1
|
Cao YY, Wu LL, Li XN, Yuan YL, Zhao WW, Qi JX, Zhao XY, Ward N, Wang J. Molecular Mechanisms of AMPA Receptor Trafficking in the Nervous System. Int J Mol Sci 2023; 25:111. [PMID: 38203282 PMCID: PMC10779435 DOI: 10.3390/ijms25010111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Synaptic plasticity enhances or reduces connections between neurons, affecting learning and memory. Postsynaptic AMPARs mediate greater than 90% of the rapid excitatory synaptic transmission in glutamatergic neurons. The number and subunit composition of AMPARs are fundamental to synaptic plasticity and the formation of entire neural networks. Accordingly, the insertion and functionalization of AMPARs at the postsynaptic membrane have become a core issue related to neural circuit formation and information processing in the central nervous system. In this review, we summarize current knowledge regarding the related mechanisms of AMPAR expression and trafficking. The proteins related to AMPAR trafficking are discussed in detail, including vesicle-related proteins, cytoskeletal proteins, synaptic proteins, and protein kinases. Furthermore, significant emphasis was placed on the pivotal role of the actin cytoskeleton, which spans throughout the entire transport process in AMPAR transport, indicating that the actin cytoskeleton may serve as a fundamental basis for AMPAR trafficking. Additionally, we summarize the proteases involved in AMPAR post-translational modifications. Moreover, we provide an overview of AMPAR transport and localization to the postsynaptic membrane. Understanding the assembly, trafficking, and dynamic synaptic expression mechanisms of AMPAR may provide valuable insights into the cognitive decline associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi-Yang Cao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Ling-Ling Wu
- School of Medicine, Shanghai University, Shanghai 200444, China;
| | - Xiao-Nan Li
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Yu-Lian Yuan
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Wan-Wei Zhao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Jing-Xuan Qi
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Xu-Yu Zhao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Natalie Ward
- Medical Laboratory, Exceptional Community Hospital, 19060 N John Wayne Pkwy, Maricopa, AZ 85139, USA;
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| |
Collapse
|
2
|
Yu Y, Zhang Z, Yu Y. Timing of Phagosome Maturation Depends on Their Transport Switching from Actin to Microtubule Tracks. J Phys Chem B 2023; 127:9312-9322. [PMID: 37871280 PMCID: PMC10759163 DOI: 10.1021/acs.jpcb.3c05647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Phagosomes, specialized membrane compartments responsible for digesting internalized pathogens, undergo sequential dynamic and biochemical changes as they mature from nascent phagosomes to degradative phagolysosomes. Maturation of phagosomes depends on their transport along actin filaments and microtubules. However, the specific quantitative relationship between the biochemical transformation and transport dynamics remains poorly characterized. The autonomous nature of phagosomes, moving and maturing at different rates, makes understanding this relationship challenging. Addressing this challenge, in this study we engineered particle sensors to image and quantify single phagosomes' maturation. We found that as phagosomes move from the actin cortex to microtubule tracks, the timing of their actin-to-microtubule transition governs the duration of the early phagosome stage before acquiring degradative capacities. Prolonged entrapment of phagosomes in the actin cortex extends the early phagosome stage by delaying the dissociation of early endosome markers and phagosome acidification. Conversely, a shortened transition from actin- to microtubule-based movements causes the opposite effect on phagosome maturation. These results suggest that the actin- and microtubule-based transport of phagosomes functions like a "clock" to coordinate the timing of biochemical events during phagosome maturation, which is crucial for effective pathogen degradation.
Collapse
Affiliation(s)
- Yanqi Yu
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405-7102, United States
| | - Zihan Zhang
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405-7102, United States
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405-7102, United States
| |
Collapse
|
3
|
Mechanical instability generated by Myosin 19 contributes to mitochondria cristae architecture and OXPHOS. Nat Commun 2022; 13:2673. [PMID: 35562374 PMCID: PMC9106661 DOI: 10.1038/s41467-022-30431-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/29/2022] [Indexed: 01/02/2023] Open
Abstract
The folded mitochondria inner membrane-cristae is the structural foundation for oxidative phosphorylation (OXPHOS) and energy production. By mechanically simulating mitochondria morphogenesis, we speculate that efficient sculpting of the cristae is organelle non-autonomous. It has long been inferred that folding requires buckling in living systems. However, the tethering force for cristae formation and regulation has not been identified. Combining electron tomography, proteomics strategies, super resolution live cell imaging and mathematical modeling, we reveal that the mitochondria localized actin motor-myosin 19 (Myo19) is critical for maintaining cristae structure, by associating with the SAM-MICOS super complex. We discover that depletion of Myo19 or disruption of its motor activity leads to altered mitochondria membrane potential and decreased OXPHOS. We propose that Myo19 may act as a mechanical tether for effective ridging of the mitochondria cristae, thus sustaining the energy homeostasis essential for various cellular functions. The structure of the mitochondrial inner membrane, or cristae, is important for functional oxidative phosphorylation and energy production. Here, the authors show that loss of myosin 19 impairs cristae structure as well as energy production, connecting motor activity to membrane potential.
Collapse
|
4
|
Wang S, Liu H, Roberts JB, Wiley AP, Marayati BF, Adams KL, Luessen DJ, Eldeeb K, Sun H, Zhang K, Chen R. Prolonged ethanol exposure modulates constitutive internalization and recycling of 5-HT1A receptors. J Neurochem 2022; 160:469-481. [PMID: 34928513 PMCID: PMC8828711 DOI: 10.1111/jnc.15564] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 11/24/2021] [Accepted: 12/13/2021] [Indexed: 02/03/2023]
Abstract
Alcohol exposure alters the signaling of the serotoninergic system, which is involved in alcohol consumption, reward, and dependence. In particular, dysregulation of serotonin receptor type 1A (5-HT1AR) is associated with alcohol intake and withdrawal-induced anxiety-like behavior in rodents. However, how ethanol regulates 5-HT1AR activity and cell surface availability remains elusive. Using neuroblastoma 2a cells stably expressing human 5-HT1ARs tagged with hemagglutinin at the N-terminus, we found that prolonged ethanol exposure (18 h) reduced the basal surface levels of 5-HT1ARs in a concentration-dependent manner. This reduction is attributed to both enhanced receptor internalization and attenuated receptor recycling. Moreover, constitutive 5-HT1AR internalization in ethanol naïve cells was blocked by concanavalin A (ConA) but not nystatin, suggesting clathrin-dependent 5-HT1AR internalization. In contrast, constitutive 5-HT1AR internalization in ethanol-treated cells was blocked by nystatin but not by ConA, indicating that constitutive 5-HT1AR internalization switched from a clathrin- to a caveolin-dependent pathway. Dynasore, an inhibitor of dynamin, blocked 5-HT1AR internalization in both vehicle- and ethanol-treated cells. Furthermore, ethanol exposure enhanced the activity of dynamin I via dephosphorylation and reduced myosin Va levels, which may contribute to increased internalization and reduced recycling of 5-HT1ARs, respectively. Our findings suggest that prolonged ethanol exposure not only alters the endocytic trafficking of 5-HT1ARs but also the mechanism by which constitutive 5-HT1AR internalization occurs.
Collapse
Affiliation(s)
- Shiyu Wang
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
| | - Haoran Liu
- Department of Biology, Wake Forest University, Winston Salem, NC 27106
| | - Jonté B. Roberts
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
| | - Aidan P. Wiley
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
| | | | - Kristen L. Adams
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
| | - Deborah J. Luessen
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
| | - Khalil Eldeeb
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
- Campbell University School of Osteopathic Medicine, Lillington, NC 27546
| | - Haiguo Sun
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
| | - Ke Zhang
- Department of Biology, Wake Forest University, Winston Salem, NC 27106
- Center for Molecular Signaling, Wake Forest University, Winston Salem, NC 27106
| | - Rong Chen
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
- Center for Molecular Signaling, Wake Forest University, Winston Salem, NC 27106
| |
Collapse
|
5
|
Chakrabarti R, Lee M, Higgs HN. Multiple roles for actin in secretory and endocytic pathways. Curr Biol 2021; 31:R603-R618. [PMID: 34033793 DOI: 10.1016/j.cub.2021.03.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Actin filaments play multiple roles in the secretory pathway and in endosome dynamics in mammals, including maintenance of Golgi structure, release of membrane cargo from the trans-Golgi network (TGN), endocytosis, and endosomal sorting dynamics. In addition, TGN carrier transport and endocytosis both occur by multiple mechanisms in mammals. Actin likely plays a role in at least four mammalian endocytic pathways, five pathways for membrane release from the TGN, and three processes involving endosomes. Also, the mammalian Golgi structure is highly dynamic, and actin is likely important for these dynamics. One challenge for many of these processes is the need to deal with other membrane-associated structures, such as the cortical actin network at the plasma membrane or the matrix that surrounds the Golgi. Arp2/3 complex is a major actin assembly factor in most of the processes mentioned, but roles for formins and tandem WH2-motif-containing assembly factors are being elucidated and are anticipated to grow with further study. The specific role for actin has not been defined for most of these processes, but is likely to involve the generation of force for membrane dynamics, either by actin polymerization itself or by myosin motor activity. Defining these processes mechanistically is necessary for understanding membrane dynamics in general, as well as pathways that utilize these processes, such as autophagy.
Collapse
Affiliation(s)
- Rajarshi Chakrabarti
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Miriam Lee
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Henry N Higgs
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA.
| |
Collapse
|
6
|
Taylor RW, Holler C, Mahmoodabadi RG, Küppers M, Dastjerdi HM, Zaburdaev V, Schambony A, Sandoghdar V. High-Precision Protein-Tracking With Interferometric Scattering Microscopy. Front Cell Dev Biol 2020; 8:590158. [PMID: 33224953 PMCID: PMC7669747 DOI: 10.3389/fcell.2020.590158] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/05/2020] [Indexed: 01/01/2023] Open
Abstract
The mobility of proteins and lipids within the cell, sculpted oftentimes by the organization of the membrane, reveals a great wealth of information on the function and interaction of these molecules as well as the membrane itself. Single particle tracking has proven to be a vital tool to study the mobility of individual molecules and unravel details of their behavior. Interferometric scattering (iSCAT) microscopy is an emerging technique well-suited for visualizing the diffusion of gold nanoparticle-labeled membrane proteins to a spatial and temporal resolution beyond the means of traditional fluorescent labels. We discuss the applicability of interferometric single particle tracking (iSPT) microscopy to investigate the minutia in the motion of a protein through measurements visualizing the mobility of the epidermal growth factor receptor in various biological scenarios on the live cell.
Collapse
Affiliation(s)
- Richard W Taylor
- Max Planck Institute for the Science of Light, Erlangen, Germany.,Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Cornelia Holler
- Max Planck Institute for the Science of Light, Erlangen, Germany.,Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Reza Gholami Mahmoodabadi
- Max Planck Institute for the Science of Light, Erlangen, Germany.,Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Michelle Küppers
- Max Planck Institute for the Science of Light, Erlangen, Germany.,Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany.,Department of Physics, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Houman Mirzaalian Dastjerdi
- Max Planck Institute for the Science of Light, Erlangen, Germany.,Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany.,Department of Computer Science, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Vasily Zaburdaev
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany.,Department of Biology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Alexandra Schambony
- Max Planck Institute for the Science of Light, Erlangen, Germany.,Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany.,Department of Biology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Vahid Sandoghdar
- Max Planck Institute for the Science of Light, Erlangen, Germany.,Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany.,Department of Physics, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
7
|
Unconventional Myosins: How Regulation Meets Function. Int J Mol Sci 2019; 21:ijms21010067. [PMID: 31861842 PMCID: PMC6981383 DOI: 10.3390/ijms21010067] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 01/24/2023] Open
Abstract
Unconventional myosins are multi-potent molecular motors that are assigned important roles in fundamental cellular processes. Depending on their mechano-enzymatic properties and structural features, myosins fulfil their roles by acting as cargo transporters along the actin cytoskeleton, molecular anchors or tension sensors. In order to perform such a wide range of roles and modes of action, myosins need to be under tight regulation in time and space. This is achieved at multiple levels through diverse regulatory mechanisms: the alternative splicing of various isoforms, the interaction with their binding partners, their phosphorylation, their applied load and the composition of their local environment, such as ions and lipids. This review summarizes our current knowledge of how unconventional myosins are regulated, how these regulatory mechanisms can adapt to the specific features of a myosin and how they can converge with each other in order to ensure the required tight control of their function.
Collapse
|
8
|
Jaykumar AB, Caceres PS, King-Medina KN, Liao TD, Datta I, Maskey D, Naggert JK, Mendez M, Beierwaltes WH, Ortiz PA. Role of Alström syndrome 1 in the regulation of blood pressure and renal function. JCI Insight 2018; 3:95076. [PMID: 30385718 DOI: 10.1172/jci.insight.95076] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/26/2018] [Indexed: 01/22/2023] Open
Abstract
Elevated blood pressure (BP) and renal dysfunction are complex traits representing major global health problems. Single nucleotide polymorphisms identified by genome-wide association studies have identified the Alström syndrome 1 (ALMS1) gene locus to render susceptibility for renal dysfunction, hypertension, and chronic kidney disease (CKD). Mutations in the ALMS1 gene in humans causes Alström syndrome, characterized by progressive metabolic alterations including hypertension and CKD. Despite compelling genetic evidence, the underlying biological mechanism by which mutations in the ALMS1 gene lead to the above-mentioned pathophysiology is not understood. We modeled this effect in a KO rat model and showed that ALMS1 genetic deletion leads to hypertension. We demonstrate that the link between ALMS1 and hypertension involves the activation of the renal Na+/K+/2Cl- cotransporter NKCC2, mediated by regulation of its endocytosis. Our findings establish a link between the genetic susceptibility to hypertension, CKD, and the expression of ALMS1 through its role in a salt-reabsorbing tubular segment of the kidney. These data point to ALMS1 as a potentially novel gene involved in BP and renal function regulation.
Collapse
Affiliation(s)
- Ankita Bachhawat Jaykumar
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Paulo S Caceres
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Keyona N King-Medina
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Tang-Dong Liao
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA
| | - Indrani Datta
- Department of Public Health Sciences and.,Center for Bioinformatics, Henry Ford Health System, Detroit, Michigan, USA
| | - Dipak Maskey
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA
| | | | - Mariela Mendez
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA
| | - William H Beierwaltes
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Pablo A Ortiz
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
9
|
Simonetti B, Cullen PJ. Actin-dependent endosomal receptor recycling. Curr Opin Cell Biol 2018; 56:22-33. [PMID: 30227382 DOI: 10.1016/j.ceb.2018.08.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/20/2018] [Accepted: 08/27/2018] [Indexed: 12/18/2022]
Abstract
Endosomes constitute major sorting compartments within the cell. There, a myriad of transmembrane proteins (cargoes) are delivered to the lysosome for degradation or retrieved from this fate and recycled through tubulo-vesicular transport carriers to different cellular destinations. Retrieval and recycling are orchestrated by multi-protein assemblies that include retromer and retriever, sorting nexins, and the Arp2/3 activating WASH complex. Fine-tuned control of actin polymerization on endosomes is fundamental for the retrieval and recycling of cargoes. Recent advances in the field have highlighted several roles that actin plays in this process including the binding to cargoes, stabilization of endosomal subdomains, generation of the remodeling forces required for the biogenesis of cargo-enriched transport carriers and short-range motility of the transport carriers.
Collapse
Affiliation(s)
- Boris Simonetti
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
| | - Peter J Cullen
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol BS8 1TD, UK.
| |
Collapse
|
10
|
Real-Hohn A, Provance DW, Gonçalves RB, Denani CB, de Oliveira AC, Salerno VP, Oliveira Gomes AM. Impairing the function of MLCK, myosin Va or myosin Vb disrupts Rhinovirus B14 replication. Sci Rep 2017; 7:17153. [PMID: 29215055 PMCID: PMC5719429 DOI: 10.1038/s41598-017-17501-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/27/2017] [Indexed: 12/19/2022] Open
Abstract
Together, the three human rhinovirus (RV) species are the most frequent cause of the common cold. Because of their high similarity with other viral species of the genus Enterovirus, within the large family Picornaviridae, studies on RV infectious activities often offer a less pathogenic model for more aggressive enteroviruses, e.g. poliovirus or EV71. Picornaviruses enter via receptor mediated endocytosis and replicate in the cytosol. Most of them depend on functional F-actin, Rab proteins, and probably motor proteins. To assess the latter, we evaluated the role of myosin light chain kinase (MLCK) and two myosin V isoforms (Va and Vb) in RV-B14 infection. We report that ML-9, a very specific MLCK inhibitor, dramatically reduced RV-B14 entry. We also demonstrate that RV-B14 infection in cells expressing dominant-negative forms of myosin Va and Vb was impaired after virus entry. Using immunofluorescent localization and immunoprecipitation, we show that myosin Va co-localized with RV-B14 exclusively after viral entry (15 min post infection) and that myosin Vb was present in the clusters of newly synthesized RNA in infected cells. These clusters, observed at 180 min post infection, are reminiscent of replication sites. Taken together, these results identify myosin light chain kinase, myosin Va and myosin Vb as new players in RV-B14 infection that participate directly or indirectly in different stages of the viral cycle.
Collapse
Affiliation(s)
- Antonio Real-Hohn
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Biociências da Atividade Física, Escola de Educação Física e Desportos, Universidade Federal Rio do Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, RJ, Brazil
| | - D William Provance
- Center for Technological Development in Health, National Institute of Science and Technology for Innovation in Diseases of Neglected Populations, Oswaldo Cruz Foundation/Fiocruz, Rio de Janeiro, Brazil
| | - Rafael Braga Gonçalves
- Departamento de Bioquímica, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, RJ, Brazil
| | - Caio Bidueira Denani
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, RJ, Brazil
| | - Andréa Cheble de Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, RJ, Brazil
| | - Verônica P Salerno
- Departamento de Biociências da Atividade Física, Escola de Educação Física e Desportos, Universidade Federal Rio do Janeiro, Rio de Janeiro, Brazil
| | - Andre Marco Oliveira Gomes
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil. .,Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
11
|
Ramos-Nascimento A, Kellen B, Ferreira F, Alenquer M, Vale-Costa S, Raposo G, Delevoye C, Amorim MJ. KIF13A mediates trafficking of influenza A virus ribonucleoproteins. J Cell Sci 2017; 130:4038-4050. [PMID: 29061883 DOI: 10.1242/jcs.210807] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 10/18/2017] [Indexed: 01/18/2023] Open
Abstract
Influenza A is a rapidly evolving virus that is successful in provoking periodic epidemics and occasional pandemics in humans. Viral assembly is complex as the virus incorporates an eight-partite genome of RNA (in the form of viral ribonucleoproteins, vRNPs), and viral genome assembly - with its implications to public health - is not completely understood. It has previously been reported that vRNPs are transported to the cell surface on Rab11-containing vesicles by using microtubules but, so far, no molecular motor has been assigned to the process. Here, we have identified KIF13A, a member of the kinesin-3 family, as the first molecular motor to efficiently transport vRNP-Rab11 vesicles during infection with influenza A. Depletion of KIF13A resulted in reduced viral titers and less accumulation of vRNPs at the cell surface, without interfering with the levels of other viral proteins at sites of viral assembly. In addition, when overexpressed and following two separate approaches to displace vRNP-Rab11 vesicles, KIF13A increased levels of vRNP at the plasma membrane. Together, our results show that KIF13A plays an important role in the transport of influenza A vRNPs, a crucial step for viral assembly.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Ana Ramos-Nascimento
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência (IGC), 2780-156 Oeiras, Portugal.,Institute of Virology, Hannover Medical School, 30625 Hannover, Germany
| | - Bárbara Kellen
- Science4U, MARL Espaços CC02 e CC03, São Julião do Tojal, 2660-421, Loures, Portugal
| | - Filipe Ferreira
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência (IGC), 2780-156 Oeiras, Portugal
| | - Marta Alenquer
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência (IGC), 2780-156 Oeiras, Portugal
| | - Sílvia Vale-Costa
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência (IGC), 2780-156 Oeiras, Portugal
| | - Graça Raposo
- Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane Compartments, 75005 Paris, France
| | - Cédric Delevoye
- Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane Compartments, 75005 Paris, France
| | - Maria João Amorim
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência (IGC), 2780-156 Oeiras, Portugal
| |
Collapse
|
12
|
Acquisition of Rab11 and Rab11-Fip2-A novel strategy for Chlamydia pneumoniae early survival. PLoS Pathog 2017; 13:e1006556. [PMID: 28787457 PMCID: PMC5560749 DOI: 10.1371/journal.ppat.1006556] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 08/17/2017] [Accepted: 07/27/2017] [Indexed: 11/30/2022] Open
Abstract
The initial steps in chlamydial infection involve adhesion and internalization into host cells and, most importantly, modification of the nascent inclusion to establish the intracellular niche. Here, we show that Chlamydia pneumoniae enters host cells via EGFR-dependent endocytosis into an early endosome with a phosphatidylinositol 3-phosphate (PI3P) membrane identity. Immediately after entry, the early chlamydial inclusion acquires early endosomal Rab GTPases including Rab4, Rab5, Rab7, as well as the two recycling-specific Rabs Rab11 and Rab14. While Rab5, Rab11 and Rab14 are retained in the vesicular membrane, Rab4 and Rab7 soon disappear. Loss of Rab7 enables the C. pneumoniae inclusion to escape delivery to, and degradation in lysosomes. Loss of Rab4 and retention of Rab11/ Rab14 designates the inclusion as a slowly recycling endosome—that is protected from degradation. Furthermore, we show that the Rab11/ Rab14 adaptor protein Rab11-Fip2 (Fip2) is recruited to the nascent inclusion upon internalization and retained in the membrane throughout infection. siRNA knockdown of Fip2 demonstrated that the protein is essential for internalization and infection, and expression of various deletion variants revealed that Fip2 regulates the intracellular positioning of the inclusion. Additionally, we show that binding to Rab11 and Fip2 recruits the unconventional actin motor protein myosin Vb to the early inclusion and that together they regulate the relocation of the nascent inclusion from the cell periphery to the perinuclear region, its final destination. Here, we characterize for the first time inclusion identity and inclusion-associated proteins to delineate how C. pneumoniae establishes the intracellular niche essential for its survival. Here, we show for the first time how Chlamydia pneumoniae an obligate intracellular pathogen establishes its intracellular niche. After EGFR-dependent endocytosis into host cells, the nascent chlamydial inclusion acquires early endosomal membrane identity and the Rab GTPases Rab4, Rab5 and Rab7, as well as the recycling-specific Rab11 and Rab14. We show that Rab5, Rab11 and Rab14 are retained in the vesicular membrane, while Rab4 and Rab7 subsequently disappear. Thus, C. pneumoniae escapes lysosomal degradation by hiding in a recycling endosome vesicle. Furthermore, we show that the Rab11/Rab14 adaptor protein Rab11-Fip2 (Fip2), together with the unconventional actin motor protein myosin Vb, is recruited to the nascent inclusion. Both are essential for internalization and infection, as they regulate the intracellular positioning of the inclusion, which is essential for intracellular transport from the cell periphery to the perinuclear region. Here, we characterize for the first time inclusion identity and inclusion-associated proteins to understand how C. pneumoniae establishes the intracellular niche, which is essential for its survival.
Collapse
|
13
|
Vale-Costa S, Amorim MJ. Clustering of Rab11 vesicles in influenza A virus infected cells creates hotspots containing the 8 viral ribonucleoproteins. Small GTPases 2016; 8:71-77. [PMID: 27337591 PMCID: PMC5464114 DOI: 10.1080/21541248.2016.1199190] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Influenza A virus is an important human pathogen causative of yearly epidemics and occasional pandemics. The ability to replicate within the host cell is a determinant of virulence, amplifying viral numbers for host-to-host transmission. This process requires multiple rounds of entering permissive cells, replication, and virion assembly at the plasma membrane, the site of viral budding and release. The assembly of influenza A virus involves packaging of several viral (and host) proteins and of a segmented genome, composed of 8 distinct RNAs in the form of viral ribonucleoproteins (vRNPs). The selective assembly of the 8-segment core remains one of the most interesting unresolved problems in virology. The recycling endosome regulatory GTPase Rab11 was shown to contribute to the process, by transporting vRNPs to the periphery, giving rise to enlarged cytosolic puncta rich in Rab11 and the 8 vRNPs. We recently reported that vRNP hotspots were formed of clustered vesicles harbouring protruding electron-dense structures that resembled vRNPs. Mechanistically, vRNP hotspots were formed as vRNPs outcompeted the cognate effectors of Rab11, the Rab11-Family-Interacting-Proteins (FIPs) for binding, and as a consequence impair recycling sorting at an unknown step. Here, we speculate on the impact that such impairment might have in host immunity, membrane architecture and viral assembly.
Collapse
Affiliation(s)
- Sílvia Vale-Costa
- a Cell Biology of Viral Infection Lab , Instituto Gulbenkian de Ciência , Oeiras , Portugal
| | - Maria João Amorim
- a Cell Biology of Viral Infection Lab , Instituto Gulbenkian de Ciência , Oeiras , Portugal
| |
Collapse
|
14
|
Abstract
Many viruses exploit specific arms of the endomembrane system. The unique composition of each arm prompts the development of remarkably specific interactions between viruses and sub-organelles. This review focuses on the viral–host interactions occurring on the endocytic recycling compartment (ERC), and mediated by its regulatory Ras-related in brain (Rab) GTPase Rab11. This protein regulates trafficking from the ERC and the trans-Golgi network to the plasma membrane. Such transport comprises intricate networks of proteins/lipids operating sequentially from the membrane of origin up to the cell surface. Rab11 is also emerging as a critical factor in an increasing number of infections by major animal viruses, including pathogens that provoke human disease. Understanding the interplay between the ERC and viruses is a milestone in human health. Rab11 has been associated with several steps of the viral lifecycles by unclear processes that use sophisticated diversified host machinery. For this reason, we first explore the state-of-the-art on processes regulating membrane composition and trafficking. Subsequently, this review outlines viral interactions with the ERC, highlighting current knowledge on viral-host binding partners. Finally, using examples from the few mechanistic studies available we emphasize how ERC functions are adjusted during infection to remodel cytoskeleton dynamics, innate immunity and membrane composition.
Collapse
Affiliation(s)
- Sílvia Vale-Costa
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, 2780-156 Oeiras, Portugal.
| | - Maria João Amorim
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, 2780-156 Oeiras, Portugal.
| |
Collapse
|
15
|
Vogel GF, Klee KMC, Janecke AR, Müller T, Hess MW, Huber LA. Cargo-selective apical exocytosis in epithelial cells is conducted by Myo5B, Slp4a, Vamp7, and Syntaxin 3. J Cell Biol 2016; 211:587-604. [PMID: 26553929 PMCID: PMC4639860 DOI: 10.1083/jcb.201506112] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The motor protein Myo5B and t-SNARE Stx3 drive cargo-selective apical exocytosis in polarized epithelial cells in a pathway dependent on v-SNARE–like Slp4a, v-SNARE Vamp7, Sec1/Munc18-like protein Munc18-2, and the Rab11/8 cascade. Mutations in the motor protein Myosin Vb (Myo5B) or the soluble NSF attachment protein receptor Syntaxin 3 (Stx3) disturb epithelial polarity and cause microvillus inclusion disease (MVID), a lethal hereditary enteropathy affecting neonates. To understand the molecular mechanism of Myo5B and Stx3 interplay, we used genome editing to introduce a defined Myo5B patient mutation in a human epithelial cell line. Our results demonstrate a selective role of Myo5B and Stx3 for apical cargo exocytosis in polarized epithelial cells. Apical exocytosis of NHE3, CFTR (cystic fibrosis transmembrane conductance regulator), and GLUT5 required an interaction cascade of Rab11, Myo5B, Slp4a, Munc18-2, and Vamp7 with Stx3, which cooperate in the final steps of this selective apical traffic pathway. The brush border enzymes DPPIV and sucrase-isomaltase still correctly localize at the apical plasma membrane independent of this pathway. Hence, our work demonstrates how Myo5B, Stx3, Slp4a, Vamp7, Munc18-2, and Rab8/11 cooperate during selective apical cargo trafficking and exocytosis in epithelial cells and thereby provides further insight into MVID pathophysiology.
Collapse
Affiliation(s)
- Georg F Vogel
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria Division of Histology and Embryology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Katharina M C Klee
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria Institute of Molecular Biology, University of Innsbruck, 6020 Innsbruck, Austria Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020 Innsbruck, Austria
| | - Andreas R Janecke
- Department of Paediatrics I, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Thomas Müller
- Department of Paediatrics I, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Michael W Hess
- Division of Histology and Embryology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Lukas A Huber
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
16
|
Gupta A, Schell MJ, Bhattacharjee A, Lutsenko S, Hubbard AL. Myosin Vb mediates Cu+ export in polarized hepatocytes. J Cell Sci 2016; 129:1179-89. [PMID: 26823605 DOI: 10.1242/jcs.175307] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 01/20/2016] [Indexed: 02/06/2023] Open
Abstract
The cellular machinery responsible for Cu(+)-stimulated delivery of the Wilson-disease-associated protein ATP7B to the apical domain of hepatocytes is poorly understood. We demonstrate that myosin Vb regulates the Cu(+)-stimulated delivery of ATP7B to the apical domain of polarized hepatic cells, and that disruption of the ATP7B-myosin Vb interaction reduces the apical surface expression of ATP7B. Overexpression of the myosin Vb tail, which competes for binding of subapical cargos to myosin Vb bound to subapical actin, disrupted the surface expression of ATP7B, leading to reduced cellular Cu(+) export. The myosin-Vb-dependent targeting step occurred in parallel with hepatocyte-like polarity. If the myosin Vb tail was expressed acutely in cells just prior to the establishment of polarity, it appeared as part of an intracellular apical compartment, centered on γ-tubulin. ATP7B became selectively arrested in this compartment at high [Cu(+)] in the presence of myosin Vb tail, suggesting that these compartments are precursors of donor-acceptor transfer stations for apically targeted cargos of myosin Vb. Our data suggest that reduced hepatic Cu(+) clearance in idiopathic non-Wilsonian types of disease might be associated with the loss of function of myosin Vb.
Collapse
Affiliation(s)
- Arnab Gupta
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Michael J Schell
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Svetlana Lutsenko
- Department of Physiology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Ann L Hubbard
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
17
|
Rosazza C, Meglic SH, Zumbusch A, Rols MP, Miklavcic D. Gene Electrotransfer: A Mechanistic Perspective. Curr Gene Ther 2016; 16:98-129. [PMID: 27029943 PMCID: PMC5412002 DOI: 10.2174/1566523216666160331130040] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 11/22/2022]
Abstract
Gene electrotransfer is a powerful method of DNA delivery offering several medical applications, among the most promising of which are DNA vaccination and gene therapy for cancer treatment. Electroporation entails the application of electric fields to cells which then experience a local and transient change of membrane permeability. Although gene electrotransfer has been extensively studied in in vitro and in vivo environments, the mechanisms by which DNA enters and navigates through cells are not fully understood. Here we present a comprehensive review of the body of knowledge concerning gene electrotransfer that has been accumulated over the last three decades. For that purpose, after briefly reviewing the medical applications that gene electrotransfer can provide, we outline membrane electropermeabilization, a key process for the delivery of DNA and smaller molecules. Since gene electrotransfer is a multipart process, we proceed our review in describing step by step our current understanding, with particular emphasis on DNA internalization and intracellular trafficking. Finally, we turn our attention to in vivo testing and methodology for gene electrotransfer.
Collapse
Affiliation(s)
| | | | | | - Marie-Pierre Rols
- Institute of Pharmacology and Structural Biology (IPBS), CNRS UMR5089, 205 route de Narbonne, 31077 Toulouse, France.
| | | |
Collapse
|
18
|
Oliveira LT, Leon GVO, Provance DW, de Mello FG, Sorenson MM, Salerno VP. Exogenous β-amyloid peptide interferes with GLUT4 localization in neurons. Brain Res 2015; 1615:42-50. [PMID: 25912430 DOI: 10.1016/j.brainres.2015.04.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 04/15/2015] [Indexed: 01/14/2023]
Abstract
Aging represents a major risk factor for numerous illnesses that are of increasing importance to society, including two of the most prevalent: diabetes and Alzheimer's disease. Studies have shown that diabetes is a risk factor for spontaneous Alzheimer's disease. While these studies suggest that diabetes can contribute to Alzheimer's disease, the implications of AD on diabetes are practically unexplored. The major mediator of the pathophysiological effects, the Aβ42 peptide, has been shown to enter neurons and lead to an alteration of the intracellular distribution of the molecular motor myosin Vb. Myosin Vb functions in memory and learning by participating in the strengthening of the long-term potentiation (LTP) of synaptic transmissions. It has also been implicated in the translocation of the glucose transporter, GLUT4, to the plasma membrane in response to insulin, a process that is defective in diabetes. Here, the effect on GLUT4 upon entry of the Aβ42 peptide into cultured chick retinal neurons was explored. The results suggest an alteration in distribution and a reduced level at the cell surface, as well as an increased colocalization with myosin Vb, which can partially explain the changes in glucose metabolism associated with AD. It is also shown that the presence of the Aβ40 peptide inhibits the internalization of the Aβ42 peptide in cultured cells. Together, the results provide additional targets for the development of therapeutics against the progression and effects of Alzheimer's disease.
Collapse
Affiliation(s)
- Leandro T Oliveira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, RJ 21944-590, Brazil; School of Physical Education and Sports, Federal University of Rio deJaneiro, RJ 21944-590, Brazil
| | - Gabbriela V O Leon
- School of Physical Education and Sports, Federal University of Rio deJaneiro, RJ 21944-590, Brazil
| | - D William Provance
- Center for the Development of Technology in Health (CDTS)/National Institute of Science and Technology for Innovation in Neglected Diseases (INCT-IDN), Oswaldo Cruz Foundation, RJ 21040-361, Brazil
| | - Fernando G de Mello
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, RJ 21944-590, Brazil
| | - Martha M Sorenson
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, RJ 21944-590, Brazil
| | - Verônica P Salerno
- School of Physical Education and Sports, Federal University of Rio deJaneiro, RJ 21944-590, Brazil.
| |
Collapse
|
19
|
Coudrier E, Almeida CG. Myosin 1 controls membrane shape by coupling F-Actin to membrane. BIOARCHITECTURE 2014; 1:230-235. [PMID: 22754614 PMCID: PMC3384575 DOI: 10.4161/bioa.18406] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cellular functions are intimately associated with rapid changes in membrane shape. Different mechanisms interfering with the lipid bilayer, such as the insertion of proteins with amphipatic helices or the association of a protein scaffold, trigger membrane bending. By exerting force on membranes, molecular motors can also contribute to membrane remodeling. Previous studies have shown that actin and myosin 1 participate in the invagination of the plasma membrane during endocytosis while kinesins and dynein with microtubules provide the force to elongate membrane buds at recycling endosomes and at the trans-Golgi network (TGN). Using live cell imaging we have recently shown that a myosin 1 (myosin 1b) regulates the actin dependent post-Golgi traffic of cargo and generates force that controls the assembly of F-actin foci and promotes with the actin cytoskeleton the formation of tubules at the TGN. Our data provide evidence that actin and myosin 1 can regulate membrane remodeling of organelles as well as having an unexpected role in the spatial organization of the actin cytoskeleton. Here, we discuss our results together with the role of actin and other myosins that have been implicated in the traffic of cargo.
Collapse
|
20
|
Mukherjea M, Ali MY, Kikuti C, Safer D, Yang Z, Sirkia H, Ropars V, Houdusse A, Warshaw DM, Sweeney HL. Myosin VI must dimerize and deploy its unusual lever arm in order to perform its cellular roles. Cell Rep 2014; 8:1522-32. [PMID: 25159143 DOI: 10.1016/j.celrep.2014.07.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/13/2014] [Accepted: 07/22/2014] [Indexed: 12/21/2022] Open
Abstract
It is unclear whether the reverse-direction myosin (myosin VI) functions as a monomer or dimer in cells and how it generates large movements on actin. We deleted a stable, single-α-helix (SAH) domain that has been proposed to function as part of a lever arm to amplify movements without impact on in vitro movement or in vivo functions. A myosin VI construct that used this SAH domain as part of its lever arm was able to take large steps in vitro but did not rescue in vivo functions. It was necessary for myosin VI to internally dimerize, triggering unfolding of a three-helix bundle and calmodulin binding in order to step normally in vitro and rescue endocytosis and Golgi morphology in myosin VI-null fibroblasts. A model for myosin VI emerges in which cargo binding triggers dimerization and unfolds the three-helix bundle to create a lever arm essential for in vivo functions.
Collapse
Affiliation(s)
- Monalisa Mukherjea
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, 415 Curie Boulevard, 700 CRB, Philadelphia, PA 19104-6085, USA
| | - M Yusuf Ali
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05405, USA
| | - Carlos Kikuti
- Structural Motility, Centre de Recherche, Institut Curie, 75248 Paris, France; CNRS, UMR144, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | - Daniel Safer
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, 415 Curie Boulevard, 700 CRB, Philadelphia, PA 19104-6085, USA
| | - Zhaohui Yang
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, 415 Curie Boulevard, 700 CRB, Philadelphia, PA 19104-6085, USA
| | - Helena Sirkia
- Structural Motility, Centre de Recherche, Institut Curie, 75248 Paris, France; CNRS, UMR144, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | - Virginie Ropars
- Structural Motility, Centre de Recherche, Institut Curie, 75248 Paris, France; CNRS, UMR144, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | - Anne Houdusse
- Structural Motility, Centre de Recherche, Institut Curie, 75248 Paris, France; CNRS, UMR144, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | - David M Warshaw
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05405, USA
| | - H Lee Sweeney
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, 415 Curie Boulevard, 700 CRB, Philadelphia, PA 19104-6085, USA.
| |
Collapse
|
21
|
Knowles BC, Roland JT, Krishnan M, Tyska MJ, Lapierre LA, Dickman PS, Goldenring JR, Shub MD. Myosin Vb uncoupling from RAB8A and RAB11A elicits microvillus inclusion disease. J Clin Invest 2014; 124:2947-62. [PMID: 24892806 DOI: 10.1172/jci71651] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 04/17/2014] [Indexed: 12/14/2022] Open
Abstract
Microvillus inclusion disease (MVID) is a severe form of congenital diarrhea that arises from inactivating mutations in the gene encoding myosin Vb (MYO5B). We have examined the association of mutations in MYO5B and disruption of microvillar assembly and polarity in enterocytes. Stable MYO5B knockdown (MYO5B-KD) in CaCo2-BBE cells elicited loss of microvilli, alterations in junctional claudins, and disruption of apical and basolateral trafficking; however, no microvillus inclusions were observed in MYO5B-KD cells. Expression of WT MYO5B in MYO5B-KD cells restored microvilli; however, expression of MYO5B-P660L, a MVID-associated mutation found within Navajo populations, did not rescue the MYO5B-KD phenotype but induced formation of microvillus inclusions. Microvilli establishment required interaction between RAB8A and MYO5B, while loss of the interaction between RAB11A and MYO5B induced microvillus inclusions. Using surface biotinylation and dual immunofluorescence staining in MYO5B-KD cells expressing mutant forms of MYO5B, we observed that early microvillus inclusions were positive for the sorting marker SNX18 and derived from apical membrane internalization. In patients with MVID, MYO5B-P660L results in global changes in polarity at the villus tips that could account for deficits in apical absorption, loss of microvilli, aberrant junctions, and losses in transcellular ion transport pathways, likely leading to the MVID clinical phenotype of neonatal secretory diarrhea.
Collapse
|
22
|
Coupling of two non-processive myosin 5c dimers enables processive stepping along actin filaments. Sci Rep 2014; 4:4907. [PMID: 24809456 PMCID: PMC4014986 DOI: 10.1038/srep04907] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/11/2014] [Indexed: 11/09/2022] Open
Abstract
Myosin 5c (Myo5c) is a low duty ratio, non-processive motor unable to move continuously along actin filaments though it is believed to participate in secretory vesicle trafficking in vertebrate cells. Here, we measured the ATPase kinetics of Myo5c dimers and tested the possibility that the coupling of two Myo5c molecules enables processive movement. Steady-state ATPase activity and ADP dissociation kinetics demonstrated that a dimer of Myo5c-HMM (double-headed heavy meromyosin 5c) has a 6-fold lower Km for actin filaments than Myo5c-S1 (single-headed myosin 5c subfragment-1), indicating that the two heads of Myo5c-HMM increase F-actin-binding affinity. Nanometer-precision tracking analyses showed that two Myo5c-HMM dimers linked with each other via a DNA scaffold and moved processively along actin filaments. Moreover, the distance between the Myo5c molecules on the DNA scaffold is an important factor for the processive movement. Individual Myo5c molecules in two-dimer complexes move stochastically in 30-36 nm steps. These results demonstrate that two dimers of Myo5c molecules on a DNA scaffold increased the probability of rebinding to F-actin and enabled processive steps along actin filaments, which could be used for collective cargo transport in cells.
Collapse
|
23
|
Granger E, McNee G, Allan V, Woodman P. The role of the cytoskeleton and molecular motors in endosomal dynamics. Semin Cell Dev Biol 2014; 31:20-9. [PMID: 24727350 PMCID: PMC4071412 DOI: 10.1016/j.semcdb.2014.04.011] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/01/2014] [Accepted: 04/03/2014] [Indexed: 12/28/2022]
Abstract
The endocytic pathway is essential for processes that define how cells interact with their environment, including receptor signalling, cell adhesion and migration, pathogen entry, membrane protein turnover and nutrient uptake. The spatial organisation of endocytic trafficking requires motor proteins that tether membranes or transport them along the actin and microtubule cytoskeletons. Microtubules, actin filaments and motor proteins also provide force to deform and assist in the scission of membranes, thereby facilitating endosomal sorting and the generation of transport intermediates.
Collapse
Affiliation(s)
- Elizabeth Granger
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Gavin McNee
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Victoria Allan
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK.
| | - Philip Woodman
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK.
| |
Collapse
|
24
|
Myosin Va associates with mRNA in ribonucleoprotein particles present in myelinated peripheral axons and in the central nervous system. Dev Neurobiol 2014; 74:382-96. [DOI: 10.1002/dneu.22155] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 11/17/2013] [Accepted: 11/19/2013] [Indexed: 11/07/2022]
|
25
|
Schafer JC, Baetz NW, Lapierre LA, McRae RE, Roland JT, Goldenring JR. Rab11-FIP2 interaction with MYO5B regulates movement of Rab11a-containing recycling vesicles. Traffic 2014; 15:292-308. [PMID: 24372966 DOI: 10.1111/tra.12146] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 12/17/2013] [Accepted: 12/24/2013] [Indexed: 12/28/2022]
Abstract
A tripartite association of Rab11a with both Rab11-FIP2 and MYO5B regulates recycling endosome trafficking. We sought to define the intermolecular interactions required between Rab11-FIP2 and MYO5B. Using a random mutagenesis strategy, we identified point mutations at S229P or G233E in Rab11-FIP2 that caused loss of interaction with MYO5B in yeast two-hybrid assays as well as loss of interaction of Rab11-FIP2(129-356) with MYO5B tail when expressed in HeLa cells. Single mutations or the double S229P/G233E mutation failed to alter the association of full-length Rab11-FIP2 with MYO5B tail in HeLa cells. While EGFP-Rab11-FIP2 wild type colocalized with endogenous MYO5B staining in MDCK cells, EGFP-Rab11-FIP2(S229P/G233E) showed a significant decrease in localization with endogenous MYO5B. Analysis of Rab11a-containing vesicle movement in live HeLa cells demonstrated that when the MYO5B/Rab11-FIP2 association is perturbed by mutation or by Rab11-FIP2 knockdown, vesicle movement is increased in both speed and track length, consistent with an impairment of MYO5B tethering at the cytoskeleton. These results support a critical role for the interaction of MYO5B with Rab11-FIP2 in stabilizing the functional complex with Rab11a, which regulates dynamic movements of membrane recycling vesicles.
Collapse
Affiliation(s)
- Jenny C Schafer
- Section of Surgical Sciences and the Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | | | | | | |
Collapse
|
26
|
Dhekne HS, Hsiao NH, Roelofs P, Kumari M, Slim CL, Rings EHHM, van Ijzendoorn SCD. Myosin Vb and Rab11a regulate phosphorylation of ezrin in enterocytes. J Cell Sci 2014; 127:1007-17. [PMID: 24413175 DOI: 10.1242/jcs.137273] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Microvilli at the apical surface of enterocytes allow the efficient absorption of nutrients in the intestine. Ezrin activation by its phosphorylation at T567 is important for microvilli development, but how such ezrin phosphorylation is controlled is not well understood. We demonstrate that a subset of kinases that phosphorylate ezrin closely co-distributes with apical recycling endosome marker Rab11a in the subapical domain. Expression of dominant-negative Rab11a mutant or depletion of the Rab11a-binding motor protein myosin Vb prevents the subapical enrichment of Rab11a and these kinases and inhibits ezrin phosphorylation and microvilli development, without affecting the polarized distribution of ezrin itself. We observe a similar loss of the subapical enrichment of Rab11a and the kinases and reduced phosphorylation of ezrin in microvillus inclusion disease, which is associated with MYO5B mutations, intestinal microvilli atrophy and malabsorption. Thus, part of the machinery for ezrin activation depends on recycling endosomes controlled by myosin Vb and Rab11a which, we propose, might act as subapical signaling platforms that enterocytes use to regulate development of microvilli and maintain human intestinal function.
Collapse
Affiliation(s)
- Herschel S Dhekne
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
27
|
Thoeni CE, Vogel GF, Tancevski I, Geley S, Lechner S, Pfaller K, Hess MW, Müller T, Janecke AR, Avitzur Y, Muise A, Cutz E, Huber LA. Microvillus inclusion disease: loss of Myosin vb disrupts intracellular traffic and cell polarity. Traffic 2013; 15:22-42. [PMID: 24138727 DOI: 10.1111/tra.12131] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 10/11/2013] [Accepted: 10/18/2013] [Indexed: 12/23/2022]
Abstract
Microvillus inclusion disease (MVID) is a congenital enteropathy characterized by loss of apical microvilli and formation of cytoplasmic inclusions lined by microvilli in enterocytes. MVID is caused by mutations in the MYO5B gene, coding for the myosin Vb motor protein. Although myosin Vb is implicated in the organization of intracellular transport and cell surface polarity in epithelial cells, its precise role in the pathogenesis of MVID is unknown. We performed correlative immunohistochemistry analyses of sections from duodenal biopsies of a MVID patient, compound heterozygous for two novel MYO5B mutations, predicting loss of function of myosin Vb in duodenal enterocytes together with a stable MYO5B CaCo2 RNAi cell system. Our findings show that myosin Vb-deficient enterocytes display disruption of cell polarity as reflected by mislocalized apical and basolateral transporter proteins, altered distribution of certain endosomal/lysosomal constituents including Rab GTPases. Together, this severe disturbance of epithelial cell function could shed light on the pathology and symptoms of MVID.
Collapse
Affiliation(s)
- Cornelia E Thoeni
- Division of Cell Biology, Biocenter Innsbruck, Medical University Innsbruck, Innsbruck, Austria; Division of Pathology, Department of Pediatric Laboratory Medicine, Hospital for Sick Children, Toronto, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Intracellular tracking of single-plasmid DNA particles after delivery by electroporation. Mol Ther 2013; 21:2217-26. [PMID: 23941812 DOI: 10.1038/mt.2013.182] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 07/25/2013] [Indexed: 01/09/2023] Open
Abstract
Electroporation is a physical method of transferring molecules into cells and tissues. It takes advantage of the transient permeabilization of the cell membrane induced by electric field pulses, which gives hydrophilic molecules access to the cytoplasm. This method offers high transfer efficiency for small molecules that freely diffuse through electrically permeabilized membranes. Larger molecules, such as plasmid DNA, face several barriers (plasma membrane, cytoplasmic crowding, and nuclear envelope), which reduce transfection efficiency and engender a complex mechanism of transfer. Our work provides insight into the way electrotransferred DNA crosses the cytoplasm to reach the nucleus. For this purpose, single-particle tracking experiments of fluorescently labeled DNA were performed. Investigations were focused on the involvement of the cytoskeleton using drugs disrupting or stabilizing actin and tubulin filaments as the two relevant cellular networks for particle transport. The analysis of 315 movies (~4,000 trajectories) reveals that DNA is actively transported through the cytoskeleton. The large number of events allows a statistical quantification of the DNA motion kinetics inside the cell. Disruption of both filament types reduces occurrence and velocities of active transport and displacements of DNA particles. Interestingly, stabilization of both networks does not enhance DNA transport.
Collapse
|
29
|
Zajac AL, Goldman YE, Holzbaur ELF, Ostap EM. Local cytoskeletal and organelle interactions impact molecular-motor- driven early endosomal trafficking. Curr Biol 2013; 23:1173-80. [PMID: 23770188 DOI: 10.1016/j.cub.2013.05.015] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 04/11/2013] [Accepted: 05/09/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND In the intracellular environment, motor-driven cargo must navigate a dense cytoskeletal network among abundant organelles. RESULTS We investigated the effects of the crowded intracellular environment on early endosomal trafficking. Live-cell imaging of an endosomal cargo (endocytosed epidermal growth factor-conjugated quantum dots) combined with high-resolution tracking was used to analyze the heterogeneous motion of individual endosomes. The motile population of endosomes moved toward the perinuclear region in directed bursts of microtubule-based, dynein-dependent transport interrupted by longer periods of diffusive motion. Actin network density did not affect motile endosomes during directed runs or diffusive interruptions. Simultaneous two-color imaging was used to correlate changes in endosomal movement with potential obstacles to directed runs. Termination of directed runs spatially correlated with microtubule-dense regions, encounters with other endosomes, and interactions with the endoplasmic reticulum. During a subset of run terminations, we also observed merging and splitting of endosomes, deformation of the endoplasmic reticulum, and directional reversals at speeds up to 10-fold greater than characteristic in vitro motor velocities. These observations suggest that endosomal membrane tension is high during directed run termination. CONCLUSIONS Our results indicate that the crowded cellular environment significantly impacts the motor-driven motility of organelles. Rather than simply acting as impediments to movement, interactions of trafficking cargos with intracellular obstacles may facilitate communication between membrane-bound compartments or contribute to the generation of membrane tension necessary for fusion and fission of endosomal membranes or remodeling of the endoplasmic reticulum.
Collapse
Affiliation(s)
- Allison L Zajac
- The Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| | | | | | | |
Collapse
|
30
|
Myosin-V Opposes Microtubule-Based Cargo Transport and Drives Directional Motility on Cortical Actin. Curr Biol 2013; 23:828-34. [DOI: 10.1016/j.cub.2013.03.068] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 02/27/2013] [Accepted: 03/28/2013] [Indexed: 11/15/2022]
|
31
|
Khandelwal P, Prakasam HS, Clayton DR, Ruiz WG, Gallo LI, van Roekel D, Lukianov S, Peränen J, Goldenring JR, Apodaca G. A Rab11a-Rab8a-Myo5B network promotes stretch-regulated exocytosis in bladder umbrella cells. Mol Biol Cell 2013; 24:1007-19. [PMID: 23389633 PMCID: PMC3608489 DOI: 10.1091/mbc.e12-08-0568] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 01/29/2013] [Accepted: 01/30/2013] [Indexed: 12/03/2022] Open
Abstract
Multiple Rabs are associated with secretory granules/vesicles, but how these GTPases are coordinated to promote regulated exocytosis is not well understood. In bladder umbrella cells a subapical pool of discoidal/fusiform-shaped vesicles (DFVs) undergoes Rab11a-dependent regulated exocytosis in response to bladder filling. We show that Rab11a-associated vesicles are enmeshed in an apical cytokeratin meshwork and that Rab11a likely acts upstream of Rab8a to promote exocytosis. Surprisingly, expression of Rabin8, a previously described Rab11a effector and guanine nucleotide exchange factor for Rab8, stimulates stretch-induced exocytosis in a manner that is independent of its catalytic activity. Additional studies demonstrate that the unconventional motor protein myosin5B motor (Myo5B) works in association with the Rab8a-Rab11a module to promote exocytosis, possibly by ensuring transit of DFVs through a subapical, cortical actin cytoskeleton before fusion. Our results indicate that Rab11a, Rab8a, and Myo5B function as part of a network to promote stretch-induced exocytosis, and we predict that similarly organized Rab networks will be common to other regulated secretory pathways.
Collapse
Affiliation(s)
- Puneet Khandelwal
- Departments of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | | | - Dennis R. Clayton
- Departments of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Wily G. Ruiz
- Departments of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Luciana I. Gallo
- Departments of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Daniel van Roekel
- Departments of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Stefan Lukianov
- Departments of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Johan Peränen
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
| | - James R. Goldenring
- Department of Surgery and Epithelial Biology Center, Vanderbilt University, Nashville, TN 37232
| | - Gerard Apodaca
- Departments of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
- Cell Biology, University of Pittsburgh, Pittsburgh, PA 15261
| |
Collapse
|
32
|
Myosin motors at neuronal synapses: drivers of membrane transport and actin dynamics. Nat Rev Neurosci 2013; 14:233-47. [DOI: 10.1038/nrn3445] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
33
|
|
34
|
Ali MY, Previs SB, Trybus KM, Sweeney HL, Warshaw DM. Myosin VI has a one track mind versus myosin Va when moving on actin bundles or at an intersection. Traffic 2012; 14:70-81. [PMID: 23046080 DOI: 10.1111/tra.12017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 10/05/2012] [Accepted: 10/09/2012] [Indexed: 11/30/2022]
Abstract
Myosin VI (myoVI) and myosin Va (myoVa) serve roles both as intracellular cargo transporters and tethers/anchors. In both capacities, these motors bind to and processively travel along the actin cytoskeleton, a network of intersecting actin filaments and bundles that present directional challenges to these motors. Are myoVI and myoVa inherently different in their abilities to interact and maneuver through the complexities of the actin cytoskeleton? Thus, we created an in vitro model system of intersecting actin filaments and individual unipolar (fascin-actin) or mixed polarity (α-actinin-actin) bundles. The stepping dynamics of individual Qdot-labeled myoVI and myoVa motors were determined on these actin tracks. Interestingly, myoVI prefers to stay on the actin filament it is traveling on, while myoVa switches filaments with higher probability at an intersection or between filaments in a bundle. The structural basis for this maneuverability difference was assessed by expressing a myoVI chimera in which the single myoVI IQ was replaced with the longer, six IQ myoVa lever. The mutant behaved more like myoVI at actin intersections and on bundles, suggesting that a structural element other than the lever arm dictates myoVI's preference to stay on track, which may be critical to its role as an intracellular anchor.
Collapse
Affiliation(s)
- M Yusuf Ali
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05405, USA.
| | | | | | | | | |
Collapse
|
35
|
Caenorhabditis elegans screen reveals role of PAR-5 in RAB-11-recycling endosome positioning and apicobasal cell polarity. Nat Cell Biol 2012; 14:666-76. [PMID: 22634595 DOI: 10.1038/ncb2508] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 04/19/2012] [Indexed: 02/08/2023]
Abstract
Apically enriched Rab11-positive recycling endosomes (Rab11-REs) are important for establishing and maintaining epithelial polarity. Yet, little is known about the molecules controlling trafficking of Rab11-REs in an epithelium in vivo. Here, we report a genome-wide, image-based RNA interference screen for regulators of Rab11-RE positioning and transport of an apical membrane protein (PEPT-1) in C. elegans intestine. Among the 356 screen hits was the 14-3-3 and partitioning defective protein PAR-5, which we found to be specifically required for Rab11-RE positioning and apicobasal polarity maintenance. Depletion of PAR-5 induced abnormal clustering of Rab11-REs to ectopic sites at the basolateral cortex containing F-actin and other apical domain components. This phenotype required key regulators of F-actin dynamics and polarity, such as Rho GTPases (RHO-1 and the Rac1 orthologue CED-10) and apical PAR proteins. Our data suggest that PAR-5 acts as a regulatory hub for a polarity-maintaining network required for apicobasal asymmetry of F-actin and proper Rab11-RE positioning.
Collapse
|
36
|
Gidon A, Bardin S, Cinquin B, Boulanger J, Waharte F, Heliot L, Salle H, Hanau D, Kervrann C, Goud B, Salamero J. A Rab11A/Myosin Vb/Rab11-FIP2 Complex Frames Two Late Recycling Steps of Langerin from the ERC to the Plasma Membrane. Traffic 2012; 13:815-33. [DOI: 10.1111/j.1600-0854.2012.01354.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 03/13/2012] [Accepted: 03/15/2012] [Indexed: 01/29/2023]
Affiliation(s)
- Alexandre Gidon
- UMR 144, Molecular Mechanisms of Intracellular Transport Laboratory; CNRS-Institut Curie; 26 rue d'Ulm; 75248; Paris cedex 05; France
| | - Sabine Bardin
- UMR 144, Molecular Mechanisms of Intracellular Transport Laboratory; CNRS-Institut Curie; 26 rue d'Ulm; 75248; Paris cedex 05; France
| | | | - Jerome Boulanger
- Cell and Tissue Imaging Facility, PICT-IBiSA & Nikon Imaging Center; UMR 144 CNRS-Institut Curie; 26 rue d'Ulm; 75248; Paris cedex 05; France
| | - François Waharte
- Cell and Tissue Imaging Facility, PICT-IBiSA & Nikon Imaging Center; UMR 144 CNRS-Institut Curie; 26 rue d'Ulm; 75248; Paris cedex 05; France
| | - Laurent Heliot
- Interdisciplinary Research Institute, Molecular Dynamics and Interaction in Living Cell; 59658; Villeneuve d'Ascq; France
| | - Henri Salle
- INSERM U 725, Biology of Human Dendritic Cells; Strasbourg; France
| | - Daniel Hanau
- INSERM U 725, Biology of Human Dendritic Cells; Strasbourg; France
| | - Charles Kervrann
- INRIA Rennes - Bretagne Atlantique. Team SERPICO; Campus de Beaulieu; 35042; Rennes cedex; France
| | - Bruno Goud
- UMR 144, Molecular Mechanisms of Intracellular Transport Laboratory; CNRS-Institut Curie; 26 rue d'Ulm; 75248; Paris cedex 05; France
| | | |
Collapse
|
37
|
Oliveira LT, Matos PA, Provance DW, de Mello FG, Andrade LR, Sorenson MM, Salerno VP. β-amyloid peptide is internalized into chick retinal neurons and alters the distribution of myosin Vb. Cytoskeleton (Hoboken) 2012; 69:166-78. [DOI: 10.1002/cm.21007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 12/22/2011] [Accepted: 01/03/2012] [Indexed: 11/07/2022]
|
38
|
Abstract
Cells use molecular motors, such as myosins, to move, position and segregate their organelles. Class V myosins possess biochemical and structural properties that should make them ideal actin-based cargo transporters. Indeed, studies show that class V myosins function as cargo transporters in yeast, moving a range of organelles, such as the vacuole, peroxisomes and secretory vesicles. There is also increasing evidence in vertebrate cells that class V myosins not only tether organelles to actin but also can serve as short-range, point-to-point organelle transporters, usually following long-range, microtubule-dependent organelle transport.
Collapse
|
39
|
Xu S, Edman M, Kothawala MS, Sun G, Chiang L, Mircheff A, Zhu L, Okamoto C, Hamm-Alvarez S. A Rab11a-enriched subapical membrane compartment regulates a cytoskeleton-dependent transcytotic pathway in secretory epithelial cells of the lacrimal gland. J Cell Sci 2011; 124:3503-14. [PMID: 21984810 DOI: 10.1242/jcs.088906] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Despite observations that the lacrimal gland has been identified as the principal source of dimeric immunoglobulin A (dIgA) in tears, the mechanism used by lacrimal gland acinar cells (LGACs) to transcytose dIgA produced by interstitial plasma cells is not well-characterized. This study identifies a transcytotic pathway in LGACs regulated by Rab11a for polymeric immunoglobulin receptor (pIgR) and dIgA. EGFP-tagged Rab11a expressed in primary LGACs labeled a unique membrane compartment of comparable localization to endogenous Rab11a beneath the apical plasma membrane. This compartment was enriched in pIgR and clearly distinct from the regulated secretory pathway. Comparison of dIgA uptake in LGACs expressing wild type and dominant negative EGFP-Rab11a showed that the rapid exocytosis of dIgA was inhibited in acini expressing the dominant-negative protein, which additionally redistributed subapical pIgR. The trafficking of EGFP-Rab11a-enriched vesicles was regulated by microtubule-based and myosin Vb motors at distinct steps. Our data suggest that Rab11a is a crucial regulator of dIgA trafficking in primary acinar secretory epithelial cells and further support a role for microtubules, cytoplasmic dynein, actin filaments and myosin Vb in the maintenance of the Rab11a compartment in this primary secretory epithelial cell.
Collapse
Affiliation(s)
- Shi Xu
- Department of Pharmacology and Pharmaceutical Sciences, 1985 Zonal Avenue, USC School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
On diphtheria toxin fragment A release into the cytosol—Cytochalasin D effect and involvement of actin filaments and eukaryotic elongation factor 2. Int J Biochem Cell Biol 2011; 43:1365-72. [DOI: 10.1016/j.biocel.2011.05.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 05/13/2011] [Accepted: 05/26/2011] [Indexed: 11/17/2022]
|
41
|
Abstract
In neuroscience, myosin V motor proteins have attracted attention since they are highly expressed in brain, and absence of myosin Va in man leads to a severe neurological disease called Griscelli syndrome. While in some cells myosin V is described to act as a vesicle transport motor, an additional role in exocytosis has emerged recently. In neurons, myosin V has been linked to exocytosis of secretory vesicles and recycling endosomes. Through these functions, it is implied in regulating important brain functions including the release of neuropeptides by exocytosis of large dense-core vesicles and the insertion of neurotransmitter receptors into post-synaptic membranes. This review focuses on the role of myosin V in (i) axonal transport and stimulated exocytosis of large dense-core vesicles to regulate the secretion of neuroactive substances, (ii) tethering of the endoplasmic reticulum at cerebellar synapses to permit long-term depression, (iii) recycling of α-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA) receptors at hippocampal synapses during long-term potentiation, and (iv) recycling of nicotinic acetylcholine receptors at the neuromuscular junction. Myosin V is thus discussed as an important modulator of synaptic plasticity.
Collapse
Affiliation(s)
- Rüdiger Rudolf
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | | | | |
Collapse
|
42
|
Myosin-Va transports the endoplasmic reticulum into the dendritic spines of Purkinje neurons. Nat Cell Biol 2010; 13:40-8. [PMID: 21151132 PMCID: PMC3403743 DOI: 10.1038/ncb2132] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Accepted: 11/17/2010] [Indexed: 02/06/2023]
Abstract
Extension of the endoplasmic reticulum (ER) into dendritic spines of Purkinje neurons (PNs) is required for cerebellar synaptic plasticity and is disrupted in animals with null mutations in Myo5a, the gene encoding myosin-Va1–3. Notably, the mechanism ensuring the ER's localization to spines has not been unraveled. While it has been proposed that animal class V myosins localize organelles by tethering them to the actin cytoskeleton4–7, we demonstrate here that myosin-Va acts as a point-to-point organelle transporter to pull ER as cargo into PN spines. Specifically, the myosin accumulates at the ER tip as the organelle moves into spines, and the myosin's ability to hydrolyze ATP is required for spine ER targeting. Moreover, myosin-Va is responsible for the vast majority of spine ER insertional events. Finally, attenuation of the myosin's ability to move along actin filaments reduces the maximum velocity of ER movement into spines, providing direct evidence that myosin-Va drives ER motility. Thus, we establish that an actin-based motor moves ER within animal cells, and we uncover the mechanism that mediates ER localization to PN spines, a prerequisite for synaptic plasticity.
Collapse
|
43
|
Bridgman PC. Myosin motor proteins in the cell biology of axons and other neuronal compartments. Results Probl Cell Differ 2010; 48:91-105. [PMID: 19554282 DOI: 10.1007/400_2009_10] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Most neurons of both the central and peripheral nervous systems express multiple members of the myosin superfamily that include nonmuscle myosin II, and a number of classes of unconventional myosins. Several classes of unconventional myosins found in neurons have been shown to play important roles in transport processes. A general picture of the myosin-dependent transport processes in neurons is beginning to emerge, although much more work still needs to be done to fully define these roles and establish the importance of myosin for axonal transport. Myosins appear to contribute to three types of transport processes in neurons; recycling of receptors or other membrane components, dynamic tethering of vesicular components, and transport or tethering of protein translational machinery including mRNA. Defects in one or more of these functions have potential to contribute to disease processes.
Collapse
Affiliation(s)
- Paul C Bridgman
- Department of Anatomy and Neurobiology, Box 8108, Washington University School of Medicine, 660 Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
44
|
Chu BB, Ge L, Xie C, Zhao Y, Miao HH, Wang J, Li BL, Song BL. Requirement of myosin Vb.Rab11a.Rab11-FIP2 complex in cholesterol-regulated translocation of NPC1L1 to the cell surface. J Biol Chem 2009; 284:22481-22490. [PMID: 19542231 DOI: 10.1074/jbc.m109.034355] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Niemann-Pick C1-like 1 (NPC1L1) plays a critical role in the enterohepatic absorption of free cholesterol. Cellular cholesterol depletion induces the transport of NPC1L1 from the endocytic recycling compartment to the plasma membrane (PM), and cholesterol replenishment causes the internalization of NPC1L1 together with cholesterol via clathrin-mediated endocytosis. Although NPC1L1 has been characterized, the other proteins involved in cholesterol absorption and the endocytic recycling of NPC1L1 are largely unknown. Most of the vesicular trafficking events are dependent on the cytoskeleton and motor proteins. Here, we investigated the roles of the microfilament and microfilament-associated triple complex composed of myosin Vb, Rab11a, and Rab11-FIP2 in the transport of NPC1L1 from the endocytic recycling compartment to the PM. Interfering with the dynamics of the microfilament by pharmacological treatment delayed the transport of NPC1L1 to the cell surface. Meanwhile, inactivation of any component of the myosin Vb.Rab11a.Rab11-FIP2 triple complex inhibited the export of NPC1L1. Expression of the dominant-negative mutants of myosin Vb, Rab11a, or Rab11-FIP2 decreased the cellular cholesterol uptake by blocking the transport of NPC1L1 to the PM. These results suggest that the efficient transport of NPC1L1 to the PM is dependent on the microfilament-associated myosin Vb.Rab11a.Rab11-FIP2 triple complex.
Collapse
Affiliation(s)
- Bei-Bei Chu
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Liang Ge
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Chang Xie
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Yang Zhao
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Hong-Hua Miao
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Jing Wang
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Bo-Liang Li
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Bao-Liang Song
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| |
Collapse
|
45
|
Woolner S, Bement WM. Unconventional myosins acting unconventionally. Trends Cell Biol 2009; 19:245-52. [PMID: 19406643 DOI: 10.1016/j.tcb.2009.03.003] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2009] [Revised: 03/26/2009] [Accepted: 03/27/2009] [Indexed: 11/26/2022]
Abstract
Unconventional myosins are proteins that bind actin filaments in an ATP-regulated manner. Because of their association with membranes, they have traditionally been viewed as motors that function primarily to transport membranous organelles along actin filaments. Recently, however, a wealth of roles for myosins that are not obviously related to organelle transport have been uncovered, including organization of F-actin, mitotic spindle regulation and gene transcription. Furthermore, it has also become apparent that the motor domains of different myosins vary strikingly in their biophysical attributes. We suggest that the assumption that most unconventional myosins function primarily as organelle transporters might be misguided.
Collapse
Affiliation(s)
- Sarah Woolner
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester, UK.
| | | |
Collapse
|
46
|
Wang Z, Edwards JG, Riley N, Provance DW, Karcher R, Li XD, Davison IG, Ikebe M, Mercer JA, Kauer JA, Ehlers MD. Myosin Vb mobilizes recycling endosomes and AMPA receptors for postsynaptic plasticity. Cell 2008; 135:535-48. [PMID: 18984164 DOI: 10.1016/j.cell.2008.09.057] [Citation(s) in RCA: 367] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Revised: 07/16/2008] [Accepted: 09/09/2008] [Indexed: 10/21/2022]
Abstract
Learning-related plasticity at excitatory synapses in the mammalian brain requires the trafficking of AMPA receptors and the growth of dendritic spines. However, the mechanisms that couple plasticity stimuli to the trafficking of postsynaptic cargo are poorly understood. Here we demonstrate that myosin Vb (MyoVb), a Ca2+-sensitive motor, conducts spine trafficking during long-term potentiation (LTP) of synaptic strength. Upon activation of NMDA receptors and corresponding Ca2+ influx, MyoVb associates with recycling endosomes (REs), triggering rapid spine recruitment of endosomes and local exocytosis in spines. Disruption of MyoVb or its interaction with the RE adaptor Rab11-FIP2 abolishes LTP-induced exocytosis from REs and prevents both AMPA receptor insertion and spine growth. Furthermore, induction of tight binding of MyoVb to actin using an acute chemical genetic strategy eradicates LTP in hippocampal slices. Thus, Ca2+-activated MyoVb captures and mobilizes REs for AMPA receptor insertion and spine growth, providing a mechanistic link between the induction and expression of postsynaptic plasticity.
Collapse
Affiliation(s)
- Zhiping Wang
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|