lipids themselves do not spontaneously phase-separate. This property could facilitate ESCRT-mediated membrane budding.
Collapse
Affiliation(s)
- Evzen Boura
- Laboratory of Molecular Biology, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
49
|
Elia N, Fabrikant G, Kozlov MM, Lippincott-Schwartz J. Computational model of cytokinetic abscission driven by ESCRT-III polymerization and remodeling. Biophys J 2012; 102:2309-20. [PMID: 22677384 DOI: 10.1016/j.bpj.2012.04.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Revised: 04/04/2012] [Accepted: 04/05/2012] [Indexed: 11/27/2022] Open
Abstract
The endosomal sorting complex required for transport (ESCRT)-III complex, capable of polymerization and remodeling, participates in abscission of the intercellular membrane bridge connecting two daughter cells at the end of cytokinesis. Here, we integrate quantitative imaging of ESCRT-III during cytokinetic abscission with biophysical properties of ESCRT-III complexes to formulate and test a computational model for ESCRT-mediated cytokinetic abscission. We propose that cytokinetic abscission is driven by an ESCRT-III fission complex, which arises from ESCRT-III polymerization at the edge of the cytokinetic midbody structure, located at the center of the intercellular bridge. Formation of the fission complex is completed by remodeling and breakage of the ESCRT-III polymer assisted by VPS4. Subsequent spontaneous constriction of the fission complex generates bending deformation of the intercellular bridge membrane. The related membrane elastic force propels the fission complex along the intercellular bridge away from the midbody until it reaches an equilibrium position, determining the scission site. Membrane attachment to the dome-like end-cap of the fission complex drives membrane fission, completing the abscission process. We substantiate the model by theoretical analysis of the membrane elastic energy and by experimental verification of the major model assumptions.
Collapse
Affiliation(s)
- Natalie Elia
- Cell Biology and Metabolism Program, Eunice Kennedy Shriver National Institute of Child Health and Development, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
50
|
Martinelli N, Hartlieb B, Usami Y, Sabin C, Dordor A, Miguet N, Avilov SV, Ribeiro EA, Göttlinger H, Weissenhorn W. CC2D1A is a regulator of ESCRT-III CHMP4B. J Mol Biol 2012; 419:75-88. [PMID: 22406677 DOI: 10.1016/j.jmb.2012.02.044] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 02/29/2012] [Accepted: 02/29/2012] [Indexed: 12/01/2022]
Abstract
Endosomal sorting complexes required for transport (ESCRTs) regulate diverse processes ranging from receptor sorting at endosomes to distinct steps in cell division and budding of some enveloped viruses. Common to all processes is the membrane recruitment of ESCRT-III that leads to membrane fission. Here, we show that CC2D1A is a novel regulator of ESCRT-III CHMP4B function. We demonstrate that CHMP4B interacts directly with CC2D1A and CC2D1B with nanomolar affinity by forming a 1:1 complex. Deletion mapping revealed a minimal CC2D1A-CHMP4B binding construct, which includes a short linear sequence within the third DM14 domain of CC2D1A. The CC2D1A binding site on CHMP4B was mapped to the N-terminal helical hairpin. Based on a crystal structure of the CHMP4B helical hairpin, two surface patches were identified that interfere with CC2D1A interaction as determined by surface plasmon resonance. Introducing these mutations into a C-terminal truncation of CHMP4B that exerts a potent dominant negative effect on human immunodeficiency virus type 1 budding revealed that one of the mutants lost this effect completely. This suggests that the identified CC2D1A binding surface might be required for CHMP4B polymerization, which is consistent with the finding that CC2D1A binding to CHMP4B prevents CHMP4B polymerization in vitro. Thus, CC2D1A might act as a negative regulator of CHMP4B function.
Collapse
Affiliation(s)
- Nicolas Martinelli
- Unit of Virus Host Cell Interactions UMI 3265, Université Joseph Fourier-EMBL-CNRS, 6 rue Jules Horowitz, 38042 Grenoble Cedex 9, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|